You are on page 1of 894

Madjid Samii

Venelin Gerganov

Surgery of
Cerebellopontine
Lesions

123
Surgery of Cerebellopontine Lesions
Madjid Samii • Venelin Gerganov

Surgery of
Cerebellopontine
Lesions
With Chapters on Microsurgical Anatomy by
A.L. Rhoton and Neuropathology by G.F. Walter
Madjid Samii Venelin Gerganov
International Neuroscience Institute INI Department of Neurosurgery
Hannover International Neuroscience Institute
Germany Hannover
Germany

ISBN 978-3-642-35421-2 ISBN 978-3-642-35422-9 (eBook)


DOI 10.1007/978-3-642-35422-9
Springer Heidelberg New York Dordrecht London

Library of Congress Control Number: 2013934833

© Springer-Verlag Berlin Heidelberg 2013


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material
supplied specifically for the purpose of being entered and executed on a computer system, for
exclusive use by the purchaser of the work. Duplication of this publication or parts thereof is
permitted only under the provisions of the Copyright Law of the Publisher’s location, in its
current version, and permission for use must always be obtained from Springer. Permissions for
use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable
to prosecution under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of
publication, neither the authors nor the editors nor the publisher can accept any legal responsibility
for any errors or omissions that may be made. The publisher makes no warranty, express or
implied, with respect to the material contained herein.

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


To my family for their support and to all my patients for their
confidence in me.
Madjid Samii
To my beloved family - my wife Lyuba for her love,
understanding and support, and our wonderful children, Zara
and Marco, and to my parents.
Venelin M. Gerganov
Authors

Madjid Samii, MD, PhD; Prof.


President, International Neuroscience Institute, Hannover, Germany
President, China International Neuroscience Institute at the Capital Medical
University, Beijing, China
President, Iran International Neuroscience Institute at Tehran University of
Medical Sciences, Tehran, Iran
Rudolf Pichlmayr Str. 4, 30625 Hannover, Germany
Telephone number: 0049 511 27092 700
Fax number: 0049 511 27092 706
E-mail address: samii@ini-hannover.de
Venelin M. Gerganov, MD, PhD; Assoc. Prof.
Consultant neurosurgeon
Department of Neurosurgery, International Neuroscience Institute
Hannover, Germany
Rudolf Pichlmayr Str. 4, 30625 Hannover, Germany
Telephone number: 0049 511 27092 835
Fax number: 0049 511 27092 706
E-mail address: vgerganov@gmail.com
Albert L. Rhoton, Jr., M.D.
R.D. Keene Family Professor and Chairman Emeritus Department of
Neurosurgery
University of Florida P.O. Box 100265
Gainesville, Florida 32610
Telephone number: 352-273-9000
Fax number: 352-392-8413
E-mail address: rhoton@neurosurgery.ufl.edu
Gerhard Franz Walter, MD, PhD, Prof.
Dean, PhD-Programme Clinical Neurosciences
International Neuroscience Institute, Hannover, Germany
Rudolf Pichlmayr Str. 4, 30625 Hannover, Germany
Telephone number: 0049 511 27092 700
Fax number: 0049 511 27092 706
E-mail address: gf.walter@ini-hannover.de

vii
Contents

1 History of Cerebellopontine Angle Surgery .............................. 1


Description of CP Angle Tumors and First Attempts
at Surgery ....................................................................................... 1
Improvement of the Surgical Outcome and Introduction
of the Standard of Complete Tumor Removal ............................... 3
Development of Operative Techniques to Preserve
the Facial Nerve ............................................................................. 4
Translabyrinthine Approach ....................................................... 4
Transpetrosal Approach ............................................................. 5
Middle Fossa Approach ............................................................. 5
Modern Period of Functional Preserving Safe CP Angle
Surgery ........................................................................................... 5
Hearing Preservation .................................................................. 6
Radiosurgery .............................................................................. 6
References ...................................................................................... 6
2 Microsurgical Anatomy of the Cerebellopontine Angle
by the Retrosigmoid Approach ................................................... 9
Posterior Cranial Fossa .................................................................. 9
The Cerebellopontine Angle .......................................................... 9
Cerebellar Surfaces .................................................................... 11
Cerebellar Brain Stem Fissures .................................................. 14
Cerebellar Arteries ..................................................................... 16
The Fourth Ventricle ...................................................................... 18
Three Neurovascular Complexes ................................................... 19
Upper Neurovascular Complex ...................................................... 19
Trigeminal Root Anatomy ......................................................... 20
Anatomy of Vascular Compression in the Upper
Neurovascular Complex ............................................................. 24
Arterial Relationships................................................................. 26
Venous Relationships ................................................................. 29
Suprameatal Extension of the Retrosigmoid Approach ............. 32
Middle Neurovascular Complex .................................................... 34
Anatomy of Acoustic Neuromas ................................................ 34
Meatal Relationships .................................................................. 36
Retrosigmoid Approach ............................................................. 37
Brain Stem Relationships ........................................................... 39
Summary: Anatomy of Acoustic Neuromas .............................. 45
ix
x Contents

Anatomy of Vascular Compression in the Middle


Neurovascular Complex ................................................................. 50
Cochlear and Vestibular Nerve Compression Syndromes ......... 55
Geniculate Neuralgia .................................................................. 58
Lower Neurovascular Complex ..................................................... 59
Neural Relationships .................................................................. 60
Anatomy of Glossopharyngeal Neuralgia .................................. 61
Vascular Relationships ............................................................... 63
Vascular Compression in the Lower
Neurovascular Complex ................................................. 63
Tumors Involving Multiple Neurovascular Complexes ................. 63
Telovelar Approach to Fourth Ventricle ......................................... 65
References ...................................................................................... 70
3 Neuropathology of the Cerebellopontine Angle
and Its Surroundings ................................................................... 73
Introductory Remarks .................................................................... 73
Pathology of the Peripheral Nervous System ................................ 73
Schwannoma .............................................................................. 74
Malignant Peripheral Nerve Sheath Tumor (MPNST) .............. 76
Pathology of the Autonomous Nervous System ............................ 77
Jugulotympanic Paraganglioma ................................................. 77
Pathology of the Meninges ............................................................ 78
Meningioma ............................................................................... 78
Hemangiopericytoma ................................................................. 81
Hemangioblastoma ..................................................................... 82
Leptomeningeal Melanocytic Lesions ....................................... 83
Meningiosis and Solid Metastatic Tumors ................................. 83
Acute Bacterial Sive Purulent Meningitis .................................. 84
Tuberculous Meningitis.............................................................. 85
Sarcoidosis ................................................................................. 86
Aspergillosis ............................................................................... 86
Cryptococcosis ........................................................................... 88
Cysticercosis............................................................................... 89
Echinococcosis ........................................................................... 89
Pathology of the Osseous Skull Base............................................. 89
Fibrous Dysplasia ....................................................................... 90
Aneurysmal Bone Cyst .............................................................. 91
Osteochondroma......................................................................... 91
Chondroblastoma ....................................................................... 91
Chondrosarcoma......................................................................... 91
Chordoma ................................................................................... 91
Endolymphatic Sac Tumor ......................................................... 92
Pathology of the Sellar Region ...................................................... 93
Pituitary Adenoma ..................................................................... 93
Pituitary Carcinoma ................................................................... 95
Spindle Cell Oncocytoma of the Adenohypophysis .................. 96
Pituicytoma................................................................................. 96
Granular Cell Tumor of the Neurohypophysis........................... 97
Contents xi

Craniopharyngioma .................................................................... 97
Xanthogranuloma of the Sellar Region ...................................... 98
Pathology of Regional Glial Tumors ............................................. 99
Pilocytic Astrocytoma ................................................................ 99
Pilomyxoid Astrocytoma ........................................................... 99
Ependymoma.............................................................................. 100
Pathology of the Choroid Plexus of the Fourth Ventricle .............. 100
Choroid Plexus Papilloma .......................................................... 100
Choroid Plexus Carcinoma ........................................................ 101
Pathology of the Hematopoietic System ........................................ 101
Primary CNS Lymphomas ......................................................... 102
Langerhans Cell Histiocytosis.................................................... 102
Non-Langerhans Cell Histiocytosis ........................................... 103
Pathology of Germ Cells ................................................................ 104
Germ Cell Tumors ...................................................................... 104
Tumorlike Dysontogenetic Cysts ............................................... 105
Pathology of Blood Vessels and Circulation .................................. 105
Arteriovenous Malformation ...................................................... 105
Saccular Aneurysm .................................................................... 107
Ischemic Brain Infarct ................................................................ 109
Hypertensive Angiopathy and Brain Hemorrhage ..................... 109
Vasculitis .................................................................................... 110
References .................................................................................. 112
4 Approaches to the Cerebellopontine Angle ............................... 115
Middle Fossa Approach ................................................................. 115
Standard MF Approach .............................................................. 115
Extended MF Approach ................................................................. 118
Translabyrinthine Approach and Its Variations ............................. 119
Operative Technique................................................................... 119
Mastoidectomy ........................................................................... 119
Labyrinthectomy ........................................................................ 120
Surgical Outcome ....................................................................... 120
Retrolabyrinthine Approach........................................................... 121
Partial Translabyrinthine Approach ............................................... 121
Transcochlear Approach ................................................................ 121
Combined Approaches ................................................................... 121
Combined Translabyrinthine–Retrosigmoid Approach ............. 122
Translabyrinthine–Transtentorial Approach .............................. 122
Retrosigmoid Approach ................................................................. 122
Benefits of the Approach ............................................................ 122
Patient Positioning and Anesthesia Considerations ................... 123
Anesthetic Considerations ............................................................. 123
The Semi-sitting Position ........................................................... 126
Standard Instrument Sets Used in All CP Angle Surgeries ....... 128
Skin Incision and Craniotomy.................................................... 128
Dura Incision .............................................................................. 131
Hemostasis, dural closure and mastoid occlusion...................... 132
Reconstruction of the Posterior Skull Base................................ 134
xii Contents

Drawbacks of the RS Approach ..................................................... 134


Limited Exposure of the Fundus ................................................ 134
Need of Cerebellar Retraction.................................................... 136
Postoperative Headache ............................................................. 136
Pneumocephalus and Tension Pneumocephalus ........................ 137
References ...................................................................................... 141
5 Vestibular Schwannomas............................................................. 147
Epidemiology ................................................................................. 147
Biology of VS ................................................................................ 148
Molecular Genetics .................................................................... 148
The NF2 Protein ......................................................................... 148
Pathogenesis of VS ........................................................................ 149
Origin of VS ............................................................................... 149
Classification .................................................................................. 152
Neuroimaging ................................................................................ 156
Clinical Presentation ...................................................................... 156
Management of Patients with VS .................................................. 162
Conservative Management ......................................................... 162
Surgical Treatment of VS ........................................................... 163
Radiosurgical Management of VS ............................................. 282
References ...................................................................................... 304
6 Neurofibromatosis Type 2 and Other Bilateral
Cerebellopontine Angle Tumors ................................................. 315
Neurofibromatosis Type 2 .............................................................. 315
Criteria for Diagnosis ................................................................. 315
Genetics ...................................................................................... 315
Clinical Presentation .................................................................. 316
General Management ................................................................. 321
Large VSs ................................................................................... 328
Smaller VS, Useful Hearing....................................................... 328
Surgery on the Only Hearing Side ............................................. 332
Personal Series ........................................................................... 337
Other Bilateral CP Angle Tumors .................................................. 352
Hearing Rehabilitation ............................................................... 364
References ...................................................................................... 371
7 Meningiomas ................................................................................ 375
Cerebellopontine Angle Meningiomas .......................................... 375
Epidemiology ............................................................................. 375
Pathogenesis and Pathology ....................................................... 375
Classification .............................................................................. 376
Neuroimaging ............................................................................. 380
Clinical Presentation .................................................................. 390
Management ............................................................................... 392
Petroclival Meningiomas ............................................................... 436
General Features......................................................................... 436
Management ............................................................................... 438
Foramen Magnum Meningiomas ................................................... 483
Contents xiii

General Features......................................................................... 483


Management ............................................................................... 483
References ...................................................................................... 502
8 Cerebellopontine Angle Epidermoids ........................................ 509
General Features ............................................................................ 509
Neuroimaging ................................................................................ 509
Clinical Presentation ...................................................................... 511
Management................................................................................... 511
Outcome ..................................................................................... 515
Recurrences ................................................................................ 515
References ...................................................................................... 545
9 Nonvestibular Schwannomas ...................................................... 547
Trigeminal Schwannomas .............................................................. 547
General Features......................................................................... 547
Neuroimaging ............................................................................. 547
Clinical Presentation .................................................................. 549
Management ............................................................................... 549
Oculomotor Nerve Schwannoma ................................................... 577
Trochlear Schwannomas ................................................................ 577
Abducens Nerve Schwannoma ...................................................... 580
Facial Schwannomas ...................................................................... 583
Cochlear Schwannomas ................................................................. 604
Lower Cranial Nerve Schwannomas.............................................. 604
References ...................................................................................... 604
10 Jugular Foramen Tumors............................................................ 609
General Features ............................................................................ 609
Neuroimaging ................................................................................ 609
Clinical Presentation and Diagnosis .............................................. 610
Management................................................................................... 611
Operative Approaches ................................................................ 611
Endoscope-Assisted or Pure Endoscopic Tumor Removal ........ 617
Extended Endoscopic Endonasal Approach .............................. 625
Postoperative Care and Outcome ................................................... 625
Lower Cranial Nerves Dysfunction............................................ 625
Postoperative Care ...................................................................... 625
Radiotherapy/Radiosurgery........................................................ 626
Jugular Foramen Schwannomas..................................................... 627
Classification .............................................................................. 627
Neuroimaging ............................................................................. 629
Management ............................................................................... 637
Hypoglossal Nerve Schwannomas ................................................. 664
Glomus Jugulare Tumors ............................................................... 664
General Features......................................................................... 664
Pathogenesis and Classification ................................................. 664
Clinical Presentation .................................................................. 666
Management ............................................................................... 668
Meningiomas of the Jugular Foramen ........................................... 686
xiv Contents

General Features......................................................................... 686


Neuroimaging ............................................................................. 686
Clinical Manifestation ................................................................ 691
Operative Treatment ................................................................... 691
References ...................................................................................... 698
11 Other Cerebellopontine Angle Tumors ...................................... 703
Intra-Axial Tumors Extending Secondarily to the
Cerebellopontine Angle ................................................................. 703
Brain Stem Gliomas ................................................................... 703
Choroid Plexus Papillomas ........................................................ 714
Ependymomas ............................................................................ 718
Subependymomas ...................................................................... 726
Medulloblastomas ...................................................................... 730
Lymphomas ................................................................................ 730
Rare Extra-axial Cerebellopontine Angle Lesions ........................ 730
Arachnoid Cysts ......................................................................... 731
Dermoids .................................................................................... 740
Hamartomas ............................................................................... 744
Lipomas ...................................................................................... 748
Neurenteric Cysts ....................................................................... 749
Hemangioblastomas ................................................................... 757
CP Angle Metastases.................................................................. 764
Collision Tumors ........................................................................ 767
Vascular Lesions......................................................................... 773
Neurocysticercosis ..................................................................... 788
Temporal Bone and Petrous Apex Lesions .................................... 788
Cholesterol Granulomas of the Petrous Apex ............................ 788
Chordomas ................................................................................. 798
Chondrosarcomas ....................................................................... 807
Chondroblastomas ...................................................................... 818
Endolymphatic Sac Tumors ....................................................... 818
Fibrous Dysplasia ....................................................................... 834
Osteomas .................................................................................... 839
Pleomorphic Adenomas ............................................................. 840
Intralabyrinthine Schwannomas ................................................. 844
References ...................................................................................... 844
12 Management of Facial Palsy ....................................................... 855
Restoration of Facial Nerve Function ............................................ 855
Principles of Nerve Anastomosis ............................................... 855
Facial Nerve Reconstruction ...................................................... 855
Facial Nerve Reanimation with a Donor Nerve
(or Indirect Facial Nerve Reconstruction).................................. 862
Plastic Surgical Procedures ............................................................ 868
Physiotherapy ................................................................................. 869
Eye Care ......................................................................................... 869
References ...................................................................................... 870
Index ..................................................................................................... 873
History of Cerebellopontine
Angle Surgery 1

During different historical phases, the tumors Description of CP Angle Tumors


originating from the eighth nerve have been and First Attempts at Surgery
referred to as perineural fibroblastomas, neuri-
lemoma, schwannomas, acoustic neurinomas, Eduard Sandifort—a professor of anatomy in
acoustic neuromas, and vestibular schwanno- Leyden—made the first description of presump-
mas (VS). The last term corresponds most pre- tive VS in 1777. He described at autopsy a small
cisely to the pathogenetic tumor features and is firm tumor adherent to the acoustic nerve, com-
currently generally accepted. Although it was pressing the brain stem and extending into the
introduced in the early twentieth century, we will IAC: an “acoustic neuroma” [1, 49]. In 1822, the
use it throughout the text, even in the histori- Scottish physician J. H. Wishart reported the first
cal part. The term “acoustic neuroma,” which is case of bilateral VSs [57]. He published a case
popular in North America, is a misnomer—the report about a patient with blindness, deafness,
tumor does not originate from the acoustic nerve facial twitching, and unrelenting vomiting and
and neuromas arise only from a cut peripheral headaches. The patient died during surgery at the
nerve. In 1902, Henneberg and Koch introduced age of 21 years. At autopsy, Wishart found sev-
the definition cerebellopontine angle (CP angle) eral skull base tumors, including two attached to
or “Kleinhirnbrückenwinkel” (in the German both seventh cranial nerves. In 1830, Sir Charles
nomenclature), when presenting two patients Bell made the first definitively accurate postmor-
with bilateral vestibular schwannomas. tem description of VS [2, 29].
The history of CP angle tumors’ surgery can During these initial years, the correct diagno-
be divided in several periods: sis of CP angle tumors was possible only at
I period—description of CP angle tumors and autopsy. Jean Cruveilhier—professor of patho-
first attempts at surgery logical anatomy in Paris—was the first to describe
II period—improvement of the surgical outcome in detail the progression of clinical symptoms in
and introduction of the standard of complete a 26-year-old patient with such tumor [8, 43]. He
tumor removal believed that the lesion was located at the skull
III period—development of operative techniques base but was unable to localize it correctly until
to preserve the facial nerve autopsy. The autopsy findings were consistent
IV period—modern period of function preserving with a VS. Oppenheim in Germany was the first
safe CP angle surgery to make the correct diagnosis antemortem based

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 1


DOI 10.1007/978-3-642-35422-9_1, © Springer-Verlag Berlin Heidelberg 2013
2 1 History of Cerebellopontine Angle Surgery

on the typical clinical symptoms in 1890 [34]. seen his first case of acoustic tumor in 1887 at
In the second half of the nineteenth century, the St. Thomas’ Hospital, although did not even think
neurologists begun to understand that patients of surgery at that time [29].
with unilateral deafness, facial numbness, and The credit for the first removal of a VS belongs
progressive blindness most probably have a CP probably to Thomas Annandale of Edinburgh
angle tumor. Only in the last two decades of the [48]. In 1895, he succeeded in removing a tumor
century, however, the diagnosis became accurate “the size of a pigeon’s egg” in a young woman
enough to justify surgery [5]. In 1910, the via a unilateral suboccipital craniotomy. Although
Swedish pathologist Folke Henschen described her clinical history and the operative findings
the involvement of the IAC by VSs [21]. suggested that the lesion must have been a VS,
Von Bergmann in Germany undertook the some authors argued later that it has been actu-
earliest attempt at CP angle surgery in 1890. He ally a cerebellar tumor.
performed an unsuccessful attempt to remove a Victor Horsley in 1905 performed a gross total
tumor, which at autopsy turned out to be VS. One removal of a VS at the National Hospital in
year later, Charles McBurney of New York, after London. The patient lived for several years but
whom the appendectomy incision is named, was seriously crippled due to a supposed brain
performed an exploratory posterior fossa craniec- stem ischemia resulting from injury to the AICA.
tomy for a presumed cerebellar tumor [28]. The In the same year, Borchardt performed the first
first successful complete removal of a CP angle “transsigmoid” VS surgery [7]. Fedor Krause
tumor was performed by Sir Charles Balance, a was chief of surgery in the Augusta Hospital in
classmate and coworker of Viktor Horsley at the Berlin and professor at Friedrich-Wilhelms
Brown Institute and the National Hospital, Queen University. The approach to the CP angle, which
Square in London. A 49-year-old woman pre- he described in 1903, was a unilateral suboccipi-
sented with 1-year history of vertigo, headache, tal osteoplastic flap and is considered the
vomiting, gait instability, progressive visual loss, predecessor of the current retrosigmoid approach
and unilateral deafness [56]. Presumptive diag- [31]. Krause reported that VSs could be removed
nosis of acoustic tumor was made and she has by surgery but described them as the most prob-
been operated in two stages: on November 19, lematic of all brain tumors [52]. Interestingly, he
1894, a right posterior fossa craniectomy was was the first to use faradic stimulation to differen-
done, and 1 week later, the tumor was approached tiate the facial from the audiovestibular nerve in a
and removed with the finger inserted in an unster- case of tinnitus when he performed an eighth cra-
ile fashion between the pons and the petrous nial nerve section [53]. Garré in 1903 and Biggs
bone. According to the original description, the in 1909 were the first to attempt surgery for bilat-
tumor was well encapsulated, firm, and had a eral VSs, with dismal outcome in both cases [1].
wide attachment to the dura of the petrous bone In 1913 at the International Congress of Medicine
[4]. The patient recovered from surgery and was in London, three of the most outstanding European
alive for at least 18 years. She had facial anesthe- surgeons—Horsley of London, von Eiselsberg of
sia and complete facial palsy, which led to ulcer- Vienna, and Krause of Berlin—reported their early
ation and ultimately to removal of her right eye. experience with VS surgery. The mortality rates
Later on, a controversy ensued on the issue of the were 67 % (in the Horsley’s series of 15 patients),
pathological nature of this tumor. Dandy was 77 % (in the Eiselsberg’s series of 17 patients), and
convinced that the tumor was an acoustic tumor, 84 % (in the Krause’s series of 31 patients) [2].
while Cushing contended that it was rather a They used similar technique of unilateral suboc-
meningioma, basing his opinion on the assump- cipital approach and the index finger or a spatula
tion that the patient had normal preoperative to remove the tumor quickly. The speed of tumor
auditory function [9, 37]. In 1923, Balance him- removal was of primary importance because of
self, as President of the Section of Otology of the the profuse bleeding and the limited possibilities
Royal Society of Medicine, related that he had for hemostasis (obtained by packing the CP angle
Improvement of the Surgical Outcome and Introduction of the Standard of Complete Tumor Removal 3

with gauze). The CP angle earned later the name 30 operated patients with VS cases in his famous
“bloody angle” by Harvey Cushing [9, 10]. Some monograph Tumors of the Nervus Acusticus and
decades later, Horrax introduced another famous the Syndrome of the Cerebellopontine Angle, in
description of VSs: the “yellow monsters” [22]. which he reported remarkably lower mortality
The survivors usually suffered severe neurological rates than those of his predecessors [9]. In 1917,
deficits. Eiselsberg and Horsley attempted later a his mortality rate was 20 % and was further
2-stage removal of VS without better success. reduced to 4 % by 1936. Cushing considered
The reasons for the poor initial results were that the safe complete removal of VS is an unat-
the limited technical abilities, lack of reliable tainable goal with the available anesthesia, diag-
anesthesia, and delayed tumor diagnosis. Most of nostic, and operative techniques and focused on
the patients presented late with very large tumors, the need for decompression of the brain stem
had severe deficits and hydrocephalus, and were and avoidance of medullary compression dur-
moribund [29]. The increased intracranial pres- ing surgical manipulations. As usual for that
sure could be hardly controlled and “the surgeon period, all patients were diagnosed late, all had
often struggled to achieve sufficient tumor expo- signs of increased intracranial pressure, and half
sure using rudimentary methods of illumination were blind. Cushing emphasized the importance
and hemostasis” [2]. Frequently, the cerebellum of controlling intracranial pressure by using a
swelled so massively that it was necessary “to ventricular tap. He performed a large curvilin-
shave off the excess.” ear incision extending between the two mastoid
Gradually, the knowledge of the anatomy and processes, followed by a midline incision down
pathology increased and new operative approaches to the spinous process of the upper cervical ver-
and techniques have been introduced. De Martel, tebrae [34]. Then, he made bilateral suboccipi-
a pioneer of French neurosurgery, introduced the tal craniectomy that allowed him to explore both
sitting position for posterior fossa surgery [29]. CP angles, considering the frequently unreliable
In 1916, F. Henschen published his major contri- diagnostic findings. Furthermore, an osseous
bution to the histology and pathogenesis of CP suboccipital decompression was achieved. The
angle tumors, in particular of a clinical series bone opening allowed him to access early the
including 245 verified cases of unilateral and 24 cerebellomedullary cistern to drain CSF and to
bilateral VSs [21]. At autopsy, he discovered that perform decompression of the tonsils. Cushing
VSs originate from the vestibular portion of the was the first to apply gentle and meticulous
acoustic nerve, lying in the porus acusticus [1]. technique and to monitor the changes of vital
H. Stenvers of Utrecht (1917) and E. Towne of signs during surgery. He operated consciously
San Francisco (1926) described the enlargement without unjustified emphasis on the speed and
of the porus of the IAC, seen on conventional paid great attention to hemostasis, using bone
X-ray examinations. wax, silver vessel clips, and electrocautery.
Despite the good early results, the intracapsular
removal was associated with an expectedly high
tumor recurrence rate and the 5-year mortality
Improvement of the Surgical Outcome rate reached 54 % [9, 10].
and Introduction of the Standard Walter Dandy, a student of Cushing, consid-
of Complete Tumor Removal ered this outcome in case of benign tumors as
unacceptable and favored total VS removal
Harvey Cushing who established new standards [11, 12]. He believed that the residual tumor cap-
of surgery in the posterior fossa and reduced to sule could be removed, thus achieving better
an acceptable level the complications and the long-term results. Dandy used initially the bilat-
mortality rates of VS surgery made the next eral suboccipital craniectomy of Cushing and
major contribution to CP angle surgery. In 1917, performed extensive intracapsular decompres-
he published a description of his experience with sion, followed by careful excision of the tumor
4 1 History of Cerebellopontine Angle Surgery

capsule. He emphasized the importance of metic- to preserve the function of the facial nerve while
ulous clipping of the veins and arteries surround- performing total VS removal. Herbert Olivecrona
ing the capsule in order to avoid hemorrhagic emphasized the importance of facial nerve pres-
complications. Dandy was the first surgeon to ervation. He routinely performed electrical stim-
achieve total removal of VS with relatively low ulation to identify the nerve and had a special
operative mortality. nurse to observe the patient for facial contrac-
This next stage in the history of VS surgery tions [28, 55]. In 1939 Olivecrona presented his
was possible due to the advances in intraopera- initial results, demonstrating that the facial nerve
tive technology and the earlier detection of such was preserved in an astonishing for the time rate
tumors: the syndrome of the CP angle was widely of 65 % of the patients [39]. Later on, he described
recognized and the introduced by Dandy in 1918 a series of 300 patients with VSs: half of the
air ventriculography allowed neurosurgeons to tumors were removed totally; the facial nerve
detect smaller tumors [2]. His preliminary report function was preserved in 69 of them [40]. The
on five patients was published in 1922 and was mortality rate in the Olivecrona series, however,
followed 3 years later by a detailed follow-up, was still very high—29 %.
including an extensive discussion on the benefits One reason for these poor results was the
of total resection [11, 12]. Dandy continued to lack of sufficient knowledge of the anatomi-
improve his technique of tumor removal and in cal structure of the CP angle, especially that of
1934 described a less invasive unilateral suboc- its vasculature. In 1949, Atkinson published his
cipital approach, actually similar to the one of study on the distribution of the AICA and—in
Krause [13]. In 1941, he reported on 46 patients particular—on its relation to VSs [3]. He stressed
with VSs, removed totally with an operative the importance of its preservation and described
mortality rate of 10.87 % [14]. the dramatic sequelae of its injury.
Interestingly, Cushing never agreed officially
with the concept of Dandy, probably due to their
untoward personal relationship. He maintained Translabyrinthine Approach
his belief that the mortality rate is too high to jus-
tify total VS resection. Horrax and Poppen applied Parallel to the development of the suboccipital
the technique of Dandy and in 1949 published approach, alternative techniques have been devel-
their experience, reporting a 5-year mortality rate oped in an attempt to avoid the complications
of 12.7 % [22]. They could preserve the facial related to cerebellar retraction and the difficulty
nerve just in one of the 46 operated cases. The to work at the long working distance with poor
technique of Dandy was refined further by illumination and visualization. The anatomy of
McKenzie and Alexander, who placed the patients the bony labyrinth was extensively studied in the
in a seated position [35]. Adson and Bucy propa- late nineteenth century and the otologists became
gated a straight lateral incision, avoiding the bulky increasingly familiar with it [37]. The first laby-
musculocutaneous flap of the Dandy [5]. rinthectomy was performed in 1895 by the
German otologist Albert Jansen for a suppurative
disease. Rudolf Panse from Dresden developed
Development of Operative the concept of using this operative approach to
Techniques to Preserve the Facial expose the CP angle [37] and published it in 1904
Nerve [41]. Although his proposal was to chisel out the
mastoid bone and to remove the cochlea, he never
W. Dandy considered the facial palsy as “a neces- performed such surgery personally. Charles
sary sequel of the operation.” The next major Elsberg of New York performed cadaver dissec-
breakthrough in CP angle tumor surgery was the tions and measurements and found that the
successful facial nerve preservation. In 1931, Sir shortest route to the CP angle started 1–2 cm
Hugh Cairns of London became the first surgeon behind the ear: the route through the mastoid.
Modern Period of Functional Preserving Safe CP Angle Surgery 5

Franciscus H. Quix of Utrecht is credited for treatment of recurrent vertigo [46], while in 1947,
the first translabyrinthine removal of a VS [47]. The Gardner used a similar technique to resect the
surgery was performed in two stages because GSPN in a patient with unilateral headache [20].
the initial operation had to be aborted due to
strong bleeding from the superior petrosal sinus.
The tumor could not be removed completely. The
patient survived the surgery but died 6 months Modern Period of Functional
later. Later on in 1911 and 1912, Werner Kummel Preserving Safe CP Angle Surgery
and Ernst Schmiegelow reported their experience
with the approach [2]. The greatest breakthrough in the surgical treat-
The translabyrinthine approach could not be ment of VS that allowed achieving the current
established at that time because of the lack of standards was the introduction of the surgical
adequate surgical instruments and technique. microscope and of the microsurgical technique. In
Furthermore, the leading neurosurgeons of the 1957, Theodore Kurze resected for the first time a
time—such as Cushing and Dandy—found VS with an operating microscope at the University
it absolutely inappropriate. They stressed on of Southern California. In 1958, William House
the poor surgical exposure provided by this began using the microsurgical technique for tem-
approach and the difficult reconstruction with poral bone dissections and applied it in 1961 dur-
the related high likelihood of meningitis [37]. ing a translabyrinthine surgery of a VS. His major
The approach was reintroduced by William F. contribution was the utilization of the operating
House in the 1960s who was unaware of the earlier microscope and high-speed drill. William House
descriptions [25]. himself learned microsurgery from Wullstein in
Siegen, who inaugurated microsurgery of oto-
sclerosis (Wullstein, personal communication
Transpetrosal Approach 1971). This new technique allowed Dr. House to
remove the required amount of the petrous bone
Moritz Borchardt of Berlin in 1905 was the first to safely and to expose widely the IAC. He refined
realize the wide exposure gained by an extensive and established the translabyrinthine approach as
removal of the petrous bone [7]. As recounted by a technique to expose tumors of the CP angle and
Ngyuen, he attempted to remove a cerebellar IAC. In 1964, he published his experience with
tumor via the suboccipital approach but had to the transtemporal bone microsurgical removal of
stop because of excessive bleeding [37]. In order VSs [24, 25]. W. House elaborated and refined
to avoid the same delicate area, he resected the the middle fossa technique in the late 1950s and
temporal bone deep to the IAC. Once again, he had described systematically the relevant anatomical
to discontinue the surgery but at the third attempt landmarks [36]. In January 1960, he operated his
was able to remove the entire tumor. Later on, von first VS using the middle fossa approach. Using
Eiselsberg of Vienna used similar combined sub- the MF approach in small tumors, House could
occipital and transpetrosal approach in one patient reduce the mortality to 5.4 % and preserve the
but could remove only a portion of the tumor. facial nerve in 95 % [23].
Pool and Pava in 1957 were the first to empha-
size the importance of tumor removal from the
Middle Fossa Approach IAC [45]. Rand and Kurze developed a technique
to remove the posterior meatal wall. In 1965, they
The middle fossa (MF) approach to the IAC presented their experience on the microsurgical
was described in 1904 by Parry who used it suboccipital transmeatal approach, achieving
for sectioning of the auditory nerve in a patient complete removal of the VS and preservation of
with vertigo and tinnitus [42]. Later on—in the cranial nerves [50]. In 1967, C. G. Drake
1938—Putman reported its utilization for the reported on his experience with the complete
6 1 History of Cerebellopontine Angle Surgery

removal of even large VS [17, 18]. He described using the microsurgical technique [51]. Rand was
his techniques for preservation of the facial nerve the first to mention hearing preservation as treat-
and for its reconstruction [17]. As stated by W. ment goal. In 1978, M. G. Yasargil reported on
Pellet, the transmeatal technique quickly gained preserved hearing in 2/164 cases. In the same
popularity among neurosurgeons as a means to year, Di Tullio et al. reported one case of hearing
achieve complete tumor removal [44, 54]. The preservation out of 79 surgeries [16].
opening or “trepanation” of the meatal wall was W. House used the MF approach to preserve
performed with a gauge or drill. hearing [44]. His first successful attempt was in
M. G. Yasargil studied the microsurgical 1964. In 1968, he reported on 7 cases of hear-
anatomy of the CP angle and showed that during ing preservation from 19 attempts [26]. However,
the extension of VSs into the CP angle, a double House was against the attempts at systematic hear-
arachnoid membrane envelops the tumor. He ing preservation in patients with VSs. Hearing
emphasized the significance of these arachnoid preservation candidates were selected carefully,
layers as a safe dissection plane that allows for considering factors such as tumor size, position
preservation of even the finest structures [58, 59]. of cranial nerves, and hearing level on the oppo-
Major breakthrough was the introduction of intra- site side. In 1982, at one conference, he reported
operative facial nerve monitoring by Delgado on a series of 200 patients operated via the MF
et al. in 1979, which opened the way for func- approach: he regarded as hearing-preservation
tional preserving CP angle surgery [15]. The later candidates only 10 and could preserve hearing
progress in the management of CP angle tumors in 4 of them. At the same conference, the senior
was related to the introduction of new diagnos- author (MS) presented his initial experience with
tic modalities (CT and MRI), which allowed 200 patients with VSs with a hearing preserva-
earlier diagnosis, precise operative planning, tion rate of approximately 30 %.
and detection of postoperative complications;
developments in neuroanesthesia and neuroin-
tensive care; establishing of reliable techniques Radiosurgery
of continuous electrophysiological monitoring of
the function of all relevant cranial nerves; increas- In 1951, Lars Leksell described a new technique
ing the knowledge of the normal and pathological for stereotactically focused irradiation as a method
anatomy of the CP angle; and establishment of to destroy intracranial targets—the gamma knife
the microneurosurgery. The competition between [32]. He and E. O. Backlund performed the first
surgeons using different approaches stimulated gamma knife procedure in 1967. In 1969, Leksell
the refinement of the operative techniques and performed the first gamma knife radiosurgical
further improvement of the outcome. procedure for a VS in a patient with NF2 [30, 33, 38].
Since then, radiation techniques and protocols
have continued to evolve; currently, radiosurgery
Hearing Preservation is increasingly used in patients with VS [6].

In 1954, Elliot and McKissick were the first to


report hearing preservation in two of three References
patients after gross VS removal via suboccipital
1. Ahn MS, Jackler RK, Lustig LR (1996) The early his-
approach [19]. Eleven years later—in 1965—
tory of the neurofibromatosis. Evolution of the con-
McKissick reported preserved hearing in 5 of 270 cept of neurofibromatosis type 2. Arch Otolaryngol
operated patients. Hullay and Tomits in 1965 Head Neck Surg 122(11):1240–1249
could preserve hearing in 1/50 cases [27]. In 2. Akard W, Tubbs RS, Seymour ZA, Hitselberger WE,
Cohen-Gadol AA (2009) Evolution of techniques for
1968, Rand and Kurze presented two cases with
the resection of vestibular schwannomas: from saving
preserved hearing after complete tumor removal life to saving function. J Neurosurg 110(4):642–647
References 7

3. Atkinson WJ (1949) The anterior inferior cerebellar with special emphasis on saving the facial nerve. J
artery; its variations, pontine distribution, and Neurosurg 26(5):554–561
significance in the surgery of cerebello-pontine angle 19. Elliott FA, McKissick KW (1954) Acoustic neuroma;
tumours. J Neurol Neurosurg Psychiatry 12(2): early diagnosis. Lancet 267(6850):1189–1191
137–151 20. Gardner WJ, Stowell A, Dutlinger R (1947) Resection
4. Ballance C (1907) Some points in the surgery of the of the greater superficial petrosal nerve in the treat-
brain and its membranes. Macmillan and Co, Ltd., ment of unilateral headache. J Neurosurg 4(2):
London 105–114
5. Bambakidis NC, Kakarla UK, Kim LJ, Nakaji P, 21. Henschen F (1916) Zur Histologie und Pathogenese
Porter RW, Daspit CP, Spetzler RF (2007) Evolution der Kleinhirnbrüchenwinkeltumoren. Arch Psychiatr
of surgical approaches in the treatment of petroclival Nervenkr 56:21–122
meningiomas: a retrospective review. Neurosurgery 22. Horrax GPJ, Poppen JL (1949) The end results of
61(5 Suppl 2):202–209 complete versus intracapsular removal of acoustic
6. Bassim MK, Berliner KI, Fisher LM, Brackmann DE, tumors. Ann Surg 130:567–575
Friedman RA (2010) Radiation therapy for the 23. House WF (1963) Middle cranial fossa approach to
treatment of vestibular schwannoma: a critical evalua- the petrous pyramid. Report of 50 cases. Arch
tion of the state of the literature. Otol Neurotol 31(4): Otolaryngol 78:460–469
567–573 24. House WF (1964) Transtemporal bone microsurgical
7. Borchardt M (1906) Ueber Operationen in der hin- removal of acoustic neuromas. Evolution of transtem-
teren Schaedelgrube inkl. der Operationen der poral bone removal of acoustic tumors. Arch
Tumoren am Kleinhirnbrueckenwinkel. Arch Klin Otolaryngol 80:731–742
Chir 81:386–432 25. House WF (1964) Transtemporal bone microsurgical
8. Cruveilhier J (1829–1842) Anatomie pathologique du removal of acoustic neuromas. Report of cases. Arch
corps humain, ou Descriptions, avec figures Otolaryngol 80:617–667
lithographiées et coloriées, des diverses alterations 26. House WF (1968) Acoustic neuroma. Case summa-
morbides dont le corps humain est susceptible, vol 2. ries. Arch Otolaryngol 88(6):586–591
Bailière, Paris. Liv 32, Pl 2, pp 19–24; Liv 38, Pl 5, 27. Hullay J (1958) Variety in the site of acoustic tumors
pp 1–4 in the pontile angle; a preliminary report. Acta
9. Cushing H (1917) Tumors of the nervus acusticus and Neurochir (Wien) 6(1–2):115–120
the syndrome of the cerebellopontine angle. WB 28. Jackler RK, Whinney D (2001) A century of eighth
Saunders Co, Philadelphia nerve surgery. Otol Neurotol 22(3):401–416
10. Cushing H (1921) Further concerning the acoustic 29. Koerbel A, Gharabaghi A, Safavi-Abbasi S, Tatagiba
neuromas. Laryngoscope 31:209–228 M, Samii M (2005) Evolution of vestibular schwan-
11. Dandy W (1922) An operation for the total extirpation noma surgery: the long journey to current success.
of tumors in the cerebello-pontine angle. A prelimi- Neurosurg Focus 18(4):e10. doi:180410 [pii]
nary report. Bull Johns Hopkins Hosp 33:344–345 30. Kondziolka D, Lunsford LD, Flickinger JC (2004)
12. Dandy W (1925) An operation for the total removal of Acoustic neuroma radiosurgery. Origins, contempo-
cerebellopontine (acoustic) tumors. Surg Gynecol rary use and future expectations. Neurochirurgie
Obstet 41:129–148 50(2–3 Pt 2):427–435
13. Dandy W (1934) Removal of cerebellopontile (acous- 31. Krause F (1903) Zur Freilegung der hinteren
tic) tumors through a unilateral approach. Arch Surg Felsenbeinflache und des Kleinhirns. Beitr Klin Chir
29:337–344 37:728–764
14. Dandy W (1941) Results of removal of acoustic 32. Leksell L (1951) The stereotaxic method and radio-
tumors by the unilateral approach. Arch Surg 42: surgery of the brain. Acta Chir Scand 102(4):
1026–1033 316–319
15. Delgado TE, Bucheit WA, Rosenholtz HR, Chrissian 33. Leksell L (1971) A note on the treatment of acoustic
S (1979) Intraoperative monitoring of facila muscle tumours. Acta Chir Scand 137(8):763–765
evoked responses obtained by intracranial stimulation 34. Machinis TG, Fountas KN, Dimopoulos V, Robinson
of the facila nerve: a more accurate technique for JS (2005) History of acoustic neurinoma surgery.
facila nerve dissection. Neurosurgery 4(5):418–421 Neurosurg Focus 18(4):e9
16. DiTullio MV Jr, Malkasian D, Rand RW (1978) A 35. McKenzie KG, Alexander E Jr (1954) Acoustic neu-
critical comparison of neurosurgical and otolaryngo- roma. Clin Neurosurg 2:21–36
logical approaches to acoustic neuromas. J Neurosurg 36. Monfared A, Mudry A, Jackler R (2009) The history
48(1):1–12 of middle cranial fossa approach to the cerebellopon-
17. Drake CG (1967) Surgical treatment of acoustic neu- tine angle. Otol Neurotol 31(4):691–696
roma with preservation or reconstitution of the facial 37. Nguyen-Huynh AT, Jackler RK, Pfister M, Tseng J
nerve. J Neurosurg 26(5):459–464 (2007) The aborted early history of the translabyrin-
18. Drake CG (1967) Total removal of large acoustic neu- thine approach: a victim of suppression or technical
romas. A modification of the McKenzie operation prematurity? Otol Neurotol 28(2):269–279
8 1 History of Cerebellopontine Angle Surgery

38. Niranjan A, Lunsford LD (2000) Radiosurgery: where 50. Rand RW, Kurze T (1965) Micro-neurosurgical resec-
we were, are, and may be in the third millennium. tion of acoustic tumors by a transmeatal posterior
Neurosurgery 46(3):531–543 fossa approach. Bull Los Angel Neuro Soc 30:17–20
39. Olivecrona H (1939) Acoustic tumors. In: Winther 51. Rand RW, Kurze T (1968) Preservation of vestibular,
KKK (ed) III Congrès Neurologique International, cochlear, and facial nerves during microsurgical
Copenhague, 21/8–25/8 1939. Comptes Rendus des removal of acoustic tumors. Report of two cases.
Séances. Einar Munksgaard, Copenhangue, pp 761–771 J Neurosurg 28(2):158–161
40. Olivecrona H (1950) Analysis of results of complete 52. Reidel C, Post KD (1993) Complications in the treat-
and partial removal of acoustic neuromas. J Neurol ment of acoustic neuromas. In: Post KD, Friedman E,
Neurosurg Psychiatry 13(4):271–272 McCormick P (eds) Postoperative complications in
41. Panse R (1904) Ein Gliom des Acusticus. Arch intracranial neurosurgery. Thieme, New York, pp
Ohrenheilkd 61:251–255 91–110
42. Parry D (1904) A case of tinnitus treated by division of 53. Rosegay H (1992) The Krause operations. J Neurosurg
the auditory nerve. J Laryngol Otol 19(19):402–406 76(6):1032–1036
43. Pearce JM (2003) Cruveilhier and acoustic neuroma. 54. Rougerie J, Guyot JF (1964) Attempted preservation
J Neurol Neurosurg Psychiatry 74(8):1015 of the facial nerve in the ablation of neurinomas of the
44. Pellet W (2008) History of vestibular schwannoma cerebellopontile angle. Neurochirurgie 10:13–21
surgery. Prog Neurol Surg 21:6–23 55. Sampath P, Holliday MJ, Brem H, Niparko JK, Long
45. Pool JL, Pava AA (1957) The early diagnosis and DM (1997) Facial nerve injury in acoustic neuroma
treatment of acoustic nerve tumors. Charles C Thomas, (vestibular schwannoma) surgery: etiology and pre-
Springfield vention. J Neurosurg 87(1):60–66
46. Putnam T (1938) Treatment of recurrent vertigo 56. Stone JL (1999) Sir Charles Ballance: pioneer British
(Meniere’s syndrome) by subtemporal destruction of neurological surgeon. Neurosurgery 44(3):610–631
the labyrinth. Arch Otolaryngol 27:161–168 57. Wishart J (1822) Case of tumours in the skull, dura
47. Quix F (1912) Ein fall von translabyrintharisch operier- mater, and brain. Edinb Med Surg J 18:393
tem tumor acusticus. Verh Dtsch Otol Ges 21:245–255 58. Yasargil M (1996) Acoustic neuromas. In: Microneuro-
48. Ramsden R (1995) ‘A brilliant surgical result, the first surgery, vol IVb. Thieme, New York, pp 100–123
recorded’: Annandale’s case, 3 May 1895. J Laryngol 59. Yasargil MG (1999) A legacy of microneurosurgery:
Otol 109:369–373 memoirs, lessons, and axioms. Neurosurgery 45(5):
49. Ramsden RT (1995) The bloody angle: 100 years of 1025–1092
acoustic neuroma surgery. J R Soc Med 88(8):
464P–468P
Microsurgical Anatomy
of the Cerebellopontine Angle 2
by the Retrosigmoid Approach

Posterior Cranial Fossa retrosigmoid approach directed through the cer-


ebellopontine angle.
The posterior cranial fossa, the largest and deepest
of the three cranial fossae, contains the most com-
plex of the intracranial anatomy. It extends from The Cerebellopontine Angle
the tentorial incisura, through which it communi-
cates with the supratentorial space, to the foramen The cerebellopontine angle is located between the
magnum, through which it communicates with superior and inferior limbs of the cerebellopon-
the spinal canal. The upper surface of the cerebel- tine fissure, a V-shaped angular cleft, formed by
lum is separated from the supratentorial space by the cerebellum folding around the pons and mid-
the tentorium cerebelli. Its intracranial surface is dle cerebellar peduncle (Fig. 2.1) [33, 38, 40].
penetrated by the jugular foramen, internal acous- The cerebellopontine angle faces the posterior
tic meatus, hypoglossal canal, the vestibular and surface of the temporal bone. The middle cerebel-
cochlear aqueducts, and several venous emissary lar peduncle fills the interval between the superior
foramina. Here, in approximately one-eighth the and inferior limbs of the angle (fissure). The apex
intracranial space, are found the pathways regulat- of the angle is located laterally where the superior
ing consciousness, vital autonomic functions, and and inferior limbs meet. The fourth through the
motor activities and sensory reception for the head, eleventh cranial nerves are located near or within
body, and extremities, in addition to the centers for the angular space between the two limbs (Figs. 2.1,
controlling balance and gait. Only two of the 12 2.2, and 2.3). The trochlear and trigeminal nerves
pairs of cranial nerves are located entirely outside are located near the angle’s superior limb, and the
the posterior fossa (Fig. 2.1). The posterior fossa glossopharyngeal, vagus, and accessory nerves
is strategically situated at the outlet of the cerebro- are located near the inferior limb. The facial and
spinal fluid flow from the ventricular system. The acousticovestibular nerves arise near the central
arterial relationships are especially complex with part of the CP angle. The abducens nerve is
the vertebral and basilar arteries having relatively located near the base of the fissure, along a line
inaccessible segments deep in front of the brain connecting the anterior ends of the superior and
stem and the major cerebellar arteries coursing in inferior limbs. The lateral recess and the foramen
relation to multiple sets of cranial nerves before of Luschka open into the middle part of the infe-
reaching the cerebellum. The most commonly rior limb. Other structures located along the mid-
utilized operative route to the posterior fossa is a dle part of the inferior limb are the flocculus, the

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 9


DOI 10.1007/978-3-642-35422-9_2, © Springer-Verlag Berlin Heidelberg 2013
10 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

Fig. 2.1 (a) The petrosal surface of the cerebellum faces retrosigmoid approach. The petrosal surface of the
forward toward the posterior surface of the temporal cerebellum faces forward toward the posterior surface of
bone. It is the surface that is elevated to reach the cerebel- the temporal bone. It is the surface that is elevated to
lopontine angle. The cerebellopontine fissure, also called reach the cerebellopontine angle. The cerebellopontine
the cerebellopontine angle, a V-shaped fissure formed by fissure, the V-shaped fissure formed by the cerebellum
the cerebellum wrapping around the pons and middle cer- wrapping around the pons and middle cerebellar pedun-
ebellar peduncle, has superior and inferior limbs that cle, has superior and inferior limbs that define the mar-
define the margins of the cerebellopontine angle. Cranial gins of the cerebellopontine angle. Cranial nerves V–XI
nerves V–XI arise in or near the cerebellopontine fissure arise in or near the cerebellopontine angle. The superior
or angle. The superior limb extends above the trigeminal limb extends above the trigeminal nerve, and the inferior
nerve, and the inferior limb passes below the flocculus limb passes below the flocculus and the cranial nerves IX,
and the nerves that pass to the jugular foramen. The supe- X, and XI that pass to the jugular foramen. The flocculus
rior and inferior limbs meet laterally at the apex located protrudes from the foramen of Luschka behind cranial
at the anterior end of the petrosal fissure that divides the nerves VII and VIII. A large superior petrosal vein with
petrosal surface of the cerebellum into superior and infe- multiple tributaries passes behind the trigeminal nerve.
rior parts. The fourth ventricle is located behind the pons The trochlear nerve courses below the superior cerebellar
and medulla. The midbrain and pons are separated by the artery. The anteroinferior cerebellar artery passes between
pontomesencephalic sulcus and the pons and medulla by the facial and vestibulocochlear nerves and turns back-
the pontomedullary sulcus. The trigeminal nerves arise ward to course along the middle cerebellar peduncle and
from the midpons. The abducens nerve arises in the cerebellopontine fissure. (d) The cerebellum has been
medial part of the pontomedullary sulcus, rostral to the removed in this specimen to expose the dura covering the
medullary pyramids. The facial and vestibulocochlear part of the posterior fossa that faces the cerebellopontine
nerves arise at the lateral end of the pontomedullary angle and is exposed in the retrosigmoid approach. The
sulcus immediately rostral to the foramen of Luschka. fourth ventricle sits on the posterior surface of the pons
The hypoglossal nerves arise anterior to the olives, and the and medulla. The flocculus projects laterally into the cer-
glossopharyngeal and vagus nerves arise posterior to the ebellopontine angle. The glossopharyngeal, vagus, and
olives. The flocculus and choroid plexus protrude from accessory nerves enter the jugular foramen. The superior
the foramen of Luschka behind to the glossopharyngeal cerebellar artery is at the upper margin of the exposure.
and vagus nerves. The foramen of Luschka opens into the The posteroinferior cerebellar artery courses around the
cerebellopontine angle below the junction of the facial glossopharyngeal, vagus, and accessory nerves. The
and vestibulocochlear nerves with the lateral end of the endolymphatic sac sits beneath the dura inferolateral to
pontomedullary sulcus. Choroid plexus protrudes from the internal acoustic meatus. (e) The supra- and infraten-
the lateral recess and foramen of Luschka behind the torial areas have been exposed while preserving the
glossopharyngeal, vagus, and accessory nerves. (b) bone at the site of the sutures. The asterion, located at the
Enlarged view of the right cerebellopontine angle. The junction of the lambdoid, occipitomastoid, and pariet-
cerebellopontine fissure, which is most commonly omastoid sutures, usually overlies the lower half of the
referred to as the cerebellopontine angle, is a V-shaped junction of the transverse and sigmoid sinuses. The verti-
fissure formed where the cerebellum wraps around the cal lateral suboccipital incision for the retrosigmoid
pons and middle cerebellar peduncle. The flocculus and approach usually crosses the asterion. The burr hole for
choroid plexus extend laterally from the foramen of elevating a suboccipital bone flap is usually placed at the
Magendie above the lower limb of the fissure. The facial lower edge of the asterion. The junction of the supramas-
and vestibulocochlear nerves join the brain stem 2 or toid crest and the squamosal suture is located at the pos-
3 mm rostral to the glossopharyngeal nerve along a line terior edge of the middle fossa and slightly anterior and
drawn dorsal to the olive along the origin of the rootlets above the junction of the transverse and sigmoid sinuses.
of the glossopharyngeal, vagus, and accessory rootlets. The supramastoid crest is an inferior continuation of the
(c) Cerebellopontine angle seen through a right superior temporal line (From Rhoton [33])
The Cerebellopontine Angle 11

c d

Fig. 2.1 (continued)

rhomboid lip, and the choroid plexus protruding parts of the brain stem, one of the three surfaces of
from the foramen of Luschka. the cerebellum, one of the three cerebellar pedun-
Optimizing operative approaches to the cer- cles, and one of the three major fissures between
ebellopontine angle requires an understanding of the cerebellum and the brain stem. In addition, each
the relationships of the cerebellar arteries, cranial neurovascular complex contains a group of cranial
nerves, brain stem, cerebellar peduncles, fissures nerves. The upper complex includes the oculo-
between the cerebellum and brain stem, and the motor, trochlear, and trigeminal nerves that are
cerebellar surfaces. When examining these relation- related to the SCA. The middle complex includes
ships, three neurovascular complexes are defined: the abducens, facial, and vestibulocochlear nerves
an upper complex related to the superior cerebellar that are related to the AICA. The lower complex
artery (SCA), a middle complex related to the antero- includes the glossopharyngeal, vagus, accessory,
inferior cerebellar artery (AICA), and a lower com- and hypoglossal nerves that are related to the PICA.
plex related to the posteroinferior cerebellar artery The structures in each complex are reviewed next.
(PICA). Other structures, in addition to the three
cerebellar arteries, occurring in sets of three in the
posterior fossa that bear a consistent relationship to Cerebellar Surfaces
the SCA, AICA, and PICA are the parts of the brain
stem (midbrain, pons, and medulla); the cerebel- The cerebellar surfaces are divided on the basis of
lar peduncles (superior, middle, and inferior); the the structures they face, or along which they may
fissures between the brain stem and the cerebellum be exposed. The first surface, the tentorial surface,
(cerebellomesencephalic, cerebellopontine, and faces the tentorium and is retracted in a supracer-
cerebellomedullary); and the surfaces of the cere- ebellar approach; the second surface, the suboc-
bellum (tentorial, petrosal, and suboccipital). Each cipital surface, is located below and between the
neurovascular complex includes one of the three lateral and sigmoid sinuses and is exposed in
12 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

Fig. 2.2 (a) Upper part of the cerebellopontine angle. to expose the dura lining the meatus. The subarcuate
A large superior petrosal vein with multiple tributaries, artery usually has to be obliterated and divided prior to
including the pontotrigeminal and transverse pontine removing the posterior meatal wall. Two bundles from
veins and the vein of the cerebellopontine fissure, passes the nervus intermedius are exposed above the vestibulo-
behind the trigeminal nerve. The trochlear nerve courses cochlear nerve. Care is taken to avoid entering the semi-
below the superior cerebellar artery. The anteroinferior circular canals and vestibule during drilling of the
cerebellar artery passes between the facial and vestibulo- posterior wall of the meatus if hearing is to be preserved.
cochlear nerves and turns medially to course along the (d) The meatal dura has been opened, the facial nerve
middle cerebellar peduncle and cerebellopontine fissure. has been elevated, and the vestibulocochlear nerve
(b) The flocculus and choroid plexus, which protrude depressed to expose the facial nerve coursing in the
from the foramen of Luschka, have been elevated to anterosuperior quadrant of the meatus. The nervus inter-
expose the junction of the facial and vestibulocochlear medius, which arises along the anterior surface of the
nerves with the brain stem. The facial nerve is exposed vestibulocochlear nerve and passes laterally to join the
below the vestibulocochlear nerve. A branch of the facial nerve, is composed of several rootlets, as is com-
anteroinferior cerebellar artery gives rise to both the sub- mon. The superior vestibular nerve passes posterior to
arcuate and labyrinthine arteries. The subarcuate artery the facial nerve, and the cochlear nerve is hidden anterior
enters the dura and bone superolateral to the meatus. The to the inferior vestibular nerve. (e) The cleavage plane
junction of the facial nerve with the brain stem is easier between the superior and inferior vestibular nerves has
to expose from below rather than above the flocculus and been developed. The superior vestibular and facial nerves
vestibulocochlear nerve in an operation for hemifacial pass above and the inferior vestibular and cochlear
spasm. This approach for decompressing the facial nerve nerves pass below the transverse crest. The facial nerve
in hemifacial spasm is referred to as an “infrafloccular courses anterior to the superior vestibular nerve, and the
approach.” A large posteroinferior cerebellar artery loops cochlear nerve is located anterior to the inferior vestibu-
upward behind the vestibulocochlear nerve. (c) The dura lar nerve. The vertical crest separates the superior ves-
lining the posterior wall of the internal acoustic meatus tibular and facial nerves at the fundus of the meatus
has been removed, and the posterior meatal wall opened (From Rhoton [33])
The Cerebellopontine Angle 13

d e

Fig. 2.2 (continued)

a b

Fig. 2.3 Retrosigmoid exposure of the nerves in the on the anterior surface of the vestibulocochlear nerve,
right cerebellopontine angle. (a) The vestibulocochlear has a free segment in the cistern and/or meatus, and
nerve enters the internal acoustic meatus with a labyrin- joins the facial nerve distally. The facial nerve is located
thine branch of the AICA. The PICA courses around anterior to the superior vestibular nerve and the cochlear
the glossopharyngeal, vagus, and accessory nerves. The nerve is anterior to the inferior vestibular nerve. (c) The
abducens nerve ascends in front of the pons. A subarcu- cleavage plane between the cochlear and inferior
ate artery enters the subarcuate fossa superolateral to vestibular nerves, which is well developed in the lateral
the porus of the meatus. Choroid plexus protrudes into end of the internal acoustic meatus, has been extended
the cerebellopontine angle behind the glossopharyngeal medially. Within the cerebellopontine angle, the supe-
and vagus nerves. (b) The posterior wall of the internal rior vestibular nerve is posterior and superior, the facial
acoustic meatus has been removed. The cleavage plane nerve anterior and superior, the inferior vestibular nerve
between the upper bundle, formed by the superior ves- posterior and inferior, and the cochlear nerve anterior
tibular nerve, and the lower bundle, formed by the infe- and inferior. (d) The superior and inferior vestibular
rior vestibular and cochlear nerves, was begun laterally nerves have been divided to expose the facial and
where the nerves have separated near the meatal fundus cochlear nerves. A labyrinthine branch of the PICA
and extended medially. The nervus intermedius arises enters the internal meatus (From Rhoton [40])
14 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

c d

Fig. 2.3 (continued)

a suboccipital craniectomy; and the third surface, Cerebellar Brain Stem Fissures
the petrosal surface, faces forward toward the pos-
terior surface of the petrous bone and is elevated to The three fissures formed by the embryological
expose the cerebellopontine angle. Each of the folding of the cerebellum around the brain stem
surfaces has the vermis in the midline and the are the cerebellomesencephalic fissure, which
hemispheres laterally (Figs. 2.4, 2.5, and 2.6). extends inferiorly between the cerebellum and

a b

Fig. 2.4 SCA. (a) Superior view. The SCA arises at the trigeminal nerve with the midpons. (d) Posterolateral
level of the midbrain and dips caudally along the pon- view. The SCA bifurcates early and both the rostral and
tomesencephalic junction to enter the cerebellomedullary caudal trunks dip into the cerebellomesencephalic fissure.
fissure and supply the tentorial surface of the cerebellum. (e) The superior lip of the cerebellomesencephalic
(b) Lateral view of the right pontomesencephalic junction fissure has been removed to expose the SCA within the fissure
showing the SCA dipping down toward the trigeminal where it gives branches that continue down the superior
nerve and dividing into a rostral and caudal trunk. (c) cerebellar peduncle to the dentate nucleus. (f) Superior
Anterior view. The SCA arises anterior to the midbrain view showing the cortical branches of the SCA supplying
and loops downward to the level of the junction of the the tentorial surface of the cerebellum
The Cerebellopontine Angle 15

c d

e f

Fig. 2.4 (continued)

a b

Fig. 2.5 AICA. (a) Anterior view. The AICA arises at surface of the middle cerebellar peduncle, and toward the
the pontine level and passes below CN VI, above the apex of the cerebellopontine fissure. Its cortical branches
flocculus, and between CNs VII and VIII to run on supply the petrosal surface of the cerebellum. (d) Posterior
the middle cerebellar peduncle, which forms the floor of the view with the posterior meatal lip removed to show the
cerebellopontine angle. (b) Enlarged view. The cerebel- AICA looping into the porus of the meatus. (e) Right ante-
lopontine angle is formed by the cerebellum folding rolateral view of the AICA passing along the cerebello-
around the pons to form an angular cleft with the flocculus pontine fissure and supplying the petrosal cerebellar
and CNs VII and VIII in the center. The fissure has upper surface. (f) Anterolateral view of another brain stem and
and lower limbs that meet at a lateral apex. (c) View cerebellum showing the AICA passing above CNs VII
through a retrosigmoid craniotomy. The AICA passes and VIII and along the cerebellopontine fissure to supply
between CNs VII and VIII, above the flocculus on the the petrosal surface
16 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

c d

e f

Fig. 2.5 (continued)

mesencephalon and is intimately related to the in each fissure. The SCA courses within the cer-
superior half of the roof of the fourth ventricle; ebellomesencephalic fissure, the AICA is related
the cerebellopontine fissure, which is formed by to the cerebellopontine fissure (also called the
the folding of the cerebellum around the lateral cerebellopontine angle), and the PICA is inti-
sides of the pons and is intimately related to the mately related to the cerebellomedullary fissure.
lateral recesses of the fourth ventricle; and the
cerebellomedullary fissure, which extends supe-
riorly between the cerebellum and the medulla Cerebellar Arteries
and is intimately related to the inferior half of the
roof (Figs. 2.4, 2.5, and 2.6). Each fissure com- The AICA is the cerebellar artery most intimately
municates with the adjacent fissure. The cerebel- related to the cerebellopontine angle, but a segment
lopontine fissures are continuous around the of the SCA and PICA commonly enters the angle
rostral surface of the middle cerebellar peduncles especially if they have a tortuous course [39].
with the caudal edges of the cerebellomesenceph- The SCA is intimately related to the cerebellom-
alic fissure and around the caudal margin of the esencephalic fissure, the superior half of the fourth
middle cerebellar peduncles with the rostral lim- ventricular roof, the superior cerebellar peduncle,
its of the cerebellomedullary fissure. The external and the tentorial surface (Fig. 2.4). The SCA arises
or cisternal surfaces of the structures forming the in front of the midbrain, usually from the basilar
roof of the fourth ventricle are intimately related artery near the apex, and passes below the oculomo-
to the fissures between the cerebellum and brain tor nerve, but may infrequently arise from the proxi-
stem. One of the three cerebellar arteries courses mal PCA and pass above the oculomotor nerve.
The Cerebellopontine Angle 17

a b

d
c

e
f

Fig. 2.6 PICA. (a) Left posterolateral view. The cerebel- the rootlets of the hypoglossal nerve. (d) Posterior view.
lum was removed by dividing the cerebellar peduncles. The PICA arises anterior to the midportion of the olive,
The PICA arises anterior to the inferior olive and courses passes posteriorly by the rootlets of CN XII, and courses
between CNs X and XI and along the posterolateral along the posterolateral medulla. (e) The right half of the
medulla and inferior cerebellar peduncle. It forms a cau- cerebellum has been removed to show the PICA looping
dal loop below the tonsil. (b) Posterior view. The AICA around the tonsil to form a cranial loop and through the
courses below CN X and along the posterolateral medulla cerebellomedullary fissure to supply the suboccipital
and inferior cerebellar peduncle. (c) Superior view of an surface. (f) Posterior view of the suboccipital cerebellar
axial section through the medulla. The PICA arises along surface. The PICA courses along the caudal margin of the
the anterolateral margin of the medulla near and distorting tonsil and supplies the cortical surface

It dips caudally and encircles the brain stem near the proximal portion courses medial to the free edge of
pontomesencephalic junction, passing below the the tentorium cerebelli, and its distal part passes
trochlear nerve and above the trigeminal nerve. Its below the tentorium. After passing above the trigeminal
18 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

nerve, it enters the cerebellomesencephalic fissure, ventricular roof, the inferior cerebellar peduncle,
where its branches make several sharp turns and give and the suboccipital surface (Fig. 2.6). The PICA,
rise to the precerebellar arteries, which pass to the by definition, arises from the vertebral artery near
deep cerebellar white matter and the dentate nucleus. the inferior olive and passes posteriorly around the
On leaving the cerebellomesencephalic fissure medulla. At the anterolateral margin of the medulla,
where its branches are again medial to the tentorial it passes rostral or caudal to or between the rootlets
edge, its branches pass posteriorly under the tento- of the hypoglossal nerve, and at the posterolateral
rial edge and are distributed to the tentorial cerebel- margin of the medulla, it courses rostral to or
lar surface. It usually arises as a single trunk, but between the fila of the glossopharyngeal, vagus,
may also arise as a double (or duplicate) trunk. The and accessory nerves. After passing the latter
SCAs arising as a single trunk bifurcate into a rostral nerves, it courses around the cerebellar tonsil,
and a caudal trunk. The SCA gives off perforating enters the cerebellomedullary fissure, and passes
branches to the brain stem and cerebellar peduncles. posterior to the lower half of the roof of the fourth
Precerebellar branches that pass along the superior ventricle. On exiting the cerebellomedullary fissure,
cerebellar peduncle to the dentate nucleus arise its branches are distributed to the vermis and hemi-
within the cerebellomesencephalic fissure. The ros- sphere of the suboccipital surface. Most PICAs
tral trunk supplies the vermian and paravermian bifurcate into a medial and a lateral trunk. The
area, and the caudal trunk supplies the hemisphere medial trunk supplies the vermis and adjacent part
on the suboccipital surface. of the hemisphere, and the lateral trunk supplies the
The AICA courses through the central part of the cortical surface of the tonsil and the hemisphere.
cerebellopontine angle near the facial and vestibulo- The PICA gives off perforating, choroidal, and cor-
cochlear nerve and is intimately related to the pons, tical arteries. The cortical arteries are divided into
lateral recess, foramen of Luschka, cerebellopontine vermian, tonsillar, and hemispheric groups.
fissure, middle cerebellar peduncle, and petrosal
cerebellar surface (Fig. 2.5). The AICA originates
from the basilar artery, usually as a single trunk, and The Fourth Ventricle
encircles the pons near the abducens, facial, and ves-
tibulocochlear nerves. After coursing near and send- The fourth ventricle is a broad, tent-shaped
ing branches to the nerves entering the acoustic midline cavity located ventral to the cerebellum,
meatus and to the choroid plexus protruding from dorsal to the pons and medulla, and medial to the
the foramen of Luschka, it passes around the cerebellar peduncles. It is connected rostrally
flocculus on the middle cerebellar peduncle to sup- through the aqueduct with the third ventricle, cau-
ply the lips of the cerebellopontine fissure and the dally through the foramen of Magendie with the
petrosal cerebellar surface. It commonly bifurcates cisterna magna, and laterally through the foram-
near the facial-vestibulocochlear nerve complex to ina of Luschka with the cerebellopontine angles.
form a rostral and a caudal trunk. The rostral trunk Most of the cranial nerves arise near its floor. It
sends its branches laterally along the middle cere- has a roof, a floor, and two lateral recesses.
bellar peduncle to the superior lip of the cerebello- The ventricular roof is tent-shaped. The roof
pontine fissure and the adjoining part of the petrosal expands laterally and posteriorly from its narrow
surface, and the caudal trunk supplies the inferior rostral end just below the aqueduct to the level of
part of the petrosal surface, including a part of the the fastigium and lateral recess, the site of its
flocculus and the choroid plexus. The AICA gives greatest height and width, and from there, it
rise to perforating arteries to the brain stem, choroi- tapers to a narrow caudal apex at the level of the
dal branches to the tela and choroid plexus, and the foramen of Magendie. The apex of the roof, the
nerve-related arteries, including the labyrinthine, fastigium, divides the roof into superior and infe-
recurrent perforating, and subarcuate arteries. rior parts. The superior part is distinctly different
The PICA is intimately related to the cerebel- from the inferior part in that the inferior part is
lomedullary fissure, the inferior half of the fourth formed largely by thin membranous layers and
Upper Neurovascular Complex 19

the superior part is formed by thicker neural The middle complex includes the AICA, pons,
structures. middle cerebellar peduncle, cerebellopontine
The lateral recesses of the fourth ventricle are fissure, petrosal surface of the cerebellum, and
narrow, curved pouches formed by the union of the abducens, facial, and vestibulocochlear
the roof and the floor that extends laterally below nerves. The AICA arises at the pontine level and
the cerebellar peduncles to open through the courses in relationship to the abducens, facial,
foramina of Luschka into the cerebellopontine and vestibulocochlear nerves to reach the surface
angles. The ventral wall of each lateral recess is of the middle cerebellar peduncle, where it
formed by the junctional part of the ventricular courses along the cerebellopontine fissure and
floor and the rhomboid lip, a sheetlike layer of terminates by supplying the petrosal surface of
neural tissue that extends laterally from the floor the cerebellum.
and unites with the tela choroidea to form a pouch The lower complex includes the PICA,
at the outer extremity of the lateral recess. The medulla, inferior cerebellar peduncle, cerebel-
rostral wall of each lateral recess is formed by lomedullary fissure, suboccipital surface of the
the caudal margin of the cerebellar peduncles. cerebellum, and the glossopharyngeal, vagus,
The inferior cerebellar peduncle courses upward accessory, and hypoglossal nerves. The PICA
in the floor ventral to the lateral recess and turns arises at the medullary level and encircles the
posteriorly at the lower part of the pons, to form medulla, passing in relationship to the
the ventricular surface of the rostral wall of the glossopharyngeal, vagus, accessory, and hypo-
recess. The peduncle of the flocculus intercon- glossal nerves to reach the surface of the infe-
necting the inferior medullary velum and the rior cerebellar peduncle, where it dips into the
flocculus of the vermis crosses in the dorsal mar- cerebellomedullary fissure and terminates by
gin of the lateral recess. The caudal wall is formed supplying the suboccipital surface of the
by the tela choroidea that stretches from the lat- cerebellum.
eral edge of the ventricular floor to the peduncle The discussion of microsurgical anatomy is
of the flocculus. The flocculus is superior to the divided in sections related to approaches to the
outer extremity of the lateral recess. The rootlets neurovascular complexes. The most common
of the glossopharyngeal and vagus nerves arise operation directed to the upper neurovascular
ventral to and the facial nerve arises rostral to the complex is the exposure of the posterior root of
lateral recess. The dorsal and ventral cochlear the trigeminal nerve for a vascular decompres-
nuclei, the site of placement of the auditory brain sion procedure for trigeminal neuralgia.
stem implant and fibers related to the vestibulo- Operations directed to the middle complex are
cochlear nerve, are in the floor of the recess. for the removal of acoustic neuromas and other
tumors and for the relief of hemifacial spasm.
Our most common operations directed to the
Three Neurovascular Complexes lower complex are for glossopharyngeal neural-
gia and tumors centered near the jugular tubercle
In summary, the upper complex includes the and foramen [40]. Tumors involving multiple
SCA, midbrain, cerebellomesencephalic fissure, complexes are also reviewed.
superior cerebellar peduncle, tentorial surface of
the cerebellum, and the oculomotor, trochlear,
and trigeminal nerves (Fig. 2.7). The SCA arises Upper Neurovascular Complex
in front of the midbrain and passes below the
oculomotor and trochlear nerves and above the The most common operation directed to the
trigeminal nerve to reach the cerebellomesen- upper neurovascular complex is the exposure of
cephalic fissure, where it runs on the superior the posterior root of the trigeminal nerve. The
cerebellar peduncle and terminates by supplying posterior trigeminal root joins the brain stem
the tentorial surface of the cerebellum. about halfway between the lower and upper
20 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

Upper complex
SCA
Midbrain
CN III, IV, V
Cerebellomesencephalic Fiss.
Superior Cerebellar Ped.
Tentorial Surface
Middle complex
AICA
Pons
CN VI, VII, VIII
Middle Cerebellar Ped.
Lower complex Cerebellopontine Fiss.,
PICA Petrosal Surface
Medulla
CN IX, X, XI, XII
Cerebellomedullary Fiss.
Inferior Cerebellar Ped.
Suboccipital Surface

Fig. 2.7 Each of the three neurovascular complexes in the cerebellar peduncle, cerebellomedullary fissure, suboc-
posterior fossa includes one of the three cerebellar arter- cipital surface, and the glossopharyngeal, vagus, acces-
ies, one of the three parts of the brain stem, one of the sory, and hypoglossal nerves. The SCA is divided into
three cerebellar peduncles, one of the three cerebellar sur- four segments: anterior pontomesencephalic (green), lat-
faces, one of the three fissures between the brain stem and eral pontomesencephalic (orange), cerebellomesenceph-
the cerebellum, and one of the three groups of cranial alic (blue), and cortical (red). Each segment may be
nerves. The upper complex is related to the SCA, the mid- composed of one or more trunks, depending on the level
dle complex is related to the AICA, and the lower com- of bifurcation of the main trunk. The AICA is divided into
plex is related to the PICA. The upper complex includes four segments: anterior pontine (green), lateral pontomed-
the SCA, midbrain, superior cerebellar peduncle, cerebel- ullary (orange), flocculonodular (blue), and cortical (red).
lomesencephalic fissure, tentorial cerebellar surface, and The PICA is divided into five segments: anterior medul-
the oculomotor, trochlear, and trigeminal nerves. The mid- lary (green), lateral medullary (orange), tonsillomedullary
dle complex includes the PICA, pons, middle cerebellar (blue), telovelotonsillar (yellow), and cortical (red). AICA
peduncle, cerebellopontine fissure, petrosal surface, and anteroinferior cerebellar artery, CN cranial nerve, Fiss.
the abducens, facial, and vestibulocochlear nerves. The fissure, Ped. peduncle, PICA posteroinferior cerebellar
lower complex includes the PICA, medulla, inferior artery, SCA superior cerebellar artery (From Rhoton [39])

borders of the pons (Figs. 2.1, 2.2, 2.3, 2.5, and Trigeminal Root Anatomy
2.8). Frequently, the lip of cerebellum projects
forward and obscures the course of the posterior The fibers from the third division remain in a cau-
root through the middle cerebellar peduncle into dolateral position in the posterior root throughout
the pons. In its intradural course, the trigeminal the interval from the ganglion to the pons, the first
nerve uniformly runs obliquely upward from the division rostromedial, with second-division fibers
lateral part of the pons toward the petrous apex. in an intermediate position (Figs. 2.8, 2.9, and
It exits the posterior fossa to enter the middle 2.10) [3, 6]. There are anastomoses between the
cranial fossa by passing forward beneath the fibers from each division in the area posterior to
tentorial attachment to enter Meckel’s cave, the ganglion (Fig. 2.8). Results of selective rhizo-
which sits in the trigeminal impression on the tomy of the posterior root indicate that somato-
upper surface of the petrous part of the temporal topic localization with the third division
bone. inferolaterally and the ophthalmic division
Upper Neurovascular Complex 21

a b

c d

Fig. 2.8 Lateral views, right trigeminal nerve. (a) Meckel’s anastomosis in the region of Meckel’s cave. (d) Four motor
cave, the cistern which extends forward from the posterior rootlets, which arise around the rostral margin of the
fossa along the posterior trigeminal root to the level of the sensory root, have been elevated to expose the anastomoses
midportion of the ganglion, has been exposed by removing between the motor and sensory roots. The cerebellar lip
the lateral dural wall of the cave. The motor root arises projects forward and may hide the junction of the sensory
rostral to the sensory root and passes through Meckel’s root with the pons in the retrosigmoid approach. (e) A
cave on the medial side of the posterior sensory root and cleavage plane has been started anteriorly and extended
ganglion. (b) The dura has been removed to expose the backward to the level of the posterior root. The first-division
posterior root and ganglion and the three trigeminal divi- fibers are rostromedial within the posterior root and the
sions. There is diffuse anastomosis between the rootlets third-division fibers caudolateral with the second division
posterior to the ganglion. (c) Enlarged view of the diffuse being in an intermediate location (From Rhoton [40])
22 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b
A
A B
B

F C
F

E D
E D

Fig. 2.9 Diagrams of 12 trigeminal nerves showing the rootlets are shown by the dark outline with clear center.
relationship of the trigeminal sensory root, motor rootlets, None are present in B and C. In A, D, E, and F, some aber-
and aberrant sensory rootlets at the site of entry into the rant rootlets arose farther from the sensory root than some
pons. The central diagrams are for orientation and show of the motor rootlets. Lines through the oval representing
the elliptical cross section of the sensory root. The large the main sensory root show portions of the nerve from
ovals (A–F) represent the sensory root and are oriented in each of the three divisions. In all diagrams, the rostrome-
the same manner as the sensory root in the central dia- dial portion was from the first division, the caudolateral
gram. The sites of origin of the motor rootlets are black. portion was from the third division, and the second divi-
(a) Nerves on the right. Only 5 motor rootlets are present sion was in an intermediate position. In all these nerves,
in B, but 13 are seen in F. The aberrant sensory rootlets except A and B in the left nerve, the second-division fibers
are shown by the dark outline with clear center. None are made up a greater portion of the medial than the lateral
present in D and F. In C and E, some aberrant rootlets portion of the sensory root. Small arteries or veins cours-
arose farther from the sensory root than some of the motor ing between the rootlets at the level of entry into the pons
rootlets. (b) Nerves on the left. Only 4 motor rootlets are are shown in all diagrams of both nerves except D in the
present in A, but there are 10 in B and C. Aberrant sensory right nerve (a) (From Gudmundsson et al. [10])

dorsomedially is well maintained posterior to and division; others are rotated nearly 70° away from
despite the prominent retrogasserian anastomoses this so that the third-division fibers will be directly
[3]. A cross section of the sensory root between caudal to those of the first division. Cutting into
the pons and the petrous apex is elliptical. In most the nerve partially from a caudolateral direction
nerves, the angle between the longest diameter of would give a significantly different pattern of sen-
this cross section and the long axis of the body is sory loss if the nerve is rotated with the third divi-
40–50°; the angle, however, can vary from 10° to sion lateral to the first, than if the third division is
80° (Fig. 2.10) [10]. An angle of 80° places the almost directly caudal to the first division.
third-division fibers almost directly lateral to those At the junction of the nerve with the pons, as
of the first division, but an angle of 10° places the many as 15 separate nerve rootlets may be spread
third-division fibers almost directly caudal to around the rostral half of the site where the main
those of the first division. The variability in the sensory cone enters the pons [10]. These rootlets
degree of rotation of the sensory root entering the are either motor or aberrant sensory rootlets. The
pons may explain some differences in the quantity aberrant sensory fibers are small rootlets that
of sensation retained after partial section of the penetrate the pons outside the main sensory root
nerve in the posterior cranial fossa. The most fre- (Figs. 2.9 and 2.11). The aberrant rootlets arise
quent pattern is for the third-division fibers to be around the rostral two-thirds of the nerve and
caudolateral to the first-division fibers; some usually join the root a short distance from the
nerves, however, are rotated so that third-division brain stem. There may be as many as eight
fibers will be almost directly lateral to the first aberrant roots. Those arising rostral to the sensory
Upper Neurovascular Complex 23

root most frequently enter the first division and


those arising more caudally enter the second or
third division. No aberrant rootlets originate
around the caudal third of the sensory root. Of 66
aberrant rootlets found in our study of 50 trigemi-
nal nerves, 49 went into the first division, 10 into
the second division, and 7 into the third division
[10]. The findings that aberrant rootlets are most
commonly related to the first division agree with
Dandy’s conclusion that when the accessory
fibers are spared, sensation in the first division
tends to be spared [3]. The aberrant rootlets
appear to be nonspecific sensory fibers separated
V1
V1 from the root by transverse pontine fibers [10, 48].
V2 V2
V3 Aberrant roots contribute mainly to the first divi-
V1
V3 sion and probably do not convey a specific
V2 sensory modality from all three divisions.
ο ο
80 V3 10 Motor rootlets also arise around the rostral part
of the nerve; however, they tend to arise further
ο
45 from the main sensory cone than do the accessory
sensory rootlets. The motor root may be composed
A C
B of 4–14 separate rootlets, each having a separate
exit from the pons (Figs. 2.8, 2.9, 2.11, and 2.12).
Fig. 2.10 Variability of the longest axis of the elliptical
cross section of the trigeminal nerve at the pons (broken The aberrant sensory fibers usually arise closer to
line) to the longitudinal axis of the body (solid line). The the main sensory root than to the motor fibers.
long axis of most nerves makes a 40–50° angle with the Some aberrant sensory fibers, however, will arise
longitudinal axis of the body (A); however, this can vary further from the main sensory root than does the
from 10° (C) to 80° (B). In B, the third division is almost
directly lateral to the first division, and in C, it is almost origin of some motor filaments; for this reason, it
directly caudal (From Gudmundsson et al. [10]) is easy to confuse aberrant sensory fibers and
motor filaments at the nerve/pons junction.
Anastomoses between the motor and sensory
roots are present in most nerves (Fig. 2.8). Those
sensory fibers associated with the motor root
from the pons to just proximal to the ganglion,
where they anastomose with the sensory root,
would be spared with a rhizotomy in the posterior
fossa. Horsley et al. [13] suspected that there
were sensory fibers in the trigeminal motor root
suggesting that the motor root be sectioned if
trigeminal neuralgia recurred after the complete
section of the sensory root. Our studies offer two
explanations for the accidental preservation of
sensation after posterior rhizotomy: (1) sparing
Fig. 2.11 Lateral view of the junction of the left trigemi-
of the aberrant sensory rootlets and (2) sparing of
nal nerve with the pons. A nerve hook is between the large the anastomotic sensory fibers that run with the
aberrant sensory rootlet and the main sensory root. The motor root at the level of the rhizotomy [10].
aberrant rootlet arises from the pons directly lateral to the Anastomosis is a more likely explanation for the
sensory root and joins the sensory root about 1 cm from
the brain stem. Four motor rootlets are seen above the sen-
accidental sensory preservation and recurrence of
sory root (From Gudmundsson et al. [10]) trigeminal neuralgia after the section of the
24 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

Fig. 2.12 (a) Lateral view of the right trigeminal nerve proved to be a motor rootlet when traced distally. This
near its junction with the pons. The arrow points to the illustrates the difficulty in telling whether an intermediate
intermediate group of fibers between the motor rootlet and group of fibers is motor or sensory unless the nerve
the sensory root. (b) The same trigeminal nerve. The bundles can be separated and examined individually
arrow points to the intermediate group of fibers that (From Gudmundsson et al. [10])

posterior root because anastomotic rootlets are craniotomy, located behind the upper half of the
present throughout the interval from the pons to sigmoid sinus exposes the edge of the junction of
the ganglion. Aberrant sensory roots are present the transverse and sigmoid sinuses in its supero-
in only one-half of the nerves. They provide lateral margin. The cerebellum is relaxed by
another explanation for the preservation of opening the arachnoid and removing cerebrospi-
sensation following the section of the main nal fluid, a maneuver made safer by the use of the
sensory root. operating microscope. A narrow brain spatula,
commonly 3 mm at the tip, is introduced parallel
and just below the superior petrosal sinus to ele-
Anatomy of Vascular Compression vate the superolateral margin of the cerebellum
in the Upper Neurovascular Complex [30]. The use of a wider spatula or a lower entry
point along the lateral cerebellum risks damaging
In 1934, Dandy postulated that arterial compres- the vestibulocochlear nerve. A bridging petrosal
sion and distortion of the trigeminal nerve might vein, which commonly blocks access to the
be the cause of trigeminal neuralgia [4]. He trigeminal nerve, is coagulated with gentle bipo-
described the SCA as affecting the nerve in lar coagulation and divided nearer its junction
30.7 % of his 215 cases of trigeminal neuralgia. with the brain than to the superior petrosal sinus.
The vascular compression theory failed to gain Unexpected bleeding encountered as the supero-
acceptance at the time, but it awaited the better lateral margin of the cerebellum is elevated, if
demonstration of these pathological changes at venous in appearance, is usually related to stretch-
surgery by Jannetta [15, 18] using magnification ing and tearing of the petrosal veins that pass
provided by the operating microscope. from the superior surface of the cerebellum to the
This upper neurovascular complex, for a venous sinus in the tentorium or, if arterial in
vascular decompression operation, is approached appearance, to tearing of the subarcuate branch
utilizing a vertical scalp incision crossing the of the AICA behind the internal auditory canal at
asterion, which usually overlies the junction of its site of penetration of the dura covering the
the lower half of the transverse and sigmoid subarcuate fossa. The trochlear nerve is identified
sinuses (Fig. 2.13). The bone opening, a small before opening the arachnoid behind the
Upper Neurovascular Complex 25

Sig. Sinus

Trans. Sinus

VIII
VII

S.C.A
V Pons

IV
Sup. Pet. Sinus

Fig. 2.13 Retrosigmoid approach to the trigeminal nerve to 3 or 5 mm at the tip to expose the site at which the
for a microvascular decompression operation. (a) (Upper trigeminal nerve enters the pons. The brain spatula is
left) The patient is positioned in the three-quarter prone advanced and aligned parallel to the superior petrosal
position. The surgeon is seated at the head of the table. sinus. The trochlear nerve is at the superior margin of the
The table is tilted so that the feet are lower than the heart. exposure, and the facial and vestibulocochlear nerves are
(b) The vertical paramedian incision crosses the asterion. at the lower margin. The dura is tacked up to the adjacent
The superolateral margin of the craniotomy is positioned muscles to maximize the exposure along the superolateral
at the junction of the transverse and sigmoid sinuses. (c) margin of the cerebellum. The main trunk of the SCA
The superolateral margin of the cerebellum is gently ele- loops down into the axilla of the trigeminal nerve (From
vated using a brain spatula tapered from 10 mm at the base Rhoton [29])
26 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

c d

Fig. 2.14 Trigeminal nerve and SCA relationships. trigeminal root at the pontine junction. The cerebellar lip
(a) The trigeminal posterior root, ganglion, and three divi- projects forward and may block access to the junction of
sions have been exposed by removing the dura from the the trigeminal nerve and pons in the retrosigmoid
lateral wall of Meckel’s cave and the cavernous sinus. The approach. (c) SCA with an early bifurcation. The rostral
posterior root enters the midpons below the SCA and is trunk loops downward and indents the upper surface of
intertwined with the branches of the superior petrosal the trigeminal nerve. (d) Another SCA passes around the
vein. (b) The SCA loops downward and, at the junction of pons and bifurcates into its rostral and caudal trunks above
the rostral and caudal trunks, contacts the posterior the trigeminal root entry zone (From Rhoton [40])

trigeminal nerve because it may be difficult to see nerve [12]. Normally, the SCA encircles the brain
the nerve after the arachnoid has been opened stem well above the trigeminal nerve. In adults,
and shrinks into thick white clumps that may hide the SCA commonly makes a shallow, caudal loop
the nerve. Usually, the trochlear nerve is several and courses inferiorly for a variable distance on
millimeters above the trigeminal nerve; it may be the lateral side of the pons (Figs. 2.13, 2.14, 2.15,
carried downward, however, if it is adherent to a and 2.16). In those cases with the most prominent
segment of the SCA that has looped into the axilla caudally projecting loop, contact between the
of the trigeminal nerve. The overhanging lip of artery and the trigeminal nerve occurs. The point
the cerebellomesencephalic fissure must be of contact with the SCA is usually on the superior
retracted gently to expose the junction of the or supermedial aspect of the nerve, and often, a
nerve with the pons. few fascicles of the nerve are distorted by an SCA
that has looped down into the axilla between the
medial side of the nerve and the pons. An arterial
Arterial Relationships loop in the axilla may not be visible from the ret-
rosigmoid view behind the trigeminal nerve if the
The most common finding at a vascular decom- SCA courses around the brain stem directly in
pression operation for trigeminal neuralgia is a front of the nerve. The loop of the SCA also may
segment of the SCA compressing the trigeminal be difficult to see if the artery passes over the
Upper Neurovascular Complex 27

b Ro.Tr d
Main Tr.

Ro.Tr
Ca.Tr
Ca.Tr

a V V

Bas.A. S.C.A

f S.C.A.
e

A.I.C.A. Pontine A.

Fig. 2.15 Sites of arterial compression of the trigeminal distally to the nerve. The nerve is compressed by the
nerve. Orientation as shown in the central diagram. (a) main trunk. (d) The SCA bifurcates prior to reaching the
Central diagram. The right trigeminal nerve is com- nerve. The nerve is compressed by both the rostral and
pressed by a tortuous basilar artery, and the left trigemi- caudal trunks. (e) The nerve is compressed by a large
nal nerve is compressed by the main trunk of the SCA. pontine artery. (f) The nerve is compressed by an AICA
(b) The SCA bifurcates into rostral and caudal trunks that has a high origin and loops upward into the medial
prior to reaching the trigeminal nerve. The nerve is com- surface of the nerve. The SCA passes around the brain
pressed by the caudal trunk. (c) The SCA bifurcates stem above the nerve (From Rhoton [29])

rostral aspect of the nerve very close to the brain visible above the nerve. These loops of the SCA,
stem, where it may be hidden by the overhanging however, always pass rostrally along the medial
lip of the cerebellomesencephalic fissure. The and superior surfaces of the nerve to reach the cer-
loop of the SCA may be seen dangling below the ebellomesencephalic fissure. The medial axilla of
lower margin of the nerve, even though it is not the nerve must be carefully explored before
28 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

Main Tr.

Ra.Tr.
Ca Tr.
c Ca.Tr.

IV

Ro Tr.
b Ca Tr. Sup.
d
Petrosal Sinus
V

S.C.V
Pons
VII
VIII

a Pontine A.

Bas.A.

e
A.I.C.A

Fig. 2.16 Sites of arterial compression of the trigeminal The trigeminal nerve is compressed by a loop of the SCA
nerve as seen through a suboccipital craniotomy. (a) Central that dangles down into the axilla of the nerve. The site of
diagram. The site of the skin incision (solid line) and the compression on the artery is at the junction of the main trunk
craniotomy (interrupted line) are shown in the insert. The with the rostral and caudal trunks. (b) The nerve is com-
superolateral margin of the cerebellum is gently retracted to pressed by the caudal trunk. (c) The nerve is compressed by
expose the trigeminal nerve and the SCA. The brain spatula the main trunk. (d) Compression by both the rostral and cau-
is advanced parallel to the superior petrosal sinus. The tro- dal trunks. (e) Compression by a pontine branch of the basi-
chlear nerve is at the superior margin of the exposure and the lar artery. (f) Compression by the AICA. (g) Compression
facial and vestibulocochlear nerves are at the lower margin. by a tortuous basilar artery (From Rhoton [29])
Upper Neurovascular Complex 29

concluding that there is no arterial loop in the caudal surface. Infrequently, the PICA may reach
axilla of the nerve. It is important to remember and groove the undersurface of the trigeminal
that the trunks do not pass directly from the side nerve. The trigeminal nerve also may be com-
of the brain stem to the superior surface of the cer- pressed by a large pontine branch of the basilar
ebellum, but, rather, that they dip into the deep artery (Figs. 2.15 and 2.16). Normally, these
fissure between the cerebellum and midbrain at pontine branches pass around and penetrate the
the posterior margin of the trigeminal nerve. The pons before reaching the trigeminal nerve. A large
SCA gives off perforating arteries that may limit pontine artery, however, may indent the medial
the degree of repositioning of the artery achiev- surface of the trigeminal nerve and then course
able in a microvascular decompression operation. rostral or caudal to the nerve to supply the pons
The most common site of compression of the behind the nerve.
trigeminal nerve on the SCA is at the junction of
the main trunk with the origin of the rostral and
caudal trunks [29]. However, other sites of com- Venous Relationships
pression are seen depending on how far distal the
artery bifurcates in relation to the trigeminal Compression and distortion of the trigeminal
nerve. If the SCA bifurcates near the basilar nerve by the surrounding veins, although less fre-
artery or if there is a duplicate configuration in quent than arterial compression, also is found in
which the rostral and caudal trunks arise directly trigeminal neuralgia (Figs. 2.17 and 2.18) [24]. It
from the basilar artery, both trunks may loop is the superior petrosal veins, which empty into
down into the axilla and compress the nerve. the superior petrosal sinus, that are most fre-
Alternatively, if the artery bifurcates before quently encountered in operative approaches to
reaching the nerve, the caudal trunk may com- the trigeminal nerve and that most commonly
press the nerve and the rostral trunk may course compress the trigeminal nerve.
well above the nerve. If the artery bifurcates dis- The superior petrosal veins are among the
tal to the nerve, only the main trunk will be largest and most frequently encountered veins in
involved in the compression. The point of bifur- the posterior fossa. The superior petrosal veins
cation of the SCA does affect the caliber of the may be formed by the terminal segment of a sin-
vessel that makes contact with the nerve. The gle vein or by the common stem formed by the
contacting vessel will be of a smaller caliber if union of several veins. The most common tribu-
the SCA bifurcates before reaching the trigemi- taries of the superior petrosal veins are the trans-
nal nerve. verse pontine and pontotrigeminal veins, the
A less frequent source of compression of the veins of the cerebellopontine fissure and the mid-
trigeminal nerve is by the AICA (Figs. 2.15 and dle cerebellar peduncle, and the common stem of
2.16). Normally, the AICA passes around the the veins draining the lateral part of the cerebellar
pons below the trigeminal nerve with the facial hemisphere. The transverse pontine veins, which
and vestibulocochlear nerves. The AICA, how- pass near the trigeminal nerve to reach the bridg-
ever, may have a high origin and loop upward to ing veins entering the superior petrosal sinus, are
indent the medial or lower surface of the trigemi- the most frequent veins to compress the trigemi-
nal nerve prior to passing downward to course nal nerve. They may course medially in the axilla
with the facial and vestibulocochlear nerves. of the nerve or they also may pass above, below,
A serpentine basilar artery also may wander lat- or lateral to the nerve and may indent any of its
erally and compress the medial side of the trigem- surfaces. The vein of the middle cerebellar
inal nerve [48]. This type of basilar artery often is peduncle may compress the lateral or medial sur-
elongated and has a fusiform configuration. More face of the trigeminal nerve before joining the
than one artery may compress the nerve. In a few petrosal veins as it ascends in the pons. The vein
cases, the SCA will compress the rostral surface of the cerebellopontine fissure may indent the lat-
of the nerve and the AICA will compress the eral margin of the trigeminal nerve as it ascends
30 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

b d

f e

Fig. 2.17 Sites of venous compression of the trigeminal above the trigeminal nerve. (b) A transverse pontine vein
nerve. (a) Central diagram. Anterior view. The veins that compresses the lateral side of the nerve and joins the veins
commonly compress the trigeminal nerve are tributaries of the middle cerebellar peduncle and cerebellopontine
of the superior petrosal vein. The tributaries that converge fissure to empty into a superior petrosal vein. (c) The
on and may compress the nerve are the transverse pontine medial side of the nerve is compressed by a tortuous trans-
and pontotrigeminal veins and the veins of the cerebello- verse pontine vein. (d) The lateral side of the nerve is
pontine fissure and middle cerebellar peduncle. The trans- compressed by the junction of the transverse pontine vein
verse pontine veins course transversely across the pons. with the veins of the middle cerebellar peduncle and the
The vein of the middle cerebellar peduncle arises in the cerebellopontine fissure. (e) The nerve is compressed on
region of the facial and vestibulocochlear nerves and the medial side by the vein of the middle cerebellar
ascends on the pons. The vein of the cerebellopontine peduncle and on the lateral side by the vein of the cerebel-
fissure arises along the cleft between the pons and the cer- lopontine fissure. (f) The lateral side of the nerve is com-
ebellum and ascends behind the trigeminal nerve. The pressed by the vein of the cerebellopontine fissure (From
pontotrigeminal vein arises on the upper pons and passes Rhoton [29])
Upper Neurovascular Complex 31

d
b

Fig. 2.18 Sites of venous compression of the trigeminal vein passes above the nerve. (b) The trigeminal nerve is
nerve as seen through a retrosigmoid craniotomy. compressed on its medial side by a transverse pontine
(a) The insert shows the site of the scalp incision (solid vein and on its lateral side by the vein of the middle
line) and the craniotomy (interrupted line). The cerebel- cerebellar peduncle. (c) The lateral side of the nerve is
lum has been elevated to expose the junction of the compressed by a transverse pontine vein. (d) The medial
trigeminal nerve with the pons. The superior petrosal side of the nerve is compressed by the junction of a
veins empty into the superior petrosal sinus. The tro- transverse pontine vein with the veins of the middle
chlear nerve is at the superior margin and the facial and cerebellar peduncle and cerebellopontine fissure. (e) The
vestibulocochlear nerves are at the lower margin of the lateral side of the nerve is compressed by the junction of
exposure. The craniotomy exposes the junction of the the transverse pontine vein with the veins of the middle
sigmoid and transverse sinuses. The trigeminal nerve is cerebellar peduncle and cerebellopontine fissure. (f) The
compressed by the junction of a transverse pontine vein medial side of the nerve is compressed by the vein of
and the vein of the middle cerebellar peduncle with the the middle cerebellar peduncle. (g) The lateral side of the
superior petrosal vein. The vein of the cerebellopontine nerve is compressed by the vein of the cerebellopontine
fissure ascends behind the nerve, and the pontotrigeminal fissure (From Rhoton [29])
32 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

toward the superior petrosal sinus, and the The neural structures that surround and limit
pontotrigeminal vein may indent the upper mar- access to the suprameatal tubercle are the cer-
gin of the nerve. ebellum posteriorly, the facial and vestibuloco-
The junction of these veins, which converge chlear nerves below, the trigeminal nerve above
and form a single trunk prior to entering the supe- and medial, and the abducens nerve medially.
rior petrosal sinus, usually is lateral to the trigem- The cerebellopontine cistern opens through the
inal nerve. This junction, however, may be located porus into Meckel’s cave. Meckel’s segment of
medial to the trigeminal nerve, in which case the the trigeminal nerve, which begins at the porus
common trunk must pass around the trigeminal and extends to the trigeminal ganglion, is dif-
nerve prior to reaching the superior petrosal ferentiated from the cavernous segment in the
sinus. These common trunks also may compress wall of the cavernous sinus. Meckel’s segment
the trigeminal nerve. is narrower adjacent to the porus and fans out
as it approaches the posterior edge of the gas-
serian ganglion, which is embedded in the dura
Suprameatal Extension just anterior to the anterior edge of Meckel’s
of the Retrosigmoid Approach cave.
The intraosseous structures, which limit the
The part of the posterior surface of the temporal extent of the drilling if they are to be preserved,
bone that forms the superior lip of the porus of are the posterior part of the superior semicircular
the internal acoustic meatus is the site of a promi- canal, the upper part of the posterior semicircular
nence, the suprameatal tubercle, which blocks canal, and the common crus of the two canals.
access to the lateral margin of the trigeminal After removal of the suprameatal tubercle, the
nerve and the prepontine cistern medial to the drilling can be extended below Meckel’s cave to
trigeminal nerve (Fig. 2.19). Removal of the supra- the edge of the petroclival fissure just in front of
meatal tubercle increases access to the region of the inferior petrosal sinus and immediately lateral
the upper neurovascular complex around the to the abducens nerve.
trigeminal nerve and may possibly avoid the need Removing the bone in this area provides
for a supratentorial craniotomy in exposing access, on average, to the posterior 10.3 mm
tumors that are located predominantly in the cer- (range, 8.0–13.0 mm) of Meckel’s cave and the
ebellopontine angle but also extend into the pos- enclosed portion of the trigeminal nerve and
terior part of the middle fossa in the region of opens a 180° window around the lateral and lower
Meckel’s cave [47]. The tubercle, the most prom- surface of the posterior trigeminal root, which
inent bony elevation around the circumference of may be used for accessing the posterior part of
the internal acoustic meatus, is defined below by the middle fossa (Fig. 2.19).
the internal acoustic meatus, above by the petrous The size of the area created by removing the
ridge, laterally by a vertical line crossing the pos- suprameatal tubercle and adjacent part of the
terior edge of the porus of the internal acoustic petrous apex by the retrosigmoid route is lim-
meatus, and medially by a vertical line crossing ited superiorly by the superior petrosal sinus
the medial edge of the trigeminal notch, a depres- and the dura covering the upper surface of the
sion in the petrous ridge located below the porus petrous bone. The superior petrosal sinus can be
of Meckel’s cave. Above and medial to the divided, and Meckel’s cave and the tentorium
suprameatal tubercle, the posteromedial part of lateral to the porus of Meckel’s cave opened to
the floor of the middle cranial fossa is the site of the provide intradural access to the posteromedial
depression underlying Meckel’s cave in which part of the middle fossa, but cannot be extended
the posterior trigeminal root sits. The most prom- forward to the horizontal portion of the petrous
inent posterior projection of the tubercle is carotid.
located above the lateral half of the porus of the The suprameatal extension of the retrosig-
internal acoustic meatus. moid approach may permit removal of some
Upper Neurovascular Complex 33

a b

c d

Fig. 2.19 Suprameatal variant of the retrosigmoid approach. lateral to the trigeminal nerve. (c) The dura over the
(a) The cerebellum has been elevated to expose the nerves in suprameatal tubercle has been removed in preparation for
the cerebellopontine angle. A large petrosal vein blocks drilling. (d) Removing the suprameatal bone, including the
access to the suprameatal area. (b) The superior petrosal tubercle, extends the exposure along the posterior trigeminal
vein has been divided to expose the suprameatal tubercle root by approximately 1 cm and increases access to the front
located above the porus of the internal acoustic meatus and of the brain stem and clivus (From Rhoton [40])
34 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

tumors that are located mainly in the posterior Middle Neurovascular Complex
fossa but that extend into the middle fossa in the
region of Meckel’s cave. The space created after The middle complex includes the AICA, pons,
drilling the suprameatal tubercle and the bone middle cerebellar peduncle, cerebellopontine
medial to the internal auditory canal and below fissure, petrosal surface of the cerebellum, and the
the trigeminal nerve was enough to extend the abducens, facial, and vestibulocochlear nerves
retrosigmoid approach as far as 13.0 mm (aver- (Figs. 2.1, 2.2, 2.3, 2.5, 2.7, 2.20, and 2.21). The
age, 10.3 mm) anterior to what could be achieved AICA arises at the pontine level and courses in
by using the retrosigmoid approach alone [47]. relationship to the abducens, facial, and vestibulo-
The extent to which the bone in the region of the cochlear nerves to reach the surface of the middle
suprameatal tubercle could be removed using the cerebellar peduncle, where it courses along the cer-
retrosigmoid approach is defined and limited by ebellopontine fissure and terminates by supplying
the neural and bony structures in the region. The the petrosal surface of the cerebellum. Operations
cerebellum, with gentle retraction, limits the directed to the middle complex are for the removal
angle at which the suprameatal tubercle can be of acoustic neuromas and other tumors and for the
drilled, although when combined with evacua- relief of hemifacial spasm. The considerations
tion of cerebrospinal fluid, it provides a space to related to acoustic neuromas will be dealt with
adequately visualize and remove lesions medial first.
to the suprameatal tubercle. A portion of this
bone is commonly removed in approaching
tumors extending into the internal acoustic Anatomy of Acoustic Neuromas
meatus. It is possible to remove approximately
270° of the circumference of the wall of the inter- Acoustic neuromas, as they expand, may involve
nal acoustic meatus when using the retrosigmoid a majority of the cranial nerves, cerebellar
approach; however, with the approach described arteries, and parts of the brain stem. On the lat-
herein, only the bone in the region of the eral side, in the meatus, they commonly expand
suprameatal tubercle is removed. The drilling on by enlarging the meatus but infrequently erode
the lateral side of the tubercle should avoid the into the vestibule and cochlea. On the medial
posterior semicircular canal and common crus of side, they compress the pons, medulla, and cere-
the posterior and superior canals if hearing is to bellum. An understanding of microsurgical anat-
be preserved, but on the medial side, it can extend omy is especially important in preserving the
through the petrous apex into the side of the facial and adjacent cranial nerves, which are the
clivus. neural structures at greatest risk during acoustic
Often, the subarcuate branch of the AICA neuroma removal. A widely accepted operative
must be obliterated to access the suprameatal precept is that a nerve involved by a tumor should
tubercle. Accessing the suprameatal tubercle be identified proximal or distal to the tumor,
often requires that the superior petrosal veins be where its displacement and distortion is the least,
obliterated and divided. This allows the drilling before the tumor is removed from the involved
to be directed medially along the lateral and lower segment of the nerve. Considerable attention has
margin of the porus of Meckel’s cave. The dura been directed to the early identification of the
along the lower and lateral margin of the porus of facial nerve distal to the tumor at the lateral part
Meckel’s cave and the tentorium lateral to the of the internal acoustic canal. Less attention has
porus trigeminus can then be opened to expose been directed to identification at the brain stem
the trigeminal nerve in the posterior part of on the medial side of the tumor. These anatomic
Meckel’s cave and the middle cranial fossa. Care considerations are divided into sections dealing
is taken to protect the trochlear nerve if the tento- with the relationships at the lateral end of the
rial incision is to be extended through the free tumor in the meatus and those on the medial end
edge. of the tumor at the brain stem [31, 37, 45].
Middle Neurovascular Complex 35

a b

c d

Fig. 2.20 Left retrosigmoid exposure. (a) The cerebel- foramen of Luschka into the cerebellopontine angle
lum has been elevated. A large AICA loops into the porus behind the glossopharyngeal and vagus nerves. A nerve
of the internal meatus. The junction of the facial nerve hook has been placed inside the rhomboid lip, a pouch of
with the brain stem is located below and slightly in front neural tissue attached along the anterior margin of the lat-
of the vestibulocochlear nerve. (b) The vestibulocochlear eral recess and extending laterally behind the glossopha-
nerve has been elevated to provide additional exposure of ryngeal and vagus nerves. (d) Enlarged view of the
the facial nerve. (c) Choroid plexus protrudes from the rhomboid lip (From Rhoton [40])
36 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

c d

e f

Fig. 2.21 Left cerebellopontine angle. (a) The AICA prominent. (d) The dura lining the internal acoustic
passes between the facial and vestibulocochlear nerves. A meatus has been opened. The transverse crest separates
dural septum separates the glossopharyngeal and vagus the superior vestibular and facial nerves above from the
nerves at the jugular foramen. (b) The vestibulocochlear inferior vestibular and cochlear nerves below. (e) Enlarged
nerve and the flocculus have been elevated to expose the view of the nerves within the meatus. The cochlear nerve
junction of the facial nerve with the brain stem. In the ret- is partially hidden anterior to the inferior vestibular nerve.
rosigmoid approach, the facial nerve junction with the (f) The cleavage plane between the superior and inferior
brain stem can be exposed below the vestibulocochlear vestibular and cochlear nerves has been started laterally
nerve. (c) The posterior wall of the internal acoustic and extended medially to expose the individual nerve
meatus has been removed. The cleavage plane between bundles (From Rhoton [40])
the superior and inferior vestibular nerves is especially

Meatal Relationships 2.21, and 2.22) [28, 32, 35, 36]. The position of
the nerves is most constant in the lateral portion
The nerves in the lateral part of the internal acous- of the meatus, which is divided into a superior and
tic meatus are the facial, cochlear, and inferior an inferior portion by a horizontal ridge, called
and superior vestibular nerves (Figs. 2.2, 2.3, either the transverse or falciform crest. The facial
Middle Neurovascular Complex 37

Superior vestibular nerve


a Superior vestibular nerve b Facial nerve
Facial nerve anterior to tumor

Transverse Transverse
crest crest

Cochlear nerve Cochlear nerve


Inferior vestibular nerve Inferior vestibular nerve

c Facial nerve
d

Facial nerve

Fig. 2.22 View of right internal acoustic meatus with the meatus. (b) The facial nerve is displaced directly anteri-
posterior lip removed to show variable direction of facial orly. This is a frequent direction of displacement with
nerve displacement by acoustic neuroma. (a) Normal neu- acoustic neuroma. (c) Another frequent direction of dis-
ral relationships with the eighth nerve dividing into its placement of the facial nerve is anterior and superior. (d)
three parts in the lateral meatus. The facial and superior The facial nerve is displaced anteriorly and inferiorly by
vestibular nerves are above the transverse crest and the tumor, which erodes the superior wall of the meatus above
cochlear and inferior vestibular nerves are below. The facial the nerves and grows into the area above the nerves, dis-
nerve occupies the anterosuperior quadrant of the lateral placing them inferiorly (From Rhoton [35])

and the superior vestibular nerves are superior to Retrosigmoid Approach


the crest. The facial nerve is anterior to the supe-
rior vestibular nerve and is separated from it at the The retrosigmoid approach to the meatus is
lateral end of the meatus by a vertical ridge of directed through a vertical scalp incision that
bone, called the vertical crest. The vertical crest is crosses the asterion. A burr hole is placed below
also called “Bill’s bar” in recognition of William the asterion, and a craniotomy is performed expos-
House’s role in focusing on the importance of this ing the lower margin of the transverse sinus supe-
crest in identifying the facial nerve in the lateral riorly, the posterior margin of the sigmoid sinus
end of the canal [14]. The cochlear and inferior laterally, and the inferior portion of the squamous
vestibular nerves run below the transverse crest part of the occipital bone inferiorly. The intradural
with the cochlear nerve located anteriorly. Thus, exposure is directed down the plane between the
the lateral meatus can be considered to be divided posterior face of the temporal bone and the petro-
into four portions, with the facial nerve being sal cerebellar surface (Figs. 2.2, 2.3, 2.20, and
anterosuperior, the cochlear nerve anteroinferior, 2.21) [30, 31, 45]. The petrosal cerebellar surface
the superior vestibular nerve posterosuperior, and usually relaxes away from the temporal bone after
the inferior vestibular nerve posteroinferior. opening the arachnoid membrane over the cisterna
38 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

magna and allowing cerebrospinal fluid to escape. artery and the segment of the AICA from which it
When removing the posterior meatal wall, it often arises will be incorporated into the dura covering
is necessary to sacrifice the subarcuate artery the subarcuate fossa [49, 50]. In this case, the dura
because it passes through the dura on the posterior and the artery will have to be separated together
meatal wall to reach the subarcuate fossa from the posterior meatal lip wall in preparation
(Figs. 2.2, 2.3, and 2.21) [22]. This artery usually for opening the meatus (Fig. 2.23). In some of the
has a sufficiently long stem that its obliteration cases, the artery will be surrounded by bone that
does not risk damage to the AICA from which it has to be drilled to free the artery and access the
arises. In a few cases, however, the subarcuate meatus.

a Subarc.A CN VII b

CN VIII

Labyr.A

AICA

c d

Fig. 2.23 Artist’s renderings showing posterior view of fossa. (d) Dura over the subarcuate fossa has been incised,
the left IAM. (a) Subarcuate artery penetrates the dura of and the dura with the adherent loop is dissected free from
the subarcuate fossa near the IAM. The labyrinthine artery the subarcuate fossa in preparation for opening the IAM.
enters the meatus with the vestibulocochlear and the facial (e) Dura over the subarcuate fossa has been incised and
nerves. (b) Laterally convex loop of the AICA is embed- remains attached to the artery. The bone surrounding the
ded in the dura covering the subarcuate fossa, where it embedded AICA loop is removed with a 2-mm diamond
gives off the subarcuate artery. (c) AICA loop is embedded drill to displace the artery medially for exposure of the
in the dura and bone (arrow) surrounding the subarcuate IAM (From Tanriover and Rhoton [50])
Middle Neurovascular Complex 39

The posterior semicircular canal and its com- Neural Relationships


mon crus with the superior canal, both of which The landmarks on the medial or brain stem side
are situated just lateral to the posterior meatal lip, of structures that are helpful in guiding the sur-
should be preserved when removing the posterior geon to the junction of the facial nerve with the
meatal wall if there is the possibility of preserv- brain stem are the pontomedullary sulcus; the
ing hearing, since hearing may be lost if damage junction of the glossopharyngeal, vagus, and spi-
occurs (Fig. 2.24). Care also is required to avoid nal accessory nerves with the medulla; the fora-
injury to the vestibular aqueduct, which is situ- men of Luschka and its choroid plexus; and the
ated inferolateral to the meatal lip, and the endo- flocculus. The facial nerve arises from the brain
lymphatic sac, which expands under the dura on stem near the lateral end of the pontomedullary
the posterior surface of the temporal bone infero- sulcus 1–2 mm anterior to the point at which the
lateral to the meatal porus (Fig. 2.24). The vestibulocochlear nerve joins the brain stem at
endolymphatic sac may be entered in removing the lateral end of the sulcus. The interval between
the dura from the posterior meatal lip. There is the vestibulocochlear and facial nerves is greatest
little danger of encountering the cochlear canali- at the level of the pontomedullary sulcus and
culus, which has a more medial course below the decreases as these nerves approach the meatus.
internal auditory canal. An unusually high pro- The facial nerve enjoys a consistent relation-
jection of the jugular bulb into the posterior wall ship to the junction of the glossopharyngeal,
of the meatus presents an anomaly that may block vagus, and spinal accessory nerves with the
access to the posterior meatal lip. Mastoid air medulla (Figs. 2.25, 2.26, and 2.27). The facial
cells commonly are encountered in the posterior nerve arises 2–3 mm above the most rostral root-
meatal lip. let contributing to these nerves. A helpful way of
After removing the posterior wall of the visualizing the point where the facial nerve will
meatus, the dura lining the meatus is opened to exit from the brain stem, even when displaced by
expose its contents (Figs. 2.3 and 2.21). The a tumor, is to project an imaginary line along the
facial nerve is identified near the origin of medullary junction of the rootlets forming the
the facial canal at the anterosuperior quadrant of glossopharyngeal, vagus, and spinal accessory
the meatus rather than in a more medial location nerves upward through the pontomedullary junc-
where the direction of displacement is variable. If tion. This line, at a point 2–3 mm above the junc-
the tumor extends into the vestibule, the latter can tion of the glossopharyngeal nerve with the
easily be exposed by drilling removing the poste- medulla, will pass through the pontomedullary
rior wall of the vestibule lateral to the meatal fun- junction at the site where the facial nerve exits
dus. The strokes of the fine dissecting instruments from the brain stem. The filaments of the nervus
along the vestibulocochlear nerve should be intermedius also are stretched around an acoustic
directed from medial to lateral rather than from neuroma.
lateral to medial, because traction medially may The structures related to the lateral recess of
tear the tiny filaments of the cochlear nerve at the the fourth ventricle that have a consistent rela-
site where these filaments penetrate the lateral tionship to the facial and vestibulocochlear nerves
end of the meatus to enter the modiolus of the are the foramen of Luschka and its choroid plexus
cochlea (Fig. 2.24). and the flocculus (Figs. 2.25, 2.26, and 2.27)
[8, 23]. The foramen of Luschka is situated at the
lateral margin of the pontomedullary sulcus, just
Brain Stem Relationships behind the junction of the glossopharyngeal
nerve with the brain stem, and immediately pos-
A consistent set of neural, arterial, and venous teroinferior to the junction of the facial and ves-
relationships at the brain stem facilitates the tibulocochlear nerves with the brain stem. The
identification of the nerves on the medial side of foramen of Luschka is infrequently well visual-
an acoustic neuroma (Figs. 2.25, 2.26, and 2.27) ized. A consistently identifiable tuft of choroid
[31, 32, 37]. plexus, however, hangs out of the foramen of
40 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

c d

e f
Middle Neurovascular Complex 41

Fig. 2.24 (a) Posterior surface of a right temporal bone. vestibule during drilling of the posterior meatal wall may
The internal meatus is located near the center and the jugu- result in a loss of hearing. (d) Enlarged view of the fundus
lar foramen at the lower edge of the posterior surface. The of the meatus after removal of the posterior wall. The upper
sigmoid sinus descends along the posterior surface of the mas- edge of the porus has been preserved. The facial nerve and
toid part of the temporal bone and turns forward on the nervus intermedius are exposed medial to the porus of the
occipital bone to pass through the sigmoid part of the jugu- meatus. The subarcuate artery enters the subarcuate fossa.
lar foramen. The inferior petrosal sinus descends along the The inferior vestibular nerve gives rise to the singular
petroclival fissure and passes through the petrosal part of branch to the posterior ampullae, plus utricular and saccular
the jugular foramen. The glossopharyngeal, vagus, and branches. The superior vestibular nerve innervates the
accessory nerves pass through the intrajugular part of the ampullae of the superior and lateral semicircular canals and
foramen between the sigmoid and petrosal part. The subar- commonly gives rise to a utricular branch. Care is taken to
cuate fossa is located superolateral and the ostium for the preserve the superior and posterior canals and the common
endolymphatic duct is positioned lateral to the internal crus, plus the endolymphatic sac in those cases in which
acoustic meatus. The trigeminal impression is a shallow there is the opportunity to preserve hearing when drilling
trough on the upper surface of the petrous part behind the the posterior wall of the meatus. (e) The petrous apex medial
foramen ovale. The arcuate eminence overlies the superior to the internal acoustic meatus has been partially removed
semicircular canal. (b) View of the fundus of the right inter- to expose the petrous segment of the internal carotid artery.
nal acoustic meatus. The transverse crest divides the meatal The lateral genu of the petrous carotid, located at the junc-
fundus into superior and inferior parts. Above the transverse tion of the vertical and horizontal segments, is situated
crest, the facial canal is anterior and the superior vestibular below and medial to the cochlea. The jugular bulb extends
area is posterior. Below the transverse crest, the cochlear upward, adjacent the posterior meatal wall, toward the ves-
area is anterior and the inferior vestibular area is posterior. tibule and semicircular canals. The inferior petrosal sinus
The singular foramen, through which the singular branch of courses along the petroclival fissure and enters the petrosal
the inferior vestibular nerve passes to innervate the posterior part of the jugular foramen. The sigmoid sinus descends in
canal ampullae, is located posterior to the inferior vestibular the sigmoid sulcus and enters the sigmoid part of the fora-
area. The inferior vestibular nerve also has a saccular and, men. The glossopharyngeal, vagus, and accessory nerves
occasionally, an utricular branch. The cochlear nerve splits pass through the central or intrajugular part of the jugular
into tiny filaments as its fibers pass through the cochlear foramen located between the sigmoid and petrosal parts. (f)
area. These filaments are easily torn, with a resulting loss of Bone has been removed along the anterior margin of the
hearing, with medially directed retraction of the cerebellum meatal fundus to open the cochlea and along the posterior
and nerve. (c) The facial and vestibulocochlear nerves enter margin to expose the vestibule. The cochlear nerve pene-
the internal acoustic meatus, and the glossopharyngeal, trates the modiolus of the cochlea where its fibers are dis-
vagus, and accessory nerves enter the jugular foramen. The tributed to the turns of the cochlear duct. The basal turn of
trigeminal nerve passes above the petrous apex and through the cochlea communicates below the modiolus with the ves-
the porus of Meckel’s cave. The posterior and superior tibule. The stapes has been removed from the oval window.
semicircular canals have been exposed. The superior semi- The promontory in the medial wall of the tympanic cavity is
circular canal is positioned below the medial edge of the located lateral to the basal turn of the cochlea. A silver fiber
arcuate eminence. The upper end of the posterior canal and has been introduced into the superior semicircular canal, a
the posterior end of the superior canal join to form a com- red fiber into the lateral canal, and a blue fiber into the pos-
mon channel, the common crus, which opens into the vesti- terior canal. The ampullated ends of the canals are located
bule. The endolymphatic duct extends downward from the at the bulbous ends of the three fibers. The common crus of
vestibule and opens into the endolymphatic sac located the superior and posterior canals is located where the tips of
beneath the dura inferolateral to the internal acoustic the blue and silver fibers cross. The superior vestibular
meatus. The endolymphatic ridge, the bridge of bone form- nerve passes to the ampullae of the superior and lateral
ing the upper lip of the endolymphatic duct, has been pre- canals. The singular branch of the inferior vestibular nerve
served. The jugular bulb can be seen through the thin bone innervates the posterior ampullae. A small black fiber has
below the internal meatus. Entering the posterior canal, been introduced into the opening of the endolymphatic duct
common crus, posterior portion of the superior canal, or the into the vestibule (From Rhoton [33])
42 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

Fig. 2.25 Neurovascular relationships on the brain stem tomedullary sulcus, cerebellomedullary fissure, middle
side of an acoustic neuroma. Anterolateral view of the cerebellar peduncle, and the retro-olivary and lateral med-
right cerebellopontine angle. (a) Neural relationships. ullary veins. The vein of the cerebellopontine fissure,
The facial and vestibulocochlear nerves arise from the which passes above the flocculus on the middle cerebellar
brain stem near the lateral end of the pontomedullary sul- peduncle, is formed by the anterior hemispheric veins that
cus, anterosuperior to the choroid plexus protruding from arise on the cerebellum. Transverse pontine and transverse
the foramen of Luschka, anterior to the flocculus, rostral medullary veins cross the pons and medulla. The median
to a line drawn along the junction of the rootlets of the anterior medullary and median anterior pontomesenceph-
glossopharyngeal, vagus, and accessory nerves with alic veins ascend on the anterior surface of the medulla
the brain stem, and slightly posterior to the rostral pole of and pons. The veins of the middle cerebellar peduncle and
the inferior olive. The cerebellopontine fissure formed by the cerebellopontine fissure and a transverse pontine vein
the cerebellum wrapping around the lateral side of the join to form a superior petrosal vein, which empties into
pons and middle cerebellar peduncle has a superior limb the superior petrosal sinus. A bridging vein passes below
that passes above the trigeminal nerve and an inferior limb the vagal rootlets toward the jugular foramen. (d)
that extends below the foramen of Luschka. The cerebel- Neurovascular relationships of an acoustic neuroma. The
lomedullary fissure, which extends superiorly between the tumor arises from the vestibulocochlear nerve and dis-
medulla and cerebellum, communicates in the region of places the facial nerve anteriorly, the trigeminal nerve
the foramen of Luschka with the cerebellopontine fissure. superiorly, and the vagus and glossopharyngeal nerves
(b) Arterial relationships. The AICA arises from the basi- inferiorly. The facial nerve, even though displaced by the
lar artery and divides into rostral and caudal trunks. The tumor, enters the brain stem along the lateral margin of the
rostral trunk, which is usually the larger of the two trunks, pontomedullary sulcus, rostral to the glossopharyngeal
courses below the facial and vestibulocochlear nerves and and vagus nerves, anterior to the flocculus, and rostral to
then above the flocculus to reach the surface of the middle the choroid plexus protruding from the foramen of
cerebellar peduncle. The PICA arises from the vertebral Luschka. The rostral trunk of the AICA, after passing
artery and passes first between the hypoglossal rootlets below the tumor, returns to the surface of the middle cer-
and then between the vagus and accessory nerves on its ebellar peduncle above the flocculus. The veins displaced
way to the cerebellar hemisphere. The SCA passes above around the medial side of the tumor are the veins of the
the trigeminal nerve. The cerebellar arteries give rise to middle cerebellar peduncle, cerebellomedullary fissure,
hemispheric branches. (c) Venous relationships. The veins cerebellopontine fissure and pontomedullary sulcus, and
that converge on the junction of the facial and vestibuloco- the retro-olivary and lateral medullary veins (From
chlear nerves with the brain stem are the veins of the pon- Rhoton [37])
Middle Neurovascular Complex 43

c d

Fig. 2. 25 (continued)

Luschka and sits on the posterior surface of the middle fossa, translabyrinthine, or posterior
glossopharyngeal and vagus nerves just inferior approaches, care is required to avoid injury to
to the junction of the facial and vestibulocochlear the AICA if it is located at or protrudes through
nerves with the brain stem. Another structure the porus.
related to the lateral recess, the flocculus, proj- In most cases, the AICA passes below the
ects from the margin of the lateral recess and facial and vestibulocochlear nerves as it encircles
foramen of Luschka into the cerebellopontine the brain stem, but it also may pass above or
angle, just posterior to where the facial and ves- between these nerves in its course around the
tibulocochlear nerves join the pontomedullary brain stem (Fig. 2.28). In the most common case,
sulcus. in which the artery passes below the nerves, the
tumor would displace the artery inferiorly. If the
Arterial Relationships artery courses between the facial and vestibulo-
The arteries crossing the cerebellopontine angle, cochlear nerves, a tumor arising in the latter nerve
especially the AICA, enjoy a consistent relation- will displace the artery forward. Tumor growth
ship to the facial and vestibulocochlear nerves, would displace the artery superiorly if it passes
the foramen of Luschka, and the flocculus as above the nerves. Atkinson pointed out that those
described elsewhere in this volume [11, 12, 21, cases of acoustic neuroma appearing at necropsy
22]. In a prior study, the author’s group found after operation frequently had occlusion of the
that the AICA formed a loop that reached the AICA [1]. In three cases presented by Atkinson,
porus or protruded into the canal in 54 % of an arterial branch coursing over the tumor cap-
the cases [22]. When opening the meatus by the sule was ligated with resulting lateral pontine,
44 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

Fig. 2.26 Relationship of the foramen of Luschka and forms the caudal part of the roof of the fourth ventricle
the lateral recess of the fourth ventricle to the junction of and has the choroid plexus attached to its inner surface.
the facial and vestibulocochlear nerves with the brain- The facial and vestibulocochlear nerves enter the brain-
stem, as seen through a suboccipital craniotomy. (a) The stem at the lateral end of the pontomedullary sulcus.
orientation, skin incision (solid line), and craniotomy (c) The tela choroidea has been opened, but the choroid
(interrupted line) are shown in the insert. The foramen of plexus, which arises on the inner surface of the tela in the
Luschka opens into the cerebellopontine angle behind the fourth ventricle, has been preserved. The fringelike chor-
glossopharyngeal and vagus nerves. The choroid plexus oid plexus extends through the foramen of Luschka
protrudes from the foramen of Luschka, slightly below slightly below and behind the junction of the facial and
and behind the facial and vestibulocochlear nerves, and vestibulocochlear nerves with the brainstem. The inferior
behind to the glossopharyngeal and vagus nerves. The cerebellar peduncle ascends on the dorsolateral margin of
flocculus protrudes into the cerebellopontine angle above the medulla. (d) Relationships of an acoustic neuroma.
the foramen of Luschka. The accessory nerve arises below The facial nerve is displaced anteriorly and superiorly in
the vagus nerve. The hypoglossal rootlets arise ventral the cerebellopontine angle and enters the brainstem at the
to the olive. The trigeminal nerve crosses in the upper part lateral end of the pontomedullary sulcus, anterosuperior
of the exposure. (b) The right cerebellar tonsil has been to the choroid plexus protruding from the foramen of
removed by dividing the tonsillar peduncle to show the Luschka, and near where the flocculus is attached along
relationship of the lateral recess to the facial and vestibu- the margin of the lateral recess. The tumor displaces the
locochlear nerves. The flocculus and choroid plexus pro- trigeminal nerve upward and the glossopharyngeal and
trude in the cerebellopontine angle behind the junction of vagus nerves downward. The AICA gives rise to a subar-
the facial and vestibulocochlear nerves with the brain- cuate artery, which enters the subarcuate fossa in the pos-
stem. The inferior medullary velum stretches from the terior wall of the internal acoustic meatus and bifurcates
lateral side of the vermis to the flocculus and is all that into a rostral and a caudal trunk. The rostral trunk courses
remains of the connection between the flocculus and the above the flocculus to reach the surface of the middle cer-
nodulus, which form the flocculonodular lobe of the cer- ebellar peduncle. [37] (From, Rhoton AL Jr: Microsurgical
ebellum. The inferior medullary velum stretches laterally anatomy of the brainstem surface facing an acoustic neu-
to form the peduncle of the flocculus. The tela choroidea roma. Surg Neurol 25:326–339, 1986.)
Middle Neurovascular Complex 45

c d

Fig. 2. 26 (continued)

tegmental, and medullary infarction in the area junction of the facial and vestibulocochlear
supplied by the AICA and death. He noted that nerves with the brain stem. These veins, which
the blood pressure rose at or near the time of cross the cerebellopontine angle to reach the
occlusion of the artery, although the hypertension superior petrosal sinus, are the ones most
often subsided by the end of the operation. These frequently occluded in the course of operations
tumors may also displace the PICA and insinuate in the cerebellopontine angle. Bridging veins
themselves between the basilar artery and the are exposed more frequently and sacrificed in
pons, stretching the perforating branches of the the rostral part of the cerebellopontine angle
basilar artery. The labyrinthine, recurrent perfo- during operations near the trigeminal nerve
rating, and subarcuate branches arise from the than during operations near the nerves entering
AICA near the facial and vestibulocochlear the internal acoustic meatus. The exposure of
nerves and are frequently stretched around a cer- an acoustic neuroma in the central part of the
ebellopontine angle tumor. cerebellopontine angle near the lateral recess
usually can be completed without sacrificing a
Venous Relationships bridging vein. If a vein is obliterated during
The veins on the side of the brain stem that have acoustic tumor removal, it is usually one of the
a predictable relationship to the facial and superior petrosal veins, which is sacrificed near
vestibulocochlear nerves are the vein of the the superior pole of the tumor during the later
pontomedullary sulcus, the veins of the cerebel- stages of the removal of a large tumor. Small
lomedullary fissure, middle cerebellar pedun- acoustic neuromas usually are removed without
cle, and cerebellopontine fissure (Figs. 2.25, sacrificing a petrosal vein. The largest vein
2.26, and 2.27) [37, 40–42]. The identification encountered around the superior pole of an
of any of these veins during the removal of the acoustic neuroma is the vein of the cerebello-
tumor makes it easier to identify the site of the pontine fissure.
46 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

Summary: Anatomy of Acoustic is stretched around the anterior half of the tumor
Neuromas capsule. Variability in the direction of the growth
of the tumor arising from the vestibular nerves
Because acoustic neuromas most frequently arise may result in the facial nerve being displaced, not
in the posteriorly placed vestibular nerves, they only directly anteriorly but also anterosuperiorly
usually displace the facial and cochlear nerves or anteroinferiorly. The nerve infrequently is
anteriorly (Figs. 2.25 and 2.29). The facial nerve found on the posterior surface of the tumor.

a b

Fig. 2.27 Neurovascular relationships on the brainstem and the transverse pontine and transverse medullary veins
side of an acoustic neuroma. Anterosuperior views. cross the pons and medulla. The vein of the cerebellopontine
(a) Neural relationships. The cerebrum and tentorium cere- fissure passes above the flocculus. The transverse pontine
belli have been removed, and the trigeminal, trochlear, and vein and the veins of the middle cerebellar peduncle and
oculomotor nerves have been divided to allow the brainstem cerebellopontine fissure join to form one of the superior pet-
to be displaced posteriorly to expose the cerebellopontine rosal veins that empty into the superior petrosal sinus. A
angle from above. The facial and vestibulocochlear nerves bridging vein passes from the side of the brainstem to the
arise at the lateral end of the pontomedullary sulcus anterior jugular foramen. The anterolateral marginal vein crosses the
to the flocculus, rostral to the glossopharyngeal, vagus, and anterolateral margin of the cerebellum. The vein of the pon-
accessory nerves, and anterosuperior to the choroid plexus tomesencephalic sulcus courses in the pontomesencephalic
protruding from the foramen of Luschka. The cerebellopon- sulcus below the oculomotor nerve. (d) Neurovascular rela-
tine fissure, formed where the cerebellum wraps around the tionships of an acoustic neuroma. The tumor arises from the
lateral side of the pons and middle cerebellar peduncle, has vestibulocochlear nerve and displaces the facial nerve ante-
superior and inferior limbs. The foramen of Luschka opens riorly, the trigeminal nerve superiorly, and the glossopharyn-
into the inferior limb near the facial and vestibulocochlear geal and vagus nerves inferiorly. The vestibulocochlear
nerves. (b) Arterial relationships. The AICA arises from the nerve disappears into the tumor. The facial nerve enters the
basilar artery, passes below the facial and vestibulocochlear brainstem along the lateral margin of the pontomedullary
nerves, gives rise to the subarcuate artery, and divides into a sulcus, rostral to the glossopharyngeal nerve, anterior to the
rostral and a caudal trunk. The rostral trunk passes above the flocculus, and rostral to the choroid plexus protruding from
flocculus to course on the middle cerebellar peduncle, and the foramen of Luschka. The AICA is usually displaced
the caudal trunk supplies the area below the flocculus. around the lower margin of the tumor. The veins displaced
(c) Venous relationships. The veins converging on the junc- around the medial side of the tumor are the veins of the pon-
tion of the facial nerve with the brainstem are the lateral tomedullary sulcus, middle cerebellar peduncle, and cere-
medullary and retro-olivary veins, and the veins of the pon- bellomedullary fissure, and the lateral medullary and
tomedullary sulcus, cerebellomedullary fissure, and middle retro-olivary veins. [37] (From, Rhoton AL Jr: Microsurgical
cerebellar peduncle. The median anterior pontomesenceph- anatomy of the brainstem surface facing an acoustic neu-
alic vein ascends on the anterior surface of the brainstem, roma. Surg Neurol 25:326–339, 1986.)
Middle Neurovascular Complex 47

c d

Fig. 2. 27 (continued)

Because the facial nerve always enters the facial tumor appearing to be tightly adherent to the neu-
canal at the anterosuperior quadrant of the meatal ral structures is not adhesions between the cap-
fundus, it usually is easiest to locate it here rather sule and surrounding tissue but, rather, the
than at a more medial location where the degree residual tumor within the capsule wedging the
of displacement of the nerve is more variable. tumor into position. As the intracapsular contents
The cochlear nerve also lies anterior to the ves- are removed, the tumor capsule folds laterally,
tibular nerve and is stretched most frequently revealing the structures on the brain stem side of
around the anterior half of the tumor. The strokes the tumor.
of the fine dissecting instruments used in remov- The landmarks that are helpful in identify-
ing the tumor should be directed along the ves- ing the facial and vestibulocochlear nerves at
tibulocochlear nerve from medial to lateral rather the brain stem on the medial side of the tumor
than from lateral to medial, because traction have been reviewed (Figs. 2.25, 2.26, 2.27, and
medially may tear the tiny filaments of the 2.29) [37]. These nerves, although distorted
cochlear nerve at the site where these filaments by the tumor, usually can be identified on the
penetrate the lateral end of the meatus to enter the brain stem side of the tumor at the lateral end
cochlea (Figs. 2.24 and 2.30). of the pontomedullary sulcus just rostral to the
The operation for a cerebellopontine angle glossopharyngeal nerve and just anterosuperior
tumor should be planned so that the tumor sur- to the foramen of Luschka, the flocculus, and
face is allowed to settle away from the neural tis- the choroid plexus protruding from the foramen
sue rather than the neural structures being of Luschka. After the facial and vestibuloco-
retracted away from the tumor (Fig. 2.30). No chlear nerves are identified on the medial and
attempt is made to see the whole tumor upon lateral sides of the tumor, the final remnants
initial exposure. The surface of the tumor then of the tumor are separated from the interven-
is opened, and the intracapsular contents are ing segment of the nerves using fine dissect-
removed. As the intracapsular contents are ing instruments (Fig. 2.30). It is especially
evacuated, the tumor shifts laterally, allowing important to preserve the segment of the cer-
more of the tumor to be removed through the ebellar arteries adherent to the tumor capsule
small exposure. The most common reason for the because a major cause of operative mortality
48 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

S.C.A
Pons Dura
Tent.
Cerebellum
V
Tumor VII
R.P.A VIII S.V.
VIII Co.
Trans.crest
Flocculus
Ch.Pl. VIII I.V.
S.A.
Post.mea.seg.
Pre.mea.seg.
IX

P.I.C.A XI X
S.C.A.
Post.mea.seg.
Mea.seg.
V I.A.A.
VIII S.V.
VII

S.C.A. VIII I.V.


VIII co.
Pre.mea.seg.
R.P.A. IX
R.P.A.
V X
Mea.seg.
Post.mea.seg. P.I.C.A.

VII
VIII S.V. I.A.A.
VIII I.V. VIII Co.
Pre.mea.seg. IX
Medulla X
P.I.C.A.
XI

Fig. 2.28 Posterior views of the direction of displacement vestibular nerves disappear into the tumor; however, the
of the AICA around an acoustic neuroma. Top left: The cochlear and facial nerves are displaced around the ante-
insert shows the direction of view. Both the premeatal and rior margin of the tumor. A subarcuate artery arises from
the postmeatal segments are in their most common loca- the premeatal segment, and a recurrent perforating artery
tions around the lower margin of the tumor. The premeatal arises from the postmeatal segment. Center right: In a less
segment approaches the meatus from anteroinferior, and common pattern of displacement of the AICA, the pre-
the postmeatal segment passes posteroinferior to the meatal and postmeatal segments are above the tumor. The
tumor. The SCA and the trigeminal nerve are above the internal auditory arteries arise from the meatal segment.
tumor, and the PICA and the glossopharyngeal, vagus, and Bottom left: Both the premeatal and the postmeatal seg-
spinal accessory nerves are below the tumor. The choroid ments are displaced anteriorly to the tumor. This occurs if
plexus protrudes into the cerebellopontine angle medial to the AICA courses between the vestibulocochlear and
the tumor. The posterior wall of the internal acoustic canal facial nerves. The tumor arises in the vestibular nerves,
has been removed to expose the transverse crest and the and tumor growth displaces both the premeatal and the
superior vestibular and inferior vestibular nerves. The postmeatal segments anteriorly (From Martin et al. [22])
Anatomy of Vascular Compression in the Middle Neurovascular Complex 49

and morbidity is the loss of perforating arteries tages to being able to separate the tumor cap-
and branches of the cerebellar arteries that may sule off the nerves beginning medially for this
be adherent to and displaced by the tumor. Any more often results in preservation of hearing
vessel that stands above or is stretched around than dissection starting laterally. Compared to
the tumor capsule should be dealt with initially the middle fossa approach, it has the advantage
as if it were an artery that runs over the tumor that the facial nerve is usually deep to the tumor
surface to supply the brain. After the tumor and often is protected by a thin veil of vestibu-
has been removed from within the capsule, an locochlear nerve, thus increasing the opportu-
attempt should be made to displace the vessel nity for facial nerve preservation. In the middle
off the tumor capsule. When dissected free of fossa approach, the facial nerve is often in the
the capsule, vessels that initially appeared to be upper part of the exposure stretched over the
adherent to the capsule often prove to be neural upper half of the tumor and much of the dis-
vessels. The number of veins sacrificed should section is directly on the surface of the nerve,
be kept to a minimum because of the undesir- which increases the risk of facial dysfunction
able consequences of their loss. Obliteration of after surgery.
the petrosal veins, which pass from the surface
of the cerebellum and the brain stem to the
superior petrosal sinus, is inescapable when Anatomy of Vascular Compression
reaching and removing some cerebellopontine in the Middle Neurovascular Complex
angle tumors. Occlusion of these veins, which
drain much of the cerebellum and the brain Compression of the facial and vestibulocochlear
stem, infrequently may cause hemorrhagic nerves by tortuous arteries is postulated to cause
edema of the cerebellum and the brain stem. dysfunction of these nerves, and cases in which
Some of these veins may need to be sacrificed if surgical liberation of the vessels from these nerves
the tumor extends into the area above the inter- has relieved the symptoms provide support for a
nal acoustic meatus. Small acoustic neuromas vascular compressive etiology (Figs. 2.31 and
and other tumors in the lower part of the cer- 2.32) [9, 16, 32]. Ectasia and elongation of the
ebellopontine angle, however, frequently may arteries are important in forcing the arteries into
be removed without sacrificing a petrosal vein. the nerves. Gardner was the first to treat hemifa-
In removing the posterior meatal lip, a commu- cial spasm by removing a compressive arterial
nication may be established between the suba- loop from the facial nerve [9]. Jannetta et al.,
rachnoid space and the mastoid air cells that using the suboccipital approach to the cerebello-
will require careful closure to prevent a cere- pontine angle, found mechanical compression
brospinal fluid leak. Laying a small pledget and distortion of the root exit zone of the facial
of crushed fat over the drilled meatal area has nerve in all of 47 patients with hemifacial spasm
been successful in minimizing this complica- [16]. The distorting vessel not only was the AICA
tion. The retrosigmoid approach is used by this and its branches, but in some cases was found to
author for most acoustic neuromas because it is be the PICA, the vertebral or basilar artery, veins,
suitable for the removal of both small and large or an arteriovenous malformation (Figs. 2.31 and
tumors. Unlike the translabyrinthine approach, 2.32) [16]. It is expected that the AICA would be
described in the section on the temporal bone, the compressing vessel in most cases because the
which is directed through the vestibule and facial nerve is located in the middle neurovascu-
semicircular canals, the retrosigmoid approach lar complex. However, a tortuous PICA is an
is not necessarily associated with hearing loss. equally frequent offending vessel in hemifacial
The retrosigmoid approach provides a broader spasm followed in order by the vertebral artery,
exposure of the small tumor than does the mid- basilar artery, veins, and a combination of these
dle fossa approach. Also, once the nerves are vessels (Figs. 2.31 and 2.32). The proximal part
identified lateral to the tumor, there are advan- of the PICA usually passes around the brain stem
50 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

d
Anatomy of Vascular Compression in the Middle Neurovascular Complex 51

Fig. 2.29 Neurovascular relationships on the brain stem vein of the cerebellopontine fissure passes above the
side of an acoustic neuroma. Posterior view through a ret- flocculus along the superior limb of the cerebellopontine
rosigmoid craniotomy. (a) Neural relationships. The ori- fissure and joins the vein of the middle cerebellar pedun-
entation, skin incision (solid line), and craniotomy site cle and a transverse pontine vein to form a superior petro-
(interrupted line) are shown in the insert. The retractor is sal vein, which empties into the superior petrosal sinus.
on the petrosal surface of the cerebellum. The facial and A bridging vein passes behind the vagus nerve. The lateral
vestibulocochlear nerves arise at the lateral end of the anterior pontomesencephalic vein ascends on the pons.
pontomedullary sulcus, anterior to the flocculus, rostral to (d) Neurovascular relationships of an acoustic neuroma.
the glossopharyngeal, vagus, and accessory nerves, and The tumor arises from the vestibulocochlear nerve and
anterosuperior to the choroid plexus protruding from the displaces the facial nerve anteriorly, the trigeminal nerve
foramen of Luschka. The cerebellopontine fissure, formed superiorly, and the glossopharyngeal and vagus nerves
where the cerebellum wraps around the lateral side of the inferiorly. The vestibulocochlear nerve disappears into the
pons and middle cerebellar peduncle, has superior and tumor. The facial nerve enters the brain stem at the lateral
inferior limbs. (b) Arterial relationships. The AICA arises margin of the pontomedullary sulcus anterior to the
from the basilar artery and divides into a rostral trunk, flocculus and rostral to the choroid plexus protruding from
which passes above the flocculus to reach the surface of the foramen of Luschka. The rostral trunk of the AICA
the middle cerebellar peduncle, and a caudal trunk, which courses below the tumor and above the flocculus to reach
supplies the area below the flocculus. The PICA arises the surface of the middle cerebellar peduncle. The veins
from the vertebral artery and passes dorsally between the displaced around the medial side of the tumor are the lat-
vagus and accessory nerves. The SCA courses above the eral medullary veins and the veins of the middle cerebellar
trigeminal nerve. (c) Venous relationships. The veins that peduncle, cerebellomedullary fissure, and pontomedul-
join near the junction of the facial and vestibulocochlear lary sulcus. The vein of the cerebellopontine fissure passes
nerves with the brain stem are the lateral medullary veins above the tumor. A recurrent perforating branch of the
and the veins of the cerebellomedullary fissure, pon- AICA passes across the tumor and supplies the brain stem
tomedullary sulcus, and middle cerebellar peduncle. The (From Rhoton [37])

below the facial and vestibulocochlear nerves. In cochlear nerves in the interval between the brain
some cerebellopontine angles, however, the prox- stem and porus acusticus, as found by Gardner
imal part of the PICA, after coursing posteriorly [9], were not the cause of hemifacial spasm, but
to the level of the hypoglossal rootlets, will loop that cross compression of the facial nerve by the
superiorly toward the facial and vestibulocochlear same arteries coursing at right angles to the
nerves before descending to pass between the nerve at the root exit zone was the essential ele-
glossopharyngeal, vagus, and spinal accessory ment [16]. The craniotomy for hemifacial spasm
nerves. is positioned behind the lower half of the
The offending arterial loop may be located sigmoid sinus.
on either the superior or the inferior aspect of The operation for hemifacial spasm is directed
the facial nerve at its exit from the brain stem. along the inferolateral margin of the cerebellum
In the most common type of hemifacial spasm, (Figs. 2.33 and 2.34). The craniotomy is located
that beginning in the orbicularis oculi muscle medial to the lower half of the sigmoid sinus. It is
and gradually spreading downward to involve not necessary to extend the bone opening down-
the lower face, the anteroinferior aspect of the ward to the foramen magnum or upward to the
nerve root exit zone will commonly be com- transverse sinus. The inferolateral margin of the
pressed. Atypical hemifacial spasm, a much cerebellum is elevated with a small brain spatula,
less common entity, beginning in the lower or and the arachnoid behind the glossopharyngeal
midface and spreading upward to involve the and vagus nerves is opened.
frontalis muscle, will be caused by the compres- This will expose the tuft of choroid plexus
sion of the posterosuperior aspect of the facial protruding from the foramen of Luschka, which
nerve at the brain stem. Jannetta and others sits on the posterior surface of the glossopharyn-
thought that the arteries frequently seen cours- geal and vagus nerves. Commonly, the flocculus
ing around or between the facial and vestibulo- is seen protruding behind the nerves and blocks
52 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

b c

d e
Anatomy of Vascular Compression in the Middle Neurovascular Complex 53

Fig. 2.30 Retrosigmoid approach for removal of small- at the lateral end of the internal auditory canal. The
or medium-sized acoustic neuromas. (a) The patient is trigeminal nerve and SCA are above the tumor, and the
positioned in the three-quarter prone position with the glossopharyngeal and vagus nerves and the PICA are
surgeon behind the head. The insert (right) shows the site below the tumor. (d) The dissection along the eighth
of the scalp incision (continuous line) and the bony open- nerve is done in a medial to lateral direction (arrows) in
ing (interrupted line). (b) The posterior wall of the inter- order to avoid tearing the tiny filaments of the cochlear
nal auditory canal is removed using an irrigating drill. nerve in the lateral end of the canal where they pass
The AICA courses around the lower margin of the tumor. through the lamina cribrosa. The transverse crest sepa-
(c) The intracapsular contents of the tumor have been rates the superior and inferior vestibular nerves in the lat-
removed. The capsule of the tumor is being separated eral end of the canal. (e) Cerebellopontine angle and
from the pons and the posterior surface of the part of the internal auditory canal after tumor removal. The facial
facial and vestibulocochlear nerves adjacent to the brain and vestibulocochlear nerves have been preserved (From
stem. The superior and inferior vestibular nerves are seen Rhoton [31])

their visualization at the junction with the brain tibulocochlear nerves because it would be easy to
stem. It also may be difficult to see the facial miss a vessel compressing the facial nerve in this
nerve that is hidden in front of the vestibuloco- location.
chlear nerve. At this time, it is important to recall Venous compression is less commonly encoun-
that the facial nerve root exits the brain stem tered. The most common venous compression is
2–3 mm rostral to the point at which the by the vein of the pontomedullary sulcus, the
glossopharyngeal nerve enters the brain stem. To retro-olivary vein, or the vein of the middle cere-
expose the nerve’s exit zone, it may be necessary bral peduncle [24]. The vein of the pontomedul-
to gently separate the choroid plexus from the lary sulcus and the retro-olivary vein commonly
posterior margin of the glossopharyngeal nerve join in the region of the facial nerve to form the
so that its junction with the brain stem can be vein of the middle cerebellar peduncle, which
seen. The brain spatula is advanced upward to ascends on the middle cerebral peduncle toward
elevate the choroid plexus away from the poste- the superior petrosal sinus. The vein of the middle
rior margin of the glossopharyngeal nerve. The cerebral peduncle commonly passes between the
exposure is then directed several millimeters facial and vestibulocochlear nerves. It is not
above the glossopharyngeal nerve to where the uncommon to encounter a bridging vein that
facial nerve will be seen joining the brain stem passes from the lateral side of the medulla to the
below and in front of the vestibulocochlear nerve. jugular bulb. At the time of elevating the cerebel-
The spatula often needs to be positioned so that it lum, it is best to obliterate this vein with gentle
elevates the lower margin of the flocculus, which bipolar coagulation.
results in the approach being referred to as an
infrafloccular approach. Care must be taken to
avoid damage to the vestibulocochlear nerve, Cochlear and Vestibular Nerve
which may be adherent to the flocculus. In my Compression Syndromes
experience, the most common offending artery is
a PICA that loops upward prior to passing Vascular compression has been reported as a
between the glossopharyngeal, vagus, and spinal cause of cochlear and vestibular nerve dysfunction
accessory nerves. After looping into the facial manifested by tinnitus, hearing loss, disequilib-
nerve exit zone, the PICA then passes distally rium, and disabling positional vertigo [18, 25,
between the rootlets of the lower cranial nerves. 27]. The site of the compressive lesion with ves-
The compressing artery may also be the pre- tibulocochlear nerve dysfunction has been
meatal or postmeatal segments of the AICA or a reported to be more peripheral along the nerve
tortuous vertebral or basilar artery. Care is taken rather than at the junction with the brain stem, as
to explore the interval between the facial and ves- commonly seen in trigeminal neuralgia and
54 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a III

IV P.C.A
Pre.Mea.Seg. S.C.A

Post.Mea.Seg. Post.Mea.Seg.
Flocculus
VIII
VIII
Mea.Seg. VII
VII Pre.Mea.Seg.
Ch.Pl.
Pons IX
P.IC.A X
VI XI

V.A Medulla V.A

b III

P.C.A S.C.A

V V
A.I.C.A. A.I.C.A.
VII
VII
VIII VIII
V.A. P.I.C.A.
Flocculus
X IX
VI

Fig. 2.31 Sites of arterial compression of the facial nerve The junction of the right facial and vestibulocochlear
in hemifacial spasm. (a) Anterosuperior view. The facial nerves with the brain stem is compressed by a tortuous
and vestibulocochlear nerves are distorted at their junc- vertebral artery. The nerves on the left side are compressed
tion with the brain stem by the right premeatal and the left by the PICA (From Martin et al. [22])
postmeatal segments of the AICAs. (b) Anterior view.

hemifacial spasm. Jannetta and others have stem, but the entire intracranial portion of the
restricted the use of the operation for vestibulo- vestibulocochlear nerve is sensitive to compres-
cochlear nerve symptoms to those patients who sion because the glial-neurilemmal junction is
are disabled and have documented unilateral dis- located at or in the internal acoustic meatus [18].
ease on neuro-otologic testing. Jannetta et al. [18] Compression by veins is less common around
and Gardner [9] have postulated that vascular the facial and vestibulocochlear nerves than in
compression of a cranial nerve is more likely to the region of the trigeminal nerve because the
be symptomatic when it is located on the nerve veins around the facial and vestibulocochlear
proximal to the Obersteiner–Redlich zone where nerves tend to be smaller. Because no large bridg-
the axons are insulated by central myelin pro- ing veins cross the subarachnoid space around
duced by oligodendroglia. It is proximal to this the facial and vestibulocochlear nerves, as are
glial-neurilemmal junction that the compression seen frequently around the trigeminal nerve, any
causes transaxonal excitement between the affer- vascular cross compression of facial and vestibu-
ent and efferent fibers. This glial-neurilemmal locochlear nerves peripheral to the brain stem is
junction on the facial and trigeminal nerves is likely to be caused by arteries that loop through
situated at the nerve root junction with the brain the cerebellopontine angle and even into the
Anatomy of Vascular Compression in the Middle Neurovascular Complex 55

Fig. 2.32 (a) The PICA arises from the vertebral artery, passes between the vagus and spinal accessory nerves.
passes between the rootlets of the hypoglossal nerve, and The PICA compresses the medulla anterior to the origin
loops superiorly under the glossopharyngeal and vagus of the glossopharyngeal and vagus nerves. The choroid
nerves before passing posteroinferiorly between the root- plexus protrudes from the foramen of Luschka posterior
lets of the vagus and spinal accessory nerves. The verte- to the glossopharyngeal nerve. The cerebellar peduncles
bral artery stretches the rootlets of the hypoglossal nerve are above the lateral recess of the fourth ventricle. (c) The
posteriorly. The AICA loops posterior to the facial and vertebral artery displaces and stretches the hypoglossal
vestibulocochlear nerves. (b) A tortuous PICA arises from rootlets so far posteriorly that they intermingle with the
the vertebral artery and passes rostrally toward the ves- rootlets of the spinal accessory nerve. The PICA descends
tibulocochlear and facial nerves. At the level of the ves- between the rootlets of the spinal accessory nerve (From
tibulocochlear nerve, it loops inferiorly and descends Lister [21])
anterior to the glossopharyngeal and vagus nerves, and
56 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

meatus. The veins at the level of the junction of complex anatomy of this small nerve which is
the facial and vestibulocochlear nerves with the hidden between the vestibulocochlear and facial
brain stem tightly hug the pontomedullary junc- nerves (Figs. 2.2, 2.3, and 2.35) [44]. The ner-
tion where they are adhere to the pial membrane vus intermedius usually is described as a com-
as described in the section on hemifacial spasm. ponent of the facial nerve. Relatively little note
has been taken of the fact that it may be closely
bound to the vestibulocochlear nerve for a vari-
Geniculate Neuralgia able distance before it enters the brain stem,
and that in the cerebellopontine angle, it may
Sectioning the nervus intermedius for genicu- consist of as many as four rootlets. The nervus
late neuralgia requires an understanding of the intermedius is found divisible into three parts: a

a b

Fig. 2.33 Infrafloccular exposure of the facial nerve in the brain stem. (b) Enlarged view. Exposing the facial
hemifacial spasm. (a) The insert shows the approach nerve exit zone from the brain stem is facilitated by direct-
along the inferolateral margin of the cerebellum and below ing the exposure along the inferolateral margin of the cer-
the flocculus. The cerebellum has been elevated to expose ebellum in the area above the glossopharyngeal nerve and
the right cerebellopontine angle. The facial nerve exit below the lower edge of the flocculus. (c) The vestibuloco-
zone from the brain stem is seen along the lower margin of chlear nerve has been depressed. This exposes the distal
the vestibulocochlear nerve. The AICA passes between the segment of the facial nerve, but does not provide access to
facial and vestibulocochlear nerve. A large tortuous PICA the junction of the facial nerve with the brain stem, which
loops upward anterior to the facial and vestibulocochlear should be visualized in dealing with hemifacial spasm.
nerves and behind the trigeminal nerve, before turning (d) The vestibulocochlear nerve has been gently elevated.
downward to reach the medulla. The flocculus and the This exposes both the rostral and caudal margins of the
choroid plexus protruding from the foramen of Luschka facial nerve at the brain stem. A rootlet of the nervus inter-
often hide the junction of the facial and vestibulocochlear medius is also exposed. The vein of the middle cerebellar
nerves with brain stem. In this case, the flocculus has been peduncle passes between the facial and vestibulocochlear
gently elevated to expose the junction of these nerves with nerve (From Rhoton [40])
Lower Neurovascular Complex 57

c d

Fig. 2. 33 (continued)

medial segment that adheres closely to the ves- It is the free segment between the facial and
tibulocochlear nerve, an intermediate segment vestibulocochlear nerve that is divided in genicu-
that lies freely between the acoustic nerve and late neuralgia. This segment, where the nervus
the motor root of the facial nerve, and a distal intermedius is free of both the facial and vestibu-
segment that joins the motor root to form the locochlear nerves, may be located in the cerebel-
facial nerve [44]. Twenty-two percent of the lopontine angle or in the meatus if the nervus
nerves were adherent to the acoustic nerve for intermedius is composed of a single rootlet. If the
14 mm or more (the entire course of the nerve in nervus intermedius is composed of more than one
the posterior cranial fossa) and could be found rootlet, however, there may be free segments both
as a separate structure only after opening the in the cerebellopontine angle and in the meatus.
internal acoustic meatus. In most instances, the Geniculate neuralgia with or without vestibulo-
nerve was a single trunk, but in some cases, it cochlear dysfunction also has been postulated to
was composed of as many as four rootlets. A be caused by vascular compression of the nervus
single large root most frequently arises at the intermedius or vestibulocochlear nerve [18, 27].
brain stem anterior to the superior vestibular
nerve and, in the meatus, lies anterior to the
superior vestibular nerve. When multiple root- Lower Neurovascular Complex
lets are present, they may arise along the whole
anterior surface of the vestibulocochlear nerve; The lower complex, which is related to the PICA,
however, they usually converge immediately includes the medulla, inferior cerebellar pedun-
proximal to the junction with the facial motor cle, cerebellomedullary fissure, suboccipital sur-
root to form a single bundle that lies anterior to face of the cerebellum, and the glossopharyngeal,
the superior vestibular nerve. vagus, spinal accessory, and hypoglossal nerves
58 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b Mea.seg.
Subarc.A.
VIII,VII

Pons Pre.,mea.seg.

Post.mea.seg. IX
VII,VIII

IX X
Flocculus
A.I.C.A.
X
XI
XI
P.I.C.A.
Vert.A. Chor.Plex.

Medulla

P.I.C.A. Vert.A.

c d
Subarc.A. Mea.seg. Labyrin.A.

Pre.mea.seg. VIII Pre.mea.seg.


VII
IX IX
Post.mea.seg.
Post.mea.seg.
X X
VIII
XI XI
VII

P.I.C.A.
Chor.Plex.
P.I.C.A.

Medulla Chor.plex.

Fig. 2.34 Arterial compression of the facial nerve in choroid plexus block the view of the junction of the facial
hemifacial spasm as viewed through a retrosigmoid cran- and vestibulocochlear nerves with the brain stem. (c) The
iotomy performed with the patient in the three-quarter flocculus and the choroid plexus have been elevated in
prone position. (a) The upper drawing shows the site of the infrafloccular approach to the root entry/exit zone of
the incision (straight line) and the location of the cran- the facial and vestibulocochlear nerves. The premeatal
iotomy for the infrafloccular approach to the facial nerve segment compresses the nerves at the junction with the
(broken line). The lower drawing shows the surgical expo- pons and the medulla. (d) The nerve root entry/exit zone
sure obtained with this approach. The AICA and the facial is compressed by the postmeatal segment. (e) A tortuous
and vestibulocochlear nerves are in the midportion of the PICA loops upward to compress the nerves at their junc-
exposure. The vertebral artery, PICA, and the glossopha- tion with the brain stem before turning inferiorly to pass
ryngeal, vagus, and spinal accessory nerves are below. (b) between the glossopharyngeal and vagus nerves. (f) A tor-
The cerebellum is elevated to expose the facial and ves- tuous vertebral artery compresses the nerve root entry/exit
tibulocochlear nerves and the premeatal, meatal, and post- zone (From Rhoton [40])
meatal segments of the AICA. The flocculus and the
Lower Neurovascular Complex 59

e f

VIII
A.I.C.A. VII A.I.C.A.

Pons IX IX
VIII
X X
VII
P.I.C.A. XI
XI Vert.A.
P.I.C.A.
P.I.C.A.

Vert.A. Vert.A.

Fig. 2. 34 (continued)

(Figs. 2.1, 2.6, 2.7, 2.19, and 2.32). The PICA level of the superior one-third of the olive. The
arises at the medullary level and encircles the spinal accessory rootlets arise along the posterior
medulla, passing in relationship to the glossopha- margin of the inferior two-thirds of the olive and
ryngeal, vagus, spinal accessory, and hypoglossal from the lower medulla and the upper segments
nerves to reach the surface of the inferior cerebellar of the cervical spinal cord. The glossopharyngeal
peduncle, where it dips into the cerebellomedul- and vagus nerves arise rostral to the level of ori-
lary fissure and terminates by supplying the sub- gin of the hypoglossal rootlets.
occipital surface of the cerebellum. The glossopharyngeal nerve arises as one or
rarely two rootlets from the upper medulla, pos-
terior to the olive, just caudal to the origin of the
Neural Relationships facial nerve. It courses ventral to the choroid
plexus protruding from the foramen of Luschka
The glossopharyngeal, vagus, spinal accessory, on its way to the jugular foramen. Frequently, a
and hypoglossal nerves arise from the medulla larger dorsal and a smaller ventral component
along the margin of the inferior olive. The can be seen at the junction with the brain stem
glossopharyngeal, vagus, and spinal accessory (Fig. 2.36) [19, 43]. The smaller ventral rootlets
nerves arise as a line of rootlets that exit the brain have been demonstrated to be motor and the
stem along the posterior edge of the olive in the larger main bundle to be sensory [5, 51].
postolivary sulcus, a shallow groove between the The larger dorsal component usually arises from
olive and posterolateral surface of the medulla the medulla as one root except in a few cases in
(Figs. 2.1, 2.20, and 2.32). The hypoglossal nerve which it will originate as two rootlets. The two
arises as a line of rootlets that exit the brain stem rootlets may remain separate throughout their
along the anterior margin of the lower two-thirds course to the dura (Fig. 2.34). The vagus nerve
of the olive in the preolivary sulcus, a groove arises below the glossopharyngeal nerve as a line
between the olive and the medullary pyramid. of tightly packed rootlets along a line 2–5.5 mm
The glossopharyngeal and vagus nerves arise at the in length posterior to the superior one-third of
60 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b N.I.

N.I.
VII
VII

VIII

VIII

VII

VIII

Fig. 2.35 View of the cerebellopontine angle from above studied. The free segment is entirely in the meatus. (c)
to show the relationship of the nervus intermedius to the The nervus intermedius consists of three free segments:
facial and vestibulocochlear nerves. (a) Most common two are the angle and one is in the meatus. The nervus
relationship. The nervus intermedius is joined to the ven- intermedius in a could be exposed in the angle without
tral surface of the vestibulocochlear nerve for a few mil- drilling off the posterior lip of the meatus. In b, the free
limeters adjacent to the brain stem, then has a free segment segment could not be found in the angle but only in the
in the cerebellopontine angle as it courses to join the facial meatus (From Rhoton and Tedeschi [45])
motor root. (b) Pattern present in 20 % of the nerves

the olive (Figs. 2.1, 2.30, and 2.34). The most several small rootlets are found originating ven-
rostral vagal fibers arise adjacent to the glossopha- tral to the majority of the vagal rootlets
ryngeal origin, from which they are sometimes (Fig. 2.36). These small ventral rootlets are con-
separated by as much as 2 mm. The vagus is sidered to be motor [5].
composed of multiple combinations of large and The accessory nerve arises as a widely sepa-
small rootlets that pass ventral to the choroid rated series of rootlets that originated from the
plexus protruding from the foramen of Luschka medulla at the level of the lower two-thirds of
on its way to the jugular foramen. Occasionally, the olive and from the upper cervical cord. The
Lower Neurovascular Complex 61

cranial rootlets of the accessory nerve arise as a Anatomy of Glossopharyngeal


line of rootlets ranging in diameter from 0.1 to Neuralgia
1 mm just caudal to the vagal fibers (Figs. 2.32
and 2.37). The cranial rootlets of the accessory Dandy [2] described endocranial sectioning of
nerve are more properly regarded as inferior the glossopharyngeal nerve for neuralgia, but
vagal rootlets since they arise from vagal nuclei because this alone did not adequately control the
[19, 43]. It may be difficult to separate the lower neuralgia, he later advocated the additional sec-
vagal fibers from the upper accessory rootlets tioning of “perhaps 1/8 to 1/6 of the vagus”
because the vagal and cranial accessory fibers (Figs. 2.20, 2.32, and 2.37). Tarlov [51, 52] sec-
usually enter the vagal meatus as a single tioned the cephalic third of the vagal–spinal
bundle. accessory group and produced analgesia of the
The upper rootlets of the spinal portion of the epiglottis but only hypalgesia over the mucosa of
accessory nerve originate several millimeters the lower pharynx and larynx. In his second case,
caudal to the lowest cranial accessory fibers and he sectioned the cephalic half of the vagal–spinal
either course to join the cranial accessory bundle accessory complex; this caused both analgesia
or enter the lower border of the vagal meatus and transient paralysis of the ipsilateral soft pal-
separate from the cranial accessory rootlets. The ate, pharynx, and larynx. In our study, the struc-
spinal accessory fibers pass superolateral from ture of the vagus nerve was variable, being
their origin to reach the jugular foramen. Although composed of all large or all small rootlets or any
the cranial and spinal portion of the accessory combination of the two. It is suggested that fewer
nerve most frequently entered the vagal meatus of the rostral rootlets be cut if the diameters of the
together, they may infrequently be separated by a upper rootlets are large rather than small;
dural septum. the diameter of the largest rootlet is 1.5 mm and
The rootlets forming the hypoglossal nerve the smallest is 0.1 mm [43].
arise from the medulla along a line that is A large glossopharyngeal nerve diameter
continuous inferiorly with the line along which might be associated with a small diameter of the
the ventral spinal roots arise (Figs. 2.1, 2.7, and upper rootlets of the vagus nerve or a large vagus
2.32). These rootlets arise in a nucleus, whose nerve might be associated with a small glossopha-
rostral part sits deep to the hypoglossal triangle in ryngeal nerve because the two nerves arise from
the floor of the fourth ventricle, and exit the the same nuclei and have a similar function [5].
medulla along the anterior margin of the caudal This idea that more fibers might be distributed to
two-thirds of the olive. The hypoglossal rootlets one nerve, leaving the other smaller, was not
course anterolateral through the subarachnoid confirmed in our studies [43]. When the diameter
space and pass behind the vertebral artery to of the dorsal root of the glossopharyngeal nerve
reach the hypoglossal canal. If the vertebral artery is compared to the mean of the upper rootlets of
is short and straight, it may not contact or distort the vagus nerve, no significant correlation is
the hypoglossal rootlets, but if the artery is tortu- found [43]. A smaller diameter of the glossopha-
ous, it may stretch the hypoglossal rootlets poste- ryngeal nerve is not commonly associated with a
riorly over its dorsal surface [32]. Infrequently, large mean diameter of the upper rootlets of the
the vertebral artery passes between the rootlets of vagus nor is a large glossopharyngeal nerve
the hypoglossal nerve [21]. Prior to entering the diameter associated with a small diameter of the
hypoglossal canal, the rootlets collect into two vagal rootlets.
bundles, and in some cases, the canal is divided The only location where the glossopharyngeal
by a bony septum that separates the two bundles. nerve can consistently be distinguished from the
After passing through the canal, the bundles unite vagus is just proximal to the dural meatus where
and the nerve lies medial to the internal jugular a dural septum separates the glossopharyngeal
vein, and the glossopharyngeal, vagus, and acces- and vagus nerves. This septum varies in width
sory nerves. from 0.5 to 4.9 mm and serves to differentiate
62 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

Fig. 2.36 (a) Lateral view of the left side of the brain nerves. One ventral glossopharyngeal and two ventral
stem as outlined by the broken line. (b) Note the ventral vagal rootlets (arrows) are seen (From Rhoton and Buza
and dorsal rootlets of the glossopharyngeal and vagus [43])

the glossopharyngeal nerve from the vagus intracranial in 32 % of 50 jugular foramina that
nerve [43]. The close medullary origin of the we examined and within or extracranial to the
glossopharyngeal and vagus nerves and the fre- foramen in 68 % [43]. The superior ganglion of
quent arachnoid adhesions between the two make the vagus could be seen intracranially in only
separation difficult in their course through the 14 % of the cases.
subarachnoid space or adjacent to the brain stem
except in the few cases in which there will be a
1–2-mm separation between their origins at the Vascular Relationships
medulla.
The superior glossopharyngeal and vagal gan- The vertebral artery courses anterior to the nerves
glia may be visible intracranially [19]. In in the lower neurovascular complex. The hypo-
glossopharyngeal neuralgia, the glossopharyn- glossal rootlets usually pass behind the vertebral
geal nerve should be sectioned proximal to the artery; however, some hypoglossal rootlets infre-
superior ganglion. The superior ganglion was quently pass anterior to the artery (Fig. 2.6). If
Lower Neurovascular Complex 63

a c a c
Left
Right

b d b d

Fig. 2.37 The broken line on the drawing of the lateral series of small rootlets. Left: Note the small ventral root-
surface of the brain stem outlines the area shown in each lets of the glossopharyngeal nerve in (a, b), and (c) and
diagram, demonstrating the brain stem origin and varia- the small ventral rootlet between the glossopharyngeal
tions of the rootlet size of the glossopharyngeal, vagus, and vagus nerves in (a). The glossopharyngeal rootlet is
and spinal accessory nerves. The large ovoid structure is larger than the rostral rootlet of the vagus nerve in all
the inferior olive. The broken-line circles outline the ori- except (d), in which the rostral vagal rootlet is larger than
gin of the facial and vestibulocochlear nerves. The most the glossopharyngeal nerve. Right: Note the wide separa-
cephalad, shaded circles indicate glossopharyngeal root- tion of the origin of the glossopharyngeal and vagus
let origins; intermediate, open circles indicate vagal nerves in (c), the small ventral rootlet of the glossopha-
rootlet origins; and caudal, black circles outline spinal ryngeal nerve in (c), and the small ventral rootlets of the
accessory rootlet origins. The glossopharyngeal nerve glossopharyngeal and vagus nerves in (a). The glossopha-
usually originates as one large rootlet, the vagus as a series ryngeal nerve is smaller than the upper vagal rootlet in (a)
of large and small rootlets, and the spinal accessory as a and (d) (From Rhoton and Buza [43])

the vertebral artery is elongated or tortuous and and distorts these nerves as it passes between
courses laterally to the olive, it will stretch the them (Fig. 2.30). The relationships of the PICA
hypoglossal rootlets over its posterior surface. and vertebral artery to these nerves are reviewed
Some tortuous vertebral arteries will stretch the in greater detail in the section on the PICA.
hypoglossal rootlets so far posterior that they
intermingle with the glossopharyngeal, vagus,
and spinal accessory nerves. The PICA has a Vascular Compression in the Lower
much more complex relationship to these nerves Neurovascular Complex
(Figs. 2.6 and 2.32). The proximal part of the
PICA passes around or between and often The close relationship of the PICA and the verte-
stretches or distorts the rootlets of the nerves in bral artery to the glossopharyngeal and vagus
the lower complex. At the anterolateral medulla, nerves makes it logical to explore these relation-
the PICA passes around or between the rootlets ships in glossopharyngeal neuralgia [20]. Both
of the hypoglossal nerve. At the posterolateral the glossopharyngeal and vagus nerves have been
margin of the medulla, it passes between the fila found to be compressed at their junction with the
of the glossopharyngeal, vagus, and spinal brain stem by the PICA and/or the vertebral artery,
accessory nerves. The PICA may be ascending, with relief following separation of the arteries and
descending, or passing laterally or medially or nerves [20]. The adverse cardiovascular effects of
may be involved in a complex loop that stretches mobilization of these nerves and the risk to
64 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

causing swallowing and vocal cord defects have ryngeal and vagus nerves and between the
led some to conclude that rhizotomy of the individual vagal rootlets usually are too narrow to
glossopharyngeal nerve and upper vagal rootlets work through unless they have been opened by the
is a reasonable alternative to vascular mobiliza- tumor; however, the interval between the lower of
tion along the lateral medulla [16–18, 27, 46]. the cranial accessory rootlets may provide access
Jannetta has proposed that compression of the to lesions in the area.
left side of the medulla by the PICA or the verte-
bral artery may be a cause of hypertension and
that diabetes mellitus may result from right lat- Telovelar Approach to Fourth
eral medullary compression as a result of vagal Ventricle
effects on pancreatic islet cell secretion [7, 17].
The fact that hypertension is a component of the Lesions of the fourth ventricle, commonly
Cushing response to lateral medullary approached through the cerebellomedullary or
compression has been well established and that cerebellopontine fissures, posed a special chal-
hypertension has been relieved following decom- lenge to neurosurgeons because of the severe
pression of the left side of the medulla supports deficits which may follow injury to the struc-
this concept. The relationship of vascular com- tures in the ventricular walls and floor. In the
pression to diabetes mellitus awaits further past, operative access to the fourth ventricle was
elucidation. obtained by splitting the cerebellar vermis or
removing part of a cerebellar hemisphere
[26, 34, 38]. In examining the clefts and walls of
Tumors Involving Multiple the cerebellomedullary fissure, we have found
Neurovascular Complexes that opening the tela alone will provide adequate
ventricular exposure in most cases without split-
Tumors in the cerebellopontine angle commonly ting the vermis (Fig. 2.39). The inferior medul-
involve more than one of the neurovascular com- lary velum, another paper-thin layer, can also be
plexes (Fig. 2.38). An especially difficult challenge opened if opening the tela does not provide
is exposing and removing the tumors that are situ- adequate exposure. Opening the tela alone pro-
ated medial to the nerves. In this case, the opera- vides access to the full length of the floor and all
tion must be directed through the interval between of the ventricular cavity except, possibly, the
the neurovascular complexes because these tumors fastigium, superolateral recess, and the superior
often will widen these intervals. Lesions in the half of the roof. Opening the inferior medullary
upper cerebellopontine angle may be exposed velum accesses the latter areas and the superior
through the interval between the lower margin of half of the roof. Extending the telar opening lat-
the tentorium and the upper edge of the trigeminal erally toward the foramen of Magendie opens
nerve. Care is needed to protect the trochlear nerve the lateral recess and exposes the peduncular
and the SCA in this area. Further inferiorly, the surfaces bordering the recess. Tumors in the
medially placed tumor may be approached through fourth ventricle may stretch and thin these two
the interval between the trigeminal nerve above semitranslucent membranes to a degree that one
and the facial and vestibulocochlear nerves below. may not be aware that they are being opened in
If the tumor has an even lower attachment near the exposing a fourth ventricular tumor. There are
jugular foramen, it can be approached through the no reports of deficits following isolate opening
interval between the lower margin of the nerves of the tela and velum. However, other structures
entering the internal meatus and the upper margin exposed in the ventricle walls and at risk for
of the glossopharyngeal nerve, or through the producing the deficits described above include
interval between the lower rootlets of the vagus the dentate nuclei, cerebellar peduncles, floor of
nerve and the upper rootlets of the spinal acces- the fourth ventricle, and the PICA. During an
sory nerve. The intervals between the glossopha- operation on the caudal part of the roof, one
Telovelar Approach to Fourth Ventricle 65

should remember that the dentate nuclei are operations on lesions involving the superior part
located just rostral to the superior pole of the of the roof above the level of the dentate tuber-
tonsils underlying the dentate tubercles in the cles; the inferior peduncles are most susceptible
posterolateral part of the roof where they are to damage in exposing lesions within the lateral
wrapped around the superolateral recesses near recess; and the middle cerebellar peduncle is
the lateral edges of the inferior medullary velum susceptible to injury in procedures near the
(Fig. 2.36). All the cerebellar peduncles con- external wall of the superior half of the roof
verge on the lateral wall and roof where they such as those in the cerebellopontine angle
may be damaged. The superior cerebellar because the middle peduncle forms a major part
peduncle is more likely to be injured during of the cisternal surface of the ventricular wall.

VII,VIII
IX

V X

IV XI

Flocculus
Ch.plex

Fig. 2.38 Tumors involving multiple neurovascular com- chlear nerves are stretched below the tumor. (c) The tumor
plexes. (a) Routes that can be taken between the cranial has been removed. The thin distorted nerves have been
nerves to expose and remove a tumor situated medial to preserved, and the remaining dural attachment is removed
and involving multiple cranial nerves. The patient is posi- or cauterized with bipolar coagulation. The basilar artery
tioned in the three-quarter prone position. The insert and abducens nerve are exposed. (d) Large meningioma
(upper left) shows the site of the vertical scalp incision arising from the clivus in the region of the inferior petro-
and craniotomy. The approach to pathology located medial sal sinus with involvement of the fourth through the elev-
to the nerves can be directed (arrows) between the enth nerves. The nerves are displaced laterally around the
trochlear nerve above and trigeminal nerve below, tumor. The tumor is removed by working through the
between the trigeminal nerve above and the facial and vesti- intervals between the nerves. (e) The meningioma has
bulocochlear nerves below, between the facial and been removed. The dural attachment has been partially
vestibulocochlear nerves above and the glossopharyngeal removed and the base is being cauterized. (f) Meningioma
nerve below, between the glossopharyngeal and vagus arising medial to the jugular bulb in the region of the jugu-
nerves, between the vagus nerve and accessory rootlets, lar tubercle and involving the lower cranial nerves. (g)
and between the widely separated rootlets of the acces- The tumor was removed by operating through the inter-
sory nerve. Tumor located medial to the nerves will often vals between the facial and vestibulocochlear nerves
widen the intervals between the nerves, depending on the above and the glossopharyngeal nerve below and between
site of origin of the tumor. Choroid plexus protrudes from the glossopharyngeal and vagus nerves (round insert). (h)
the foramen of Luschka. (b) Meningioma attached lateral Large epidermoid tumor being removed by working
to the trigeminal nerve in the region of the superior petro- through the intervals between the nerves. (i) Distorted
sal sinus. The trochlear nerve is elevated, the trigeminal nerves after the removal of the epidermoid tumor (From
nerve is pushed medially, and the facial and vestibuloco- Rhoton [40])
66 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

Sup.pet.sinus
VII,VIII

Tent. A.I.C.A.
Sig.sinus
Tumor
IX ,X
XI
IV
S.C.A. Vert.A.

Flocculus
XII

P.I.C.A.
VII,VIII
Ch.Plex

A.I.C.A.

VII,VIII
IX ,X

VI

Tent.
S.C.A. V

IV
Vert.A.

Midbrain

Medulla

Bas.A.
P.I.C.A.
Pons

Fig. 2. 38 (continued)
Telovelar Approach to Fourth Ventricle 67

VII,VIII
d

Sig.sinus

Sup.pet.sinus IX,X

XI,XI
V
S.C.A.
Tumor Vert.A.
IV

A.I.C.A.

P.I.C.A.
Ch.Plex.

e VII,VIII IX
Dural Edge
X

V XI

IV XII

S.C.A.
Vert.A.
Tent.

A.I.C.A. P.I.C.A.
Ch.plex.

Fig. 2. 38 (continued)
68 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

f
VI VII,VIII
V

Tumor
S.C.A.
Sig. sinus
IV IX,X,XI

Tent. Vert.A.
Medulla

Pons
P.I.C.A.
A.I.C.A.

IX
g
VII,VIII

VII,VIII VI

A.I.C.A. A.I.C.A. XI
IV

V
IX XII
X

X
XI
S.C.A.
P.I.C.A.
Tent.

IV

Bas.A.
Vert.A.

P.I.C.A.

Fig. 2. 38 (continued)
Telovelar Approach to Fourth Ventricle 69

h VII,VIII Tumor Sig. sinus

Sup.pet.sinus XII
V XI

IV
Tent. Vert.A.
S.C.S.

X
A.I.C.A.

P.I.C.A.
IX

i IV VII,VIII IX

X
XII
V

XI

Tent.

IV Vert.A.

S.C.A.

Pons
P.I.C.A.
Ch.plex.
A.I.C.A.

Fig. 2. 38 (continued)
70 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

a b

c d

e f

Fig. 2.39 Telovelar approach to the fourth ventricle and arises, is hidden deep to the uvula. (c) The uvula has been
lateral recess. (a) The suboccipital cerebellar surface is retracted to the right and the tonsil to the left to expose the
located below and between the sigmoid and lateral sinuses inferior medullary velum and the tela choroidea forming the
and is the surface that is exposed in a suboccipital craniec- lower half of the roof of the ventricle. The tela choroidea has
tomy. The vermis sits in a depression, the posterior cerebel- been opened extending from the foramen of Magendie to
larincisura,betweenthehemispheres.Thecerebellomedullary the junction with the inferior medullary velum to expose the
fissure extends superiorly between the cerebellum and ventricular floor and aqueduct. (d) The left half of the infe-
medulla along the inferior half of the ventricular roof and rior medullary velum has been divided to expose the supero-
lateral recess. The vallecula extends upward between the lateral recess and the ventricular surface formed by the
tonsils and communicates through the foramen of Magendie superior and inferior peduncles. (e) The cerebellar tonsil has
with the fourth ventricle. The posteroinferior cerebellar been elevated to expose the tela forming the lower part of
arteries loop above the tonsil and exit the fissure to supply the roof of the lateral recess. (f) The tela has been opened to
the suboccipital surface. (b) Both tonsils have been retracted expose the lateral recess. The opening extends laterally to
laterally to expose the inferior medullary velum and tela the foramen of Luschka. The choroid plexus and flocculus
choroidea that form the lower half of the ventricular roof. are exposed in the cerebellopontine angle behind the
The nodule of the vermis, on which the inferior medullary glossopharyngeal nerve (From Rhoton [34])
References 71

References 17. Jannetta PJ, Gendell HM (1979) Clinical observations


on etiology of essential hypertension. Surg Forum
30:431–432
1. Atkinson WJ (1949) The anterior inferior cerebellar
18. Jannetta PJ, Moller MB, Moller AR, Sekhar LN
artery; its variations, pontine distribution, and significance
(1986) Neurosurgical treatment of vertigo by micro-
in the surgery of cerebello-pontine angle tumours. J
vascular decompression of the eighth cranial nerve.
Neurol Neurosurg Psychiatry 12(2):137–151
Clin Neurosurg 33:645–665
2. Dandy WE (1927) Glossopharyngeal neuralgia (tic
19. Katsuta T, Rhoton AL Jr, Matsushima T (1997) The
douloureux): its diagnosis and treatment. Arch Surg
jugular foramen: microsurgical anatomy and operative
15:198–214
approaches. Neurosurgery 41(1):149–201; discussion
3. Dandy WE (1929) An operation for the cure of tic
201–202
douloureux: partial section of the sensory root at the
20. Laha RK, Jannetta PJ (1977) Glossopharyngeal
Pons. Arch Surg 18:687–734
neuralgia. J Neurosurg 47(3):316–320. doi:10.3171/
4. Dandy WE (1934) Concerning the cause of trigeminal
jns.1977.47.3.0316
neuralgia. Am J Surg 24:447–455
21. Lister JR, Rhoton AL Jr, Matsushima T, Peace DA
5. DuBois FSFJ, Foley JO (1936) Experimental studies
(1982) Microsurgical anatomy of the posterior infe-
on the vagus and spinal accessory nerves in the cat.
rior cerebellar artery. Neurosurgery 10(2):170–199
Anat Rec 64:285–307
22. Martin RG, Grant JL, Peace D, Theiss C, Rhoton AL
6. Emmons WF, Rhoton AL Jr (1971) Subdivision of the
Jr (1980) Microsurgical relationships of the anterior
trigeminal sensory root. Experimental study in the
inferior cerebellar artery and the facial-vestibuloco-
monkey. J Neurosurg 35(5):585–591. doi:10.3171/
chlear nerve complex. Neurosurgery 6(5):483–507
jns.1971.35.5.0585
23. Matsushima T, Rhoton AL Jr, Lenkey C. (1982).
7. Fein JM, Frishman W (1980) Neurogenic hyperten-
Microsurgery of the fourth ventricle I: Microsurgical
sion related to vascular compression of the lateral
anatomy. Neurosurgery, 11, 631–667
medulla. Neurosurgery 6(6):615–622
24. Matsushima T, Rhoton AL Jr, de Oliveira E, Peace D
8. Fujii K, Lenkey C, Rhoton AL Jr (1980) Microsurgical
(1983) Microsurgical anatomy of the veins of the
anatomy of the choroidal arteries. Fourth ventricle
posterior fossa. J Neurosurg 59(1):63–105.
and cerebellopontine angles. J Neurosurg 52(4):
doi:10.3171/jns.1983.59.1.0063
504–524. doi:10.3171/jns.1980.52.4.0504
25. Moller MB, Moller AR, Jannetta PJ, Sekhar L (1986)
9. Gardner WJ (1962) Concerning the mechanism of
Diagnosis and surgical treatment of disabling posi-
trigeminal neuralgia and hemifacial spasm. J Neurosurg
tional vertigo. J Neurosurg 64(1):21–28. doi:10.3171/
19:947–958. doi:10.3171/jns.1962.19.11.0947
jns.1986.64.1.0021
10. Gudmundsson K, Rhoton AL Jr, Rushton JG (1971)
26. Mussi AC, Rhoton AL Jr (2000) Telovelar approach to
Detailed anatomy of the intracranial portion of the
the fourth ventricle: microsurgical anatomy. J Neurosurg
trigeminal nerve. J Neurosurg 35(5):592–600.
92(5):812–823. doi:10.3171/jns.2000.92.5.0812
doi:10.3171/jns.1971.35.5.0592
27. Ouaknine GE, Robert F, Molina-Negro P, Hardy J
11. Hardy DG, Peace DA, Rhoton AL Jr (1980)
(1980) Geniculate neuralgia and audio-vestibular dis-
Microsurgical anatomy of the superior cerebellar
turbances due to compression of the intermediate and
artery. Neurosurgery 6(1):10–28
eighth nerves by the postero-inferior cerebellar artery.
12. Hardy DG, Rhoton AL Jr (1978) Microsurgical
Surg Neurol 13(2):147–150
relationships of the superior cerebellar artery and the
28. Pait TG, Harris FS, Paullus WS, Rhoton AL Jr (1977)
trigeminal nerve. J Neurosurg 49(5):669–678.
Microsurgical anatomy and dissection of the temporal
doi:10.3171/jns.1978.49.5.0669
bone. Surg Neurol 8(5):363–391
13. Horsley V, Taylor J, Coleman WS (1981) Remarks on
29. Rhoton AL Jr (1990) Microsurgical anatomy of
the various surgical procedures devised for the relief
decompression operations on the trigeminal nerve.
or cure of trigeminal neuralgia (tic douloureux). Brit
In: Rovit RLMR, Murali R, Jannetta PJ (eds)
Med J 2:1139–1143, 1191–1193, 1249–1252
Trigeminal neuralgia. Williams and Wilkins,
14. House WF (1979) Translabyrinthine approach. In:
Baltimore, pp 165–200
House WFLC, Luetje CM (eds) Acoustic tumors, II
30. Rhoton AL Jr (1993) Instrumentation. In: Apuzzo MLJ
management. University Park Press, Baltimore, pp
(ed) Brain surgery: complication, avoidance and man-
43–87
agement. Churchill-Livingstone, New York, pp
15. Jannetta PJ (1967) Arterial compression of the trigem-
1647–1670
inal nerve at the pons in patients with trigeminal neu-
31. Rhoton AL Jr (1993) Microsurgical anatomy of
ralgia. J Neurosurg 26(1 Suppl):159–162. doi:10.3171/
acoustic neuromas. In: Sekhar LN, Janecka IP (eds)
jns.1967.26.1part2.0159
Surgery of cranial base tumors. Raven Press, Ltd,
16. Jannetta PJ, Abbasy M, Maroon JC, Ramos FM, Albin
New York, pp 687–713
MS (1977) Etiology and definitive microsurgical
32. Rhoton AL Jr (1993) Microsurgical anatomy of
treatment of hemifacial spasm. Operative techniques
posterior fossa cranial nerves. In: Barrow DL (ed)
and results in 47 patients. J Neurosurg 47(3):321–328.
Surgery of the cranial nerves of the posterior fossa:
doi:10.3171/jns.1977.47.3.0321
72 2 Microsurgical Anatomy of the Cerebellopontine Angle by the Retrosigmoid Approach

neurosurgical topics. American Association of 45. Rhoton AL Jr, Tedeschi H (1992) Microsurgical anat-
Neurological Surgeons, Chicago, pp 1–103 omy of acoustic neuroma. Otolaryngol Clin North
33. Rhoton AL Jr (2007) Cerebellopontine angle and the Am 25(2):257–294
retrosigmoid approach. Neurosurgery 61(S4):175–192 46. Segal R, Gendell HM, Canfield D, Dujovny M,
34. Rhoton AL Jr (2007) Telovelar approach to the fourth Jannetta PJ (1979) Cardiovascular response to pulsa-
ventricle. Neurosurgery 61(S4):193–210 tile pressure applied to ventrolateral medulla. Surg
35. Rhoton AL Jr (1974) Microsurgery of the internal Forum 30:433–435
acoustic meatus. Surg Neurol 2(5):311–318 47. Seoane E, Rhoton AL Jr (1999) Suprameatal exten-
36. Rhoton AL Jr (1976) Microsurgical removal of acous- sion of the retrosigmoid approach: microsurgical
tic neuromas. Surg Neurol 6(4):211–219 anatomy. Neurosurgery 44(3):553–560
37. Rhoton AL Jr (1986) Microsurgical anatomy of the 48. Sunderland S (1948) Neurovascular relations and
brainstem surface facing an acoustic neuroma. Surg anomalies at the base of the brain. J Neurol Neurosurg
Neurol 25(4):326–339 Psychiatry 11(4):243–257
38. Rhoton AL Jr (2000) Cerebellum and fourth ventricle. 49. Tanriover N, Abe H, Rhoton AL Jr, Kawashima M,
Neurosurgery 47(3 Suppl):S7–S27 Sanus GZ, Akar Z (2007) Microsurgical anatomy of the
39. Rhoton AL Jr (2000) The cerebellar arteries. superior petrosal venous complex: new classifications
Neurosurgery 47(3 Suppl):S29–S68 and implications for subtemporal transtentorial and ret-
40. Rhoton AL Jr (2000) The cerebellopontine angle and rosigmoid suprameatal approaches. J Neurosurg
posterior fossa cranial nerves by the retrosigmoid 106(6):1041–1050. doi:10.3171/jns.2007.106.6.1041
approach. Neurosurgery 47(3 Suppl):S93–S129 50. Tanriover N, Rhoton AL Jr (2005) The anteroinferior
41. Rhoton AL Jr (2000) The posterior fossa cisterns. cerebellar artery embedded in the subarcuate fossa: a
Neurosurgery 47(3 Suppl):S287–S297 rare anomaly and its clinical significance. Neurosurgery
42. Rhoton AL Jr (2000) The posterior fossa veins. 57:314–319
Neurosurgery 47(3 Suppl):S69–S92 51. Tarlov I (1937) Structure of the nerve root. II: differ-
43. Rhoton AL Jr, Buza R (1975) Microsurgical anatomy entiation of sensory from motor roots; observations
of the jugular foramen. J Neurosurg 42(5):541–550. on identification of function in roots of mixed cranial
doi:10.3171/jns.1975.42.5.0541 nerves. Arch Neurol Psychiatry 37:1338–1355
44. Rhoton AL Jr, Kobayashi S, Hollinshead WH (1968) 52. Tarlov I (1942) Section of the cephalic third of the
Nervus intermedius. J Neurosurg 29(6):609–618. vagus-spinal accessory complex: clinical and histo-
doi:10.3171/jns.1968.29.6.0609 logic results. Arch Neurol Psychiatry 47:141–148
Neuropathology
of the Cerebellopontine Angle 3
and Its Surroundings

Introductory Remarks For tumors as the most important pathological


entities with regard to neurosurgical interven-
In this chapter, the properties of the most typical tions, the relevant international reference for
and relevant tumors and other pathologies of the classification is the series of classification books
cerebellopontine angle (CPA) and its surround- edited by the World Health Organization (WHO).
ings are dealt with from a neuropathological For skull base tumors, three of the books are of
point of view in order to present a quick orienta- special interest, the WHO Classification of
tion for clinicians interested in macroscopic and Tumours of the Central Nervous System [25], the
microscopic pathomorphology. It is a main inten- WHO Classification of Head and Neck Tumours
tion to provide a broad scale of pathomorphologi- [4], and the WHO Classification of Tumours of
cal images enabling clinicians to get impressions Soft Tissue and Bone [12].
on the relevant macropathology of the CPA on For non-tumorous pathological entities, the stan-
the one hand and to better interpret the value and dard handbook Greenfield’s Neuropathology is rec-
the limitations of histopathological findings on ommended for further reading [26]. The description
the other hand. of non-tumorous pathological entities in the CPA is
For practical reasons, I have chosen to divide restricted to some of those entities, which might be
the chapter in the following subchapters, all of encountered more commonly by the neurosurgeon.
them introduced by an overview of the described
entities:
• Pathology of the peripheral nervous system Pathology of the Peripheral Nervous
• Pathology of the autonomous nervous System
system
• Pathology of the meninges Schwannoma
Cellular schwannoma
• Pathology of the osseous skull base
Plexiform schwannoma
• Pathology of the sellar region
Melanotic schwannoma
• Pathology of regional glial tumors
Malignant peripheral nerve sheath tumor (MPNST)
• Pathology of the choroid plexus
Epithelioid MPNST
• Pathology of the hematopoietic system MPNST with mesenchymal differentiation
• Pathology of germ cells Malignant Triton tumor
• Pathology of blood vessels and circulation Glandular MPNST

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 73


DOI 10.1007/978-3-642-35422-9_3, © Springer-Verlag Berlin Heidelberg 2013
74 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

Schwannoma Vestibular schwannomas arise from the supe-


rior or the inferior branches of the vestibular and
Age distribution: All ages with a peak in the not from the cochlear portion of the vestibuloco-
fourth to sixth decades. chlear nerve. The tumor growth regularly com-
Sex distribution: Male to female ratio of 1:2. mences in the internal auditory canal, later
Epidemiology: 8 % of intracranial tumors; occupying the cerebellopontine angle and reaching
85 % of cerebellopontine angle tumors. Ninety a diameter of a few centimeters, thereby producing
percent solitary and sporadic, 5 % multiple and a deformation of the brainstem and the adjacent
associated with NF2, 5 % multiple and unassoci- cerebellum as well as stretching and compressing
ated with NF2. the neighboring cranial nerves.
Grading: WHO grade I (non-melanotic); Histologically, schwannomas are usually
about 10 % of melanotic schwannomas follow a encapsulated and composed of elongated cells in
malignant course. interlacing fascicles (Antoni A-areas) often inter-
Schwannomas derive from the Schwann cells mingled with extracellular clear areas of fatty
which form the myelin sheath in peripheral degeneration and more variable cell shapes (Antoni
nerves. With the exception of the olfactory and B-areas). The arrangement of tumor nuclei in pali-
optic nerves being entirely invested by central sades is a common feature. Verocay bodies in form
myelin formed by oligodendrocytes, the periph- of parallel nuclei surrounding zones with few
eral portion of the other cranial nerves is invested nuclei can infrequently be found. So-called ancient
by peripheral myelin formed by Schwann cells. schwannomas show a distinct nuclear polymor-
The vast majority of schwannomas of the skull phism as well as hyalinized vessels indicating
base occur in the cerebellopontine angle arising regressive changes but not malignancy. Also the
from the eighth cranial nerve, the vestibular variant of cellular schwannoma with a high cellu-
schwannomas. Nonvestibular schwannomas of larity remains benign (Figs. 3.1 and 3.2).
the trigeminal nerve are rare; all other locations Bilateral vestibular schwannomas are pathog-
are extremely rare. In German-speaking coun- nomonic for neurofibromatosis 2 (NF2), an
tries, the synonym acoustic neurinoma is com- autosomal dominant disorder additionally char-
monly used; the synonym neurilemoma is almost acterized by multiple peripheral schwannomas
everywhere abandoned. including plexiform cutaneous schwannomas,

a b

Fig. 3.1 (a) Small vestibular schwannoma in an early trigeminal micro-schwannoma expanding within the
stage extending from the internal auditory canal (arrow). nerve. Luxol fast blue. (e) Vestibular schwannoma with
(b) Large vestibular schwannoma protruding into the cer- dense fascicular Antoni A-areas and clear Antoni B-areas
ebellopontine angle (arrow). (c) Transition between the by fatty degeneration (arrows). (f) Encapsulated schwan-
central oligodendroglial portion (turquoise) and the noma compressing the adjacent eighth nerve; note the
peripheral Schwann cell portion (dark violet) of the acous- compressed red myelin sheaths. Masson trichrome
tic nerve. Luxol fast blue–PAS staining. (d) Incipient
Pathology of the Peripheral Nervous System 75

c d

e f

Fig. 3.1 (continued)

a b

c d

Fig. 3.2 (a) Loose fascicular pattern of elongated tumor polymorphism but less cell density than in cellular
cells with spindle-shaped monomorphous nuclei (Antoni schwannoma. HE. (d) Cellular schwannoma forming a
A). HE. (b) Typical palisading of tumor cell nuclei. HE. fascicular pattern with high cell density. HE
(c) Ancient schwannoma with prominent nuclear
76 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

Fig. 3.3 (a) Bilateral vestibular schwannoma in NF2 note the decompression of the pons after unilateral surgical
before surgical removal of the smaller one. After surgical tumor removal. (c) Plexiform schwannoma with lobular
tumor removal, the patient died of pulmonary embolism. pattern in NF2. HE. (d) Within the lobules, palisading of
(b) Remaining larger vestibular schwannoma in autopsy; the nuclei can be observed. HE

dysplastic lesions of Schwann cells (schwan- schwannomas may rarely affect cranial nerves such
nosis), meningiomas, gliomas (astrocytomas, as the acoustic nerve [31]. However, there is a dis-
ependymomas), and glial microhamartomas as cussion if melanocytic tumors of the cerebellopon-
well as cerebral calcifications and posterior lens tine angle mimicking vestibular schwannomas
opacities. NF2 vestibular schwannomas may might actually be malignant melanomas [37].
entrap nerve fibers of the facial nerve (Fig. 3.3).
Melanotic schwannomas occur in non-psam-
momatous and psammomatous varieties grossly Malignant Peripheral Nerve Sheath
appearing pigmented, because the Schwann cells Tumor (MPNST)
contain melanosomes immunohistochemically
reactive for melanoma markers. About 50 % of Age distribution: Primarily adults in the third to
psammomatous melanotic schwannomas are sixth decades; in NF1-associated cases in the
accompanied by the autosomal dominant Carney third to fourth decades.
complex which is characterized by lentiginous Sex distribution: No sex predilection; in NF1-
facial pigmentation, cardiac myxoma, and endo- associated cases slightly more common in males.
crine disorders including Cushing’s syndrome and/ Epidemiology: No accurate incidence rates for
or acromegaly. Melanotic non-psammomatous tumors arising from intracranial nerves.
trigeminal schwannoma as the first manifestation Grading: WHO grade II, III, or IV; applied
of Carney complex is reported postulating that the following the criteria of sarcoma grading [13].
absence of psammoma bodies or cranial localiza- MPNSTs of the vestibular nerve are rare
tion does not exclude this diagnosis [8]. Melanotic [21, 29, 32]. Extremely rare variants with
Pathology of the Autonomous Nervous System 77

a b

c d

Fig. 3.4 (a) Glomus jugulare tumor presenting as the chief cells of the “Zellballen.” Synaptophysin. (d)
circumscribed vascularized soft mass. (b) Jugulotympanic Ultrastructure of a chief cell with cytoplasmic dark round
paraganglioma with “Zellballen” arrangement of chief granules containing noradrenaline (arrows). Transmission
cells. HE. (c) Strong positivity for synaptophysin within electron microscopy

a potentially malignant course comprise the Epidemiology: No accurate incidence rates


malignant Triton tumor with rhabdomyoblas- available.
tic cells [5, 14] reminiscent of the Triton tumor Grading: WHO grade I.
observed in neurofibromatosis 1 (NF1). Paragangliomas derive from sympathetic
or parasympathetic autonomous nervous cell
complexes producing catecholamines. In
Pathology of the Autonomous the literature, frequently also the synonym
Nervous System chemodectoma is used. Intracranial paragan-
gliomas are rare. At the skull base, they may
Jugulotympanic paraganglioma (chemodectoma) be situated either in relation to the jugular
Glomus jugulare tumor
bulb (glomus jugulare tumor) or under the
Glomus tympanicum tumor
mucosa of the middle ear (glomus tympani-
cum tumor). Though the jugular variant may
infiltrate the petrous bone, the course normally
Jugulotympanic Paraganglioma remains benign. Histologically, paragan-
gliomas are composed of epithelioid uniform
Age distribution: More common in Caucasians chief cells and sustentacular cells character-
with a peak in the fifth and sixth decades. istically forming clusters or “Zellballen.”
Sex distribution: Strong female predilection; Immunohistochemically, the epithelioid chief
the familial type with gene mutation on chromo- cells express synaptophysin and chromogra-
some 11 occurs predominantly in men. nin (Fig. 3.4).
78 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

Pathology of the Meninges 10 %, anaplastic meningiomas approximately


1–3 % of all meningiomas.
Meningioma Grading: WHO grade I, II, or III (see text
Meningothelial
below).
Fibrous (fibroblastic)
The histogenesis of meningiomas is both neu-
Transitional (mixed)
roectodermal and mesenchymal. They derive on
Psammomatous
the one hand from neuroectodermal cells of the
Angiomatous
Microcystic
arachnothelial layer, the arachnoidal cap cells,
Secretory and on the other hand from mesenchymal cells of
Lymphoplasmacyte-rich subarachnoidal blood vessels and from fibroblasts
Metaplastic of the subarachnoidal connective tissue. The
Chordoid changing composition of these histological ele-
Clear cell ments is responsible for the different subtypes of
Atypical meningiomas. Thus, meningiomas are tumors
Papillary with both neuroectodermal and mesenchymal
Rhabdoid histogenesis. Since the first description by
Anaplastic (malignant) Cushing and Eisenhardt, the correlation between
Hemangiopericytoma pregnancy and rapid increase in neurological
Anaplastic symptoms is known.
Hemangioblastoma At the skull base, the typical nodular spherical
Leptomeningeal melanocytic lesions
or dumbbell-shaped growth type of meningiomas
Diffuse melanocytosis
can be seen but also an “en plaque”-growth type
Melanomatosis
expanding in a diffuse carpet-like fashion.
Melanocytoma
Especially meningiomas en plaque tend to per-
Malignant melanoma
Metastatic meningiosis and solid metastastic tumors
meate adjacent bony structures causing hyperos-
Meningiosis carcinomatosa tosis. All types of the histologically manifold
Meningiosis sarcomatosa meningiomas can occur.
Meningiosis lymphomatosa WHO grade I subtypes comprise the following:
Meningitis Meningothelial meningiomas form syncytial
Bacterial meningitis cell clusters. Few whorls or psamomma bodies
Acute bacterial sive purulent meningitis may be encountered. The oval nuclei contain little
Tuberculous meningitis chromatin and are characteristically pale. Fibrous
Sarcoidosis meningiomas show a conspicuous formation of
Parasitic meningitis fibroblasts between elongated strands of menin-
Cysticercosis gothelial tissue. Transitional meningiomas are
Fungal meningitis characterized by a pronounced tendency to form
Aspergillosis whorls. Psammomatous meningiomas contain
Cryptococcosis
large amounts of calcified round psammoma bod-
ies. Angiomatous meningiomas exhibit many
Meningioma large and small blood vessels, often with a fibrous
hyaline thickening of the vessel wall. Microcystic
Age distribution: All ages with a peak in the fifth meningiomas form microcysts containing pale-
to seventh decades. eosinophilic mucinous fluid. Secretory menin-
Sex distribution: Female patients predominate giomas contain small round PAS-positive hyaline
in benign meningiomas, whereas no sexual predi- inclusions, the pseudopsammoma bodies.
lection in malignant variants. Lymphoplasmacyte-rich meningiomas show lym-
Epidemiology: 25–30 % of primary intracra- phocytic infiltrates of chronic inflammatory char-
nial tumors; atypical meningiomas approximately acter. Metaplastic meningiomas exhibit focal
Pathology of the Meninges 79

a b

c d

e f

Fig. 3.5 (a) Dura (blue collagenous fibers) with incipient (d) Transitional meningioma with abundant whorl forma-
meningioma. Masson trichrome. (b) Meningothelial men- tion. Masson trichrome. (e) Psammomatous meningioma
ingioma with syncytially arranged tumor cells. Masson with abundant calcified psammoma bodies. Masson
trichrome. (c) Fibrous meningioma characterized by a trichrome. (f) Angiomatous meningioma characterized by
strong proliferation of collagenous fibers (blue) in between its predominant blood vessels. Masson trichrome
the meningothelial tumor cells (violet). Masson trichrome.

mesenchymal components such as xanthomatous, hyperplasia of lymph nodes [20]. Clear cell
lipomatous, osseous, or cartilaginous areas, sin- meningiomas show a patternless histology with
gly or in combination (Figs. 3.5, 3.6, and 3.7). rather uniform glycogen-rich cells which, in con-
WHO grade II subtypes comprise the ventional histology, appear “clear” when the gly-
following: cogen is washed out. Interestingly, clear cell
Chordoid meningiomas resemble the histologi- meningiomas have a certain predilection for the
cal features of chordoma. In some cases, chordoid cerebellopontine angle (Fig. 3.6d). Atypical men-
meningioma is associated with Castleman’s dis- ingiomas combine increased mitotic activity of
ease, a benign local or generalized angiofollicular four or more mitoses per ten high-power fields
80 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

Fig. 3.6 (a) Microcystic meningioma. Masson trichrome. ingioma. Note the lymphocytic infiltration (arrow) Masson
(b) Secretory meningioma with pseudopsammoma bodies trichrome. (d) Clear cell meningioma with glycogen-rich
rich in glycogen. PAS. (c) Lymphoplasmacyte-rich men- cytoplasm; the glycogen is washed out by fixation. HE

a b

c d

Fig. 3.7 (a) Xanthomatous metaplasia with foci of xan- Osteoblastic metaplasia with bone formation. Masson
thoma cells. Masson trichrome. (b) Lipoblastic metapla- trichrome. (d) Chondroblastic metaplasia with foci of car-
sia with scattered lipocytes. Masson trichrome. (c) tilage. Masson trichrome
Pathology of the Meninges 81

a b

c d

Fig. 3.8 (a) Chordoid meningioma with strands of tumor ingioma with focal necrosis. HE. (d) Anaplastic
cells in a mucous-rich matrix. HE. (b) Atypical menin- (malignant) meningioma characterized by polymorphous
gioma with increased mitotic rate. HE. (c) Papillary men- cells with increased mitotic rate. HE

with increased cellularity and other signs of his- Hemangiopericytoma


tological pleomorphism. Chordoid, clear cell,
and atypical meningiomas are classified with Age distribution: Relatively rare, mainly occur-
WHO grade II because of a much higher rate of ring in younger adults.
recurrence than WHO grade I meningiomas Sex distribution: Slightly more often in men
(Fig. 3.8a, b). than in women.
WHO grade III subtypes comprise the Epidemiology: 0.4 % of primary CNS tumors.
following: Grading: WHO grade II; anaplastic hemangi-
Papillary meningioma is a variant with a pre- opericytoma WHO grade III.
dominant perivascular pseudopapillary pattern. The meningeal hemangiopericytoma is a
The young patients, including children, suffer highly cellular and vascularized mesenchymal
from local invasion in the brain, recurrences, and tumor almost always attached to the dura.
even metastases mostly to the lung. The rare Histologically, the hemangiopericytoma is com-
rhabdoid meningioma is characterized by plump posed of closely packed tumor cells with a rich
rhabdoid cells with eccentric nuclei and often network of reticulin fibers and with numerous
undergo an aggressive clinical course. The ana- slit-like vascular channels and branching stag-
plastic (malignant) meningioma exhibits malig- horn-type vessels (Fig. 3.9a, b).
nant histological features and a markedly elevated The anaplastic hemangiopericytoma has a
mitotic activity of 20 or more mitoses per 10 high tendency to recur and to metastasize outside
high-power fields (Fig. 3.8c, d). the central nervous system. Histologically, it
82 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

e f

Fig. 3.9 (a) Hemangiopericytoma with high cellularity to its rich lipid content and abundant capillaries.
and jumbled arrangement of spindle cells. Masson (d) Hemangioblastoma forming a typically large central
trichrome. (b) Hemangiopericytoma with abundant reti- cyst (arrow). Nissl. (e) Hemangioblastoma with large
culin fibers and staghorn-type vessel (arrow). Gomori vacuolated stromal cells and vascular cells. Masson
reticulin impregnation. (c) Hemangioblastoma extending trichrome. (f) Hemangioblastoma with accumulation of
into the fourth ventricle and appearing red yellow owing lipid droplets (red) within stromal cells. Oil red

shows nuclear atypia, high mitotic activity, necro- Grading: WHO grade I.
ses, and hemorrhage. The hemangioblastoma is a benign, relatively
rare, highly vascular tumor of angio-mesenchy-
mal origin [40] localized in the cerebellum,
Hemangioblastoma brain stem, or spinal cord. Histologically, the
hemangioblastoma is characterized by two
Age distribution: Adults (sporadic), young adults, components, namely, large, vacuolated stromal
and juveniles (VHL). cells containing lipid droplets and abundant
Sex distribution: No sex predilection. thin-walled small vessels. Therefore, it is also
Epidemiology: No accurate incidence rates referred to as capillary hemangioblastoma
available. (Fig. 3.9c–f).
Pathology of the Meninges 83

In association with von Hippel–Lindau syn- The melanocytoma is a solitary circumscribed


drome (VHL), one or multiple hemangioblastomas tumor deriving from leptomeningeal melano-
occur in significantly younger patients. VHL is an cytes. It does not invade surrounding structures.
autosomal dominant disorder caused by germline The primary leptomeningeal malignant mela-
mutations of the VHL gene on chromosome 3p. noma has to be distinguished from metastatic
Different mutations of the VHL gene are correlated malignant melanoma. It is a highly aggressive
with different phenotypes. In VHL, next to heman- tumor and may metastasize along the CSF path-
gioblastomas of the central nervous system and the ways and to remote organs.
retina, extraneural manifestations such as clear cell Histologically, leptomeningeal melanocytic
renal carcinoma, pheochromocytoma and paragan- lesions are composed of spindle cells or epithe-
gliomas, epididymal cystadenoma, neuroendocrine lioid cells similar to melanocytic tumors arising
tumors and microcystic adenomas of the pancreas, in other sites (Fig. 3.10a, b). It is important to
and endolymphatic sac tumors can be observed. distinguish melanocytic lesions from histogeneti-
Morbidity and mortality for VHL patients is high. cally different tumor entities undergoing
Yearly lifetime screening with familial screening melanization such as schwannoma, medulloblas-
and counseling is recommended. toma, or paraganglioma [6].

Leptomeningeal Melanocytic Lesions Meningiosis and Solid Metastatic


Tumors
Age distribution: Children (melanocytosis and
melanomatosis linked with neurocutaneous mel- Age distribution: Depending from the primary
anosis); all ages with a peak in the fifth decade tumor.
(melanocytoma and malignant melanoma). Sex distribution: Depending from the primary
Sex distribution: No sex predilection (melano- tumor.
cytosis and melanomatosis); male to female ratio Epidemiology: Depending from the primary
of 1:1.5 (melanocytoma). tumor.
Epidemiology: 0.1 % of brain tumors; one per Grading: Malignant.
ten million population per year (melanocytomas); Metastatic tumor cells may diffusely spread
0.5 per ten million population per year (primary into the arachnoid forming meningiosis carcino-
CNS melanoma). matosa, sarcomatosa, or lymphomatosa. A men-
Grading: Benign (melanocytosis, melanocy- ingeal metastatic spread of malignant melanoma
toma); malignant (melanomatosis, malignant is possible, but the differential diagnosis of a dif-
melanoma). fuse benign meningeal melanocytosis or a malig-
Primary melanocytic lesions of the meninges are nant meningeal melanomatosis has to be kept in
rare. They arise from leptomeningeal melanocytes. mind. Rarely a carcinosis of the dura with meta-
Diffuse and circumscribed, benign and malignant, static islands can be observed (Fig. 3.10c, d).
and melanotic and amelanotic variants occur. Of course, all kinds of metastatic tumors
The diffuse melanocytosis involves the spreading from carcinomas, sarcomas, melano-
leptomeninges including those of the skull base mas, or lymphomas can be encountered in all
by a proliferation of leptomeningeal melanocytes parts of the central nervous system including
within the Virchow–Robin spaces but without the cerebellopontine angle and the adjacent
invasion of the central nervous system. It may be osseous skull base. Mainly circumscribed solid
associated with the congenital nevus of Ota [2]. metastases with more or less distinct borders
The malignant change to melanomatosis is char- toward the surrounding brain tissue occur. But
acterized by CNS parenchymal invasion. Both also a continuous tumor growth of different
diffuse leptomeningeal melanocytosis and mela- primary tumors of the surrounding tissues
nomatosis are strongly linked with neurocutane- toward the cerebellopontine angle is possible
ous melanosis, a phakomatosis of childhood. (Fig. 3.10e, f).
84 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

d
c

Fig. 3.10 (a) Melanocytoma with loose nests of slightly of the dura with small islands of carcinoma cells sur-
pigmented spindle cells. HE. (b) Epithelioid malignant rounded by fibrous dural tissue. Masson trichrome. (e)
melanoma with polymorphous tumor cells heavily laden Pons and cerebellum with metastasis of bronchial carci-
with melanin pigment. HE. (c) Thickened and infiltrated noma. (f) Continuous growth of a squamous epithelium
arachnoid in meningiosis lymphomatosa. (d) Carcinosis carcinoma of the paranasal sinus toward the skull base

Acute Bacterial Sive Purulent sub-Saharan Africa; pneumococcal meningitis at


Meningitis the extremes of age.
Sex distribution: More prevalent in males than
Age distribution: Age relation of various bacteria, females.
for example, menigococcal meningitis in chil- Epidemiology: 1–5 per 100,000 population
dren in Europe and North America and adoles- per year in Europe and North America; endemic
cents or young adults in the “meningitis belt” of in the “meningitis belt” of sub-Saharan Africa. H.
Pathology of the Meninges 85

influenzae meningitis due to vaccination virtually allows a reliable morphological diagnosis by


eliminated in developed countries. 0.5–6 % post- pleiocytosis up to 10,000 and more cells/mm3
operative meningitis in neurosurgical patients. and predominance of PMNs (Fig. 3.11).
Pathogens: Streptococcus pneumoniae
(Pneumococcus), Neisseria meningitidis
(Meningococcus), and Haemophilus influenzae Tuberculous Meningitis
cause approximately 80 % of cases, worldwide.
Bacterial meningitis may complicate neurosur- Age distribution: All ages, more indolent course
gical procedures such as craniotomy for the in adults than in children.
resection of tumor and shunting; the most com- Sex distribution: More prevalent in males than
mon bacteria causing postoperative meningitis females.
include Escherichia coli, Klebsiella pneumo- Epidemiology: Rare in developed countries
niae, Pseudomonas aeruginosa, Acinetobacter but still important in developing countries; more
spp., and Staphylococcus aureus. frequent in people with impaired immunity (alco-
Morphological changes are indistinguishable, holism, HIV-AIDS, intravenous drug abuse, long-
irrespective of the bacterial cause, thus term steroid or immunosuppressive therapy) and
unspecific. In hyperacute cases, the Waterhouse– in residents in closed facilities (nursing homes,
Friderichsen syndrome characterized by bilat- chronic care facilities, prisons, refugee camps).
eral adrenal hemorrhage, centrilobular necroses Pathogens: Mycobacterium tuberculosis,
of the liver, and disseminated intravascular Mycobacterium bovis (rare infection acquired by
coagulation may cause death within 24 h. Up to ingestion of contaminated milk).
15 % of patients with meningococcal sepsis Tuberculous meningitis is the most common
develop a Waterhouse–Friderichsen syndrome. form of tuberculosis of the central nervous sys-
In the acute stage, after 2 days or longer, the tem. Macrosopically, a thick pale-green exudate
migration of polymorphonuclear neutrophilic fills the basal cisterns, covers the cerebellopon-
leukocytes (PMNs) into the leptomeninges tine angle and the front of the pons, and enters the
becomes visible in form of a layer of pus cover- Sylvian fissures. Histologically, specific granulo-
ing the vertex and/or the base of the brain. In the mas, the tubercles, consist of central caseous
subacute stage, PMNs become less numerous in necrosis surrounded by epithelioid cells, among
the exudates, and lymphocytes and a few plasma them multinucleated Langhans’ giant cells, and a
cells appear. Lumbar puncture for CSF cytology peripheral ring of lymphocytes. In CSF cytology,

a b

Fig. 3.11 (a) Bilateral adrenal hemorrhage in neonatal perivascular infiltration in a Virchow–Robin space (arrow)
Waterhouse–Friderichsen syndrome. (b) Hyperacute stage by PMNs, no infiltration of the brain cortex. Masson
of neonatal bacterial meningitis with excessive numbers trichrome. (e) Acute stage of bacterial meningitis with
of Streptococcus pneumoniae bacteria in CSF cytology. massive pleocytosis of PMNs in CSF cytology. May–
May–Grünwald–Giemsa. (c) Acute stage of bacterial Grünwald–Giemsa. (f) Subacute stage of bacterial menin-
meningitis with pus layer covering the brain surface. (d) gitis with less numerous PMNs, lymphocytes, and plasma
Massive infiltration of the leptomeninges, discrete cells (arrows) in CSF cytology. May–Grünwald–Giemsa
86 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

c d

e f

Fig. 3.11 (continued)

pleocytosis usually below 500 cells/mm3 and Sarcoid granulomas most often affect the
predominance of transformed lymphocytes are lungs, but virtually any organ can be affected
observed. The identification of acid-fast bacilli is including the CNS. The most frequent manifesta-
diagnostic (Fig. 3.12a–e). tion of neurosarcoidosis is characterized by sar-
coid granulomas in the meninges, in individual
cases also without an evidence of systemic dis-
Sarcoidosis ease. The posterior fossa, the cerebellopontine
angle, and the suprasellar region are preferentially
Age distribution: Young adults with a peak in the involved. Histologically, the sarcoid granulomas
third decade, a second peak in women over 50 have a specific pattern and consist of a central
years of age. mass of epithelioid cells and few multinucleated
Sex distribution: Slightly prevalent in females giant cells surrounded by a ring of lymphocytes.
than males. Central caseous necrosis as seen in tuberculous
Epidemiology: CNS affected in about 5 % of meningitis does not occur (Fig. 3.12f).
cases.
Pathogen: Unknown etiology.
Sarcoidosis (synonym Besnier–Boeck– Aspergillosis
Schaumann disease) is a granulomatous multi-
system disorder with worldwide distribution. Age distribution: All ages.
Sarcoidosis may be oligosymptomatic and com- Sex distribution: No sex predilection.
monly improves or clears up spontaneously. Epidemiology: People with impaired immu-
About 50 % of the patients have relapses, about nity, cerebral infection in 10–20 % of cases with
10 % of the patients develop serious disability. invasive aspergillosis.
Pathology of the Meninges 87

a b

c d

e f

Fig. 3.12 (a) Tuberculous meningitis with pale-greenish lymphocytes as well as Langhans’ giant cells (arrows).
pus layer covering the basal meninges including the cere- HE. (d) Langhans’ giant cell in CSF cytology. May–
bellopontine angles. (b) Tuberculous granuloma of the Grünwald–Giemsa. (e) Acid-fast bacilli of Mycobacterium
basal meninges with central caseous necrosis surrounded tuberculosis. Ziehl–Neelsen. (f) Meningeal sarcoid granu-
by a wall of inflammatory cells (arrow). Elastica-van loma with a central mass of epithelioid cells surrounded
Gieson. (c) Detail of a tuberculous granuloma with central by lymphocytes. There is no caseous necrosis. Masson
caseous necrosis and the wall of epithelioid cells and trichrome

Pathogens: Aspergillus fumigatus (majority of Aspergillus ssp., infections caused by the fungus
cases), Aspergillus flavus, Aspergillus terreus, rarely occur in people with a competent immune
Aspergillus niger. system.
The fungus Aspergillus ssp. is commonly Aspergillosis is a typical example of a mold-
found growing on decaying vegetation such as like fungal infection. Brain infection usually
dead leaves, stored grain or compost piles. manifests in form of granulomatous fungal
Although most people are frequently exposed to abscesses surrounded by fungal hyphae
88 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

e f

Fig. 3.13 (a) Aspergilloma with central necrosis and Cryptococcus yeasts with clear capsule as example of
radiated fungal hyphae arranged in a starburst pattern. HE. typical yeastlike mycosis. HE. (e) Multiple cysticercosis
(b) Close-up of septate aspergillus hyphae within necrosis cysts at the brain base including the cerebellopontine
as example of typical mold like mycosis. PAS. (c) angle (arrows). (f) Multiple proto-scolices in a hydatid
Meningeal cryptococcus granuloma (arrows). HE. (d) cyst in echinococcosis. Masson trichrome

arranged in a starburst pattern. In rare cases, a countries, and 15–45 % of HIV-AIDS patients in
subdural manifestation may be observed sub-Saharan Africa have cryptococcal CNS
(Fig. 3.13a, b). infection.
Pathogens: Cryptococcus neoformans (majority
of cases), Cryptococcus gattii, Cryptococcus
Cryptococcosis grubii.
The fungus Cryptococcus ssp. is commonly
Age distribution: All ages. found in the soil and the feces of birds.
Sex distribution: No sex predilection. Cryptococcosis (synonym torulosis) is a typi-
Epidemiology: People with impaired immu- cal example for a yeastlike fungal infection.
nity; 5–10 % of HIV-AIDS patients in developed CNS infection manifests as meningitis or
Pathology of the Osseous Skull Base 89

meningoencephalitis. Macroscopically, the lep- Echinococcosis


tomeninges appear as granular milky texture.
An infiltration of the brain parenchyma along Age distribution: All ages.
the Virchow–Robin spaces is possible. Sex distribution: No sex predilection.
Histologically, the fungus forms specific Epidemiology: Echinococcus granulosus
budding, round or oval yeasts surrounded by a endemic worldwide, more common in the Middle
broad mucoid polysaccharide capsule East and Eastern Africa; Echinococcus multiloc-
(Fig. 3.13c, d). ularis restricted to Central Europe, Russia,
Turkey, China, and North America; Echinococcus
vogeli and Echinococcus oligarthus are rare spe-
Cysticercosis cies limited to Central and South America.
Pathogens: Dog tapeworm Echinococcus
Age distribution: All ages. granulosus (classic hydatid disease), fox tape-
Sex distribution: No sex predilection. worm Echinococcus multilocularis (alveolar
Epidemiology: Sporadic in Europe, more com- hydatid disease), Echinococcus vogeli,
mon in the tropics, the highest rates being in Latin Echinococcus oligarthus (the two latter causing
America, prevalence of neurocysticercosis up to polycystic echinococcosis).
3–4 %. In Mexico, Brazil, and India, the most Dogs and other canines are definitive hosts of
common space-occupying brain lesion. Echinococcus granulosus. Humans as intermedi-
Pathogen: Cysticerci are the larvae of the pig ate hosts acquire Echinococcus granulosus clas-
tapeworm Taenia solium. sic hydatid disease by ingesting food contaminated
During the past 30 years, neurocysticercosis by canine feces containing eggs of the tapeworm.
has been increasingly recognized as a major The eggs hatch in the duodenum into oncospheres
cause of neurologic disease worldwide and an that burrow into the gut wall and reach the liver.
important problem among immigrant populations The oncospheres grow into the hydatid cysts that
in the United States. may spread to other organs including the brain.
Cysticercosis is the most common of all para- Macroscopically, cysts in the cerebrum or cere-
sitic infections to cause CNS disease. The human bellum can reach a diameter of up to 10 cm, con-
normally is the definitive host in which the pig sisting of one cyst or several cysts. Histologically,
tapeworm develops to maturity producing eggs within the cysts, 200 mm-long proto-scolices with
which are passed out in the feces. Excreted eggs four spherical suckers and two rows of hooklets
of the tapeworm can therefore be found in fecally can be observed (Fig. 3.13f).
contaminated water or food as well as on the The fox is the definitive host of Echinococcus
hands of the infected person. In humans as inter- multilocularis. Cerebral infections are rare and
mediate hosts, autoinfection occurs most fre- present as multiseptate, partly solid, partly cystic
quently by hand-to-mouth infection with the space-occupying lesion mimicking a cerebral
person’s own feces. Excreted eggs are ingested tumor because the cyst wall penetrates into the
by humans and hatch in the gut into oncospheres. surrounding brain parenchyma.
The oncospheres burrow into the gut wall and are
disseminated by blood vessel to any organ includ-
ing brain and meninges. There they develop into Pathology of the Osseous Skull Base
cystic larvae, the cysticerci.
Macroscopically, the cysticerci appear as Fibrous dysplasia
Aneurysmal bone cyst
fluid-filled bladders of 0.5–2 cm in diameter;
Osteochondroma
their number can vary from a few to hundreds
Chondroblastoma
(Fig. 3.13e). Cysts may degenerate and resolve
Chondrosarcoma
into calcified nodules. Histologically, the cysts
Chordoma
contain one proto-scolex, the head of the future Endolymphatic sac tumor
tapeworm.
90 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

Fig. 3.14 (a) Fibrous dysplasia with C-shaped bony type giant cells (dark violet). Goldner. (c) Chondroblas-
spicules (red) within a hypocellular fibrous stroma. toma with well-defined cytoplasmic borders of the tumor
Masson trichrome. (b) Aneurysmal bone cyst with cystic cells. Some nuclei display indentations (arrows). HE.
spaces separated by septa of connective tissue, reactive (d) Chondrosarcoma with ill-defined cytoplasmic borders
woven bone (turquois), and multinucleated osteoclast- of the tumor cells. HE

Fibrous Dysplasia one bone or polyostotic (PFD) involving several


bones. The tympanic, mastoid, squamous, or
Age distribution: Children and young adults. petrous temporal bones may be involved.
Sex distribution: There is no general sex predi- Unusual sites include the internal auditory canal,
lection in monostotic fibrous dysplasia (MFD); the lateral semicircular canal, and the ossicles.
however, with regard to the skull base manifesta- PFD may manifest in infants in the setting of
tion, there is a predilection for males. To the con- McCune–Albright syndrome caused by muta-
trary for polyostotic fibrous dysplasia (PFD) male tions in the GNAS1 gene and associated with
to female ratio of 1:3. café-au-lait spots and hyperfunctioning
Epidemiology: one with FD per 15,000–30,000 endocrinopathies.
population per year. MFD six times more common The bone shows local grayish and firm
than PFD. About 45 % of FD arise in the craniofa- expansions. Histologically, the bone marrow is
cial region (skull, mandible, facial bones). replaced by an intramedullary proliferation of
Grading: Benign; malignant transformation trabecular woven bone admixed with fibrous
may occur in McCune–Albright syndrome, but tissue, and typical C-shaped bony spicules
rarely. within the fibrous matrix. The lesion arises
Fibrous dysplasia is a medullary fibro-osseous abruptly from a bland spindle cell stroma
lesion and may be monostotic (MFD) involving (Fig. 3.14a).
Pathology of the Osseous Skull Base 91

Aneurysmal Bone Cyst Grading: Benign.


Within the craniofacial region, the temporal
Age distribution: All age groups, most common bone is most frequently affected. Macroscopically,
in the first two decades with a median age of a whitish, partly cystic tumor tissue is seen.
approximately 13 years. Histologically, sheets of uniform round to polyg-
Sex distribution: No sex predilection. onal chondroblasts with well-defined cytoplasmic
Epidemiology: 0.15 per million population per borders are characteristic. The round
year. nuclei frequently display grooves and indenta-
Grading: Benign, potentially locally recurrent. tions (Fig. 3.14c).
Though aneurysmal bone cysts usually arise
in the metaphysis of long bones, any bone includ-
ing the skull base can be affected. The well-cir- Chondrosarcoma
cumscribed lesion is composed of blood-filled
cystic spaces separated by septa of connective Age distribution: Adults with a peak in the fifth to
tissue, reactive woven bone, and multinucleated seventh decades.
osteoclast-type giant cells (Fig. 3.14b). Sex distribution: Slight male predominance.
Approximately 70 % of the cystic lesions arise Epidemiology: Extremely rare in craniofacial
de novo, thus forming primary aneurysmal bone bones.
cysts. Secondary aneurysmal bone cysts develop Grading: Malignant; grade 1, few cells with
most commonly in association with benign bone no variation in size and shape; grade 2, hypercel-
tumors, for example, giant cell tumor, osteoblas- lularity with variation in size and shape; grade 3,
toma, chondroblastoma, and fibrous dysplasia, high cellularity with prominent pleomorphic
but may also complicate malignant bone tumors appearance, atypia, and mitoses.
that have undergone hemorrhagic cystic change, Chondrosarcomas display a pure hyaline car-
especially osteosarcoma. tilage differentiation. Macroscopically, they have
a translucent, blue-gray or white color and often
a lobular growth pattern. Histologically, the carti-
Osteochondroma lagineous tumor cells with ill-defined borders
permeate between preexisting bony trabeculae.
Age distribution: Juvenile and young adults. Myxoid changes, calcification, or ossification
Sex distribution: No sex predilection. may be present (Fig. 3.14d). Secondary chondro-
Epidemiology: Osteochondromas of the skull sarcoma may arise in a benign precursor, for
base are rare [15, 36, 39]. example, an osteochondroma.
Grading: Benign.
Osteochondroma is a cartilage-capped bony
projection arising on the external surface of bone Chordoma
containing a marrow cavity that is continuous
with that of the underlying bone. Usually osteo- Age distribution: Adults with a peak in the sixth
chondromas are well demarcated. Histologically, decade.
an outer layer of fibrous perichondrium underly- Sex distribution: Male to female ratio of 1.8:1.
ing cartilage and bone can be differentiated. Epidemiology: 1–4 % of primary malignant
bone tumors.
Grading: Intermediate.
Chondroblastoma Small remnants of the embryonic notochord
may be found in form of the ecchordosis physali-
Age distribution: Patients with skull and temporal phora. Chordomas are bone tumors that recapitu-
bone involvement present at 40–50 years of age. late notochord. With regard to the skull base, they
Sex distribution: Male predominance. are located in the spheno-occipital region, typi-
Epidemiology: Less than 1 % of bone tumors. cally beginning in the clivus. A small proportion
92 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

e f

Fig. 3.15 (a) Ecchordosis physaliphora (arrow). (b) from the clivus. (e) Chordoma lobule with cords of physa-
Ecchordosis physaliphora. Masson trichrome. (c) liphorous tumor cells in a myxoid matrix. HE. (f)
Chordoma masses protruding in the skull base. (d) Chordoma exhibiting a strong positivity of cytokeratin
Expansively growing chordoma of the skull base arising within the tumor cells. Pancytokeratin AE1/3

may involve the nasopharynx and/or paranasal toid chondroma with cell-dense sarcomatoid
sinuses. They extend into the surrounding struc- areas are rare variants.
tures and may often form large tumor masses.
Metastases may occur. Histologically, chordomas
are lobulated tumors with tumor cells arranged in Endolymphatic Sac Tumor
sheets or cords within an abundant myxoid
stroma. The classic physaliphorous tumor cells Age distribution: Adults with a peak at 50 years
have a pale vacuolated cytoplasm (Fig. 3.15). of age without von Hippel–Lindau disease
The chondroid chordoma with areas mimick- (VHL), and with a peak of 30 years of age with
ing cartilage and the “dedifferentiated” sarcoma- VHL.
Pathology of the Sellar Region 93

Sex distribution: No sex predilection without Pituitary Adenoma


VHL; male to female ratio of 1:2 with VHL.
Epidemiology: Extremely rare in general pop- Age distribution: Adults; growth hormone-con-
ulation; prevalence in VHL 1–10 %, VHL occurs taining tumors causing gigantism in children.
at a rate of about 1:40 000 population. Sex distribution: No sex predilection; ACTH-
Grading: Intermediate; slow growing, locally containing adenomas causing Cushing’s disease
invasive but not metastasizing; a differential more often in women than men with a male to
grading system was proposed by Bambakidis female ratio of 1:2.3.
et al. [3]. Epidemiology: 15–25 % of intracranial tumors;
Endolymphatic sac tumors (synonyms: cere- 1 per 1,000 adult population per year.
bellopontine angle ceruminoma, Heffner tumor, Grading: Benign.
aggressive papillary middle ear adenoma, and Pituitary adenomas derive from adenohypo-
low-grade adenocarcinoma of endolymphatic sac physeal cells and are the most common tumors of
origin) are destructive, papillary hypervascular the adenohypophysis. Their prevalence increases
lesions that arise from the temporal bone retro- with advancing age. Although classified as
labyrinthine region. At an early stage, the tumor benign, these lesions can be highly invasive and
is located within the endolymphatic sac, at a later may cause major complications due to infiltration
stage, it destroys much of the petrous bone and into functionally delicate neighboring structures.
involves the cerebellopontine angle. Most endo- Also metabolic effects of hormone excess syn-
lymphatic sac tumors occur sporadically, rarely dromes can occur. A minority of patients with
bilaterally. In association with von Hippel–Lindau large tumors may have acute bleeding into the
disease (VHL), the tumors are more frequently tumor (pituitary apoplexy), leading to an abrupt
bilateral and less advanced as opposed to the non- increase in size and causing the sudden onset of
VHL patients [24]. Mutations in the VHL gene headache, visual loss, double vision, and/or pitu-
have been demonstrated in endolymphatic sac itary failure. Multiple endocrine neoplasia type 1
tumor. For more information on VHL, see above (MEN 1) is a rare condition characterized by
in the paragraph on hemangioblastoma. simultaneous tumors of the pituitary, pancreas,
Histologically, endolymphatic sac tumors and parathyroid glands. Pituitary adenomas
show a papillary adenomatous architecture, the develop in 25 % of patients with MEN 1.
papillary processes infiltrating the surrounding In rare cases, ectopic pituitary adenomas can
connective tissue and bone. The tumor may con- arise in embryonic remnants of Rathke’s cleft,
tain areas of hemorrhage, hemosiderin, and cho- that is, the rare ectopic pharyngeal pituitary gland
lesterol clefts with scattered inflammatory giant (Fig. 3.16a, b). They may be found in the parapha-
cell reactions. Dilated glands may contain secre- ryngeal area, sphenoid sinus, middle nasal
tion, which resembles colloid. meatus, petrous temporal bone, clivus, hypothal-
amus, and third ventricle.
Whereas microadenomas (less than 1 cm in
diameter) are still growing within the intra-
Pathology of the Sellar Region sellar adenohypohysis, macroadenomas (larger
than 1 cm in diameter) grow upward, the tumor
Pituitary adenoma can elevate and compress the optic chiasm
Pituitary carcinoma
(Fig. 3.16c, d).
Spindle cell oncocytoma of the adenohypophysis
Histologically, the traditional classification of
Pituicytoma
chromophobic, acidophilic, and basophilic ade-
Granular cell tumor of the neurohypophysis
nomas has largely been abandoned or at least has
Craniopharyngioma
Adamantinomatous craniopharyngioma
to be completed by an immunohistochemical
Papillary craniopharyngioma (Xanthogranuloma of classification based on hormone content. For a
the sellar region) reliable morphological determination of the
94 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

Fig. 3.16 (a) Ectopic pharyngeal pituitary gland (arrow) growth. (d) Circumscribed pituitary microadenoma
HE. (b) Detail of the ectopic pharyngeal pituitary gland. surrounded by non-tumorous pituitary tissue contained to
HE. (c) Pituitary macroadenoma with infra- and suprasellar the sella turcica. Elastica-van Gieson

hormone excretion, transmission electron causing thyroid-stimulating hormone (TSH) excess


microscopy is indispensable. The ultrastructure are thyrotroph adenomas, adenomas causing adre-
reveals densely granulated adenomas, thus, ade- nocorticotroph hormone (ACTH) excess comprise
nomas with a high number of hormone granules densely or sparsely granulated corticotroph ade-
within the cytoplasm, and after enhanced extru- nomas, and finally adenomas causing gonadotropin
sion of hormone granules, sparsely granulated (FSH/LH) excess are gonadotroph adenomas.
adenomas (Fig. 3.17), the latter often negative in Plurihormonal adenomas, especially GH- and
immunohistochemical staining. PRL-containing or GH-, PRL-, and TSH-containing
The most effective way is the use of a clinico- adenomas, may be observed. PRL-containing ade-
pathological classification in functioning and nomas (prolactinomas) are the most common type
nonfunctioning adenomas [11] using both mor- followed by GH- and ACTH-containing adenomas.
phological and clinical features (Fig. 3.18). Thyrotroph and gonadotroph adenomas are rare.
Functioning adenomas with growth hormone Nonfunctioning adenomas account for approx-
(GH) excess comprise densely or sparsely imately one-third of all pituitary adenomas. They
granulated somatotroph adenomas and mammoso- may be extremely sparsely granulated silent soma-
matotroph adenomas, adenomas causing hyperpro- totroph, lactotroph, thyrotroph, corticotroph
lactinemia (PRL) comprise lactotroph adenomas (except a subtype of silent but densely granulated
and acidophilic stem cell adenomas, adenomas corticotroph adenoma), or gonadotroph adenomas
Pathology of the Sellar Region 95

a b

c d

e f

Fig. 3.17 (a) Non-tumorous pituitary gland. Masson sparsely granulated (arrow) somatotroph adenoma.
trichrome. (b) Chromophobic adenoma. Masson trichrome. Transmission electron microscopy. (f) Nonfunctioning
(c) Eosinophilic adenoma. Masson trichrome. (d) extremely sparsely granulated “silent” somatotroph ade-
Basophilic adenoma. Masson trichrome. (e) Functioning noma. Transmission electron microscopy

on the one hand or true null cell adenomas on the Epidemiology: 0.2 % of pituitary tumors.
other hand. Oncocytomas represent silent gonado- Grading: Malignant.
troph adenomas with extensive oncocytic change. Pituitary carcinomas are rare and character-
ized by marked polymorphism and mitoses. The
majority of carcinomas are secretory, usually
Pituitary Carcinoma arising from corticotroph tumors or prolactino-
mas, but all histological types and secretory pat-
Age distribution: Adults. terns are represented [19]. They have to be
Sex distribution: No sex predilection. differentiated from polymorphous, in some cases
96 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

Fig. 3.18 (a) Border of a chromophobic adenoma, to the to growth hormone. (c) Border of an eosinophilic adenoma,
left compressed preexistent non-tumorous pituitary tissue to the left preexistent non-tumorous pituitary tissue. Masson
with light-blue proliferated connective tissue fibers. Masson trichrome. (d) The same case as in c with growth hormone
trichrome. (b) The same case as in a with no hormone expression within the adenoma (somatotroph adenoma)
expression within the adenoma (null cell adenoma) and and growth hormone expression within the scattered preex-
growth hormone expression within the scattered preexistent istent eosinophilic cells. Immunohistochemistry with anti-
eosinophilic cells. Immunohistochemistry with antibodies bodies to growth hormone

calcified benign adenomas with regressive adenomas. Histologically, the tumor is composed
changes. Pituitary carcinomas can locally invade of interlacing fascicles of eosinophilic epithelioid
surrounding tissue or spread through the venous cells with oncocytic cytoplasm.
system and the cerebrospinal fluid pathways
forming distant metastases (Fig. 3.19a, b).
Pituicytoma

Spindle Cell Oncocytoma Age distribution: Adults in the fifth to seventh


of the Adenohypophysis decades.
Sex distribution: Male to female ratio of 1.6:1.
Age distribution: Adults (mean 56 years). Epidemiology: Less than 30 cases described.
Sex distribution: No sex predilection. Grading: WHO grade I.
Epidemiology: No accurate incidence rates The pituicytoma is an extremely rare, circum-
available; 0.4 % of sellar tumors. scribed, and solid tumor that originates in the neu-
Grading: WHO grade I. rohypophysis or infundibulum [7]. Histologically,
The spindle cell oncocytoma of the adenohy- the glial tumor consists of elongate spindle cells
pophysis is a rare non-endocrine tumor that mac- arranged in interlacing fascicles or in a storiform
roscopically is indistinguishable from pituitary pattern.
Pathology of the Sellar Region 97

a b

c d

Fig. 3.19 (a) Regressive pituitary adenoma with focal neurohypophysis. HE. (d) Granular cell tumor of the
calcifications (arrow). HE. (b) Pituitary carcinoma with neurohypophysis showing the granular eosinophilic
extensive cellular polymorphism. HE. (c) Nodular cytoplasm. HE
arrangement of a granular cell tumor of the

Granular Cell Tumor Craniopharyngioma


of the Neurohypophysis
Age distribution: Adults above 40 years of age; a
Age distribution: Adults; exceptionally in second peak in children (adamantinomatous
children. craniopharyngioma).
Sex distribution: Male to female ratio of 1:2. Sex distribution: No sex predilection.
Epidemiology: No accurate incidence rates Epidemiology: 1–5 % of intracranial tumors;
available. in children 5–10 % of intracranial tumors. 0.5–
Grading: WHO grade I. 2.5 per million population per year;
The granular cell tumor is a rare tumor that more frequent in Japanese children and in
arises from the neurohypophysis or infundibu- Nigerian.
lum. In the literature, synonyms such as Grading: WHO grade I.
Abrikossoff tumor, choristoma, granular cell Craniopharyngiomas presumably derive
myoblastoma, and granular cell neuroma can be from remnants of Rathke’s pouch epithelium
found. The usually lobulated and well-circum- and the craniopharyngeal duct. They have supra-
scribed tumor occurs in a sellar and suprasellar sellar localization with a minor intrasellar
anatomic location. Histologically, the tumor cells component.
are arranged in a nodular pattern and are charac- The adamantinomatous craniopharyngioma
terized by an abundant granular eosinophilic is characterized by strands and lobules of
cytoplasm (Fig. 3.19c, d). squamous epithelium bordered by tumor cell
98 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

e f

Fig. 3.20 (a) Well-circumscribed cystic craniopharyn- tin” representing remnants of pale nuclei embedded within
gioma expanding into the third ventricle. (b) Poorly a keratinous mass. Masson trichrome. (e) Calcified xan-
demarcated craniophyaryngioma invading the third ven- thogranuloma of the sellar region. (f) Xanthogranuloma
tricle and the basal ganglia. (c) Adamantinous craniophar- with hemosiderin deposits (brownish green) and choles-
yngioma with squamous epithelium bordered by cell terol clefts surrounded by some multinucleated giant cells.
palisades. Masson trichrome. (d) Nodules of “wet kera- Masson trichrome

palisades. Nodules of “wet keratin” are typical. Xanthogranuloma of the Sellar Region
Cystic degeneration and calcifications may be
encountered. The papillary craniopharyngioma Age distribution: All ages.
is composed of papillae of squamous epithelium Sex distribution: No sex predilection.
lacking surface maturation. Calcifications are Epidemiology: No accurate incidence rates
absent (Fig. 3.20a–d). available.
Pathology of Regional Glial Tumors 99

Grading: Benign. The pilocytic astrocytoma is a relatively cir-


The xanthogranuloma of the sellar region is a cumscribed, often cystic tumor occurring in
rare tumor, not yet fully defined as a separate children and young adults. Preferred sites at the
entity [18]. Transitional cases to craniopharyn- skull base include the optic nerve and the chias-
gioma occur. Histologically, xanthogranulomas matic and hypothalamic region as well as brain
show xanthoma cells, hemosiderin deposits, cho- stem and cerebellum. There is no sex predilec-
lesterol clefts occasionally surrounded by multi- tion. Histologically, the tumor exhibits an often
nucleated giant cells, and lymphocytic infiltrates biphasic pattern with proportions of bipolar cells
(Fig. 3.20e, f). with Rosenthal fibers on the one hand and loose-
textured multipolar cells with microcysts and
eosinophilic granular bodies on the other hand
Pathology of Regional Glial Tumors (Fig. 3.21a, b).

Pilocytic astrocytoma
Pilomyxoid astrocytoma
Pilomyxoid Astrocytoma
Ependymoma
Papillary
Age distribution: Very young children (mean 10
Clear cell
months); rarely in older children.
Tanycytic
Cellular
Sex distribution: No sex predilection.
Anaplastic Epidemiology: No accurate incidence rates
available.
Grading: WHO grade II.
The pilomyxoid astrocytoma is closely related
Pilocytic Astrocytoma to pilocytic astrocytoma. The chiasmatic and
hypothalamic region is the most common loca-
Age distribution: First two decades; rare in tion. Histologically, the tumor is characterized
adults. by a markedly myxoid matrix. The bipolar tumor
Sex distribution: No sex predilection. cells show a predominantly angiocentric arrange-
Epidemiology: 5–6 % of gliomas; 0.8 per ment around blood vessels (Fig. 3.21c, d). Due to
100,000 population per year. the slow tumor growth, regressive changes can
Grading: WHO grade I. be seen.

a b

Fig. 3.21 (a) Pilocytic astrocytoma of the optic chiasm. prominent myxoid matrix. (d) Pilomyxoid astrocytoma
(b) Pilocytic astrocytoma with bipolar hairlike piloid cells with tumor cells in a myxoid matrix. Masson trichrome.
showing numerous red Rosenthal fibers. Masson (e) Ependymoma of the fourth ventricle. (f) Ependymoma
trichrome. (c) Large pilomyxoid astrocytoma with with typical perivascular pseudorosettes. HE
100 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

c d

e f

Fig. 3.21 (continued)

in fascicles, the latter variant is most common in


Ependymoma the spinal cord.
Variants with a semibenign or semimalignant
Age distribution: Infratentorial ependymomas in behavior comprise the cellular ependymoma with
children with a mean age of 6.4 years. a conspicuous increased cellularity, and the ana-
Sex distribution: No sex predilection. plastic ependymoma, which is relatively frequent
Epidemiology: 6–12 % of intracranial tumors in children, particularly in the posterior fossa
in children; up to 30 % of intracranial tumors in location.
children younger than 3 years of age; 0.2–0.3 per
100,000 population per year.
Grading: WHO grade I; cellular ependymoma Pathology of the Choroid Plexus
WHO grade II; anaplastic ependymoma WHO of the Fourth Ventricle
grade III.
Ependymomas originate from ependymal Choroid plexus papilloma
Atypical choroid plexus papilloma
cells and are well demarcated. Histologically, the
Choroid plexus carcinoma
momomorphic tumor cells have round to oval
nuclei. Typical features are perivascular pseudor-
osettes with tumor cells arranged radially around
blood vessels with perivascular anuclear zones, Choroid Plexus Papilloma
and ependymal rosettes (Fig. 3.21e, f). Variants
comprise the papillary ependymoma forming Age distribution: Children and young adults
pseudopapillae, the clear cell ependymoma with (mean 22.5 years).
an oligodendroglia-like appearance, and the Sex distribution: Male to female ratio of 1.5:1
tanycytic ependymoma with tumor cells arranged (fourth ventricle).
Pathology of the Hematopoietic System 101

a b

c d

Fig. 3.22 (a) Circumscribed cauliflower-like plexus papil- trichrome. (c) Plexus carcinoma of the cerebellopontine
loma of the cerebellopontine angle. (b) Papillomatous pattern angle with necrotic areas. (d) Papillomatous pattern of poly-
of monomorphous cuboidal tumor cells cover a connective morphous tumor cells with distinct nuclear polymorphism
tissue matrix (blue) with blood vessels (red). Masson cover a connective tissue matrix with blood vessels. HE

Epidemiology: 10–20 % of brain tumors in Choroid Plexus Carcinoma


children in the first year of life, 2–4 % of brain
tumors in children under 15 years. 0.3 per one Age distribution: Children and young adults.
million population per year. Sex distribution: Male to female ratio of 1.5:1
Grading: WHO grade I; WHO grade II (atypi- (fourth ventricle).
cal choroid plexus papilloma, about 15 % of Epidemiology: Less than 20 % of choroid
choroid plexus papillomas). plexus tumors.
Relevant for the cerebellopontine angle are the Grading: WHO grade III.
choroid plexus papillomas of the fourth ventricle, Choroid plexus carcinomas exhibit all signs of
which comprise about 40 % of this entity (50 % malignancy such as frequent mitoses of five or
lateral ventricles, 5 % third ventricle, 5 % more more per HPF, increased cellular density, nuclear
than one ventricle). However, a primary manifesta- polymorphism, necrotic areas, and also often a
tion in the cerebellopontine angle near the foramina diffuse invasion into the neighboring brain tissue
of the fourth ventricle is uncommon. Circumscribed (Fig. 3.22c, d).
cauliflower-like tumors are well delineated from
brain tissue. Histologically, a single layer of cuboi-
dal cells cover a connective tissue matrix. Mitotic Pathology of the Hematopoietic
activity is extremely low (Fig. 3.22a, b). System
Atypical choroid plexus papillomas show an
increased mitotic activity of two or more mitoses Primary CNS lymphomas
Langerhans cell histiocytosis
per high-power field (HPF), increased cellularity,
Non-Langerhans cell histiocytosis
and nuclear polymorphism.
102 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

Primary CNS Lymphomas the WHO classification respecting the Revised


European–American Lymphoma (REAL)
Age distribution: All ages with a peak in immu- classification [16]. B-cell non-Hodgkin lympho-
nocompetent patients in the sixth and seventh mas constitute up to 98 % of PCNSL, most of
decades of life. In cases of immunodeficiency, them being diffuse large B-cell lymphomas. A
young patients may be involved. primary CNS presentation of Hodgkin’s disease
Sex distribution: Male to female ratio of 3:2. is rare [10].
Epidemiology: 1–7 % of primary intracranial
tumors; incidence highly increased in AIDS
patients who develop in 2–12 % primary CNS Langerhans Cell Histiocytosis
lymphomas (PCNSL).
Grading: Malignant. Age distribution: Children under 15 years of age
Lymphoma is most often encountered in the (mean 12 years).
central nervous system as secondary spread from Sex distribution: No sex predilection.
another site. Extra-nodal malignant lymphomas Epidemiology: 0.5 per 100,000 children per
arising primarily in the central nervous system year.
also affecting the skull base or occurring as pri- Grading: Benign (unifocal); intermediate to
mary leptomeningeal lymphomas must be differ- malignant (multifocal).
entiated from secondary involvement. However, In 1997, the WHO Committee on Histiocytic/
given the absence of lymphoid tissue in the CNS, Reticulum Cell Proliferations and the
it remains possible that the PCNSL is rather a Reclassification Working Group of the
systemic lymphoma with a restricted tropism for Histiocyte Society proposed a new classification.
the CNS. Approximately 13 % of PCNSL involve With regard to skull base manifestations
the posterior fossa. A secondary leptomeningeal Langerhans cell histiocytosis (LCH) was previ-
spread in form of meningiosis lymphomatosa is ously referred to as histiocytosis X embracing
observed in 30–40 % of PCNSL. In many cases, skull and brain manifestations such as eosino-
a diagnosis by means of CSF cytology is possible philic granuloma and Hand–Schüller–Christian
(Fig. 3.23a, b). Surgery should be restricted to disease.
stereotactic biopsy. The most common form is a solitary unifocal
Histologically as in other anatomic locations, (monostotic) osteolytic lesion of the skull in form
malignant lymphomas are classified according to of an eosinophilic granuloma. The cerebral LCH

a b

Fig. 3.23 (a) CSF cytology of a malignant non-Hodgkin dif- (c) Eosinophilic granuloma of the skull as unifocal LCH
fuse large B-cell lymphoma. Note the large nuclei and the manifestation. (d) Hypothalamic LCH manifestation com-
numerous mitoses. May–Grünwald–Giemsa. (b) CSF cytol- pressing the optic chiasm. (e) LCH composed of plasma
ogy of a diffuse large B-cell lymphoma with immunocy- cells, lymphocytic, eosinophilic, and Langerhans histiocytic
tochemically positive B-cell marker (red). CD20, APAAP. tumor cells. HE. (f) Multinucleated Touton giant cell. HE
Pathology of the Hematopoietic System 103

c d

e f

Fig. 3.23 (continued)

with principal involvement of the hypothalamus Epidemiology: one per one million children
and posterior pituitary leads to diabetes insipidus per year.
often associated with signs of hypothalamic dys- Grading: Benign to malignant.
function (obesity, hypogonadism, growth retar- Non-LCH variants such as Rosai–Dorfman
dation), optic atrophy with visual field defects, disease, Erdheim–Chester disease, and juve-
and raised intracranial pressure. Multifocal (poly- nile xanthogranuloma are characterized by
ostotic) lesions of the bone with hypothalamic macrophage differentiation. Histologically,
involvement have been referred to as Hand– emperipolesis is typical, that is, well-preserved
Schüller–Christian disease. While unifocal LCH lymphocytes and plasma cells taken in the cyto-
surgically can be removed without recurrence or plasm of macrophages.
even spontaneously recover, multifocal manifes- Intracranial Rosai–Dorfman disease in adults
tations have a poor outcome with a mortality rate shows dural-based solitary or multiple masses.
up to 20 %. Intrasellar lesions and intracranial extension from
Histologically, tumor cell infiltrates are com- an orbital mass or from the nasal cavity and para-
posed of Langerhans cells, macrophages, lym- nasal sinuses may occur.
phocytes, and plasma cells. A variable fraction of Intracranial Erdheim–Chester disease in adults
eosinophils and Touton giant cells may occur may involve cerebellum, cerebellopontine angle,
(Fig. 3.23c–d). choroid plexus, pituitary, meninges, and orbit.
An extracerebral subdural manifestation of
Erdheim–Chester disease associated with a giant
Non-Langerhans Cell Histiocytosis adenoma of the pituitary was reported [42].
Intracranial juvenile xanthogranuloma in
Age distribution: Children under 15 years of age. young children may arise in the brain or the
Sex distribution: No sex predilection. meninges with or without cutaneous lesions.
104 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

Pathology of Germ Cells teratomas benign, immature teratomas intermedi-


ate, teratomas with malignant transformation
Germ cell tumors malignant.
Germinoma
In human embryos, the primordial germ cells
Embryonal carcinoma
are produced in the fetal yolk sac and migrate
Yolk sac tumor
along the midline to the gonadal folds, thereby
Choriocarcinoma
passing the central nervous, mediastinal, and ret-
Teratoma
Mature teratoma
roperitoneal regions [44]. Along this way, scat-
Immature teratoma tered primordial germ cells in different stages of
Teratoma with malignant transformation differentiation can lead to dysontogenetic tumors
Mixed germ cell tumor or tumorlike lesions.
Tumorlike dysontogenetic cysts In principle, the same undifferentiated or
Epidermoid cyst poorly differentiated primary germ cell tumors
Dermoid cyst can be encountered in the brain as, for example,
in the gonads. The primordial germ cells them-
selves give rise to germinoma (identical with
Germ Cell Tumors seminomas of the testicles and dysgerminomas
of the ovaries). Cells with early embryonic dif-
Age distribution: Children and young adults with ferentiation give rise to embryonal carcinoma,
a peak in the second decade. whereas extraembryonal cells deriving from the
Sex distribution: Strong male predilection, yolk sac or the placenta may develop to yolk sac
especially in the pineal region; certain female tumor (syn. endodermal sinus tumor) or chorio-
predilection in the suprasellar region. carcinoma, respectively. Mixed tumors, for
Epidemiology: In Far East Asia 2–3 % of example, germinoma with placenta-derived syn-
intracranial tumors, 8–15 % of intracranial cytiophoblastic giant cells, may be attributed to a
tumors in children, 0.17 per 100,000 population differentiation of the single totipotential germ
per year; in Western countries 0.3–0.6 % of cell into different cell lines. All these tumors are
intracranial tumors; 0.09 per 100,000 population highly malignant. They are rare but may be
per year. encountered along the midline of the brain and
Grading: Primordial, embryonal, extraembry- the pineal region and also extend to the skull base
onal, and mixed tumors malignant; mature (Fig. 3.24).

a b

Fig. 3.24 (a) Germinoma with biphasic appearance of HE. (d) Choriocarcinoma with predominantly placenta-
large tumor cells and small lymphocytes due to a typical derived cytotrophoblastic cells. HE. (e) Yolk sac tumor with
inflammatory reaction. HE. (b) Germinoma mixed with reticular pattern in form of an allantois-like meshwork. HE.
placenta-derived syncytiotrophoblastic giant cells. HE. (c) (f) Yolk sac tumor with endodermal sinus pattern (Schiller–
Embryonal carcinoma with cohesive group of tumor cells. Duval body) forming a distinctive cell wreath (arrow). HE
Pathology of Blood Vessels and Circulation 105

c d

e f

Fig. 3.24 (continued)

Teratomas and rare teratoma-like tumor exam- sheen like silver or like a pearl of the finest water,”
ples deriving from all three germ layers or only therefore also the synonym of pearly tumor. The
one or two ones comprise a varying tissue mixture cerebellopontine angle is a common location.
of all kind of immature and mature tissue, even The dermoid cyst usually occurs in the poste-
small well-structured bones or teeth (Fig. 3.25). rior fossa and exhibits cysts with squamous epi-
Although the vast majority of these tumors are thelial-lined wall and derivatives from skin
benign, teratomas with malignant transformation appendices such as hair follicles with hairs and
of one of the tissue components can occur. sebaceous glands (Fig. 3.26).

Tumorlike Dysontogenetic Cysts Pathology of Blood Vessels


and Circulation
Age distribution: All ages.
Sex distribution: No sex predilection. Arteriovenous malformation
Saccular aneurysm
Epidemiology: 0.2–2 % of intracranial tumors;
Ischemic brain infarct
dermoid cysts rarer than epidermoid cysts.
Hypertensive angiopathy and brain hemorrhage
Grading: Benign.
Vasculitis
The epidermoid cyst is the main representative
of the differentiated dysontogenetic cysts. It
derives from differentiated cells of the ectoder-
mal germ layer forming cysts with squamous Arteriovenous Malformation
epithelial-lined wall. In the first description of an
epidermoid cyst by Cruveilhier [9], he painted a Age distribution: All ages.
poetic picture of the outer surface of “metallic Sex distribution: No sex predilection.
106 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

e f

Fig. 3.25 (a) Teratoid lipoma including a small mature epithelium, glands, bone, and connective tissue. HE.
bone (arrow). Note the dysraphic malformation of the (d) Tooth-anlage in a mature teratoma within the brain.
cerebellum. (b) Detail of the mature bone with compacta, HE. (e) Numerous small but mature teeth within a brain
bone marrow, and cartilage within the lipoma. Masson teratoma. (f) Detail showing mature molars and an
trichrome. (c) Mature teratoma with mixture of squamous incisor

Epidemiology: In a consecutive series of 4,530 well as treatment options and recommendations


autopsies, 4.3 % AVMs were found, among them for the clinical management of arteriovenous
only 0.5 % became symptomatic [28]. In 30–80 % malformations (AVMs), were formulated by the
of the patients, the possibly life-threatening hem- AVM Study Group [41] (AMSG) and a special
orrhage is the leading clinical manifestation [27] writing group of the Stroke Council of the
with a lethality of 10–15 % and a morbidity of up American Stroke Association [34].
to 50 % [30, 38, 43]. The occurrence of AVM- Morphologically, AVMs are a complex tangle
related hemorrhage counts for about 1 % among of abnormal arteries and veins linked by one or
all strokes [13]. often more fistulae. Connecting capillaries are
Epidemiological data, current concepts on missing. It is supposed that AVMs are caused by
diagnostic procedures, and grading systems, as a prenatal disorder of primordial vascular
Pathology of Blood Vessels and Circulation 107

a b

c d

Fig. 3.26 (a) Surface of an epidermoid cyst shining like tin slivers. Masson trichrome. (d) Dermoid cyst with
a cluster of pearls. (b) Epidermoid cyst of the CPA filled squamous epithelial-lined wall, hair follicle (arrow), and
with densely packed white keratin lamellae. (c) Epidermoid sebaceous glands (asterisk). Masson trichrome
cyst with squamous epithelial-lined wall filled with kera-

channels when the normal development to venous system. Deep noncortical AVMs that lie
mature arteries, capillaries, and veins is inhib- in the white matter are supplied by both surface
ited [33, 35]. Alternatively, since juvenile and and deep arteries, those in the basal ganglia, thal-
adult AVMs show some different features, also a amus, and brain stem by the small perforating
possible postnatal development was proposed arteries, mainly lenticulostriate and thalamoper-
[22, 23]. For their tumorlike appearance, in forant arteries. Drainage is by means of the deep
German literature AVMs are often described as veins. Deep AVMs count for 15 % of all AVMs.
arteriovenous hemangiomas, differentiating Choroid plexus AVMs are fed by choroidal arter-
macroangiomas with a diameter of several cen- ies and subependymal branches of the arteries at
timeters and microangiomas with a diameter of the base of the brain. They are drained exclu-
a few millimeters up to one centimeter sively by means of the deep veins.
(Fig. 3.27a–d). However, AVMs may increase
their volume, remain static, or may even sponta-
neously shrink [1]. Saccular Aneurysm
Superficial cortical AVMs lie in a sulcus or in
the cortex. They may extend subcortically and Age distribution: Adults with a peak in the sixth
are then wedge-shaped with the base in the cor- decade; rare in children (less than 5 % under the
tex. The apex may reach the ventricular wall age of 20 years).
(corticoventricular or transcerebral AVMs). They Sex distribution: Male to female ratio of 2:3;
are fed by cortical branches of major cerebral males predominate in the younger age groups.
arteries and drained by superficial veins, in tran- Epidemiology: 2–5 % prevalence in the gen-
scerebral AVMs often also by means of the deep eral population.
108 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

a b

c d

e f

Fig. 3.27 (a) Ruptured arteriovenous malformation vessel wall. (d) AVM with convolute of vessels. Elastica-
(AVM) of the cerebellum with massive bleeding involving van Gieson. (e) Saccular (berry) aneurysm of the basilar
the cerebellopontine angle. (b) Rupture of a larger vessel artery. (f) Note the collagenous wall of the aneurysm
within an AVM with an atomic mushroom-like bleeding. (arrow) without internal elastic lamina (red) and muscular
Masson trichrome. (c) AVM characterized by a convolute media compared to the adjacent normal vessel. Masson
of larger and smaller vessels of varying thickness of the trichrome

Almost all saccular or berry aneurysms arise are giant aneurysms. Histologically, the wall of
at the bifurcations of the main intracranial arter- the aneurysm consists only of fibrous connective
ies and have a propensity to spontaneous rupture tissue and is characterized by the absence of both
leading to primary nontraumatic subarachnoid the muscular tunica media and the internal elastic
hemorrhage. Only about 5–10 % of saccular lamina, both ending abruptly at the neck of the
aneurysms involve the posterior cerebral arteries aneurysm (Fig. 3.27e, f).
and the vertebrobasilar arteries, thus directly Different syndromes with gene mutations are
involving the CPA area. Aneurysms with a diam- linked with saccular aneurysms such as Marfan’s
eter over 25 mm are called giant aneurysms; syndrome, Ehlers–Danlos syndrome type IV, auto-
interestingly 30–40 % of aneurysms of children somal dominant polycystic kidney disease, and
Pathology of Blood Vessels and Circulation 109

hereditary hemorrhagic telangiectasia. In about severe enough and lasts for long enough, perma-
12–15 % of patients, the aneurysms are familial. nent nerve cell damage ensues, clinically appear-
Familial aneurysms were more often multiple and ing as stroke. Stroke is defined as an abrupt onset
have a tendency to rupture at a smaller size. of focal or global neurological symptoms caused
by ischemia or hemorrhage.
After 4–12 h in stage I of ischemic brain
Ischemic Brain Infarct infarct, the tissue macroscopically turns pale, and
histologically Nissl bodies and nuclei disappear
Age distribution: Age dependent, increasing with (Fig. 3.28b); after 24 h, neutrophil leukocyte
higher age, in children under 15 years of age 1 infiltration begins. After 48 h in stage II, mac-
per 100,000 population, in adults 85 years and rophages which histologically appear as foam
older 1 per 33 population. cells invade the infarct and start with phagocytiz-
Sex distribution: Males predominate. ing the damaged tissue (Fig. 3.28c–e). After 5
Epidemiology: Because of higher proportion days, the neutrophil leukocyte infiltration ceases.
of elderly people, high incidence rates in Western In larger infarcts, the phagocytosis may last for
countries and Japan; white people have lower months. Finally in stage III, the finished phago-
rates than nonwhite people. cytosis results in lacunae in small infarcts or in
The cause of ischemia of brain areas is mani- extended cysts in larger infarcts, both surrounded
fold. However, most cases are related to athero- by a layer of proliferated astrocytes. When, for
sclerosis of brain arteries often in combination instance, the precentral brain cortex is irrevers-
with thrombosis at sites of atherosclerotic ibly destroyed by an infarct, not only the area of
endothelial ulcerations (Fig. 3.28a). Another infarction is destroyed but also the entire pyrami-
important cause is emboli that break loose from dal tract will become atrophic (Fig. 3.28f).
thrombi of extracranial atherosclerotic arteries or
the heart. In rare cases, tumor emboli or fat embo-
lism may cause ischemia. Vasculitides (see Hypertensive Angiopathy and Brain
below) may lead to stenosis of arteries. Cerebral Hemorrhage
vasospasm after severe head injury and subarach-
noidal hemorrhage is correlated with ischemic Age distribution: Age dependent, increasing with
damage and may complicate neurosurgical inter- higher age.
ventions in such cases. When the ischemia is Sex distribution: Males predominate.

a b

Fig. 3.28 (a) Severe atherosclerosis of all arteries of II with macrophages (arrow) phagocytizing lipids within
Willis’ circle. Note the fusiform atherosclerotic aneurysm the infarction. Nissl. (e) Macrophages (foam cells) phago-
of the basilar artery. (b) Ischemic infarct stage I of the cytizing lipids within the infarct. Nissl. (f) Secondary uni-
cerebellum with loss of tissue staining (arrow). Nissl. (c) lateral degeneration of the pyramidal tract after ischemic
Ischemic infarct stage II of the pons with loss of myelin infarct stage III in the precentral cortex
sheaths. Heidenhain–Woelcke. (d) Ischemic infarct stage
110 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

c d

e f

Fig. 3.28 (continued)

Epidemiology: 10–30 per 100,000 population; Vasculitis


higher frequency in black people and Far Eastern
populations due to higher prevalence of hyper- Age distribution: Adults over 55 years of age
tension; hypertensive brain hemorrhage 50 % of (GCA); young adults (Takayasu); all ages with a
brain hemorrhages; 13 % of hypertensive brain peak in the fifth decade (PAN).
hemorrhages in the cerebellum, 7 % in the pons. Sex distribution: No sex predilection (GCA);
Chronic hypertension on the one hand aggra- female predilection (Takayasu); male to female
vates atherosclerotic changes, on the other hand ratio of 2:1 (PAN).
induces a hypertensive angiopathy characterized Epidemiology: 20 per 100,000 population over
by reduplication of the basal lamina, and fibrosis 50 years of age (GCA); 1 per 3,000 population in
of the arterial vessel wall where fibrotic connec- Japan, rare outside Japan (Takayasu); CNS
tive tissue replaces smooth muscle cells. In later involvement in 10–50 % of generalized PAN;
stages, arteries with homogenous fibrinoid necro- CNS involvement in 50–75 % of generalized
sis of the vessel wall are prone to rupture. The SLE.
subsequent arterial bleeding, the hypertensive Inflammatory diseases of CNS blood vessels
brain hemorrhage, may result in a considerable are caused by infections or immunological pro-
size of the blood clot which displaces the sur- cesses, respectively, and may lead to ischemic
rounding brain tissue (Fig. 3.29a, b). tissue damage. Histologically, inflammatory
Other etiologies of nontraumatic brain hemor- infiltrates in the vessel wall in various cell com-
rhage comprise cerebral amyloid angiopathy, arterio- positions are diagnostic. The inflammatory
venous malformations, aneurysms, tumor bleedings, changes are often focal (focal granulomas) which
and any cause of impaired blood coagulation, for means that a negative histological result in a
example, by anticoagulants and other drugs. biopsy cannot completely rule out the possibility
Pathology of Blood Vessels and Circulation 111

a b

c d

e f

Fig. 3.29 (a) Hypertensive angiopathy characterized by Churg–Strauss characterized by predominance of eosino-
fibrinoid necrosis (red) destroying the smooth muscle phils (red) within the inflammatory infiltrate of the vessel
cells of the vessel wall. Masson trichrome. (b) Hypertensive wall. Masson trichrome. (e) Panarteritis nodosa character-
hemorrhage of the pons. (c) Arteritis temporalis Horton ized by the lymphocytic inflammatory infiltrate and focal
characterized by numerous multinucleated giant cells fibrinoid necrosis (red). Masson trichrome. (f) Panarteritis
within the inflammatory infiltrate of the vessel wall. nodosa with extended fibrinoid necrosis of intracerebral
Masson trichrome. (d) Eosinophilic (allergic) arteritis vessels. HE

of a vascultitis, for example, giant cell arteritis of of the pathogen in question. For examples of
the temporal artery. Therefore, if possible, a microbial infections, see the paragraph on the
biopsy of 3–5 cm length is recommended. different forms of meningitis above.
Direct microbial infections with bacterial For vasculitides caused by immunological
(e.g., due to streptococci, mycobacteria, spiro- processes, a classification of three groups based
chetes), viral (e.g., due to herpes zoster), fungal, on pathogenetic mechanisms includes cell-medi-
or protozoal etiologies have different age and sex ated, immune-complex-mediated, and antineu-
distributions as well as epidemiology depending trophil cytoplasmic antibody (ANCA)-mediated
112 3 Neuropathology of the Cerebellopontine Angle and Its Surroundings

inflammations [17]. Cell-mediated inflammations 10. Deckert-Schluter M, Marek J, Setlik M, Markova J,


comprise giant cell arteritis (GCA, synonym Pakos E, Fischer R, Wiestler OD (1998) Primary
manifestation of Hodgkin’s disease in the central ner-
temporal arteritis Horner) which is the most com- vous system. Virchows Arch 432(5):477–481
mon form in Europe (Fig. 3.29c), Takayasu’s 11. Ellison DW, Perry A, Rosenblum M, Asa S, Reid R,
arteritis which is very common in Japan, primary Louis DN (2008) Tumours: non-neuroepithelial
angiitis of the central nervous system, and tumours and secondary effects. In: Love S, Louis DN,
Ellison DW (eds) Greenfield’s neuropathology, 8th
Kawasaki’s disease. Immune-complex-mediated edn. Edward Arnold, London, pp 2088–2106
inflammations comprise panarteritis nodosa 12. Fletcher DM, Unni KK, Meterns F (eds) (2002) World
(PAN) manifesting as systemic or limited form Health Organization classification of tumours.
(Fig. 3.29e, f), systemic lupus erythematosus Pathology and genetics of tumours of soft tissue and
bone. IARC Press, Lyon
(SLE), Behçet syndrome, and some infection- or 13. Furlan AJ, Whisnant JP, Elveback LR (1979) The
drug-induced, or malignancy-related vasculiti- decreasing incidence of primary intracerebral hem-
des. ANCA-mediated inflammations comprise orrhage: a population study. Ann Neurol 5(4):
Churg–Strauss syndrome, which is also called 367–373
14. Han DH, Kim DG, Chi JG, Park SH, Jung HW, Kim
eosinophilic or allergic angiitis (Fig. 3.29d), YG (1992) Malignant triton tumor of the acoustic
Wegener’s granulomatosis, and some drug- nerve. Case report. J Neurosurg 76(5):874–877.
induced vasculitides. A mixed immunological doi:10.3171/jns.1992.76.5.0874
etiology is observed in Sjögren’s syndrome. 15. Ito U, Hashimoto K, Inaba Y (1974) Osteochondroma
of the posterior clinoid process. Report of a case with
special reference to its histogenesis. Acta Neuropathol
27(4):329–335
16. Jaffe ES, Harris NL, Stein H, Vardiman JW (2001)
References World Health Organization classification of tumours.
Pathology and genetics of tumours of haematopoietic
1. Abdulrauf SI, Malik GM, Awad IA (1999) Spontaneous and lymphoid tissues. IARC Press, Lyon
angiographic obliteration of cerebral arteriovenous 17. Jennette JC, Falk RJ, Milling DM (1994) Pathogenesis
malformations. Neurosurgery 44(2):280–287; discus- of vasculitis. Semin Neurol 14(4):291–299
sion 287–288 18. Jung CS, Schanzer A, Hattingen E, Plate KH, Seifert
2. Balmaceda CM, Fetell MR, O’Brien JL, Housepian V (2006) Xanthogranuloma of the sellar region. Acta
EH (1993) Nevus of Ota and leptomeningeal melano- Neurochir (Wien) 148(4):473–477
cytic lesions. Neurology 43(2):381–386 19. Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder
3. Bambakidis NC, Megerian CA, Ratcheson RA (2004) M, Grossman AB (2005) Clinical review: diagnosis
Differential grading of endolymphatic sac tumor and management of pituitary carcinomas. J Clin
extension by virtue of von Hippel-Lindau disease sta- Endocrinol Metab 90(5):3089–3099
tus. Otol Neurotol 25(5):773–781 20. Kepes JJ, Chen WY, Connors MH, Vogel FS (1988)
4. Barnes L, Eveson JW, Reichart P, Sidransky D (eds) “Chordoid” meningeal tumors in young individuals
(2005) World Health Organization classification of with peritumoral lymphoplasmacellular infiltrates
tumours. Pathology and genetics of head and neck causing systemic manifestations of the Castleman syn-
tumours. IARC Press, Lyon drome. A report of seven cases. Cancer 62(2):391–406
5. Best PV (1987) Malignant triton tumour in the cere- 21. Kudo M, Matsumoto M, Terao H (1983) Malignant
bellopontine angle. Report of a case. Acta Neuropathol nerve sheath tumor of acoustic nerve. Arch Pathol
74(1):92–96 Lab Med 107(6):293–297
6. Brat DJ, Giannini C, Scheithauer BW, Burger PC 22. Lasjaunias P (1997) Vascular diseases in neonates,
(1999) Primary melanocytic neoplasms of the central infants and children: interventional neuroradiology
nervous systems. Am J Surg Pathol 23(7):745–754 management. Springer, Berlin/Heidelberg/New York
7. Brat DJ, Scheithauer BW, Staugaitis SM, Holtzman 23. Lasjaunias PTBK (1996) Vascular diseases in chil-
RN, Morgello S, Burger PC (2000) Pituicytoma: a dren. Springer, Berlin/Heidelberg/New York
distinctive low-grade glioma of the neurohypophysis. 24. Lonser RR, Kim HJ, Butman JA, Vortmeyer AO,
Am J Surg Pathol 24(3):362–368 Choo DI, Oldfield EH (2004) Tumors of the endolym-
8. Carrasco CA, Rojas-Salazar D, Chiorino R, Venega phatic sac in von Hippel-Lindau disease. N Engl J
JC, Wohllk N (2006) Melanotic nonpsammomatous Med 350(24):2481–2486
trigeminal schwannoma as the first manifestation 25. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK
of Carney complex: case report. Neurosurgery (eds) (2007) WHO classification of tumours of the
59(6):E1334–E1335; discussion E1335. 00006123- central nervous system. IARC Press, Lyon
200612000-00022 26. Love S, Louis DN, Ellison DW (eds) (2008)
9. Cruveilhier L (1829) Anatomie pathologique du corps Greenfield’s neuropathology, vol 1 and 2, 8th edn.
humain, vol tome 1, livre II. Baillière, Paris Hodder Arnold, London
References 113

27. Mast H, Mohr JP, Osipov A, Pile-Spellman J, Marshall 35. Padget DH (1956) The cranial venous system in man
RS, Lazar RM, Stein BM, Young WL (1995) ‘Steal’ is in reference to development, adult configuration, and
an unestablished mechanism for the clinical presenta- relation to the arteries. Am J Anat 98(3):307–355
tion of cerebral arteriovenous malformations. Stroke 36. Padhya TA, Athavale SM, Kathju S, Sarkar S, Mehta
26(7):1215–1220 AR (2007) Osteochondroma of the skull base.
28. McCormick W (1978) Classification, pathology, and Otolaryngol Head Neck Surg 137(1):166–168
natural history of angiomas of the central nervous sys- 37. Piedra MP, Scheithauer BW, Driscoll CL, Link MJ
tem. Weekly update. Neurol Neurosurg 14:2–7 (2006) Primary melanocytic tumor of the cerebellopon-
29. McLean CA, Laidlaw JD, Brownbill DS, Gonzales MF tine angle mimicking a vestibular schwannoma: case
(1990) Recurrence of acoustic neurilemoma as a malig- report. Neurosurgery 59(1):E206; discussion E206
nant spindle-cell neoplasm. Case report. J Neurosurg 38. Samson DSBH (1991) Preoperative evaluation of the
73(6):946–950 risk/benefit ratio for arteriovenous malformations of the
30. Michelsen WJ (1979) Natural history and pathophysi- brain. In: Williams RH, Rengachary SS (eds) Neurosurgery
ology of arteriovenous malformations. Clin Neurosurg update II: vascular, spinal pediatric, and functional neuro-
26:307–313 surgery. MacGraw-Hill, New York, pp 129–133
31. Miller RT, Sarikaya H, Sos A (1986) Melanotic 39. Sato K, Kodera T, Kitai R, Kubota T (1996)
schwannoma of the acoustic nerve. Arch Pathol Lab Osteochondroma of the skull base: MRI and histo-
Med 110(2):153–154 logical correlation. Neuroradiology 38(1):41–43
32. Mrak RE, Flanigan S, Collins CL (1994) Malignant 40. Stein AA, Schilp AO, Whitfield RD (1960) The histo-
acoustic schwannoma. Arch Pathol Lab Med genesis of hemangioblastoma of the brain. A review
118(5):557–561 of twenty-one cases. J Neurosurg 17:751–761
33. Nelson MD Jr, Gonzalez-Gomez I, Gilles FH (1991) 41. The AMSG (1999) Arteriovenous malformations of
Dyke award. The search for human telencephalic ven- the brain in adults. N Engl J Med 340:1812–1818
triculofugal arteries. AJNR Am J Neuroradiol 12(2): 42. Thorns V, Zajaczek J, Becker H, Walter GF, Hori A
215–222 (2003) Extracerebral subdural manifestation of Chester-
34. Ogilvy CS, Stieg PE, Awad I, Brown RD Jr, Erdheim disease associated with a giant adenoma of
Kondziolka D, Rosenwasser R, Young WL, the pituitary. Clin Neuropathol 22(5):246–251
Hademenos G (2001) AHA scientific statement: rec- 43. Wilkins RH (1985) Natural history of intracranial vas-
ommendations for the management of intracranial cular malformations: a review. Neurosurgery 16(3):
arteriovenous malformations: a statement for health- 421–430
care professionals from a special writing group of the 44. Witschi E (1948) Migration of germ cells of human
Stroke Council, American Stroke Association. Stroke embryos from the yolk sac to the primitive gonadal
32(6):1458–1471 folds. Contrib Embryol 32:67–80
Approaches to the Cerebellopontine
Angle 4

In this chapter, the three main operative advantage of the MF approach, according to these
approaches to the internal auditory canal and CP authors, is that the lateral end of the IAC is
angle—the retrosigmoid, the translabyrinthine, exposed adequately, which ensures removal of
and the middle fossa approach—are discussed. the entire tumor [4, 11, 104]. With the MF
An overview of the other approaches and tech- approach, however, the transverse crest may hin-
niques to the CP angle is presented in the chapter der the visualization of the inferior half of the
on the “anatomy of the CP angle.” fundus—of the so-called inferior recesses [18,
24]. The approach has several additional
drawbacks: limited exposure of the CP angle,
Middle Fossa Approach especially of its lower portion, which restricts the
control of possible bleeding, and the potential
The MF approach to the IAC was described by major morbidity related to the retraction of the
Parry in 1904 who used it to section the auditory temporal lobe or to damage to the vein of Labbé
nerve in a patient with vertigo and tinnitus [67]. It [7, 45, 46]. Furthermore, it is related to a risk of
was W. F. House who popularized the approach injuring the facial nerve during opening of the
as a reliable technique initially for the treatment meatal dura and tumor removal because the nerve
of otosclerosis and then for removing of small is located in the anterosuperior part of the IAC,
VSs [60]. Currently the approach is utilized in sometimes immediately beneath the dura. Thus,
case of small tumors, in which there is an oppor- the surgeon must work around the nerve to
tunity to preserve hearing, especially in those remove the tumor, increasing the risk of its dam-
located mainly laterally within the IAC [9, 46, age during the surgical maneuvers.
90, 93]. It is used rarely in other tumors, such as
small meningiomas located at the lip of the IAC.
The MF approach provides a direct exposure Standard MF Approach
of the IAC including its fundus from a superior
trajectory [7, 95, 104]. The facial nerve can be The patient is placed supine on the operating
exposed from its cisternal to its tympanic seg- table, and the head is rotated so that the plane of
ment. Moreover, the facial and the cochlear the temporal bone becomes parallel to the floor.
nerves can be identified early in the fundus region, Alternatively, obese patients or those with comor-
which facilitates safe tumor dissection. Many bidities can be placed in lateral or modified
authors assert that the location of the posterior park-bench position. A lumbar drainage may be
semicircular canal and of the vestibule may limit inserted to relax the brain and enable safe tempo-
the lateral exposure if the RS approach is used ral lobe elevation. The skin incision begins just in
and hearing is to be preserved [95]. The main front of the ear and is vertical, curved, inverted

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 115


DOI 10.1007/978-3-642-35422-9_4, © Springer-Verlag Berlin Heidelberg 2013
116 4 Approaches to the Cerebellopontine Angle

“U,” or question mark shaped. Then, the temporal The labyrinthine segment of the facial nerve is then
bone is exposed, and a craniotomy is performed, followed to the lateral end of IAC. This direct
located generally one-third posterior and two- approach to the facial nerve may be used to access
thirds anterior to the external auditory meatus facial nerve tumors or for reconstruction of the
[20]. Importantly, sufficient bone has to be facial nerve [7, 11]. The technique of U. Fisch is
removed basally in order to acquire a flat plane of based on the relation of the superior semicircular
view across the middle cranial fossa floor and canal to the IAC [30]. A line is drawn over the long
avoid excessive retraction of the temporal lobe. axis of the arcuate eminence (lying over the supe-
Next, the dura is dissected and elevated from rior semicircular canal), and a second line is drawn
the floor of middle cranial fossa in a posterior- 60° to the first one. The IAC is located along the
to- anterior direction until the arcuate eminence second line. This technique is still applied in case
and the greater petrosal nerve are identified of complex temporal bone fractures. Garcia-Ibanez
[97]. Thus, inadvertent injury of the greater [32] introduced the method of direct identification
superficial petrosal nerve (GSPN), which lies in of the IAC and applied it initially to perform ves-
the major petrosal groove and is covered by a tibular neurectomy. The angle between GSPN and
thin layer of periosteum, is avoided. Injury to arcuate eminence is 120°, and the bisecting line
this nerve leads to “dry eye” syndrome due to of the angle determines the position of IAC.
impaired lacrimation. In rare cases, though, the Advantages of this method are the lower risk of
nerve has to be sacrificed in order to prevent facial nerve injury because the technique requires
traction injury to the geniculate ganglion [103]. less dissection along the nerve; secondly, the early
The middle meningeal artery at the foramen and more extensive drilling to locate the IAC
spinosum, the greater superficial petrosal nerve, enhances the subsequent exposure of the tumor.
and the arcuate eminence (overlying the superior Whatever method is utilized, however, the surgeon
semicircular canal) are identified. Meticulous should account for the individual anatomical vari-
care should be taken to maintain the integrity of ability. Currently the neuronavigatory guidance, in
the dura in order to avoid CSF leak or iatrogenic particular the fusion of bone-window CT and MRI
temporal lobe injury. This might be more datasets, allows for direct demonstration of these
difficult in the elderly who have a thin dura that structures with high precision. Its usefulness is
may be easily torn. A possible dehiscent genicu- especially high in case of absent normal anatomy,
late ganglion has to be recognized early [7]. for example, in reoperations or in case of extensive
According to the findings from one study, in bone destruction (Fig. 4.1).
15 % of the examined cases, all or part of the The IAC, which lies 3–4 mm under the middle
geniculate ganglion and the genu of the facial cranial fossa floor, is unroofed using progres-
nerve were exposed in the floor of middle cra- sively smaller diamond burrs, and its dura is
nial fossa [72]. Importantly, the petrous segment exposed from the porus to the fundus. The verti-
of the carotid artery may lie in the floor of the cal crest (Bill’s bar) and the entrance of the facial
middle fossa deep to the greater petrosal nerve nerve into the fallopian canal are the lateral limit
without bony covering [73, 92]. The medial of the dissection. Drilling in the angle between
limit of dural elevation is the trigeminal impres- the GSPN and the IAC should be avoided because
sion. An important step is to identify reliably of risk of injury to the underlying cochlea [103].
the superior petrous ridge, along which the Brackmann [7] proposed to decompress the prox-
superior petrosal sinus lies. imal labyrinthine portion of the facial nerve for
The identification of IAC is traditionally based approximately 3 mm to compensate for any
on anatomical relationships of the middle cranial swelling that may be caused during dissection.
fossa structures. With the technique described by The dura is incised along the long axis of the
House [45], the GSPN is identified and followed IAC and the cranial nerves are identified. The
up to the facial hiatus and to the geniculate gan- VS, especially if it originates from the inferior
glion by progressively drilling the middle fossa. vestibular nerve, tends to displace the facial
Middle Fossa Approach 117

a b

c d

Fig. 4.1 (a, b) Navigation screenshots of the initial expo- spinosum, foramen ovale, trigeminal nerve (V3), and mid-
sure of the MCF base and of the IAC; (c, d) respective dle meningeal artery
intraoperative images, demonstrating the foramen

nerve superiorly. It lies frequently immediately all non-involved structures, such as the supe-
underneath the dura, between the tumor and the rior and inferior vestibular nerves, should not
surgeon. Tumor removal follows the general be cut: their preservation may help to maintain
principles: an initial debulking has to be done the cochlear blood supply [7]. The importance
in larger VSs, followed by dissection of the of the medial-to-lateral dissection technique for
capsule from the cranial nerves. Smaller VSs hearing preservation has been frequently high-
are removed piecemeal or—more rarely—as lighted. It avoids the risk of avulsion of the very
a single mass. The removal of the most lateral fragile fibers that enter the modiolus. Further, the
tumor portion, especially if it extends inferior blood vessels when retracted laterally have some
the transverse crest, may require blind dis- elasticity from their medial origins but not from
section with an angled curette [24]. Of utmost their foraminal terminations.
importance is to preserve the blood supply to the The opened IAC has to be sealed off—either
cochlea, which usually runs between the facial with muscle or fat pieces—in order to prevent
and cochlear nerves. Some authors state that CSF leak. The major surgical complications with
118 4 Approaches to the Cerebellopontine Angle

the MF approach are related to damage of the CP angle. The upper size limit of the extracanali-
temporal lobe (edema, ischemia, or hemorrhage cular tumor portion for the extended MF approach
that may lead to functional deficit or seizures), is stated to be even 1.6–2 cm. Its application in
injury to the vein of Labbé, and sub/epidural larger tumors is rather controversial despite the
hematomas. fact that some good results have been reported.
The temporal lobe has to be retracted for a longer
period, and the facial nerve may be more difficult
Extended MF Approach to dissect due to its location in front of the tumor.
Importantly, the access and the possibility to con-
The classical MF approach was modified by two trol the inferior aspect of the CP angle are also
teams—in Germany and in Japan—and applied limited [84].
successfully in case of VSs with a substantial Satar et al. [84] examined the issue, which
component in the CP angle [91, 109]. The major VSs may profit from the extended MF approach.
difference to the standard MF approach is the These authors evaluated the facial nerve func-
amount of petrous bone removal: with the classi- tion and hearing outcome in case of intracanali-
cal technique, the IAC is just unroofed, while the cular VSs, VSs extending 1–9 mm into the CP
extended MF approach provides additional expo- angle extension, and VSs with 10–18-mm CP
sure by extensive extradural removal from the angle extension. They found that the functional
petrous apex anteriorly to the superior semicircu- outcome was similar in the first two groups:
lar canal posteriorly [92, 107, 108]. With the functional hearing preservation rate was 62.2 and
technique described by Wigand, the inner ear 63.1 %, respectively, and good FN outcome had
structures are preserved [107]. The author 93.7 % vs. 97.6 %. For tumors of 10–18-mm CP
reported on his experience with this approach in angle extension, however, the hearing preserva-
a series of 735 patients with VS growing more tion rate was lower (34 %). Patients with >10-
than 2 cm into the CP angle [35]. Twenty-two mm tumors should be advised that choosing the
percent of the tumors were intracanalicular, 42 % MF approach for hearing preservation carries a
had less than 1-cm extracanalicular extension, somewhat higher risk of persistent FN dysfunction,
29 % had 1–1.9-cm extracanalicular extension, concluded the authors.
and 7 % had larger than 2-cm extracanalicular The MF approach has been modified in order
extension; complete VS removal was achieved in to access lesions located in the anterior CP angle,
97.1 %. Depending on the tumor size, normal and in the petroclival area and anterior brain stem [3,
near-normal facial nerve function ranged from 83 22, 44, 48–50]. The initial operative steps are
to 99 %. Hearing preservation rate at or near the similar to the described above. The superior pet-
preoperative level was preserved in 60.2, 48.2, rosal sinus, however, has to be ligated and cut.
23.9, and 17.6 %, respectively. Once this is performed and the key structures—
The extensive bone removal is key to expose GSPN, foramen ovale, mandibular nerve, IAC,
the tumor portion protruding into the CP angle. and vestibulocochlear nerves—are identified, the
At the porus, the exposure should be approxi- medial petrous bone lying within the Kawase
mately three-quarters of the circumference of the rhomboid is removed [49, 50]. It should be
canal. It is not possible to achieve this degree of remembered that the cochlea is located at the
exposure in the lateral portion of the IAC: the posterolateral corner of this rhomboid. The limits
bone removal is limited posteriorly by the supe- of the extended MF approach are the inferior pet-
rior semicircular canal, while the anteromedial rosal sinus inferiorly, the middle ear ossicles lat-
edge of the exposure is limited by the cochlea [70, erally, the gasserian ganglion anteriorly, and the
71]. Therefore, at the fundus the IAC is exposed superior semicircular canal posteriorly [103].
only 90° to avoid injury to these structures. The Kawase approach is similar to the extended
The extended MF approach is currently used MF approach with the same extent of medial
in VSs with less than 1-cm extension into the petrous bone removal. The main difference is the
Translabyrinthine Approach and Its Variations 119

additional transverse incision of the tentorium to The size of the VSs is not a contraindication
the incisura. Elevating the divided tentorial edges for selecting this approach, and even tumors mea-
provides wide exposure of the petroclival region suring 4 cm or more can be exposed and removed
down to the midclivus level. Thus, the basilar successfully [2, 21, 53]. As pointed by Day et al.
artery, the posterior cerebral artery, the superior [21], when the bone removal is adequate at
cerebellar artery, and the anterior inferior cere- superficial levels, the corridor of access is more
bellar artery, as well as the cranial nerves III to than sufficient regardless of tumor size. Although
VIII, are well visualized. some anatomical variations, such as a low-lying
tegmen, anterior sigmoid sinus, or high jugular
bulb may complicate the removal of large tumors,
Translabyrinthine Approach various techniques to overcome these obstacles
and Its Variations have been described [31].

The translabyrinthine approach (TL approach)


was refined and popularized by WF House and Operative Technique
WE Hitselberger in the early 1960s (see Chap. 1).
The approach is performed usually by a team of Patients are usually positioned supine with the
neurotologist (translabyrinthine temporal bone head turned opposite the side of the lesion. Rarely,
dissection) and neurosurgeon (microsurgical the lateral position is preferred. A retroauricular
tumor removal). curved skin incision is made, extending approxi-
The TL approach is used widely for removal mately 1 cm behind the body of the mastoid. The
of VSs when hearing preservation is not an issue. incision starts above the pinna and extends inferi-
In combination with other approaches, it is used orly to the mastoid tip. The temporal fascia and
in various cranial base tumors [8, 10, 21, 56, 74]. pericranium are incised and elevated as a flap.
Candidates for this approach are VSs of any size
with poor hearing, but it is most frequently
applied for those >2–2.5 cm [5, 83, 102]. Mastoidectomy
Assuming that in tumors larger than 2.5 cm the
possibility of hearing preservation is “extremely After the mastoid is exposed, the spine of Henle,
unlikely,” some authors state that this consider- the external auditory meatus, the mastoid ridge,
ation should not influence the choice of the the supramastoid crest, and the mastoid tip are
surgical approach [21, 111]. They recommend identified [71, 100]. The cortical bone of the mas-
the TL approach in patients with such tumors toid is drilled off and the underlying air cells are
even if they have good preoperative hearing—a exposed. The mastoid antrum lies approximately
concept that is rather disputable, as discussed in 15 mm deep to the surface below the spine of
the section on large VSs operated via the retrosig- Henle. Once the antrum is opened, the bony
moid route. semicircular canals are identified. Sufficient bone
Advantages of the TL approach are shorter at the margins of the mastoidectomy has to be
distance between the surface and the neoplasm, removed not to restrict the exposure of the deep
allowing the surgeon to work significantly closer structures [21, 53].
to the tumor; early identification of facial nerve Then, the sigmoid sinus, the middle fossa dura,
using a constant bony landmark (Bill’s bar); min- and the upper surface of the jugular bulb are skel-
imal, if any, cerebellar and brain stem retraction; etonized. The facial nerve must be identified early
ready ability to perform a direct facial nerve in the procedure: it lies below the lateral semicir-
reconstruction, if necessary; low incidence of cular canal. The inferior limit of the approach is
postoperative headaches; and easy access to the the jugular bulb. Anteriorly only a thin layer of
tumor bed in case of postoperative complications bone should be left at the posterior aspect of the
[17, 29, 56, 106]. external auditory canal. This bone layer and the
120 4 Approaches to the Cerebellopontine Angle

mastoid segment of the facial nerve form the Medical) to occlude the Eustachian tube and
anterior limit of the exposure. Posteriorly the antrum decreases the risk of CSF leaks.
bone overlying the sigmoid sinus is completely
exposed. In order to enhance the exposure of large
tumors, the bone removal should go beyond the Surgical Outcome
sinus, allowing its slight retraction [17].
The most frequent surgical complication related
to the TL approach is CSF leak. Its rate ranges
Labyrinthectomy between 1.4 and 20 % and is higher than with the
alternative approaches [14, 15, 58, 75, 82]. The
The labyrinthectomy is then performed, which risk is related to tumor size: in larger tumors it is
includes removing of the semicircular canals and more than 15 % [53, 56]. The reported rates of
vestibule to expose the dura of the IAC [41]. The meningitis range from 1.6 to 4 % [53, 56, 75].
posterior, superior, and inferior walls of the IAC Other complications include arterial injuries (of
(two-thirds of its circumference) must be the AICA or of the internal auditory artery), injury
removed. The canal is opened widely until the to veins or sinuses, postoperative hemorrhages,
transverse and vertical crests (Bill’s bar) are cerebellar edema, and pneumocephalus. The rate
exposed to help in locating the facial nerve later- of complete tumor removal in experienced hands
ally. The cochlear aqueduct is the medial is >95 %, and the recurrence rate is very low: less
landmark of the inferior bone removal. In order than 2 % [53, 56, 83, 89]. The rate of facial nerve
to obtain adequate inferior exposure in case of preservation with the TL approach is 88–94 %
high jugular bulb, the bulb should be skeleton- [14, 41, 53, 96, 99, 101]. In intracanalicular
ized and the entire inferior lip of the IAC removed. tumors, the rate is 97–100 %; in VS <2 cm, 92.5–
Inferior exposure is obtained by skeletonizing the 95 %; and in VS >2 cm, 63 % [10, 13, 53].
posterior aspect of the jugular bulb [53, 71]. The refinement of the TL technique allows
As outlined by A. Rhoton [71], the two main achieving very good results even in large VSs.
goals are exposure of the dura of Trautman’s tri- Briggs et al. [14] presented the outcome of treat-
angle on the posterior surface of the temporal ment of 167 patients with VS >4 cm: complete
bone facing the CP angle and removal of enough removal was achieved in 95 %, and the mortality
bone to identify the nerves lateral to the tumor in rate was 0 %. The rate of anatomical facial nerve
the fundus area. The dura lining the IAC and the preservation was 91 %. With an approximate 2
dura of the Trautman’s triangle are opened, con- years follow-up, 42 % of the patients had HB
sidering the possibility to come upon the subarcu- grade I–II function. The authors stressed the
ate artery or the AICA in the dura of the triangle. significance of wide bone exposure in case of
The superior vestibular nerve is the first structure large tumors. Thus, removal of the part of the tem-
encountered when the IAC is opened due to its poral squama allows extending the rostro-caudal
location posterosuperiorly within the canal, which working angle. The complete jugular bulb skele-
decreases the risk of injury to the facial nerve. tonization increases the access to the caudal tumor
As with other approaches, once the critical part. Lanman et al. [53] evaluated the outcome of
structures are identified, tumor debulking is per- TL removal of such VSs in the same clinic in a
formed; then, its capsule is dissected from the later period. Increased experience allowed increas-
neural structures in the arachnoid plane. In case ing the rates of complete tumor removal to 96.3 %
the anatomical integrity of facial nerve is lost and of anatomical facial nerve preservation to
during surgery, the exposed proximal and distal 93.7 %. The facial nerve functional outcome was
segments facilitate its repair: either with a direct also improved: 1 year after discharge, excellent
anastomosis or with a cable graft. Following function (House–Brackmann grade I or II) was
tumor removal and hemostasis, the dura is closed present in 52.6 % of the patients. This outcome
and fat is packed into the mastoid. Packing with was possible with a relatively low morbidity rate:
muscle, fat, or Tabotamp® (Johnson&Johnson CSF leakage rate was 14 % but required surgical
Combined Approaches 121

repair only in 1.1 %; eight patients had ninth cra- most instances [103]. The approach includes a
nial nerve deficit, seven had tenth cranial nerve meticulous drilling of either the superior or the
deficit, and seven showed diplopia secondary to posterior semicircular canal, depending on the
either fourth or sixth cranial nerve palsies. In the tumor location. The transmastoid transpetrous
series there were no mortalities. approach with partial labyrinthectomy is used in
The main disadvantages of the approach are small VSs with functional hearing preoperatively
that it sacrifices hearing, restricts the exposure, in [37, 87].
particular the access to the caudal tumor portion
and the tumor/brain stem interface, and is related
to a higher CSF leak rate [12, 17]. Further, the Transcochlear Approach
facial nerve is at risk of injury during the exten-
sive petrosal resection. The venous sinuses may The transcochlear approach is primarily an anter-
be injured during bone drilling. The wide expo- omedial extension of the translabyrinthine
sure and manipulation of these sinuses may cause approach [8, 42, 43, 71, 103]. It requires division
late thrombosis. Roche et al. [74] pointed out of the GSPN, transposition of the facial nerve,
that the rate of complications related to sinuses and resection of the cochlea. The initial part of
injury has never been specifically addressed the dissection is the same as for the translabyrin-
for this approach. In one study, however, 143 thine approach, except that the external auditory
patients operated via the presigmoid transpetro- canal is closed and the tympanic membrane and
sal approach were evaluated [64]: seven patients ossicles are removed [71].
(5 %) had sigmoid sinus occlusion due to direct The facial nerve is skeletonized completely
sinus injury or compression by bone particles. from its geniculate genu to the stylomastoid fora-
men. The GSPN is transected, and the facial
nerve is transposed posteriorly, which allows for
Retrolabyrinthine Approach further bone drilling through the facial canal and
the cochlea to the petrous apex. The bone removal
Compared to the TL, this approach requires less extends to the edge of the clivus medially, expos-
bone removal, provides a possibility to preserve ing the dura between the inferior and superior
hearing, and is related to a lower risk of injury to petrosal sinuses. The wide bony opening creates
the facial nerve. The approach provides good direct access to the anterior CP angle, the middle
exposure of the IAC and of the posterior CP angle third of clivus, and the anterior brain stem and
but a limited one to the anterior CP angle and to allows visualizing the abducens nerve, the lower
the petroclival area [19]. The initial part—mas- margin of the trigeminal nerve, the nerves enter-
toidectomy—is performed as described in the TL ing the jugular foramen, as well as a part of the
approach section. The three semicircular canals basilar artery and of the AICA [71].
are skeletonized but preserved. In case the sigmoid
sinus is located more anteriorly or is larger than
normal, the access to the CP angle may be restricted Combined Approaches
[103]. Bento et al. [6] pointed that the approach
allows for high rate of facial nerve preservation. In Combined Translabyrinthine–
case the exposure is insufficient, it can be extended Retrosigmoid Approach
to a complete translabyrinthine approach [27].
The TL approach may also be combined with
a retrosigmoid or with a supra- and infratento-
Partial Translabyrinthine Approach rial presigmoid approach. Glasscock et al. [36]
used four different approaches in 616 cases,
This approach increases the exposure to CP angle selecting each one on the basis of tumor size
and petroclival area, if compared to the retrolaby- and hearing level. They recommended the com-
rinthine approach. Hearing can be preserved in bined translabyrinthine–suboccipital approach
122 4 Approaches to the Cerebellopontine Angle

for tumors larger than 3 cm. H. E. Maddox [55] transpetrosal partial labyrinthectomy approach
used a lateral approach, which is a combination is another combined technique, promoted by
of the translabyrinthine and retrosigmoid routes Sekhar et al. [87].
in a series of 106 consecutive cases with facial The use of two different approaches, however,
nerve preservation rate of 88 %. Anderson et al. does not only provide operative advantages; the
[1] recommend the use of a combined translab- risks of approach-related morbidity are also
yrinthine–retrosigmoid approach in very large essentially higher, and such combined techniques
tumors. The translabyrinthine exposure allowed are rarely necessary.
for early identification of the facial nerve distally,
while the exposure posterior to the sigmoid sinus
was found to be especially beneficial in case of Retrosigmoid Approach
abnormally high jugular bulb that restricts the
vertical exposure. Benefits of the Approach

The retrosigmoid approach (RS approach) is a


Translabyrinthine–Transtentorial safe, elegant, and relatively simple technique.
Approach With small modifications, described in the corre-
sponding chapters of this book, it provides access
This technique was introduced by Morrison and to all four levels of the CP angle and their related
King in 1973 [62] and has the advantage of a bet- structures:
ter rostral tumor exposure, which is achieved by 1. Petroclival area, Meckel’s cave, and part of the
ligating the superior petrosal sinus and incision middle cranial fossa
of the tentorium. The authors achieved complete 2. Internal auditory canal
VS removal in 91.7 % and preservation of the 3. Posterior part of the petrous bone
anatomical integrity of the facial nerve in 80.8 %. 4. Jugular foramen
The approach-related morbidity, however, was 5. Craniocervical junction
unacceptably high and included aphasia, epilep- The technique is related to very low procedure-
tic seizures, and CSF leaks [51]. Sluyter et al. related morbidity rate and therefore is among the
[94] used this combined approach in a series of most frequently utilized operative routes in neu-
120 patients with VS <2 cm and achieved com- rosurgery [26, 65, 70, 110]. It is a hearing-pre-
plete tumor removal in 91.7 % and anatomical serving technique that offers an excellent
facial nerve preservation in 80.8 % of the cases. panoramic visualization to the whole CP angle
They could reduce the morbidity rate to accept- and a wide exposure of the tumor, whatever its
able levels, with the exception of the relatively size and pathological type. The dissection at all
high rate of patients with postoperative epilepsy stages is performed under direct visual control,
that requires medical management—3.3 %. Tator and the cranial nerves can be identified early both
and Nedzelski [99] found that the combined in their proximal and distal ends, thus increasing
middle fossa translabyrinthine–transtentorial the chances for their preservation. If the RS
approach provides very clear visualization of the approach is combined with an intradural resection
CP angle and is especially suitable for large of the petrous apex with/without incision of the
tumors with anterior extensions. tentorium, petroclival lesions can be adequately
The transpetrosal retrosigmoid approach exposed [78]. This retrosigmoid suprameatal
is used in tumors larger than 4 cm that extend approach (RSMA), which was developed and
superiorly, well into the tentorial notch [37, 81, introduced by the senior author in 1982, provides
87]. The wide exposure affords an anterolateral access to meningiomas of the Meckel’s cave, the
access to the lateral brain stem surface from petroclival area, and even the posterior cavernous
the jugular foramen to the trochlear nerve and sinus [16, 80]. It evades the risks related to alterna-
multiple working angles. The transmastoid tive approaches, such as extensive petrous bone
Anesthetic Considerations 123

resection or retraction of the temporal lobe with or venous drainage. Compression or traction
the associated risks of damage to neural and vas- injury to the peripheral nerves should be
cular structures. Wide access to the IAC—if avoided.
needed—can be gained by drilling of its poste- Having experience with all patient positions,
rior wall. Lesions involving the inframeatal area we favor the semi-sitting one (Fig. 4.2). Its major
of the petrous bone—inferior and/or anterome- advantages are:
dial to the IAC—may be accessed via the so- • The spontaneous outflow of CSF, blood, and
called retrosigmoid inframeatal approach. This fluids used for irrigation is facilitated by
technique was elaborated by the senior author gravity.
and utilized for the first time in 2004. The • There is no need of constant suction to keep
approach provides access to the area inferior and the operative field clear, and the surgeon can
anteromedial to the internal auditory canal, the work bimanually.
infralabyrinthine region, and the petrous apex • The continuous irrigation of the operative field
and has been used successfully for removal of performed by the assistant obviates the need
chondrosarcoma, cholesterol granuloma, and for frequent coagulation during tumor
chordoma (see Chap. 11, section “Temporal Bone removal.
and Petrous Apex Lesions”). The RS approach • The surgeon can easily change his line of
provides excellent access to the jugular foramen sight, for example, to the brain stem or to the
area: lesions extending into the canal can be IAC, by slightly changing his position without
accessed by removing its dorsal wall (see the the need of rotation of the table from side to
corresponding chapter). Tumors extending to the side.
foramen magnum level can be removed via a ret- • Importantly, there is less engorgement of the
rosigmoid craniotomy and a C1 hemilaminectomy/ cerebellum, the venous outflow is promoted,
laminectomy (see the section “Foramen Magnum and the venous bleeding is less.
Meningiomas”) [79].

Anesthetic Considerations
Patient Positioning and Anesthesia
Considerations In all patients operated in the semi-sitting posi-
tion, central venous line is required to monitor
The position of the patient—semi-sitting, prone, the central venous pressure, as well as to apply
three-quarter lateral (or park bench), lateral- catecholamines or hyperosmolar fluids, if needed.
oblique, supine-oblique, concorde, or supine with A slightly elevated central venous pressure with
the head rotated 90° to the contralateral side—is normo- or slight hypervolemia is aimed. Almost
a matter of surgeons’ experience and institutional all surgeries in the semi-sitting position are per-
tradition. With the supine position, the head is formed with a positive PEEP. Important fact to
turned to the contralateral side and a pillow consider, however, is that the PEEP does not
(sandbag or bolster) is placed under the ipsilat- decrease the risk of venous air embolism. The
eral shoulder to rotate the shoulders and chest incidence of air embolism is similar in patients
into a semilateral position. The advantages of operated with a PEEP of 10 cm H20 and with
this position are that it is simple, fast, and not PEEP of 3 cm H20 [33]. The tip of the central
related to major complications. Besides the risks venous line has to be positioned exactly at the
to the cervical spine, however, the rotation of the transition between the superior caval vein and the
head can lead to venous outflow impediment with right atrium, which allows aspiration of air if
resultant increased intracranial pressure and venous air embolism occurs. Arterial line is
parenchymal bleeding. Alternatively, the patient required to measure the blood pressure invasively
can be positioned in the three-quarter lateral posi- and for blood gas analysis. Patients are monitored
tion, which does not endanger the cervical spine throughout the surgery with precordial Doppler.
124 4 Approaches to the Cerebellopontine Angle

a b

c d

Fig. 4.2 The semi-sitting position: the patients’ head is extended, flexed, and slightly rotated to the contralateral side.
The right leg of the first patient (e) is prepared for sural nerve harvesting
Anesthetic Considerations 125

e f

i
j

Fig. 4.2 (continued)


126 4 Approaches to the Cerebellopontine Angle

Transesophageal echocardiography is not applied Rarely, in some elderly or compromised patients,


routinely in our department. systemic hypotension may be more difficult
to control. Care is taken to protect all pressure
points in order to avoid peripheral nerve pal-
The Semi-sitting Position sies. Antithrombotic stockings or an intermittent
pneumatic compression device is used to prevent
The semi-sitting position differs from the sitting deep vein thrombosis.
one by the smaller angle of flexion of the operat- Some authors defined certain contraindications
ing table (Fig. 4.2). The operating table is flexed, for the semi-sitting position, such as age, arterial
and the legs of the patient are elevated to the level hypertension, obstructive lung disease, and diag-
of the right atrium of the heart, which elevates the nosed persistent foramen ovale [69]. The issue if
venous pressure and decreases the risk of venous these conditions should be regarded as a relative
air embolism. The head is immobilized with a or absolute contraindication is related to the expe-
three-point head fixation frame. Under the con- rience of the team—the anesthesiological and the
tinuous feedback control from the SSEP mea- surgical ones. An assessment of the benefit–risk
surement, the head is slightly extended, flexed, ratio should be made in each individual case.
and rotated to the involved side (Fig. 4.2). The The risks related to the semi-sitting position are
degree of rotation depends on the location of venous air embolism, paradoxical air embolism,
the tumor. In case of pure intracanalicular VS, tension pneumocephalus, circulatory instability,
the rotation is approximately 30° because the tra- and cervical spine injury, which may cause even
jectory is from medial to lateral; in tumors with quadriplegia. In the literature the sitting and semi-
larger extracanalicular portion, it should be >30° sitting positions are frequently discussed together.
to allow adequate visualization of the brain stem. Many of the risks related to the sitting position,
Hyperflexion that can lead to occlusion of the however, are avoided in the semi-sitting one.
jugular venous outflow, airway obstruction, or Venous air embolism occurs when an open
complications related to the cervical spine should venous structure draws in air due to the negative
be avoided: the distance between the manubrium venous pressure gradient between the operative
and the chin should be at least two fingerbreadths. site and the right atrium. It has been reported to
In case of deterioration of the SSEP waveform, occur in 10–45 %, even in 100 %, of the patients
the head should be repositioned and the degree of depending on the sensitivity of the monitor-
its flexion should be reduced. In the literature ing device [57, 66]. Venous air embolism can
some cases of postoperative quadriplegy or quad- lead to severe cardiopulmonary complications.
riparesis in patients operated in the semi-sitting However, if the anesthetic and operative prin-
position have been reported [61]. In order to ciples are followed strictly, such complications
avoid such outcome, we perform routinely in all can be avoided. In experienced hands the venous
patients cervical spine X-ray in flexion and exten- air embolism is not related to any lasting mor-
sion before surgery to detect potential instability bidity. Transesophageal echocardiography is the
and/or severe degenerative spine disease. In most specific and sensitive method in detection
patients with NF-2 cervical spine, MRI should of such embolism. An alternative method is the
also be done to exclude the presence of tumors, continuous monitoring of the pulmonary artery
compressing the spinal cord. If these examina- pressure. Our experience has shown that the
tions prove that a patient is at risk, the head combined monitoring of end-tidal carbon dioxide
flexion should be less. and precordial Doppler echocardiography yields
An acute drop in systolic blood pressure of similar results, and we rely exclusively on the
more than 20 mmHg may occur after position- combined use of these two techniques. An accu-
ing but is easy to manage by increasing the rate rately placed precordial Doppler is so sensitive
of intravenous infusion or by the administra- that it can detect the clinically irrelevant volume
tion of 1-mg adrenalin or noradrenalin [112]. of 0.015 ml air/kg/min [34].
Anesthetic Considerations 127

In patients with persistent foramen ovale, Venous air embolism should be detected early,
there is certain risk of paradoxical embolism in and immediate measures should be taken to detect
the arterial system that could cause cerebral and/ and occlude its source/s. The most common
or heart ischemia [28]. The condition is diag- sources of major venous air embolism are the
nosed in 25–30 % of the normal population [39, transverse and sigmoid sinuses and the jugular
59]. Persistent foramen ovale is demonstrated bulb, which—due to their dural attachments—do
directly using transthoracic echocardiography or not collapse if opened. Air entry may also occur
Color Doppler study, which shows the flow across via emissary veins and diploic veins of the occip-
the atrial septum. Transesophageal echocardiog- ital bone. Veins in the posterior cervical region
raphy is preferred in case the atrial septum is are generally small and collapse easily; still, they
inadequately visualized by transthoracic echocar- can also be the source of air entry. The risk of air
diography. A strong relationship between embolism is not confined to the initial part of sur-
persistent foramen ovale and clinically significant gery. It can develop even during tumor removal
paradoxical air embolism, however, has not been and—besides the aforementioned causes—can
established [28]. In the experience of the senior be caused by an undetected rupture of a bridging
author with approximately 10,000 cases, oper- vein.
ated in the semi-sitting position, major lasting If a change in the Doppler signal is noticed,
complications related to paradoxical air embo- especially in case a concomitant decrease in end-
lism have never been observed. Preoperative tidal carbon dioxide is observed, air embolism
examinations for persistent foramen ovale are should be suspected. A direct confirmation is the
therefore warranted only in children or in patients aspiration of air bubbles through the central
with suspicious clinical history. In case an opened venous catheter. Nonsurgical causes of air embo-
foramen is diagnosed, the decision how to per- lism, such as untight connections or incidental air
form the surgery should be made considering infusion, should be excluded. Ventilation with a
carefully the potential benefits and risks of the FiO2 of 1.0 is recommended to prevent a poten-
semi-sitting position and discussing the pros and tial hypoxemia. If nitrous oxide is used, it should
cons with the anesthesiological team. During the be stopped. Venous air embolism can cause acute
last 10 years, we selected the supine position just strain to the right heart and acute right heart fail-
in one child with a significant right-to-left shunt, ure. Later on, left heart failure may follow. Volume
while all other cases were operated in the semi- therapy or catecholamines application may be
sitting position. Lasting complications due to required to keep an adequate systemic and coro-
venous air embolism have not been registered. nary perfusion pressure. Aspiration of air via the
Other authors have also shown that the morbidity central venous catheter might be of help.
due to venous air embolism is similar in both the The surgeon has to be informed immediately at
semi-sitting and supine positions [25]. Zeilstra first signs of venous air embolism and both jugu-
et al. [112] studied a group of 108 patients and lar veins should be compressed manually. This
recorded 23 instances of venous air embolism in stops further air entry and helps the surgeon to
20 patients. It did not lead, however, to any seri- identify its entry point. If the patient is in the
ous hemodynamic changes. Duke et al. [25] semi-sitting position, a venous lesion, even a tear
reviewed 432 consecutive operations: 222 per- of the sinus, may not cause obvious bleeding. The
formed with the patients in the sitting position assistant should continuously irrigate the surgical
and 210 with the patients in the supine position. field. In case a vein in the soft tissues is bleeding,
The incidence of venous air embolism was 28 % it can be coagulated; in case of bleeding from
in the first group and 5 % in the second group. emissary veins, bone wax is applied. If the origin
Their analysis showed that although the inci- of bleeding is a tear in the sinuses, a small piece of
dence of detected venous air embolism is higher sponge, for example, Gelita (Johnson & Johnson
in sitting patients, the morbidity is not statisti- Medical), is used to seal it. In the rare case the
cally greater. source remains unclear, the craniotomy area is
128 4 Approaches to the Cerebellopontine Angle

covered temporarily with multiple wet sponges, characteristics. In patients with more bulky
and a wet gauze is laid. In case of continuous air subcutaneous soft tissue, the incision line
entry that causes hemodynamic problems, the should be more medial to not obstruct the
upper part of the surgical table has to be lowered mediolateral optical view of the surgeon to the
and the end-expiratory pressure increased. The fundus of IAC. The superficial muscle layer
surgery is continued when the end-tidal carbon (composed of the trapezius and sternocleido-
dioxide stops to fall because the complete recov- mastoid muscles) and the intermediate muscle
ery of this value is a slower process [28]. The layer (splenius capitis, longissimus capitis, and
degree of blood gas abnormality dictates the need semispinalis capitis muscles) of the lateral neck
of postoperative ventilator support. are cut in line with the incision and carefully
separated from their attachments to the bone
with subperiosteal dissection. The suboccipital
Standard Instrument Sets Used muscles and the soft tissues between the occiput,
in All CP Angle Surgeries the posteromedial end of the mastoid, and the
posterior arch of the atlas should be dissected
The standard set of microinstruments used for the carefully in order to avoid injury to the underly-
intradural part of the retrosigmoid tumor removal ing vertebral artery.
is presented on Fig. 4.3. During VS surgeries The asterion is the converging point of the
there is seldom need of additional instruments. In lambdoid, parietomastoid, and occipitomastoid
case the endoscope-assisted microsurgical tech- sutures. This landmark overlies the junction of
nique is implemented, however, some other the transverse and sigmoid sinuses and tradition-
instruments are required, for example, angled ally guided the placement of the initial burr hole.
forceps or suction tips. It has been recommended to place the burr hole
behind the transverse–sigmoid junction at the
asterion or below it [52, 70]. Some recent stud-
Skin Incision and Craniotomy ies, however, have shown that the asterion is not
absolutely a reliable anatomic landmark and is
Different skin incisions have been proposed: lin- variable both in the cranial–caudal plane and in
ear vertical incision, inverted J-shaped incision, the anterior–posterior plane [23]. The superior
lazy S-shaped incision, and semi-curved or nuchal line has been recommended as a guide for
C-shaped incision (Fig. 4.4). Principally, the inci- the location of the transverse sinus but is also
sion should be designed so that it allows adequate variable. The course of the sigmoid sinus is less
bone exposure and does not restrict the viewing variable: it descends along an axis defined by the
line and access to all parts of the CP angle. mastoid tip and the squamosal–parietomastoid
Optimizing wound healing and obtaining good suture junction or over the mastoid groove. We
cosmetic result are further considerations. prefer to place the burr hole 2–2.5 cm below the
In case of CP angle tumors, we perform a superior nuchal line, two-thirds behind and one-
slightly curved skin incision, 2.5–3.5 cm behind third in front of the occipitomastoid suture. In
the mastoid (Fig. 4.4b). The inferior part of the case an emissary vein is seen in the area or sus-
incision should be slightly curved laterally to pected on the bone-window CT images, the
allow approach to the mastoid tip, which is placement of the burr hole should be modified
essential to allow for an unrestricted access by accordingly in order to avoid injury to the vein.
the soft tissues to the cerebellomedullary cis- The surgeon should be aware that less venous
tern and lower part of the CP angle (the lateral- bleeding during the incision and craniotomy, as
to-medial perspective). The length and position well as a slack dura, could be related to a low
of the incision in relation to the mastoid should venous blood pressure, respectively, to a low
be adapted to the patient’s individual anatomic intracranial pressure, which increases the risk of
Anesthetic Considerations 129

a h

b k

c l

m
d

e
o

f p

q
g

Fig. 4.3 (a) Standard microinstrument set (Aeskulap sizes. (h) Microscissor. (i) Angled microhook, large size.
AG). (b–r) Close view of these microinstruments. Their (j) Angled microhook, small size. (k) Microdissectors.
use is described in the text. (b) Bayonet forceps. (c) (l) Platelet knives of various sizes. (p–r) Angled micro-
Bayonet forceps (sharp). (d–g) Tumor forceps of various hook of various sizes

venous air embolism. The anesthesiologist should ence has shown that the craniotomy might be a
be informed and adequate volume therapy initi- more dangerous procedure due to the related
ated or the PEEP should be elevated. high risk of injury to underlying sinuses and
The issue of whether to make a craniectomy tearing of the emissary vein or dura by the cran-
or craniotomy is a matter of debate. Our experi- iotome. Therefore, we avoid making one-piece
130 4 Approaches to the Cerebellopontine Angle

a b

Fig. 4.4 (a) Lazy S skin incision (the initial operation viewing angle is limited by the overlying soft tissues.
has been performed elsewhere): the access to the lateral (b) C-shaped incision that we favor
cerebellomedullary cistern and the lateral-to-medial

craniotomy in the RS approach, although we of the sinuses, approximately 2–3 mm, have to be
performed it earlier. The bone is removed step- exposed (Fig. 4.7).
wise with bone rongeurs and/or high-speed drill The size of the tumor does not generally
after the underlying dura has been detached from dictate the size of craniotomy: the edges of the
the bone. sinuses should be exposed always, and the bone
The exposure of the sigmoid sinus may require opening has to extend caudally to the floor (the
opening of mastoid air cells (Fig. 4.5). horizontal plane) of the posterior fossa
Nevertheless, the bone removal should proceed (Fig. 4.8). This affords direct access to the lat-
to the sinus edge. The opened air cells are sealed eral cerebellomedullary cistern without the
off with fat pieces and fibrin glue at the end of need of excessive cerebellar retraction. Further,
surgery. the dural incision can be extended basally to
The location of the emissary vein/veins should allow continuous egress of CSF throughout the
be studied on the thin-sliced bone-window CT surgery. Small tumors of the petrous apex are
(Fig. 4.6). Excessive traction to the vein during an exception. They are approached via a supe-
the craniectomy could cause its rupture from the rolateral route (the cerebellum is retracted inf-
sinus and sinus laceration. The vein should be ero-medially) and, therefore, the extent of
hence skeletonized with a diamond drill along inferior bone opening may be less (see section
their length until it is free of any bony encase- “Petroclival Meningiomas”). The medial extent
ment and can be safely coagulated close to their of the bone opening should be large enough to
entrance in the sinus. provide access to the lateral part of IAC (the
The dura overlying the sigmoid sinus is fre- angle of access to the fundus) even in small
quently very fragile; in order to preserve the last, intracanalicular VSs. In case of pure CP angle
millimeters of overlying bone are removed with a tumors, the foramen magnum should not be
diamond drill. Extensive exposure of the trans- exposed. In craniocervical tumors or CP angle
verse and sigmoid sinuses is unnecessary and tumors with such caudal extension, however, it
might lead to sinus laceration or desiccation, with might be necessary to open the foramen
the risk of subsequent thrombosis: just the edges magnum.
Anesthetic Considerations 131

a b

Fig. 4.5 Bone-window CTs, demonstrating the medial pneumatization has been found to correlate with the risk
extension of the mastoid air cell system. Still, the cells of postoperative CSF leak [96]. Therefore, the occlusion
have to be opened, and sufficient bone should be removed of the opened air cells with fat pieces and fibrin glue in
to expose the sigmoid sinus. The extent of petrous bone such cases should be especially meticulous

Dura Incision too close to the sinuses, its suturing may be more
difficult. On the contrary, if it is incised far from
The dura is opened under the operating micro- the sinuses, the abundant dura restricts the over-
scope. Once again, a large variety of dural inci- view and the access to the CP angle. The medial
sions have been proposed: vertical, curvilinear, dural flap is left as a protective layer on the cere-
or stellate, with or without additional relaxing bellum, while the lateral dural part is retracted
incisions or division of the dural flap into triangles. with two stay sutures—one is placed in the area of
The curvilinear dural incision favored by us is the sinus knee and another in the mid-sigmoid
placed approximately 1.5–2 mm medial to the sig- sinus level (Fig. 4.9b). The slight traction of these
moid and inferior to the transverse sinus (Fig. 4.9a). sutures and of the dura enlarges additionally the
It allows for unrestricted access to the CP angle CP angle exposure. Importantly, the traction does
and IAC and—importantly—for a primarily water- not lead to venous stasis because the overlying
tight closure. Auxiliary dural incisions are not nec- bone protects the sinuses. A wide brain spatula is
essary. Thus, the need of dural reconstruction can used to elevate slightly the lower lateral cerebel-
be avoided in almost all cases. If the dura is incised lum (Fig. 4.9c) and to visualize the lateral cerebel-
132 4 Approaches to the Cerebellopontine Angle

a b

c d

Fig. 4.6 Bone-window CT in four patients, demonstrat- veins are not visible on the CT scan but may be recog-
ing the emissary veins (arrow). The emissary veins are nized at surgery
highly variable in size, location, and number. Some small

lomedullary cistern. The arachnoid of the cistern is bridging veins and prevent any venous outflow
opened as much as necessary to allow sufficient disturbance.
amount of CSF to be drained until the cerebellum Microsurgical dissection proceeds as the
relaxes away from the petrous bone. Although lesion type and location dictate and is discussed
some authors recommend inserting the tip of a in the corresponding chapters of this book.
small catheter or cottonoid in the cistern in order to
ensure continuous CSF outflow throughout the
procedure [65], in the semi-sitting position, this is Hemostasis, dural closure and mastoid
not necessary. Care should be taken to recognize occlusion
bleeding from the supracerebellar bridging veins
that may tear during CSF draining, especially in During tumor removal, brisk bleeding may occur;
patients with slightly atrophic brain. A quadran- the bleeding vessel should be exposed and
gular flat cottonoid is then laid over the cerebel- coagulated. In case of diffuse bleeding, however,
lum, and a self-retaining retractor is placed coagulation would not be helpful. Moreover,
that should support and protect the cerebellar uncontrolled coagulation must be avoided to
hemisphere, instead of compressing it. An prevent thermal injury to the vulnerable neural
attempt should be made to preserve the major structures. The irrigation performed by the
Anesthetic Considerations 133

a b

Fig. 4.7 Exposure of the sigmoid (SS) and transverse with methyl methacrylate (Palacos ®). (d) Postoperative
sinuses (TS). The preoperative bone-window CT is CT, demonstrating the extent of the bony removal (arrow-
presented on a (arrow: sigmoid sinus). (b) Intraoperative heads), reconstructed with Palacos. The long arrow points
image, showing the standard retrosigmoid bone opening at the sigmoid sinus, located in a bony groove. Note that
with exposure of 2–3 mm of the transverse (TS) and sig- only a small part of this bone covering has been removed
moid sinuses (SS). (c) The skull has been reconstructed and the sinus remained protected throughout the procedure

assistant clears the operative field and allows with a cottonoid as long as necessary to control
continuing the dissection. Final hemostasis the bleeding. If this is unsuccessful, the vessel
should be done only following tumor removal should be dissected and mobilized from the nerve
and reliable identification of all neural structures. and only then coagulated at a very low current.
If the source of bleeding is a vessel along or too Blood clothes that have been accumulated in the
close to the nerves or to the brain stem, coagula- basal cisterns should be meticulously removed
tion is prohibited. A piece of Tabotamp ® is put after tumor removal because they may hide an
onto the bleeding point and compressed gently active bleeding point.
134 4 Approaches to the Cerebellopontine Angle

epidural space. Use of bone wax is not advisable.


The wax may cause severe infectious complica-
tions and we avoid it, except to control bleeding
from the bone edges. We use multiple small fat
pieces harvested from the appropriate subcutane-
ous layer at the incision site. These pieces should
not simply cover the mastoid air cells—they
should be plugged in the bone openings and
sealed with fibrin glue.

Reconstruction of the Posterior


Skull Base

This stage is essential to avoid postoperative


complications, such as pseudomeningocele, head-
ache, or poor cosmetic result. A large piece of
Gelita sponge is placed over the dura, and the
posterior skull base is reconstructed with methyl
methacrylate. The still soft compound is poured
into the bone defect on the sponge, formed to the
desired shape, and then allowed to harden
(Fig. 4.7c). It should optimally overlap slightly
Fig. 4.8 Retrosigmoid craniotomy in a patient with a the adjacent bone edges (Fig. 4.10a). In case the
small VS. The edges of the transverse and sigmoid sinuses bone implant remains mobile, it can be fixed with
are exposed. Note the widely opened mastoid air cells miniplates or with CranioFix® (Aesculap AG),
similarly to the supratentorial craniotomy flaps
The lower intracranial pressure and lack of (Fig. 4.10b). The wound is closed in layers.
venous stasis are beneficial factors for surgery
with the patient in the semi-sitting position.
However, they increase the risk of venous bleed- Drawbacks of the RS Approach
ing when the patient resumes his normal posi-
tion in the bed. The source of such bleeding When discussing the drawbacks of a given
typically is undetected ruptured and collapsed operative technique, one should be cautious to
bridging vein/s. The elevation of venous pres- generalize his experience. Frequently, a given
sure, most easily by performing jugular veins approach is blamed for the bad outcome, while
compression, readily identifies such veins. The the application of suboptimal operative technique
jugular veins compression is performed while or lack of sufficient knowledge/experience is not
the retractor is still in place and a second time considered (Fig. 4.11).
after its removal.
Two adapting dural sutures are placed and the
dura is closed with a continuous suture. In case Limited Exposure of the Fundus
watertight closure is not possible or the dura is
extremely thinned, fat pieces and fibrin glue are The exposure of the most lateral part of the IAC
additionally placed. (its fundus) may be limited by the location of
The opened mastoid air cells have to be semicircular canals and vestibule (Fig. 4.12). One
occluded to prevent a CSF leak (if a defect in study has shown that in approximately 30 % of
the dura exists) and to prevent air collecting in the the cases, the posterior semicircular canal, the
Drawbacks of the RS Approach 135

a b

Fig. 4.9 Intraoperative images. (a) The dura has been (asterisk) and the retractor blade have been placed, thus
incised along the transverse and the sigmoid sinuses, and exposing the tumor in the depth of the CP angle. (c) View
a wide spatula is used to elevate the cerebellum and expose of the VS under higher magnification
the lateral cerebellomedullary cistern. (b) Two stay sutures

crus communis, and even the vestibule are posi- crest in VS arising from the inferior vestibular
tioned along the visual axis between the surgeon nerve. With the MF approach, the hearing is often
and the lateral third of the IAC [98]. In order to lost exactly during tumor dissection from the fun-
avoid damage to the labyrinth in such cases, the dus of the IAC [24, 38, 84].
removal of the most lateral tumor part must be This limitation of the RS approach, however,
accomplished blindly. An important fact to con- can be overcome with the technical modifications
sider, however, is that similar problem exists with that we use (described in the Chap. 5). The very
the MF approach: frequently the transverse crest low rate of incomplete VS removal and of recur-
blocks the visualization of the inferior half of the rences, as well as the functional outcome in our
fundus and of the inferior recess [18]. This is and in other large published series, is a proof that
especially important in case of deep falciform this limitation is rather theoretical.
136 4 Approaches to the Cerebellopontine Angle

a b

Fig. 4.10 Postoperative bone-window CT scans, demonstrating the reconstruction of the occipital bone with methyl
methacrylate. The implant may be fixed with miniplates or with CranioFix® (Aeskulap AG), as shown on b

Need of Cerebellar Retraction Postoperative Headache

This is another frequently mentioned “drawback” The RS approach is frequently blamed for caus-
of the RS approach. Indeed, uncontrolled retrac- ing postoperative occipital headache. Rates
tion of the cerebellar hemisphere can lead to between 2.8 and 83 % have been reported, but
damage of the cerebellum or of the draining veins most authors accept a rate of 3–10 % as more
and can cause lasting neurological dysfunction. realistic [18, 40, 47, 54]. Indeed some studies
To avoid such retraction, the lateral cerebelom- demonstrated that this complaint occurs more fre-
edullary cistern has to be opened immediately after quently and is typically more severe after RS
the dural incision, and sufficient CSF should be approach than after translabyrinthine or MFA [76,
allowed to egress. Some surgeons state that the RS 86]. Staecker et al. [95] registered such headache
approach does not permit a direct access to the in 16 % of their patients operated via the middle
arachnoid cisterns, thus restricting the CSF outflow, fossa corridor and in 46 % of those operated via
especially in large tumors. In our experience, nei- the RS approach. In most current series of ret-
ther tumor size nor the presence of intracranial rosigmoid tumor removal, however, the headache
hypertension has precluded the access to this cis- rate is from 0 to 5 % [63, 65, 77, 88, 105].
tern, provided that sufficient amount of bone has The pathogenetic factors related to the occur-
been removed along the lower one-third of the sig- rence of headache are not clearly identified.
moid sinus. The self-retaining retractor should be Possible causes are adhesions between the richly
placed only when the cerebellar pressure is reduced innervated dura to the nuchal muscles or to sub-
sufficiently: to protect rather than to compress the cutaneous tissues with subsequent traction during
hemisphere. If a stronger retraction is needed, then neck motion, dural tension in cases of direct dural
the amount of CSF drained is still insufficient. closure, chemical meningitis from bone dust
Excessive cerebellar retraction may also lead contaminating the subarachnoid space during
to traction injury of the vulnerable cochlear intradural drilling, and use of fibrin glue that can
nerve and—more rarely—of the facial nerve. also lead to aseptic meningitis [40, 47, 85, 105].
The continuous monitoring of BAEP allows for Direct or indirect injury—during retraction—to
early recognition of any cochlear nerve conduc- the greater and lesser occipital nerves has also
tive dysfunction. This feedback allows adapting been proposed as a possible mechanism [68].
the tension on the retractor and the duration of Probably the headache is provoked by a combi-
retraction. nation of these mechanisms.
Drawbacks of the RS Approach 137

a b

Fig. 4.11 MR images (a, b) of a patient with VS, oper- (a). The tumor had been approached via a transcerebellar
ated in another neurosurgical facility via the “retrosigmoid route (b), and just a small part of the tumor has been
approach.” A paramedian suboccipital craniotomy had removed. The tumor was removed completely at our insti-
been performed that did not reach the sigmoid sinus tute via the “preformed” route. (c) Postoperative CT

Different techniques have been introduced to and neck muscles. In our experience this tech-
prevent postoperative headache such as prevent- nique is related to optimal outcome: less than
ing the use of fibrin glue, limiting the extent of 1 % of the patients had persistent headaches.
drilling of the posterior aspect of the IAC or gen-
erous irrigation and suction of the debris during
intradural drilling, duraplasty instead of primary Pneumocephalus and Tension
dural closure, and replacement of the bone flap at Pneumocephalus
the end of surgery or cranioplasty [88, 105]. The
methyl methacrylate cranioplasty offers the pos- Air entry in the subdural space, usually bifrontal
sibility of achieving a better cosmetic result and or more rarely intraventricular, subarachnoid, or
prevents scar tissue formation between the dura in the tumor bed area (Fig. 4.13) is a common
138 4 Approaches to the Cerebellopontine Angle

a b

Fig. 4.12 (a, b) The location of the semicircular canals prevents complete exposure of the fundus of the IAC
(a-arrow)

finding after surgery in the semi-sitting posi- Patients with symptomatic tension pneumoceph-
tion and is asymptomatic in most of the patients. alus and radiological evidence of more severe
We have never observed any serious or last- compression of the ventricles may require burr
ing complications due to pneumocephalus. In hole trepanation and evacuation of the air collec-
case of larger volume, the air can cause tran- tion. Such additional surgery had to be performed
sient headache, nausea, and vomiting. Tension very rarely in our series: in 0.15 % of all patients
pneumocephalus with prolonged emergence operated in the semi-sitting position (Figs. 4.14
from anesthesia or drowsiness is a rare finding. and 4.15).
Drawbacks of the RS Approach 139

a b

c d

Fig. 4.13 Various forms of pneumocephalus: typical condition, and only then a decision for the management
subdural bifrontal air collection (a, b), subarachnoid (c), should be taken. None of these patients required surgery
in the tumor bed area (d), and intraventricular (e, f). The for evacuation of the air
radiological findings have to be correlated to the patients’
140 4 Approaches to the Cerebellopontine Angle

e f

Fig. 4.13 (continued)

a b

Fig. 4.14 Massive intraventricular air collection after had strong headache and nausea. Hence, an EVD had to
removal of a very large CP angle meningioma, extending be inserted (b)
to the foramen magnum (a). The patient was drowsy and
References 141

a b

Fig. 4.15 Massive intraventricular, subarachnoid, and cisternal air collection in a patient, operated for a right-sided
epidermoid tumor. The patient was treated with an EVD for 4 days

6. Bento RF, De Brito RV, Sanchez TG, Miniti A (2002)


References The transmastoid retrolabyrinthine approach in ves-
tibular schwannoma surgery. Otolaryngol Head Neck
1. Anderson DE, Leonetti J, Wind JJ, Cribari D, Fahey K Surg 127(5):437–441. doi:10.1067/mhn.2002.129824,
(2005) Resection of large vestibular schwannomas: S0194599802002243 [pii]
facial nerve preservation in the context of surgical 7. Brackmann D (1979) Middle cranial fossa approach.
approach and patient-assessed outcome. J Neurosurg In: House WF, Luetje CM (eds) Acoustic tumors, vol
102(4):643–649. doi:10.3171/jns.2005.102.4.0643 II, Management. University Park Edn., Baltimore,
2. Angeli RD, Piccirillo E, Di Trapani G, Sequino G, pp 5–41
Taibah A, Sanna M (2011) Enlarged translabyrinthine 8. Brackmann D (1993) Translabyrinthine/transcochlear
approach with transapical extension in the manage- approaches. In: Sekhar LN, Janecka IP (eds) Surgery
ment of giant vestibular schwannomas: personal of cranial base tumors. Raven, New York, pp
experience and review of literature. Otol Neurotol 351–365
32(1):125–131. doi:10.1097/MAO.0b013e3181ff7562 9. Brackmann DE (1991) Acoustic neuroma: surgical
3. Arriaga MA, Brackmann DE, Hitselberger WE (1993) approaches and complications. Ann Acad Med
Extended middle fossa resection of petroclival and Singapore 20(5):674–679
cavernous sinus neoplasms. Laryngoscope 103(6): 10. Brackmann DE, Green JD Jr (2008) Translabyrinthine
693–698. doi:10.1288/00005537-199306000-00020 approach for acoustic tumor removal. 1992. Neurosurg
4. Arts HA, Telian SA, El-Kashlan H, Thompson BG Clin N Am 19(2):251–264. doi:10.1016/j.
(2006) Hearing preservation and facial nerve outcomes nec.2008.02.007, S1042-3680(08)00008-9 [pii], vi
in vestibular schwannoma surgery: results using the 11. Brackmann DE, House JR 3rd, Hitselberger WE
middle cranial fossa approach. Otol Neurotol 27(2): (1994) Technical modifications to the middle fossa
234–241. doi:10.1097/01.mao.0000185153.54457.16, craniotomy approach in removal of acoustic neuro-
00129492-200602000-00016 [pii] mas. Am J Otol 15(5):614–619
5. Bennett M, Haynes DS (2007) Surgical approaches and 12. Brennan JW, Rowed DW, Nedzelski JM, Chen JM
complications in the removal of vestibular schwanno- (2001) Cerebrospinal fluid leak after acoustic neuroma
mas. Otolaryngol Clin North Am 40(3):589–609. surgery: influence of tumor size and surgical approach
doi:10.1016/j.otc.2007.03.007, S0030-6665(07)00042-4 on incidence and response to treatment. J Neurosurg
[pii], ix–x 94(2):217–223. doi:10.3171/jns.2001.94.2.0217
142 4 Approaches to the Cerebellopontine Angle

13. Briggs RJ, Fabinyi G, Kaye AH (2000) Current man- 26. Ebersold MJ, Harner SG, Beatty CW, Harper CM Jr,
agement of acoustic neuromas: review of surgical Quast LM (1992) Current results of the retrosigmoid
approaches and outcomes. J Clin Neurosci 7(6): approach to acoustic neurinoma. J Neurosurg
521–526. doi:10.1054/jocn.2000.0728, S0967- 76(6):901–909. doi:10.3171/jns.1992.76.6.0901
5868(00)90728-8 [pii] 27. Enee V, Guerin J, Bebear JP, Darrouzet V (2003)
14. Briggs RJ, Luxford WM, Atkins JS Jr, Hitselberger Acoustic neuroma surgery. Results and complications
WE (1994) Translabyrinthine removal of large acous- in 348 cases. Rev Laryngol Otol Rhinol (Bord)
tic neuromas. Neurosurgery 34(5):785–790; discus- 124(1):45–52
sion 790–791 28. Engelhardt M, Folkers W, Brenke C, Scholz M,
15. Bryce GE, Nedzelski JM, Rowed DW, Rappaport JM Harders A, Fidorra H, Schmieder K (2006)
(1991) Cerebrospinal fluid leaks and meningitis in Neurosurgical operations with the patient in sitting
acoustic neuroma surgery. Otolaryngol Head Neck position: analysis of risk factors using transcranial
Surg 104(1):81–87 Doppler sonography. Br J Anaesth 96(4):467–472.
16. Chanda A, Nanda A (2006) Retrosigmoid intradural doi:10.1093/bja/ael015, ael015 [pii]
suprameatal approach: advantages and disadvantages 29. Fagan PA, Sheehy JP, Chang P, Doust BD, Coakley D,
from an anatomical perspective. Neurosurgery 59(1 Atlas MD (1998) The cerebellopontine angle:
Suppl 1):ONS1–ONS6. doi:10.1227/01.NEU. does the translabyrinthine approach give adequate
0000220673.79877.30, 00006123-200607001-00001 access? Laryngoscope 108(5):679–682. doi:10.1097/
[pii]; discussion ONS1–ONS6 00005537-199805000-00010
17. Chen TC, Giannotta SL, Brackmann DE (1993) 30. Fisch U (1970) Transtemporal surgery of the internal
Acoustic neuromas. Translabyrinthine approach. In: auditory canal. Report of 92 cases, technique, indica-
Apuzzo MLJ (ed) Brain surgery. Complication avoid- tions and results. Adv Otorhinolaryngol 17:203–240
ance and management. Churchill Livingstone, 31. Friedman RA, Brackmann DE, van Loveren HR,
New York, pp 1772–1800 Hitselberger WE (1997) Management of the con-
18. Colletti V, Fiorino F (2003) Middle fossa versus ret- tracted mastoid in the translabyrinthine removal of
rosigmoid-transmeatal approach in vestibular schwan- acoustic neuroma. Arch Otolaryngol Head Neck Surg
noma surgery: a prospective study. Otol Neurotol 123(3):342–344
24(6):927–934 32. Garcia-Ibanez E, Garcia-Ibanez JL (1980) Middle
19. Darrouzet V, Guerin J, Aouad N, Dutkiewicz J, fossa vestibular neurectomy: a report of 373 cases.
Blayney AW, Bebear JP (1997) The widened retro- Otolaryngol Head Neck Surg 88(4):486–490
labyrinthe approach: a new concept in acoustic neu- 33. Giebler R, Kollenberg B, Pohlen G, Peters J (1998)
roma surgery. J Neurosurg 86(5):812–821. Effect of positive end-expiratory pressure on the inci-
doi:10.3171/jns.1997.86.5.0812 dence of venous air embolism and on the cardiovascu-
20. Day JD (2011) The middle fossa approach and lar response to the sitting position during neurosurgery.
extended middle fossa approach: technique and oper- Br J Anaesth 80(1):30–35
ative nuances. Neurosurgery. doi:10.1227/NEU. 34. Gildenberg PL, O’Brien RP, Britt WJ, Frost EA
0b013e31823583a1 (1981) The efficacy of Doppler monitoring for the
21. Day JD, Chen DA, Arriaga M (2004) Translabyrinthine detection of venous air embolism. J Neurosurg
approach for acoustic neuroma. Neurosurgery 54(1):75–78. doi:10.3171/jns.1981.54.1.0075
54(2):391–395; discussion 395–396 35. Gjuric M, Wigand ME, Wolf SR (2001) Enlarged
22. Day JD, Fukushima T, Giannotta SL (1994) middle fossa vestibular schwannoma surgery: experi-
Microanatomical study of the extradural middle fossa ence with 735 cases. Otol Neurotol 22(2):223–230;
approach to the petroclival and posterior cavernous discussion 230–231
sinus region: description of the rhomboid construct. 36. Glasscock ME 3rd, Kveton JF, Jackson CG, Levine
Neurosurgery 34(6):1009–1016; discussion 1016 SC, McKennan KX (1986) A systematic approach to
23. Day JD, Kellogg JX, Tschabitscher M, Fukushima T the surgical management of acoustic neuroma.
(1996) Surface and superficial surgical anatomy of the Laryngoscope 96(10):1088–1094
posterolateral cranial base: significance for surgical 37. Gormley WB, Sekhar LN, Wright DC, Kamerer D,
planning and approach. Neurosurgery 38(6):1079– Schessel D (1997) Acoustic neuromas: results of cur-
1083; discussion 1083–1084 rent surgical management. Neurosurgery 41(1):50–
24. Driscoll CL, Jackler RK, Pitts LH, Banthia V (2000) 58; discussion 58–60
Is the entire fundus of the internal auditory canal vis- 38. Haberkamp TJ, Meyer GA, Fox M (1998) Surgical
ible during the middle fossa approach for acoustic exposure of the fundus of the internal auditory canal:
neuroma? Am J Otol 21(3):382–388 anatomic limits of the middle fossa versus the ret-
25. Duke DA, Lynch JJ, Harner SG, Faust RJ, Ebersold MJ rosigmoid transcanal approach. Laryngoscope 108(8
(1998) Venous air embolism in sitting and supine Pt 1):1190–1194
patients undergoing vestibular schwannoma resection. 39. Hagen PT, Scholz DG, Edwards WD (1984) Incidence
Neurosurgery 42(6):1282–1286; discussion 1286–1287 and size of patent foramen ovale during the first 10
References 143

decades of life: an autopsy study of 965 normal hearts. 55. Maddox HE 3rd (1977) The lateral approach to acous-
Mayo Clin Proc 59(1):17–20 tic tumors. Laryngoscope 87(9 Pt 1):1572–1578.
40. Harner SG, Beatty CW, Ebersold MJ (1993) Headache doi:10.1288/00005537-197709000-00017
after acoustic neuroma excision. Am J Otol 56. Mamikoglu B, Wiet RJ, Esquivel CR (2002)
14(6):552–555 Translabyrinthine approach for the management of
41. House W (1979) Translabyrinthine approach. In: large and giant vestibular schwannomas. Otol Neurotol
House WF, Luetje CM (eds) Acoustic tumors, 23(2):224–227
Management. University Park Press, Baltimore, pp 57. Mammoto T, Hayashi Y, Ohnishi Y, Kuro M (1998)
43–87 Incidence of venous and paradoxical air embolism in
42. House WF, De la Cruz A, Hitselberger WE (1978) neurosurgical patients in the sitting position: detection
Surgery of the skull base: transcochlear approach to by transesophageal echocardiography. Acta
the petrous apex and clivus. Otolaryngology Anaesthesiol Scand 42(6):643–647
86(5):ORL-770–ORL-779 58. Mangham CA (1988) Complications of translabyrin-
43. House WF, Hitselberger WE (1976) The transcochlear thine vs. suboccipital approach for acoustic tumor
approach to the skull base. Arch Otolaryngol surgery. Otolaryngol Head Neck Surg 99(4):396–400
102(6):334–342 59. Matjasko MJ, Hellman J, Mackenzie CF (1987)
44. House WF, Hitselberger WE, Horn KL (1986) The Venous air embolism, hypotension, and end-tidal
middle fossa transpetrous approach to the anterior- nitrogen. Neurosurgery 21(3):378–382
superior cerebellopontine angle. Am J Otol 7(1):1–4 60. Monfared A, Mudry A, Jackler R (2009) The history of
45. House WF, Shelton C (1992) Middle fossa approach middle cranial fossa approach to the cerebellopontine angle.
for acoustic tumor removal. Otolaryngol Clin North Otol Neurotol. doi:10.1097/MAO.0b013e3181c0e98e
Am 25(2):347–359 61. Morandi X, Riffaud L, Amlashi SF, Brassier G (2004)
46. House WF, Shelton C (2008) Middle fossa approach Extensive spinal cord infarction after posterior fossa
for acoustic tumor removal. 1992. Neurosurg Clin N surgery in the sitting position: case report. Neurosurgery
Am 19(2):279–288, S1042-3680(08)00010-7 [pii], vi 54(6):1512–1515; discussion 1515–1516
47. Jackson CG, McGrew BM, Forest JA, Hampf CR, 62. Morrison AW, King TT (1973) Experiences with a
Glasscock ME 3rd, Brandes JL, Hanson MB (2000) translabyrinthine-transtentorial approach to the cere-
Comparison of postoperative headache after retrosig- bellopontine angle. Technical note. J Neurosurg
moid approach: vestibular nerve section versus ves- 38(3):382–390. doi:10.3171/jns.1973.38.3.0382
tibular schwannoma resection. Am J Otol 63. Mosek AC, Dodick DW, Ebersold MJ, Swanson JW
21(3):412–416 (1999) Headache after resection of acoustic neu-
48. Kanzaki J, Shiobara R, Toya S (1991) Classification roma. Headache 39(2):89–94. doi:10.1046/j.
of the extended middle cranial fossa approach. Acta 1526-4610.1999.3902089.x, HEDhed3902089 [pii]
Otolaryngol Suppl 487:6–16 64. Ohata K, Haque M, Morino M, Nagai K, Nishio A,
49. Kawase T, Shiobara R, Toya S (1991) Anterior transpet- Nishijima Y, Hakuba A (1998) Occlusion of the sig-
rosal-transtentorial approach for sphenopetroclival moid sinus after surgery via the presigmoidal-trans-
meningiomas: surgical method and results in 10 patients. petrosal approach. J Neurosurg 89(4):575–584.
Neurosurgery 28(6):869–875; discussion 875–876 doi:10.3171/jns.1998.89.4.0575
50. Kawase T, Shiobara R, Toya S (1994) Middle fossa 65. Ojemann RG (2001) Retrosigmoid approach to acous-
transpetrosal-transtentorial approaches for petroclival tic neuroma (vestibular schwannoma). Neurosurgery
meningiomas. Selective pyramid resection and radi- 48(3):553–558
cality. Acta Neurochir (Wien) 129(3–4):113–120 66. Papadopoulos G, Kuhly P, Brock M, Rudolph KH,
51. King TT, Morrison AW (1980) Translabyrinthine and Link J, Eyrich K (1994) Venous and paradoxical air
transtentorial removal of acoustic nerve tumors. embolism in the sitting position. A prospective study
Results in 150 cases. J Neurosurg 52(2):210–216. with transoesophageal echocardiography. Acta
doi:10.3171/jns.1980.52.2.0210 Neurochir (Wien) 126(2–4):140–143
52. Lang J Jr, Samii A (1991) Retrosigmoidal approach to 67. Parry D (1904) A case of tinnitus treated by division of
the posterior cranial fossa. An anatomical study. Acta the auditory nerve. J Laryngol Otol 19(19):402–406
Neurochir (Wien) 111(3–4):147–153 68. Pedrosa CA, Ahern DK, McKenna MJ, Ojemann RG,
53. Lanman TH, Brackmann DE, Hitselberger WE, Subin Acquadro MA (1994) Determinants and impact of
B (1999) Report of 190 consecutive cases of large headache after acoustic neuroma surgery. Am J Otol
acoustic tumors (vestibular schwannoma) removed 15(6):793–797
via the translabyrinthine approach. J Neurosurg 69. Porter JM, Pidgeon C, Cunningham AJ (1999) The
90(4):617–623 sitting position in neurosurgery: a critical appraisal.
54. Levo H, Blomstedt G, Hirvonen T, Pyykko I (2001) Br J Anaesth 82(1):117–128
Causes of persistent postoperative headache after sur- 70. Rhoton AL Jr (2000) The cerebellopontine angle and
gery for vestibular schwannoma. Clin Otolaryngol posterior fossa cranial nerves by the retrosigmoid
Allied Sci 26(5):401–406, 492 [pii] approach. Neurosurgery 47(3 Suppl):S93–S129
144 4 Approaches to the Cerebellopontine Angle

71. Rhoton AL Jr (2000) The temporal bone and tran- cerebellopontine angle component. Laryngoscope
stemporal approaches. Neurosurgery 47(3 112(8 Pt 1):1500–1506. doi:10.1097/00005537-
Suppl):S211–S265 200208000-00031
72. Rhoton AL Jr, Pulec JL, Hall GM, Boyd AS Jr (1968) 85. Schaller B, Baumann A (2003) Headache after removal
Absence of bone over the geniculate ganglion. J of vestibular schwannoma via the retrosigmoid
Neurosurg 28(1):48–53. doi:10.3171/jns.1968. approach: a long-term follow-up-study. Otolaryngol
28.1.0048 Head Neck Surg 128(3):387–395. doi:10.1067/
73. Rhoton AL Jr, Tedeschi H (2008) Microsurgical anat- mhn.2003.104, S0194599802233136 [pii]
omy of acoustic neuroma. 2002. Neurosurg Clin N 86. Schessel DA, Rowed DW, Nedzelski JM, Feghali JG
Am 19(2):145–174. doi:10.1016/j.nec.2008.02.005, (1993) Postoperative pain following excision of
S1042-3680(08)00006-5 [pii], v acoustic neuroma by the suboccipital approach: obser-
74. Roche PH, Pellet W, Moriyama T, Thomassin JM vations on possible cause and potential amelioration.
(2008) Translabyrinthine approach for vestibular Am J Otol 14(5):491–494
schwannomas: operative technique. Prog Neurol Surg 87. Sekhar LN, Schessel DA, Bucur SD, Raso JL, Wright
21:73–78. doi:10.1159/000156708, 156708 [pii] DC (1999) Partial labyrinthectomy petrous apicec-
75. Rodgers GK, Luxford WM (1993) Factors affecting tomy approach to neoplastic and vascular lesions of
the development of cerebrospinal fluid leak and men- the petroclival area. Neurosurgery 44(3):537–550;
ingitis after translabyrinthine acoustic tumor surgery. discussion 550–552
Laryngoscope 103(9):959–962. doi:10.1288/ 88. Sepehrnia A, Knopp U (2001) Osteoplastic lateral
00005537-199309000-00003 suboccipital approach for acoustic neuroma surgery.
76. Ryzenman JM, Pensak ML, Tew JM Jr (2004) Patient Neurosurgery 48(1):229–230; discussion 230–231
perception of comorbid conditions after acoustic neu- 89. Shelton C (1995) Unilateral acoustic tumors: how
roma management: survey results from the acoustic often do they recur after translabyrinthine removal?
neuroma association. Laryngoscope 114(5):814–820. Laryngoscope 105(9 Pt 1):958–966. doi:10.1288/
doi:10.1097/00005537-200405000-00005, 00005537- 00005537-199509000-00016
200405000-00005 [pii] 90. Shelton C, Brackmann DE, House WF, Hitselberger
77. Samii M, Gerganov V, Samii A (2006) Improved pres- WE (1989) Middle fossa acoustic tumor surgery:
ervation of hearing and facial nerve function in ves- results in 106 cases. Laryngoscope 99(4):405–408.
tibular schwannoma surgery via the retrosigmoid doi:10.1288/00005537-198904000-00009
approach in a series of 200 patients. J Neurosurg 91. Shiobara R (1980) A modified extended middle cra-
105(4):527–535. doi:10.3171/jns.2006.105.4.527 nial fossa approach for acoustic tumors (author’s
78. Samii M, Gerganov VM (2008) Surgery of extra-axial transl). Neurol Med Chir (Tokyo) 20(2):173–182
tumors of the cerebral base. Neurosurgery 62(6 Suppl 92. Shiobara R, Ohira T, Inoue Y, Kanzaki J, Kawase T
3):1153–1166. doi:10.1227/01.neu.0000333782. (2008) Extended middle cranial fossa approach for
19682.76, 00006123-200806001-00024 [pii]; discus- vestibular schwannoma: technical note and surgical
sion 1166–1168 results of 896 operations. Prog Neurol Surg 21:65–72.
79. Samii M, Klekamp J, Carvalho G (1996) Surgical doi:10.1159/000156595
results for meningiomas of the craniocervical junction. 93. Slattery WH 3rd, Brackmann DE, Hitselberger W
Neurosurgery 39(6):1086–1094; discussion 1094–1095 (1997) Middle fossa approach for hearing preserva-
80. Samii M, Tatagiba M, Carvalho GA (2000) tion with acoustic neuromas. Am J Otol
Retrosigmoid intradural suprameatal approach to 18(5):596–601
Meckel’s cave and the middle fossa: surgical tech- 94. Sluyter S, Graamans K, Tulleken CA, Van Veelen CW
nique and outcome. J Neurosurg 92(2):235–241. (2001) Analysis of the results obtained in 120 patients
doi:10.3171/jns.2000.92.2.0235 with large acoustic neuromas surgically treated via the
81. Sampath P, Holliday MJ, Brem H, Niparko JK, Long translabyrinthine-transtentorial approach. J Neurosurg
DM (1997) Facial nerve injury in acoustic neuroma 94(1):61–66. doi:10.3171/jns.2001.94.1.0061
(vestibular schwannoma) surgery: etiology and pre- 95. Staecker H, Nadol JB Jr, Ojeman R, Ronner S,
vention. J Neurosurg 87(1):60–66. doi:10.3171/ McKenna MJ (2000) Hearing preservation in acoustic
jns.1997.87.1.0060 neuroma surgery: middle fossa versus retrosigmoid
82. Sanna M, Falcioni M, Rohit (2003) Cerebro-spinal approach. Am J Otol 21(3):399–404
fluid leak after acoustic neuroma surgery. Otol 96. Sterkers JM, Desgeorges M, Corlieu P, Sterkers O
Neurotol 24(3):524 (1984) Translabyrinthine ablation of voluminous
83. Sanna M, Russo A, Taibah A, Falcioni M, Agarwal M acoustic neuromas. Ann Otolaryngol Chir Cervicofac
(2004) Enlarged translabyrinthine approach for the 101(1):9–14
management of large and giant acoustic neuromas: a 97. Tanriover N, Sanus GZ, Ulu MO, Tanriverdi T, Akar
report of 175 consecutive cases. Ann Otol Rhinol Z, Rubino PA, Rhoton AL Jr (2009) Middle fossa
Laryngol 113(4):319–328 approach: microsurgical anatomy and surgical tech-
84. Satar B, Jackler RK, Oghalai J, Pitts LH, Yates PD nique from the neurosurgical perspective. Surg Neurol
(2002) Risk-benefit analysis of using the middle fossa 71(5):586–596. doi:10.1016/j.surneu.2008.04.009,
approach for acoustic neuromas with >10 mm S0090-3019(08)00408-4 [pii]; discussion 596
References 145

98. Tatagiba M, Samii M, Matthies C, el Azm M, 105. Wazen JJ, Sisti M, Lam SM (2000) Cranioplasty in
Schonmayr R (1992) The significance for postopera- acoustic neuroma surgery. Laryngoscope 110(8):1294–
tive hearing of preserving the labyrinth in acoustic 1297. doi:10.1097/00005537-200008000-00013
neurinoma surgery. J Neurosurg 77(5):677–684. 106. Whittaker CK, Luetje CM (1985) Translabyrinthine
doi:10.3171/jns.1992.77.5.0677 removal of large acoustic tumors. Am J Otol
99. Tator CH, Nedzelski JM (1982) Facial nerve preser- 6(Suppl):155–160
vation in patients with large acoustic neuromas 107. Wigand ME, Haid T, Berg M (1989) The enlarged
treated by a combined middle fossa transtentorial middle cranial fossa approach for surgery of the tem-
translabyrinthine approach. J Neurosurg 57(1):1–7. poral bone and of the cerebellopontine angle. Arch
doi:10.3171/jns.1982.57.1.0001 Otorhinolaryngol 246(5):299–302
100. Tedeschi H, Rhoton AL Jr (1994) Lateral approaches 108. Wigand ME, Haid T, Berg M, Schuster B, Goertzen
to the petroclival region. Surg Neurol 41(3):180– W (1991) Extended middle cranial fossa approach
216, 0090-3019(94)90123-6 [pii] for acoustic neuroma surgery. Skull Base Surg
101. Tos M, Thomsen J (1979) Translabyrinthine surgery 1(3):183–187
of acoustic neurinoma. Acta Otolaryngol Suppl 109. Wigand ME, Rettinger G, Haid T, Berg M (1985)
360:45–47 Removal of acoustic neuromas of the cerebellopon-
102. Tos M, Thomsen J, Harmsen A (1988) Results of tile angle with transtemporal approach by the middle
translabyrinthine removal of 300 acoustic neuromas cranial fossa. HNO 33(1):11–16
related to tumour size. Acta Otolaryngol Suppl 110. Yasargil M (1996) Acoustic neuromas, vol IVb,
452:38–51 Microneurosurgery. Thieme, New York
103. Tummala RP, Coscarella E, Morcos JJ (2005) 111. Yates PD, Jackler RK, Satar B, Pitts LH, Oghalai JS
Transpetrosal approaches to the posterior fossa. (2003) Is it worthwhile to attempt hearing preserva-
Neurosurg Focus 19(2):E6, 190206 [pii] tion in larger acoustic neuromas? Otol Neurotol
104. Wackym PA, Andrews JC (1995) Middle cranial 24(3):460–464
fossa approach. In: Cheatham M, Becker DP, Samii 112. Zeilstra DJ, Groen RA (1999) Venous air embolism
M (eds) Atlas of cranial base surgery. W. B. Saunders, in sitting and supine patients undergoing vestibular
Philadelphia, pp 26–31 schwannoma resection. Neurosurgery 44(2):426
Vestibular Schwannomas
5

Epidemiology asymptomatic VSs is 570/100,000 (0.57 %).


A study of 240 temporal bones performed at
VSs comprise 6–8 % of all intracranial tumors, Johns Hopkins University Hospital showed an
25–33 % of the posterior cranial fossa, and incidence of 2.7 % [90]. These rates are, how-
80–94 % of the CP angle tumors [32, 58, 126, ever, generally evaluated as too high. According
246, 254]. Meningiomas are the second most to recent MRI studies, the incidence of asymp-
common CP angle tumor (3–10 %), followed by tomatic VSs is 0.07 % or even 0.2 % [5, 251].
epidermoids (2–4 %). Much more rare tumors are Most of these tumors never become symptom-
the schwannomas of other cranial nerves: trigem- atic, probably due to arrested or extremely slow
inal nerve, facial nerve or lower cranial nerves, growth [271].
paragangliomas, metastases, lipomas, osteogenic The clinical incidence of VSs (or the incidence
sarcomas, osteomas, or tumors extending sec- of diagnosed VSs) has been estimated to range
ondarily into the CP angle [13, 20]. from 0.2 to 5/100,000/year; most authorities
The average patients’ age at diagnosis is 45–55 agree that it is between 1 and 2/100,000/year [67,
years [22, 32, 83, 145]. VSs are more frequent in 90, 94, 117, 242, 261]. The consensus statement
females, and the female-to-male relation ranges by the NIH in 1991 estimated that in the USA
from 1:0.8 to 1.5:1. Frohlich and Sutherland [70] 2,000 to 3,000 VSs are diagnosed each year,
reported that the incidence is similar in both gen- which corresponds to a symptomatic incidence of
der but age differences exist: males with VS are 1/100,000 [187]. The rate of detection of VSs was
usually 30–39 years old, while in female the peak found to correspond to the distribution of radio-
is in the period 60–69 years. The incidence of logical facilities and to the awareness of the doc-
sporadic VSs in children is very low [3]. tors: a gradual increase of the annual incidence of
VSs can be divided into three groups: asymp- diagnosed tumors from 3.1 per million in 1976 to
tomatic tumors (occult or silent VS without 22.8 per million in 2004 in Denmark paralleled
subjective or objective symptoms), symptomatic the wider distribution of CT and MRI [225, 247].
but not diagnosed tumors, and diagnosed tumors In 2008 the incidence decreased to 19.4 per mil-
[246]. It is impossible to determine the actual lion, while the mean tumor size decreased from
incidence of VSs because of the unknown inci- 30 mm in 1979 to 10 mm in 2008.
dence of asymptomatic and symptomatic undiag- VSs occur in all races but some authors believe
nosed tumors in the population. The incidence of that racial differences exist [32]. The incidence
VSs, according to pathological examination of was found to be 1.7/100,000/year in the white
temporal bones, is between 0.57 and 2.7 % [85, population and just 0.1/100,000/year in the black
203, 222, 244]. Schuknecht [208] in a study of population of South Africa [210]. Preston-Martin
1,400 pyramids found that the incidence of et al. [177] found that the incidence among the

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 147


DOI 10.1007/978-3-642-35422-9_5, © Springer-Verlag Berlin Heidelberg 2013
148 5 Vestibular Schwannomas

Afro-Americans from Los Angeles is approxi- methylation [116]. Other potential mechanisms
mately 0.2/100,000/year. It is hypothesized that of gene inactivation are the posttranscriptional
these differences may be due to the varying muta- alternative splicing and differential polyadenyla-
tion rate of the NF2 gene in different races. tion, as well as influence of a modifier gene [27].
Another plausible explanation is the lower rate of The clinical response to different mutations is
diagnosing of the tumor due to differences in the heterogeneous. No clear correlation between the
social status and accessibility of MRI devices. genotype and phenotypic features of VSs, such as
tumor growth rate and cyst formation, could be
yet established [102, 165, 229, 260]. Even within
Biology of VS a given NF-2 family with the same mutation,
phenotypic variability has been seen. Patients
Nowadays the molecular cascade of events lead- with missense mutations have statistically greater
ing to VS formation is much better understood, survival than those with nonsense and frameshift
but this knowledge is still insufficient to allow mutations [14]. Mutations causing truncation of
precise prediction of the individual tumor evolu- the NF-2 protein, as a result of insertions or dele-
tion. Correlation between the tumor’s biological tions that lead to disruption of the merlin open
characteristics and its clinical behavior has yet reading frame (frameshift mutations) or of pre-
not been found. Detailed understanding of the mature termination codons (nonsense mutations),
molecular biology and genetics of VSs should have been reported to cause more severe clinical
ultimately allow developing a targeted molecular phenotype [103, 165, 236]. These findings, how-
therapy that will stop tumor progression or even ever, could not be confirmed in all studies.
eradicate preexisting tumors. Notably, not all VSs exhibit inactivation of the
NF2 gene, showing that this gene may not be the
only determinant of their origin [236]. Mutation
Molecular Genetics of the NF2 gene is found in 60 % of sporadic VSs
[183]. Tumorigenesis in the remaining cases
Both sporadic and NF-2-associated VSs are could be explained by the influence of other
caused by loss of or mutation of the NF2 tumor mechanisms, for example, of epigenetic factors.
suppressor gene on chromosome 22q12. The NF2 Using microarray technology, Caye -Thomasen
gene was independently identified by Troffater et al. [31] identified 78 genes that were
et al. [251] and Rouleau et al. [188] in 1993 and significantly up- or downregulated in tumor tis-
encodes for a cell membrane-related protein. The sue. Further studies are necessary to identify
majority of NF2 mutations are truncating muta- which of them act together with the NF2 gene.
tions that cause the synthesis of a smaller and
probably nonfunctional protein product. Mutations
within the NF2 gene have been identified in The NF2 Protein
50–60 % of spontaneous meningiomas and a small
proportion of spontaneous ependymomas [190]. The NF2 gene encodes a 69-kDa protein consist-
The occurrence of any VS requires inactiva- ing of 595 amino acids that was named merlin
tion of both allelic NF2 genes (double hit model), (Moesin–ezrin–radixin-like protein) or schwan-
which is caused by mutation, loss of the long nomin (derived from “schwannoma”) [188, 251].
arm, or loss of the whole chromosome 22. The This protein is similar to the 4.1 superfamily of
frequency, type, and distribution of these muta- proteins—the ERM family—which are involved
tions are different in the sporadic and in the in cellular remodeling and link the actin cytoskel-
familial NF-2 tumors [260]. In NF-2 patients the eton to the plasma membrane [251]. All proteins
most common cause is point mutations, whereas of this family consist of a highly homologous
small deletions account for the majority of muta- N-terminal globular domain (the FERM domain),
tions in sporadic VSs [101, 102]. In 20–40 % of an a-helical stretch, and a charged C-terminus
the sporadic tumors, the genes are inactivated by [40]. The ERM proteins bind to actin in the
Pathogenesis of VS 149

cytoskeleton via their C-terminal domain and via case of deficient merlin is probably due to destabi-
a second actin-binding site in the N-terminal lized adherent junctions [123]. Merlin’s role in
domain. The FERM region interacts with a num- stabilizing these junctions might involve erbin—a
ber of molecules of the plasma membrane, includ- PDZ protein that binds indirectly to merlin [179].
ing membrane receptors. Interestingly, the merlin As pointed by Hanemann and Evans [89],
molecule contains a unique 7-amino acid domain there is accumulating evidence that merlin has an
that has been hypothesized to mediate its effects important role in the coordination of two relevant
[158]. Merlin is a part of two pathways, the pRb- and interdependent processes: cellular adhesion
CDK pathway and the PI3K/Akt/mTOR pathway, and growth factor receptor response. Although
which have a crucial role in cell-cycle regulation. merlin/schwannomin is expressed in all tissues
Many hypotheses have been put forward to affected in NF-2, it is also expressed in unaf-
explain the mechanism of tumorigenesis after fected tissues. Obviously, other proteins compen-
merlin loss. The molecules with which merlin sate for the lost function, or a differential
interacts are transmembrane proteins, scaffold- expression of important merlin/schwannomin
ing/adapter proteins, signaling molecules/ modifying molecules occurs in different tissues.
kinases, cytoskeletal proteins, as well as differ- Nuclear proliferation markers have been stud-
ent other proteins of unknown function [89]. ied for correlation to various clinical parameters.
Merlin plays an important role not only in actin Although some studies demonstrated a correla-
cytoskeleton-mediated processes, which are tion of tumor growth with an increase in Ki-67
highly relevant in myelination, but also in regu- index [160], the data so far are not conclusive.
lating cell proliferation, cell movement, and There is no consensus on the role of cellular
communication [50, 236]. Pelton et al. [168] factors on tumor growth in both sporadic VSs and
found that schwannoma cells from NF-2 tumors NF-2-related tumors. The influence of various
show major alterations in actin cytoskeleton and hormonal factors is also controversial. It is known
display abnormalities in cell spreading reversible that pregnancy and delivery can accelerate VS
on the reintroduction of merlin. Experiments growth. Cushing was the first to report the accel-
with NF2 knockout mouse and in vitro schwan- eration of VS growth during pregnancy in 1917.
noma preparations showed that loss of merlin Some studies have shown an increased concentra-
causes loss in contact inhibition, which conse- tion of estrogen and progesterone receptors on the
quently leads to tumorigenesis [236]. tumor cell surface and suggested a correlation
Merlin suppresses cellular growth via a yet between tumor behavior and expression levels of
unknown mechanism. Its overexpression in hormone receptors [30]. A recent study of Patel
mouse fibroblasts or rat schwannoma cells can et al. [166] showed that these hormonal receptors
limit cell growth [137]. Furthermore, the cell were upregulated in most VSs. Other authors
response to environmental growth regulatory fac- argue that the concentration of these receptors is
tors is supposed to be mediated by merlin. not higher in VS cells and is gender specific [54].
Consequently, its loss may lead to increased cell
growth and tumor formation [236].
Merlin is found primarily in actin-rich cellular Pathogenesis of VS
protrusions and in cell–cell and cell–matrix con-
tact sites [65, 123]. It may be involved also in cell Origin of VS
cycle-dependent nucleus shuttling [155]. Cells
lacking merlin show deregulated adhesion to VSs originate from the vestibular nerve. The exact
extracellular matrix [253]. Several studies demon- site of origin along the nerve, however, is unknown,
strated that merlin binds to adhesion molecules and their predilection for this particular nerve is
such as b1 integrin and layillin, as well as to still unexplained. Several theories have been pro-
molecules, which are part of the focal adhesion posed, but neither of them elucidates satisfactory
complexes, such as paxillin and focal adhesion all aspects of VS pathogenesis. Schwannomas
kinase [65, 105]. The impaired cell–cell contact in may arise anywhere from the glial–Schwann cell
150 5 Vestibular Schwannomas

junction to the termination of the nerve sheath at Site of Origin


the end organ. Most frequently, however, they Most VSs arise within the IAC: typically in its
arise from the transition zone between the central medial portion and rarely in its lateral end. VSs
and peripheral myelin, also known as glial– grow medially toward the CP cistern and to a lesser
Schwann cell junction or Obersteiner–Redlich degree laterally toward the fundus. Exceptionally,
zone [226]. This hypothesis explains the various the tumor can originate in the inner ear [66]. The
sites of tumor origin along the nerve because the category of “medial VS” has been introduced to
location of the junction is not constant. According designate those tumors that originate from the cis-
to the findings of one study, the distribution of ternal part of the nerve in the CP angle cistern
Schwann cells was nearly uniform among the (Fig. 5.1)—they arise just outside the porus and
facial, cochlear, superior, and inferior vestibular show no extension into the IAC [120, 248]. The
nerves [238]. It remains unclear why schwanno- incidence of such VSs is less than 2 % [96].
mas generally originate from the vestibular nerve However, if those VSs that have minimal extension
and spare the cochlear one. The amount of into the IAC are also defined as “medial VS,” the
Schwann cells was found to be higher along the incidence rises to 12 % [230]. These less strict cri-
distal vestibular nerve—a finding that can explain teria explain the observation of Strauss et al. [230]
the higher incidence of tumors originating in that that normal IAC had one-third of cases, porus
region [219]. According to other studies, however, enlargement had 20 %, and a funnel-shaped canal
their density along the distal portions of the ves- had almost 50 % of the patients with medial VS.
tibular nerves or at the glial–Schwann cell junc- MG Yasargil hypothesized that VS originate
tion was not increased [136, 226]. Another epiarachnoidally in the IAC and gradually push
hypothesis states that VSs originate in the area of the arachnoid membrane of the IAC medially
the vestibular (Scarpa’s) ganglion. The glial– into the CP angle. At this stage a single arachnoid
Schwann cell junction most often lies in the vicin- layer covers the tumor [268]. In the CP angle the
ity of vestibular ganglion, implying that both tumor and the brain stem are covered by their
theories might be correct. own arachnoid membrane. Therefore, a double
An interesting hypothesis—the embryologic arachnoid layer surrounds VSs reaching the brain
theory—has been formulated by Sterkers et al. stem. This theory has been questioned by Tarlov
[226], who suppose that VSs arise from imma- [241], Ohata et al. [162], and Lescane et al. [130].
ture cells existing in the adult superior and inferior Still, it has a major practical implication: tumor
vestibular ganglia. dissection should be performed within the arach-
noid space, leaving a layer of arachnoid on the
Nerve of Origin tumor and a layer on the pons.
VSs originating from the superior vestibular nerve, Very rarely VS originate from the distal seg-
which innervates the lateral semicircular canal, ment of the vestibular nerve and develop in the
show much more frequently a reduced caloric area of the vestibule or semicircular canals
response. Origin from the inferior vestibular nerve without extension into the IAC. These intralaby-
may be suspected by recording the rotatory nystag- rinthine schwannomas are regarded as a separate
mus [148]. At a certain stage, however, the tumor clinical entity from the more common VSs aris-
affects the function of both nerves, and the tests ing within the IAC and extending laterally to
become unreliable. The exact site and nerve of ori- involve the labyrinth.
gin can be determined at surgery and is reliable only
in case of small VS. According to some authors, Growth Pattern and Growth Rate
70–91.4 % of VSs originate from the inferior ves- VSs are typically slow-growing tumors, but their
tibular nerve [115, 120, 183]. Other authors point growth rate may be irregular over the time, even
out that the superior vestibular nerve is the more in a single patient. Some tumors enlarge rapidly,
common origin [141]. The currently dominating whereas others remain stable or even regress.
opinion, however, is that the tumor originates from The natural evolution of VSs cannot be predicted.
both nerves in approximately similar rates [44, 45]. At least five growth patterns of VSs have been
Pathogenesis of VS 151

a b

Fig. 5.1 MRI study of a patient with a “medial VS”: the IAC is completely free from tumor. (a, b) T1-weighted
sequences after gadolinium application. (c, d) CISS sequences

outlined: continuous growth (40 %), no measur- [135]. Longer follow-up studies showed that the
able growth (18 %), no measurable growth duration of follow-up correlates with a higher
followed by continuous growth (18 %), negative rate of tumors that show progressive growth ten-
growth (8 %), and various types of positive tumor dency [221]. Resumption of growth even after a
growth or irregular growth (16 %) [34]. The period of quiescence can occur.
reported rates of VSs that grow further, do not The annual growth rate of VSs is between 1
change, or even show involution or “negative and 2.4 mm/year [12, 48, 83, 94, 260]. It is known
growth” vary widely. Progressive tumor growth that cystic and the NF-2-related tumors have a
is observed in 54–91 % of the cases, no change of higher growth rate: up to 3.2 mm/year [34]. A sub-
tumor size in 30–40 %, and tumor involution in group of VSs, however, which do not differ
0–18 % [17, 157, 187]. The spontaneous involu- macroscopically or radiologically from the rest,
tion of some VSs may be explained by tumor have a propensity for more rapid growth. In the
necrosis caused by intratumoral thrombosis series evaluated by Bakkouri et al. [12], the average
152 5 Vestibular Schwannomas

growth rate was 1.15 ± 2.4 mm/year. In 10.2 % of The outcome of treatment of two series of patients
all VSs, however, tumor growth was ³3 mm/year. with “large VS” operated in the same institution
Tumors with extrameatal extension generally have was presented in 1994 Briggs et al. [26] and in
a higher growth rate [12, 87, 224], although not all 1999 [125]. In the first only VSs ³4 cm were
studies support this correlation [250]. Despite the included, while in the second all VSs ³3 cm were
large number of studies on the VS growth, the included. The reported outcome was respectively
clinical applicability of the findings, as pointed by better in the later series. The effect of the learning
Charabi et al. [36], remains limited. curve on outcome, however, is also responsible to
a certain extent for the differences.
At the Consensus Meeting on Systems for
Classification Reporting Results in Vestibular Schwannoma
held in Tokyo in 2001, an agreement was reached
Various methods of measurement of VS size are that VS size should defined based on measure-
still in use, confusing the comparison of the results ment of the linear tumor dimensions and not on
between different series. While some authors rely its volumetric measurements [47, 110]. The size
on measurement of maximal tumor diameter in of the intracanalicular and of the extracanalicular
the axial plane, others use the product of the cis- parts should be reported separately; however,
ternal diameters, anteroposterior × mediolateral; only the extracanalicular diameter should be
yet others recommend volumetric measurements. applied for the determination of tumors’ size. At
Controversial is also the inclusion of the intrac- the conference the following scale has been
analicular tumor part, which generally is between accepted (Table 5.1).
8 and 12 mm. The definition of “large” or “giant” The extent of tumor extension in CP angle is
VS also varies. the main factor determining the clinical presenta-
The classification of VS is based either on tions, the operative difficulty, and the outcome.
their maximal diameter or on the degree of exten- Therefore, we believe that any classification of VS
sion in the CP angle. There are several widely should consider this parameter. VSs of the same
accepted classification schemas. The classification absolute size may lead to different clinical picture.
of W. T. Koos is based on the maximal diameter Thus, in young patients with small subarachnoid
of VS [120]. Classifications, frequently applied space, they might reach and compress the brain
by neurootologists, are the one of D. E. Brackmann stem, while in elderly with significant brain atro-
[21] and Tos and Thomsen [249]. The classification phy, VS of the same size may still be located
proposed by L. Sekhar is similar [212]: entirely in the CP cistern (Fig. 5.2). The Hannover
• 0 stage—intracanalicular VS tumor extension system, introduced by the senior
• I stage—<1 cm author, is based on this criterion: tumor extension.
• II stage—1–2.5 cm This classification system has been modified
• III stage—2.5–4 cm recently: giant VSs are now considered as a sepa-
• IV stage—>4 cm rate fifth category (Fig. 5.3). Previous radiosurgi-
The complexity is made even higher by the use cal treatment renders the surgical tumor removal
of different criteria in different publications of the more difficult. In this set of patients, specific
same authors. Gormley and Sekhar [83] defined problems tend to appear (see the corresponding
VS ³4 cm as large, while Sekhar and Tzortidis chapter). Hence, this has to be specified explicitly
[212] defined as large those between 2.5 and 4 cm. (Table 5.2).

Table 5.1 Consensus meeting on Intracanalicular tumor


systems for reporting results in vestibular
Grade 1 Small 1–10 mm extrameatal
schwannoma classification [47, 110]
Grade 2 Medium 11–20 mm extrameatal
Grade 3 Moderately large 21–30 mm extrameatal
Grade 4 Large 31–40 mm extrameatal
Grade 5 Giant > 40 mm extrameatal
Classification 153

a b

Fig. 5.2 Examples of VSs with similar size but different not contact the brain stem. This issue—whether the tumor
relation to brain stem. In the first and second examples (a, contacts the brain stem or not—has a major clinical
b), the tumor reaches the brain stem, while in the third (c) significance: it determines the operative difficulty and,
the subarachnoid space is much larger and the tumor does respectively, the outcome

a b

Fig. 5.3 Axial and coronal MR images of VS of various application). Grade T1: (a, b). Grade T2: (c, d). Grade
extensions, according to the INI (Samii) classification of T3a: (e, f). Grade T3b: (g, h). Grade T4a: (i, j). Grade
VS extension (T1-weighted sequences after contrast T4b: (k, l). Grade T5: (m, n)
154 5 Vestibular Schwannomas

c d

e f

g h

Fig. 5.3 (continued)


Classification 155

i j

k l

m n

Fig. 5.3 (continued)


156 5 Vestibular Schwannomas

Table 5.2 INI (or Samii) Tumor extension


classification of VS extension grade Tumor extension in the IAC and CP angle
T1 Purely intracanalicular
T2 Intra-/extrameatal
T3a Filling the CPA cistern
T3b Reaching the brain stem
T4a Compressing the brain stem
T4b Severe compression and dislocation of brain stem and fourth
ventricle; obstructive hydrocephalus
T5 Giant tumors (maximal diameter >4 cm), extension over the
midline
In case the VS has been previously treated with radiosurgery, this is designated
with an additional “+R,” for example, stage “T4 a+R”

Neuroimaging T1-weighted MRI following gadolinium admin-


istration, they also appear as small areas of con-
VSs are isodense on native CT imaging; follow- trast enhancement. In contrast to VSs, however,
ing contrast administration, they enhance strongly. which are globular and more sharply delineated,
The bone-window high-resolution CT imaging is the inflammatory lesions show typically a more
highly sensitive in regard to the bony changes of linear or ill-defined enhancement [6]. Further
IAC. Furthermore, it demonstrates the normal clue is the absence of T2 abnormality and/or the
structures of the petrous bone, which are essen- size discrepancy between the lesions observed on
tial for surgery: emissary vein/s, jugular bulb, T1 and T2 sequences.
position of the labyrinth, etc. MRI is the gold Some more advanced neuroimaging modali-
standard for diagnosing VSs: at T1-weighted ties have been evaluated recently. Okamoto et al.
MRI images, VSs are isointense to slightly have demonstrated that a small focus of high sig-
hypointense, and on T2-weighted images, they nal T2 intensity may be observed in the dorsal
are hyperintense [19]. They enhance intensely brain stem ipsilateral to the tumor. They hypoth-
after contrast administration. The three patterns esized that this signal results presumably from
of contrast enhancement are homogeneous (in the degeneration of the vestibular nucleus associ-
50–60 %), heterogeneous (in 30–40 %), and cys- ated with VS [163]. The relative tumor blood vol-
tic (in 5–15 %) [38, 51]. Cystic VSs have to be ume (rCBV) is reported to be significantly lower
distinguished from CP angle epidermoids and in VSs than in meningiomas [88]. The presence
subarachnoid cysts, which are hypointense in T1 of microhemorrhages on T2-weighted gradient-
and hyperintense in T2 and do not enhance after echo sequences may be useful to differentiate VS
contrast administration [53]. from CP angle meningiomas [243]. Using proton
In contrast to CP angle meningiomas, VSs are MR spectroscopy, VSs could be differentiated
centered at the widened IAC, cause typically ero- from meningiomas by the presence of prominent
sion and/or dilatation of the IAC (in 70–90 % of myoinositol peak at 3.55 ppm; in contrast alanine
the cases), extend almost always into the IAC, is found only in meningiomas [41].
form acute angle to the posterior surface of the
petrous bone, and show intratumoral calcifications
or hyperostotic bone changes extremely rarely. Clinical Presentation
Although VSs lack the typical for meningiomas
“dural tail sign,” in rare cases enhancement of the The typical CP angle syndromes have been well
adjacent meninges is possible [85]. recognized since the end of the nineteenth century.
Small intracanalicular VSs have to be differ- Neurological dysfunction is a result of direct com-
entiated from some inflammatory or infectious pression of neurovascular structures, brain stem
conditions, such as viral neuropathy or arach- compression, obstruction of CSF flow, and intrac-
noiditis, following viral infection [94]: at ranial hypertension. The clinical presentation of
Clinical Presentation 157

patients with CP angle tumors is related to multi-


ple factors: type of the lesion, origin and extension
pattern, size, and growth rate.
The growth of VS follows a certain pattern
that traditionally has been divided into four
stages: intracanalicular (intrameatal), cisternal,
brain stem compressive, and hydrocephalic.
Each stage correlates closely with corresponding
clinical symptoms. VSs originate within the IAC
and gradually fill it; then, they expand into the
CP angle and occupy the CP angle cistern, dis-
placing the seventh and the eighth CNs, as well
as the AICA. During its further growth, the
tumor reaches, compresses, and then displaces
a
the brain stem and cerebellum. Depending on the
degree of tumors’ cranial and caudal extension,
the fifth and the lower cranial nerves become
involved. The hydrocephalic stage begins with
the obstruction of fourth ventricle and its outlets.
This four-stage concept, however, does not
reflect entirely the high variability of VS growth
and extension patterns; furthermore, the individ-
ual anatomical variability is also not considered
(Figs. 5.4 and 5.5).
The degree of bony changes of the IAC caused
by the growing VS differs: some VSs expand
along the direction of least resistance toward the
CP angle and lead to minimal or even no bone
changes; other VSs cause dilatation and even ero- b
sion of the IAC, which may be out of proportion
to overall tumor size (Figs. 5.6, 5.7, 5.8, and 5.9).
Yamakami et al. [265] showed that the extent of
IAC bony changes does not correlate with the
overall tumor size. In their material, widened
IAC had 46 % of the patients, extensively eroded
IAC, 17 %, and normal IAC, 36 %. Preoperative
hearing level was significantly more severely dis-
turbed in patients with extensive erosion of the
IAC compared to those with normal or only wid-
ened canal [265]. Badie et al. [9] demonstrated
that the pressure within the IAC is significantly
elevated in most patients and correlates to the
extent of tumor in the IAC and not to the total
tumor size. Their findings explain the frequently c
observed lack of correlation between the preop-
erative hearing function and tumor size. Thus, Fig. 5.4 MR images demonstrating the varying exten-
some patients with small intracanalicular VS sion pattern of the tumor in the CP angle. (a) Predominant
have profound auditory dysfunction, while others anteroposterior extension. (b) Predominant mediolateral
extension (the mediolateral tumor diameter is greater than
with large CP angle tumor extension have normal the anteroposterior one). (c) Round tumor shape
158 5 Vestibular Schwannomas

hearing. The enlargement of the IAC has proved


to be an unfavorable factor for postoperative
hearing preservation [45, 206]. The elevated pres-
sure on the cochlear nerve and on the labyrinthine
artery may increase their vulnerability during
tumor dissection.
The degree of filling of the IAC is also vari-
able (Fig. 5.10) and correlates with the probabil-
ity for hearing preservation [152, 222]. In an MRI
study of 106 patients, Mohr et al. [152] found
that the IAC was filled partially in 59 % (>1/2 in
21 % and >1/2 in 38 %) and completely in 41 %.
Incomplete filling of the IAC was an independent
a favorable factor in hearing preservation. Some
tumors even spare the IAC entirely—the so-
called medial VS—which were discussed earlier
in the text.
From a surgical point of view, the key issue is
not the absolute tumor size but the degree of brain
stem compression [145, 198].
Patients with intracanalicular VS present with
vestibulocochlear nerve dysfunction: hearing loss,
tinnitus, and/or vestibular dysfunction. Hearing
loss is usually of insidious onset and is the most
frequent symptom, observed in up to 95 % of
patients [145]. Audiograms reveal typically high-
frequency sensorineural hearing loss. Speech dis-
crimination is affected out of proportion to
b pure-tone hearing loss. Several theories have been
proposed to explain how VS causes hypacusis:
mechanical injury to the cochlear apparatus; bio-
chemical changes of inner ear fluid; ischemic
injury resulting from occlusion or spasm of the
labyrinthine artery; tumor compression, stretch-
ing, or infiltration of the cochlear nerve; or eleva-
tion of the intracanalicular pressure [122, 127,
207]. As Lapislava et al. [127] point, however, the
mechanism of auditory dysfunction is most likely

Fig. 5.5 MRI of VS with various growth patterns in


relation to the IAC. (a) The tumor is centered at the IAC
c and causes compression of both the brain stem and cere-
bellum. (b) The main tumor bulk is anterior to the IAC
and causes compression of the brain stem and middle
cerebellar peduncle (but not of cerebellum). (c) The main
tumor bulk is posterior to the IAC: the tumor compresses
mainly the middle cerebellar peduncle and the cerebellum
(minimal contact to brain stem)
Clinical Presentation 159

a b

c d

Fig. 5.6 Relatively small VS (grade 3a) causing severe dilatation of the IAC, which does not correspond to the overall
tumor size

multifactorial. The incidence of VS that presents extension. Vestibular symptoms (vertigo, dizziness,
with sudden hearing loss ranges from 3 to 26 % in and unsteadiness) frequently are not recognized by
various series [145, 207]. Interestingly, only the patients but are always discovered with special
1–2 % of all patients presenting with sudden hear- testing. The caloric test, which was performed
ing loss will be diagnosed to have a VS. widely before the wide distribution of MRI
Tinnitus is the second most common present- facilities, detects even mild vestibular changes.
ing symptom. Its incidence is 48–74 % of the During the cisternal stage, hearing deteriorates
patients and has an inverse correlation with tumor further and disequilibrium gradually replaces the
160 5 Vestibular Schwannomas

a
a

b
b
Fig. 5.7 VS grade T3b: the right IAC is not widened by
the tumor and is approximately similar to the contralateral
one Fig. 5.8 VS grade T3a with moderate enlargement of the
IAC (left side)

vertigo. The CP angle tumor portion is always


centered at the IAC; the structure and the resis- appear. Obstructive hydrocephalus results from
tance of surrounding arachnoid membranes prob- occlusion of CSF outflow at the stage of severe
ably influence its shape. The shape may be oval brain stem and fourth ventricle compression. Late
(if the shortest diameter, determined in the axial symptoms are contralateral long-tract signs,
plane of MRI, is less than 90 % of the longest severe gait disturbance, visual loss, and signs of
diameter), round (if this relation is equal or intracranial hypertension.
greater than 90 %), or polycyclic [77]. Oval VS Symptom presentation is related to VS size.
with larger mediolateral diameter may compress In a study on the clinical presentation of 1,000
the brain stem and lead to corresponding neuro- VSs, the senior author and C. Matthies [145]
logical symptoms to a degree, which seems out of showed that in small tumors the mean patients’
proportion to their overall size. Oval tumors with age was significantly higher; trigeminal hypes-
larger anteroposterior diameter may attain rela- thesia, dizziness, and subjective deafness dura-
tively large size with minimal brain stem com- tion were shorter; while tinnitus duration was
pression [99]. Later on trigeminal symptoms, longer. The most frequently involved CN was
headache, lower cranial nerve palsies, and ataxia the cochlear—90–97 % of the patient had some
Clinical Presentation 161

b c

Fig. 5.9 (a) The figure demonstrates a patient with NF-2


and bilateral VSs: although the right-sided tumor is larger
than the left-sided, its intrameatal part is smaller.
Respectively, the right IAC (b) is less dilated than the left
one (c)
e

hearing loss with an average duration of 3.7 Fig. 5.10 Examples of VS with varying extension in the
years. Deaf before surgery were 3 %. Headaches IAC (coronal CISS MRI sequences). (a, b) The tumor
extends up to the fundus. (c–e) The most lateral part of the
in patients with VS are typically in the occipital IAC is free of tumor. The arrows in c–e point at the CSF
region (12 % of patients in the senior authors’ lateral to the VS
series), unless related to an intracranial hyper-
tension syndrome. The patient’s subjective frequently indicates that the tumor actually is
awareness of trigeminal and facial nerve dys- schwannoma originating from the facial nerve.
function is lower than their actual rates, assessed Facial spasm and trigeminal neuralgia are very
objectively. Symptoms of trigeminal nerve dys- unusual symptoms in patients with VS.
function were reported by 8 %, whereas 19 % Symptoms of vestibular dysfunction had 61 %,
had trigeminal signs at examination; facial nerve and vestibular signs had 45–59 % of all patients.
weakness was noted by 6 % (in half of them due The lower cranial nerves were affected in 3 %
to previous surgery) but objectively found in (glossopharyngeal paresis in 2 % and hypoglos-
17 %. Preoperative facial weakness in previ- sal paresis in 1 %) and the abducens nerve in
ously untreated cases is exceptional and 1.8 % of the patients.
162 5 Vestibular Schwannomas

Management of Patients with VS a

Conservative Management

The follow-up or “wait-and-scan” strategy has


become an acceptable management option due to
the constantly growing number of incidentally
found VSs. An increasing number of patients
with small and asymptomatic tumors are being
currently managed conservatively, and the ratio-
nale is the lack of progress of some VSs. Current
MRI facilities allow for earlier VS detection,
when they are still asymptomatic and small in
size. Furthermore, MRI is a reliable means for
control of their growth (Fig. 5.11). b
The decision on the individual management
plan should always be taken together with the
patient and his family, considering not only the
tumors’ features and patient condition but also his
expectations and professional and daily activities.
We believe that initial observation is an appropri-
ate option in elderly or somatic unstable patients
with small tumors and mild stable symptoms, in
patients with small tumors and complete hearing
loss, in patients with VS on the only hearing side,
or in those unwilling to undergo surgery.
Some surgeons are still not inclined to advice
surgery until the tumor extends into the CP angle
or neurological deficits develop. The goal of mod-
Fig. 5.11 MR images of VS that shows a certain growth
ern VS surgery is preservation of functions. Surgery tendency (a, b). The second image (b) is made approxi-
at an earlier stage, in patients with smaller tumors mately 18 months after the first one. The patient was ini-
with available useful hearing and good BAEP tially considered as good candidate for hearing
response, allows for its preservation in high num- preservation. Shortly before the second MRI, he experi-
enced sudden loss of hearing on the left side
bers (see section “Hearing”). Many studies have
demonstrated that a significant risk of loss of use-
ful hearing during observation exists even in stable percent of the patients had to be operated due to
nongrowing VS—between 50 and 67 % [34, 82, tumor enlargement, while 6 % died of brain stem
87, 256]. Hearing deterioration is not always slowly herniation induced by tumor compression.
progressive; sudden hearing loss may develop in a Interestingly, 28 patients were assessed initially
single or repeated episodes. Thus, a hearing- as candidates for hearing preservation surgery;
preserving surgery might become impossible. during the observation period, however, this
In a widely cited study performed by the group option became impossible in 75 % of them due to
of Charabi et al. [34], 123 patients (127 tumors) tumor growth and/or deterioration of hearing.
were followed with a “wait-and-scan” policy over The further follow-up of the patients in this series
a 20-year period. Tumor growth was observed in was presented in a later paper, and the results were
74 %; 18 % of the tumors did not grow, and nega- even more convincing: 89 % of the candidates
tive tumor growth occurred in 8 %. Twenty seven for hearing preservation surgery lost eligibility
Management of Patients with VS 163

because of tumor growth or deterioration of


hearing level, or both [37].
It is still impossible to predict the tumor
growth rate in an individual patient, but in
10–50 %, the conservative treatment fails and
active management is necessary [12, 82, 256,
257, 263]. As pointed by Sughrue et al. [233],
this wide range can be explained by the small to
modest size of the reported series, the varying
methods for growth measurement, the lack of sta-
tistical power, and the surgeon’s bias.
If the hearing is already lost, the decision to
follow the tumor further or to remove it depends
a
on the individual patients’ wish. Slight tumor
enlargement does not necessarily indicate that
immediate treatment is required. No reliable pre-
dictive factors of the tumor growth pattern that
could support the therapeutic decision-making
have been identified up to now [87]. Tumors with
extrameatal extension have a higher tendency
for future growth [180, 257, 263]. According to
other studies, however, the overall growth rate
did not differ significantly between intrameatal
and extrameatal tumors [12, 34, 67]. Younger
age is also not consistently associated with more
rapid growth [12, 82]. Most authors did not find
any correlation between tumor growth rate and b
patients’ sex or age, tumor size, and hearing sta-
tus at presentation [67, 180]. The only predic- Fig. 5.12 Example of a growing VS (T1-weighted MRI
tive factor of tumor growth is the delay between study). The interval between the two images is 2 years
the appearance of the first symptoms and time
of diagnosis: a shorter delay in diagnosis cor- Surgical Treatment of VS
relates with the need of active therapy at some
point during the observation [12, 34]. Cystic At present surgery of VSs should be safe for the
tumors may display sudden and dramatic growth patient. Major morbidity rate should be reduced to
and are not suitable candidates for observational very low levels. Preservation of normal facial
management [72]. nerve function should be the rule, and useful hear-
A correlation between VS growth in the first ing preservation should be the goal. Another issue
year or years of follow-up and the risk of later to consider when discussing the management
growth has been found in many studies options is the preservation and even improvement
(Fig. 5.12) [67, 224, 263]. The tumor may of the patients’ quality of life. The complete
enlarge, however, at varying intervals during removal of these benign tumors is curative and is
follow-up; a previously “inactive” tumor may to be strived in every case. Still, avoidance of
still exhibit tumor growth after many years. major deficits and preservation of facial nerve
Hence, the radiological control—even in non- function have highest priority. Exceptions to com-
growing tumors—should not be restricted only plete removal hence are all those cases in which
to the first years of observation. the surgeons—due to the characteristics of a given
164 5 Vestibular Schwannomas

tumor or due to less experience—feel that contin- preferences and institutional tradition, are
uation of surgery may be too dangerous for the applied for removal of VS and are discussed in
patient. In case that further tumor dissection would the Chap. 4:
result in anatomic discontinuity of the facial nerve, • Translabyrinthine approach and its
a thin veil of tumor capsule attached to the most modifications: translabyrinthine–transtentorial
adherent portion of the nerve might be left. Tumor approach, extended translabyrinthine approach,
remnants are followed radiologically, and if a transmastoid transpetrosal partial labyrinthec-
growth tendency is shown, active treatment should tomy, retrolabyrinthine approach, combined
be considered: either with new surgery or with translabyrinthine–retrosigmoid approach
gamma knife, depending on the experience of the • Middle fossa approach and extended middle
surgeon. The size of the remaining tumor is an fossa approach, which allows removing of
important predictor of the risk for its future growth: VSs extending up to 2 cm into the CP angle
a significant difference exists in case the tumor • Retrosigmoid suboccipital approach
removal is near total (>95 % resection), subtotal Some surgeons select the approach depend-
(>90 %), or partial (<90 %) [18, 57, 213]. Other ing on tumor size, extension into the IAC, and
exceptions to the rule of complete tumor removal preoperative hearing level. We believe that the
are patients with NF-2, such as those with bilateral results depend more on the individual surgeons’
VSs and surgery on the only hearing side (see sec- experience rather than on the advantages or dis-
tion “Neurofibromatosis Type 2 and Other advantages of a particular approach. Excellent
Bilateral Cerebellopontine Angle Tumors” for results in terms of rate of complete removal and
detailed discussion). functional preservation of the facial nerve func-
Planned subtotal tumor removal with follow- tion have been achieved in experienced hands
up or immediate radiosurgery of the remnant was with each of these approaches. The reported rates
promoted as an option to reduce postoperative of complete tumor removal are 90–99 %, and the
morbidity [111, 113, 133, 217]. Considering the recurrence rates are 0.5–5 % [7, 22, 80, 125, 194,
high probability of growth of the remnant, espe- 211, 244, 264]. The rates of anatomic and func-
cially after longer follow-up and the much higher tional preservation of the facial nerve also appear
morbidity rate and risks of damage to the facial to be similar and are related mainly to tumor size.
nerve at reoperations, the rationale of this concept Preservation of the facial nerve anatomical integ-
remains elusive. Exceptions are the rare cases of rity is possible in 93–99 % of all patients, and
lifesaving decompressive surgeries (10 cases = good facial nerve function is achieved in 52–93 %
>1 % in the series of the senior author) [197]. of them. In regard to hearing preservation, how-
Recently, another concept has become popular: ever, a major difference exists between the dif-
to perform initially intracapsular decompression ferent approaches: only the middle fossa and the
of the VS and then to treat the remnant with radio- retrosigmoid approaches provide the opportu-
surgery [71, 98]. The evaluation of the general and nity for hearing preservation. The retrosigmoid
functional outcome following such management, approach is the only one that allows for hearing
however, shows that it is not superior to that after preservation in case of large VSs.
radical microsurgical removal in one stage. The The advantages and disadvantages of these
patient is exposed to two procedures, each with its techniques are widely discussed in the literature.
specific risk of morbidity and only tumor “growth Comparisons of the outcomes of surgery with
control” is achieved. Moreover, the phenomenon different approaches have been frequently made.
of radiosurgical failure should be considered: in We believe, however, that they are unreliable and
some patients the tumor grows further despite the have low scientific and practical value: optimal
treatment (see section “Radiosurgery” of VS). results are achieved with that approach the sur-
Different operative approaches and techniques, geon is best familiar with. Notably, neurophysi-
reflecting individual surgeons’ experience and ological monitoring should be an integral part of
Management of Patients with VS 165

each VS surgery, whatever operative route or is the most important hearing functional test.
technique is preferred. Speech discrimination is important in determin-
Staged removal of VS is a concept that at ing the usefulness of hearing.
present is considered obsolete. It has been intro-
duced in the early years of VS surgery—in Electrophysiological Studies
1925—by W. Dandy [49]. He performed the Electronystagmography assesses the superior
operation in 2 stages and found that at the second vestibular nerve, which innervates the horizon-
stage the residual tumor was softer, more necrotic, tal semicircular canal. Importantly, the vestibu-
and less vascular. Later on, similar technique has lar nerve may function until almost all fibers are
been applied in case of tumors with large size destroyed. The vestibular evoked potential test
and major brain stem compression, particularly provides information on the function of infe-
those that are deeply embedded within the sub- rior vestibular nerve that innervates the saccule.
stance of the brain stem and cerebellum that were Brain stem auditory evoked potential (BAEP)
difficult to remove completely during the initial testing correlates with the chances of hearing
surgery: the tumor is removed in 2 or even in 3 preservation.
stages [46, 167, 215]. Major disadvantages of
this strategy are the need of two separate surger- Retrosigmoid Approach in VS
ies with their cumulative risks and the develop- The general principles of the RS approach have
ment of adhesions and scar tissue, which render been outlined in the Chap. 4. Here we will
the second surgery much more difficult. describe the steps specific for VS surgery.
The philosophy of the senior author is that the Once the CP angle has been exposed, the sur-
goal of VS surgery—complete tumor removal geon should identify the main anatomical land-
with preservation of all neural functions—can be marks that would be helpful for the precise
best achieved with the retrosigmoid approach orientation. The size of the tumor dictates the
with the patient in the semi-sitting position and subsequent steps. In most VS the intrameatal
has been applied successfully in more than 4,000 tumor part is approached initially in order to
such surgeries. identify the distal intracanalicular segments of
facial and cochlear nerves, which benefits their
Preoperative Examinations subsequent dissection from the tumor.
Neuroimaging Furthermore, the initial drilling of the posterior
Neuroimaging (discussed in detail earlier), wall of the IAC prevents spillage of bone dust
besides establishing the diagnosis VS, provides throughout the basal cisterns. Exceptions are the
data important for the operative planning. very large VSs that not only fill the CP angle but
also “hang” on the posterior surface of the pyra-
Audiometric and Audiological Examinations mid. IAC in such cases can be opened only after
The evaluation of hearing is important for the partial removal of the CP angle tumor part (see
decision-making process. Pure-tone audiometry Case Illustration 5.1).
166 5 Vestibular Schwannomas

Case Illustration 5.1 revealed trigeminal hypesthesia (V2, V3) and


Large VS Removal with Primary Debulking severe hearing loss (more than 70-dB loss in
Forty-two-year-old male patient presenting the main speech area) on the right side and
with a history of hearing loss and taste distur- severe gait ataxia. MRI examination demon-
bance since several years and gait instability strated a large VS on the right side with a larg-
since less than 1 year. During the last few est diameter of 4.5 cm, severe compression of
months, he complained of strong headache and the brain stem, and hydrocephalus (a, b).
visual problems. His neurological examination details of surgery are shown on c-j

a b
c d

Preoperative MR scans of the patient demonstrating a (d). Then, the posterior wall of the IAC could be
tumor in the right CP angle with growth pattern and exposed. The overlying dura was excised in the stan-
imaging characteristics, typical for a VS. (a) dard technique (e), and the bone was drilled off (f).
T1-weighted contrast-enhanced sequence. (b) CISS The intracanalicular segment of the facial nerve was
sequence. Intraoperative images illustrating the stages exposed distal to the tumor (g) and could be dissected
of tumor removal. (c) Initial overview of the tumor and medially to the area of the porus of the IAC (h).
the CP angle, demonstrating the relations to the tumor (i) Dissection of the proximal segment of the facial
to surrounding structures: the tumor is attached to the nerve from the tumor. (j) Complete tumor removal and
posterior petrous bone, and the porus of the IAC is not preservation of facial nerve (Tu tumor, 7 facial nerve,
visible. Hence, debulking had to be performed initially BS brain stem)
Management of Patients with VS 167

In the early postoperative period, the patient rapidly. At discharge the patient had complete
complained of headache and was drowsy due to right-sided hearing loss and HB grade IV facial
a moderate pneumocephalus. Within several palsy. One year later the facial nerve function
days he recovered completely. His gait improved was HB grade II and the gait was normal.

e f

g h

i j
168 5 Vestibular Schwannomas

Opening of the IAC


The important anatomical landmarks are the
tentorium, the superior petrosal sinus, and the
caudal cranial nerves. In smaller tumors the porus
and the seventh-eighth cranial nerves can be
directly seen. During the IAC opening, the struc-
tures that are at highest risk are the common crus,
the vestibular aqueduct, and the posterior semi-
circular canal: if hearing is to be preserved, these
structures should not be destroyed. The bone
structure of this area of the pyramid, the degree
of its pneumatization, and the location of semi-
circular canals are highly variable. Since no ana-
a
tomical landmarks exist to indicate their location
during surgery [86], these variations can be best
appreciated when studying the high-resolution
bone-window thin-slice CT images (Figs. 5.13,
5.14, and 5.15). CT-based neuronavigation guid-
ance has been shown to be useful for the intraop-
erative localization of the pyramid structures. Its
accuracy is highest if bone-implanted reference
markers are used [193]. Although neuronaviga-
tion support has been recommended during the
early phases of the learning curve [171], its prac-
tical value is rather limited.
The dura behind the IAC is incised circumfer-
entially, stripped off the petrous bone, and b
excised (Fig. 5.16a). If the dura is not removed
adequately, drilling may tear it and cause rupture Fig. 5.13 Variable position of the labyrinth in relation to
of the venous sinuses. The incision starts imme- the IAC
diately below the superior petrosal sinus. In
order to avoid injury to the endolymphatic sac however, BAEP monitoring shows that the con-
and duct, the dura should not be incised inferior duction of the cochlear nerve might be disturbed
to the level of the small bone ridge, projecting during bone removal, regardless how careful it
along the lower edge of the IAC (Fig. 5.16a). has been performed. The pathophysiological
High-speed pneumatic drill provides safe and mechanism behind this indirect functional
controlled bone removal. It is advisable to begin disturbance is still unknown, and its evolution
the drilling with a large-sized diamond burr cannot be predicted. In most cases, however, it
(4–5 mm in diameter) and then change to smaller recovers spontaneously. The last bone layer or
burrs. Highly experienced surgeons might prefer lamina may be safely removed with a small
to begin with a cutting burr in order to spare platelet knife.
time. Irrigation is used for cooling and for The extent of lateral opening of the IAC in the
removing the bone dust during drilling. The uti- direction of the fundus is determined by the
lization of a suction–irrigation device has proven extension of the tumor, as estimated at surgery.
to be very helpful throughout the surgery. The canal is opened or “unroofed” over 180° of
Removing the bone layer by layer in 1-mm incre- its circumference in order to obtain sufficient
ments under direct visual control helps to avoid access (Fig. 5.16): an unobstructed view of supe-
injury to underlying structures. Not infrequently, rior and inferior tumor poles should be provided.
Management of Patients with VS 169

a b

c d

e f

Fig. 5.14 Variable structure of the pyramid, posterior to recognized before beginning the bone removal in order to
the IAC (arrows). The bone layer that has to be drilled off avoid inadvertent injury to the IAC components. The air
can vary from very thick (a) to moderate one (b, c). Note cells have to be differentiated from the jugular bulb: this is
the left–right difference of the posterior wall thickness on possibly due to their different CT characteristics (compare
image a. It may contain air cells (d–f), which have to be Figs. 5.14e and 5.21)
170 5 Vestibular Schwannomas

a b

Fig. 5.15 CT (a, b) and MR (c, d) (images of a patient scan, demonstrating the amount of bone removal from the
with a moderately large VS on the left side (a, b). posterior wall of the IAC. See Fig. 5.16 for intraoperative
(a) Preoperative and (b) Postoperative bone-window CT details

In case that only a narrow trough is made over the cranial nerves function is discussed later in
the posterior aspect of the IAC, the identification this chapter.
and preservation of the cranial nerves are The amount of bone removal is not equal in
extremely difficult. The distance to the fundus is the medial and lateral parts of the canal: the
estimated intermittently by palpating with an medial part is opened wider. The superior and
angled microhook (Fig. 5.16c). As discussed inferior lips of the porus of the IAC should be
earlier in the text, MRI gives only an approxi- drilled off (Figs. 5.15 and 5.16). Ideally, the dura
mate estimation of the extent of lateral extension covering the intracanalicular tumor part should
of the VS and is not a fully reliable guidance. If be preserved during bone removal.
hearing-preserving surgery is planned and the In order to prevent CSF leaks at the end of sur-
position of the labyrinth precludes further lateral gery, the IAC is filled with multiple small fat
bone removal, part of the tumor may remain hid- pieces, harvested from the subcutaneous fat layer
den (Figs. 5.17 and 5.18). The technique to of the incision, and sealed with fibrin glue. Our
remove such tumors completely and to preserve experience has shown that the application of fat
Management of Patients with VS 171

a a

b b

Fig. 5.17 Bone-window CT demonstrating the extent of


lateral opening of the IAC. (a) The position of the semicir-
cular canals allow for safe opening of the IAC up to the
fundus. (b) In this case the most lateral part of the poste-
rior IAC wall was not removed in order to avoid injury to
the labyrinth: a small bone lip hinders the direct visualiza-
tion of the fundus. Its tiny size, however, did not hinder
the safe and complete tumor removal. (c) Wide opening of
the IAC lateral to the fundus: the bony wall of the lateral
semicircular canal wall has been slightly opened (the
patient had no useful hearing prior to surgery)

Fig. 5.16 Opening of the posterior wall of the IAC—


intraoperative images. (a) The dura behind the IAC is
excised widely, taking care not to damage the endolym-
c phatic sac and duct. The arrow points at the inferior extent
of dural excision, which is usually marked by a small bone
ridge. SPS superior petrosal sinus. (b) Bone removal should
be approximately 180° of the circumference of the IAC.
Note the wide removal of the superior and inferior lips of the
porus of the IAC (asterisks). (c) The distance to the fundus
can be determined by palpating with an angled microhook
172 5 Vestibular Schwannomas

in the IAC area should be explicitly stated in the


discharge letters to avoid misinterpretations at
follow-up MR imaging (Fig. 5.19). Not
infrequently, the fat signal is considered as tumor
remnant or recurrence.

High Jugular Bulb


An important consideration during the approach
planning is the jugular bulb position. The wall of
the bulb is thinner and more vulnerable if com-
pared to that of the sigmoid sinus, the latter being
composed by two layers [100]. Inadvertent injury
to the jugular bulb can lead to massive bleeding
and air embolism. Normally, the bulb is situated
c at the inferior surface of the pyramid, inferopos-
terior to the IAC [214]. The position of the bulb
Fig. 5.17 (continued)

a b

c d

Fig. 5.18 In this patient the VS did not extend up to the (b) Bone-window CT before surgery. (c) MR examination
fundus, and it was not necessary to remove the whole pos- following the complete tumor removal (T2-weighted
terior wall of the IAC. Therefore, a targeted bone removal sequence). (d) Postoperative bone-window CT
was performed. (a) Preoperative CISS MRT sequence.
Management of Patients with VS 173

a b

c d

Fig. 5.19 Postoperative MR images demonstrating the T-1 weighted sequence. (b) T-1 weighted sequence fol-
fat plug in the area of the right IAC. The fat has typical lowing contrast administration. (c) T-2 weighted sequence.
signal characteristics in various MRI sequences (a) Native (d) T2-CISS sequence)

is variable but can be precisely estimated on the (jugular bulb and IAC are seen concomitantly
preoperative bone-window CT. In case the jugu- in two axial CT slices).
lar fossa is above the plane of the inferior border • Grade III: The jugular bulb is more than
of the IAC, the jugular bulb is considered as 4.0 mm above the inferior border of IAC (jug-
“high.” In a previous study, we found high ular bulb and IAC are seen in three or more
jugular bulb in 9 % of all patients [214]. axial CT slices).
The classification of the high jugular bulbs’ The position of the bulb has never been an
grade that we apply currently is presented below obstacle for sufficient opening of the IAC and—
and is illustrated on Fig. 5.20: if the drilling is performed carefully—does not
• Grade I: The jugular bulb is less than 2 mm increase the morbidity rate. In order to avoid
above the inferior border of IAC (the jugular complications, the opening of the IAC should be
bulb and IAC are seen concomitantly at only planned carefully according to the CT findings
one axial CT slice of 2 mm). (Fig. 5.21).
• Grade II: The jugular bulb is between 2 and In patients with highly located jugular bulb,
4.0 mm above the inferior border of the IAC corresponding to grade I or II, drilling is
174 5 Vestibular Schwannomas

IAC
IAC IAC

JB

JB

JB

Grade I Grade II Grade III

Fig. 5.20 Illustration of the 3 grades of bulb position (Reprinted from Shao et al. [214]. Copyright © 1993; with
permission from Wolters Kluwer Health)

performed superior to the jugular fossa; thus, the Vascular Considerations


jugular bulb can be avoided (Figs. 5.22, 5.23, and The location of the CP angle vessels cannot be
5.24). In grade III cases, opening of the jugular inferred from the diagnostic images with certainty.
fossa is usually necessary in order to open the Good knowledge of the normal microanatomy
IAC sufficiently. The drilling is continued in 0.1- and the possible relations of the main arteries and
mm increments until the jugular bulb is identified veins to the tumor are a prerequisite for successful
under a thin shell of bone. Performing jugular VS surgery.
vein compression at the neck enhances its The AICA is the artery most closely related to
identification. The last bone layer is removed VS (see Chap. 2). According to the study of
carefully, and the bony wall of the jugular fossa Sunderland [237], in 64 % of all cases the artery
is opened. The jugular bulb is then mobilized is intimately related to the seventh-eighth nerve
slightly from all sides and compressed with a complex within or at the orifice of the IAC. The
microdissector. Its shrinkage creates more space growing VS usually displaces the artery inferi-
to proceed with the drilling around it (Figs. 5.25 orly and posteriorly. In more than 91 % of large
and 5.26). In case bleeding from the bulb occurs, VSs, it is adherent to the tumor capsule [202]. In
it can be controlled with Tabotamp (or Surgicel); some cases a loop of the artery may be covered
occasionally, it has to be sealed off with fat pieces by the dura of the pyramid or even protrude deep
and fibrin glue. into the IAC. If this is not recognized timely and
Evaluation of the surgical outcome, in terms tumor removal proceeds as usual, the vessel may
of completeness of tumor removal and func- be damaged, which has usually catastrophic con-
tional results, did not show any major differ- sequences. The vessel should be initially dis-
ences between patients with low and high bulb sected free from the arachnoidea and from the
position [214]. dura and only then mobilized. A cuff of dura can
Management of Patients with VS 175

be left around it to avoid any perforator injury.


The dissection of the artery may require occa-
sionally partial tumor debulking and drilling off a
part of the petrous bone (Fig. 5.27).
Infrequently, bleeding can occur from a small
artery under the dura or in the petrous bone—the
subarcuate artery—that enters the subarcuate
fossa on the posterior wall of the IAC. This artery
can be safely coagulated. The internal auditory
artery or the “labyrinth artery” originates from
the anteroinferior cerebellar artery. It runs close
to the seventh-eighth nerve complex, usually
along the inferior surface of the cochlear nerve,
a
and gives several small feeding vessels to these
nerves and supplies the dura and bony labyrinth.
Injury to the labyrinth artery may cause hearing
loss or rarely facial nerve palsy.
The PICA is located inferior to the tumor and
may also be strongly adherent to its capsule. In large
tumors the PICA or its branches may pass through
the tumor. Recurrent perforating branches—from
AICA or PICA—that supply the brain stem may
run toward the IAC initially and then turn medially;
see Fig. 5.28. It is of utmost importance to identify
and preserve all these vessels in order to prevent
major neurological dysfunction, such as the lateral
b medullary syndrome.
Although VSs have variable degree of vascu-
larization, most tend to bleed strongly during
surgery. Hypervascular variants, demonstrated on
MRI as multiple flow voids in and around the
tumor, however, are rarely encountered. Normal
vessels should be differentiated from pathologi-
cal tumor vessels, which may be safely coagu-
lated. End arteries that terminate in the tumor can
also be coagulated but have to be differentiated
form en passage vessels. If the patient is operated
in the semi-sitting position, coagulation during
tumor removal is seldom required: only in case of
bleeding from larger vessel. The frequent irriga-
c tion, performed by the assistant, ensures a clear
operative field. Thus, the risk of thermal injury to
Fig. 5.21 Bone-window CT of three patients demon- the cranial nerves can be avoided. Coagulation is
strating high position of the jugular bulb (arrow). If the reserved for final hemostasis.
IAC is opened in the standard technique (dotted line on a),
The bridging petrosal vein/s are easy to pre-
both the jugular bulb and the integrity of the labyrinth
would be endangered serve in small VS. In large tumors they may be
176 5 Vestibular Schwannomas

a b

c d

e f

Fig. 5.22 High jugular bulb: grade I, preoperative CT (a–c) and MRI (d–f), in a patient with VS grade 3. The
intraoperative findings are presented on Fig. 5.23
Management of Patients with VS 177

a b

c d

Fig. 5.23 High jugular bulb, grade I. Intraoperative angle part is debulked with an ultrasonic aspirator (h).
images illustrating the consecutive steps of IAC opening The tumor capsule is dissected from the cochlear nerve (i;
and tumor removal. (a) Initial view of the CP angle; dot- CN cochlear nerve) and the facial nerves (j; 7 facial nerve).
ted line is area of the pyramid, posterior to the IAC, that (k) At the end the most adherent part of the capsule close
will be drilled off. (b) The dura overlying this area has to the porus of the IAC is removed. (l) Complete tumor
been excised. (c) A few mm from the bone have been removal with preservation of the anatomical integrity of
removed, and the dome of the jugular bulb has been the cranial nerves. (m) Schematic presentation of the tech-
exposed (arrow). (d) Further drilling with a diamond burr, nique of IAC opening in case of grade I high jugular bulb
wider exposure of the bulb (e and f arrows), and careful (IAC internal auditory canal, JB jugular bulb, DD dia-
mobilization with a dissector (arrowhead). The IAC is mond burr) (m: Reprinted from Shao et al. [214].
then opened by drilling superior to the jugular bulb (g), Copyright © 1993; with permission from Wolters Kluwer
the intracanalicular tumor part is removed, and the CP Health)
178 5 Vestibular Schwannomas

e f

g h

i j

Fig. 5.23 (continued)


Management of Patients with VS 179

k l

IAC IAC

JB

JB

Fig. 5.23 (continued)


180 5 Vestibular Schwannomas

a b

c d

Fig. 5.24 High jugular bulb, grade I: postoperative CT (a, b) and MRI (c–e) images of the patient from Fig. 5.23
Management of Patients with VS 181

a b

c d

IAC IAC

JB

JB
DD

e f

Fig. 5.25 High jugular bulb, grade III. (a–c) Bone- auditory canal, JB jugular bulb, DD diamond burr). (g)
window CT demonstrating the high jugular bulb (grade Postoperative CT demonstrating the amount of petrous
III). (d) T1-weigheted contrast-enhanced MRI of the VS. bone opening, which allowed for safe and complete tumor
(e) T2-CISS. (f) Schematic presentation of the technique removal (h) (f: Reprinted from Shao et al. [214]. Copyright
of IAC in case of grade III high jugular bulb (IAC internal © 1993; with permission from Wolters Kluwer Health)
182 5 Vestibular Schwannomas

g h

Fig. 5.25 (continued)

a b

Fig. 5.26 CT scans of a patient with a facial nerve The high jugular bulb (grade III) was not an obstacle to
schwannoma, located in the CP angle and protruding deep the complete opening of the canal up to its fundus (b)
into the IAC. (a) Preoperative. (b) Postoperative images.
Management of Patients with VS 183

a b

c d

Fig. 5.27 VS grade 3a on the right side (a–c). A loop of exposing the underlying artery (e). Then, the tumor could
AICA was seen to enter the IAC (d). The posterior wall of be dissected from the cochlear and facial nerves (f) and
the canal was removed and the dura incised carefully, removed completely (g)
184 5 Vestibular Schwannomas

e f

Fig. 5.27 (continued)


Management of Patients with VS 185

a b

c d

Fig. 5.28 VS grade 4b on the left side (a and b: the tumor capsule (Tu VS, arrows branch of the AICA).
T1-weighted axial and coronal images following gadolin- (g) In order to avoid injury to the vascular structures or cra-
ium administration. c: T2- weighted sequence. d: CISS nial nerves, “controlled aspiration” with the ultrasonic aspi-
sequence). (e–i) Intraoperative images illustrating the dis- rator (CUSA or Sonoca) is performed: the VS is held with
section of the tumor from the vascular structures and their tumor forceps (left hand), while careful aspiration is per-
preservation. (e) Initial view of the tumor. The dura overly- formed with the right hand. (h) Gradually the facial nerve
ing the posterior wall of the IAC has been excised. (f) A was identified (FN). (i) Complete removal of the tumor with
large vessel, which later on proved to be a branch of the preservation of the facial nerve (FN), the AICA (arrow), the
AICA, was strongly displaced posteriorly and adherent to petrous vein (asterisk), and the trigeminal nerve (5)
186 5 Vestibular Schwannomas

e f

g h

Fig. 5.28 (continued)


Management of Patients with VS 187

dislocated or adherent. Still, every effort should In some VSs this part is not adherent to the nerves
be made to save them because their interruption and can be easily mobilized and removed. In case
might have unpredictable consequences. of intense adhesions, however, the dissection
should be performed with utmost care not to
Removal of the Intracanalicular Tumor injure the nerves.
Portion
The meatal dura is incised parallel to the inferior Removal of the Extrameatal Tumor Part
lip of the IAC, stripped off the underlying tumor, Tumor growth in the CP angle dislocates the
and excised. The dural attachments of the VS in adjacent cranial nerves in different directions. In
the area of the superior and inferior lips of the a study of 1,006 patients, Sampath et al. [202]
porus should be incised, which allows for its found that the facial nerve is most commonly
greater degree of mobilization. The nerves in located on the anterior middle third and on the
their intracanalicular segment are with preserved anterior superior third of the tumor capsule,
anatomical shape almost always: the facial nerve regardless of tumor size. Much more rarely it is
is displaced typically anterior to the tumor, supe- located on the superior tumor pole, on its anterior
rior to the cochlear nerve. An attempt is made to inferior third, or on the inferior pole. The authors
identify the facial nerve between the superior found that in some patients (<3 %) the nerve was
tumor pole and the upper wall of the IAC. If this located posterior. The senior author (MS), how-
is not possible without undue traction, the tumor ever, had never encountered a facial nerve poste-
mass can be removed layer by layer with an rior to a VS. Sampath et al. [202] showed that the
angled hook or microdissector until the nerve is facial nerve passed through the tumor in 2.9–
seen. In case the tumor is of harder consistency, it 3.4 % of the cases, with similar frequency in
may be necessary to debulk it initially with an small and in large tumors. We believe this can be
ultrasonic aspirator (CUSA or Sonoca). The observed in tumors infiltrating the nerve, facial
cochlear nerve is identified below the facial nerve schwannomas or in multilobulated tumors, asso-
at the anteroinferior or inferior part of the IAC. In ciated with NF-2. The cochlear nerve has a more
larger tumors the vestibular nerves are partially constant location: it is usually splayed along the
or completely destroyed by the tumor. In tumors anterior inferior third or the inferior surface of
that do not fill the entire IAC, they can be the tumor [41, 197].
identified: either proximal or distal to the tumor. The location of the nerves in relation to the
These nerves are incised several millimeters away tumor may be directly visualized preoperatively
from the tumor attachment, at a point that is com- in patients with small VS with the constructive
pletely free from tumor infiltration. In small interference in steady-state (CISS) MRI study. In
tumors the healthy vestibular nerve can be pre- large tumors that fill the CP angle cistern, the
served. The tumor is dissected and mobilized location of the nerves cannot be determined. In
from the facial and cochlear nerves in a lateral- such VSs the facial nerve can run rostrally—even
to-medial direction (Figs. 5.29 and 5.30). In case reach the axilla of the trigeminal nerve—and then
the tumor is very adherent to the nerves, this step curve down and laterally toward the IAC [43].
is performed later, following the identification of DTI-based fiber tracking appears to be a promis-
the nerve in the CP angle. The tumor part that ing technique for the depiction of the facial nerve
extends to the fundus is removed with an angled in tumors filling the CP angle cistern [76].
microhook. Performing gentle palpations with The first step of removal of the extrameatal
the hook even tiny tumor parts can be identified. tumor, regardless of its size, is tumor debulking
This lateral part of the IAC can be directly or internal decompression (Fig. 5.32a). Slight
inspected with a mirror or an angled endoscope. pulling of the remaining tumor part into the
188 5 Vestibular Schwannomas

a b

Fig. 5.29 Bone-window CT (a), T1-weighted MR image following gadolinium administration (b axial, c coronal
slices), demonstrating a small right-sided VS with minimal protruding into the CP angle (see Fig. 5.30)

operative field and dissection of its capsule in the in all directions. Creation of a deep and narrow
arachnoid plane follows. In large and giant hole should be avoided. If the debulking is con-
tumors, these steps are repeated alternatively in tinued too far anteriorly, the tumor capsule can be
all directions. Debulking is best performed with penetrated and the facial nerve can be damaged.
an ultrasonic aspirator (CUSA or Sonoca— The main principle is that attempt to mobilize the
Fig. 5.31) with a middle- or small-sized tip, remaining tumor and to perform dissection should
selected according to tumor size. The ultrasonic be made only after sufficient internal decompres-
aspirator allows for gentle and controlled removal sion has been achieved: if the remaining tumor is
and avoids excessive pressure on the underlying not easily mobilized, further debulking should be
structures. Alternatively, a platelet-shaped knife performed.
can be used to debulk the tumor. Tumor debulking Optimally the facial and cochlear nerves are
should proceed very carefully and systematically identified medial to the tumor close to their entry
Management of Patients with VS 189

a b c

d e f

Fig. 5.30 Intraoperative images illustrating the steps of facial and cochlear nerves and removed completely. (e) The
tumor removal (Patient from previous figure). (a) Initial view anatomical and functional integrity of the nerves has been
of the CP angle and the seventh-eighth nerve complex. (b) preserved, as proven by the electrical stimulation of the facial
Dural incision around the posterior wall of the IAC. (c) The nerve and the preservation of the BAEP wave amplitude and
IAC has been opened and its dura has been excised, exposing latency. (f) The dura was used to seal the IAC—a practice that
the intracanalicular tumor. (d) The tumor is dissected from the has been abandoned by the senior author since a long time

or exit zone on the brain stem surface by slightly tional condition. The integrity of the facial nerve is
pulling the tumor in a superior direction. In large monitored throughout the surgery by continuous
VSs this is possible only after considerable reduc- facial electromyography and facial nerve motor
tion of the tumor volume. The identification of the evoked potentials recording.
nerves medially is guided by their relationship to The nerve–tumor interface should be clearly
the anatomical landmarks (see Chap. 2). The identified, and the dissection should proceed
nerve electrical stimulator is applied frequently: always in the arachnoid cleavage plane
for mapping of the nerve or for testing its func- (Fig. 5.32b–d). The three-hand technique, which
190 5 Vestibular Schwannomas

a b

Fig. 5.31 Ultrasonic aspirators that we use for tumor debulking: (a) CUSA (Integra Neurosciences) and (b) Sonoca
(Söring)

is possible if the patient is in the semi-sitting dissect the facial nerve from superior (pulling the
position, is extremely useful: the surgeon uses tumor inferior) may be very dangerous for the
both his hands for tumor preparation and removal, nerve and should be avoided. Another important
while the assistant irrigates the operative field. consideration is that the dissection should be
The operative field is thus cleared and coagula- performed from medially to laterally even if both
tion can be avoided. The dissection is performed ends of the facial nerve are well visible. The
bimanually: the arachnoid layer on the tumor sur- attempt to dissect the tumor from the facial nerve
face is gripped and gently peeled off the tumor in a lateral-to-medial direction—from the IAC
with microforceps. At the same time the second toward the brain stem—is also related to a high
hand is holding the tumor. In case the dissection risk of nerve transection.
is not in the correct plane, the fragile nerves can In the area near the porus, the nerves are espe-
be destroyed or their vascular blood supply, which cially fragile and adherent to the tumor capsule.
is in the subarachnoid space, can be interrupted. This portion is dissected at the end when both the
The dissection proceeds in a methodical fash- proximal and distal portions of the facial nerve
ion from the brain stem toward the IAC. It should have been freed from the tumor. Following tumor
be performed in alternative directions, and over- removal the facial nerve is stimulated at its most
stretching in one direction should be avoided. proximal region of the brain stem root exit zone
Importantly, the tumor rest should be elevated in to verify its functional condition.
a superior direction, which allows visualizing the Tumor variables influencing the surgical strategy:
lower margin of the facial nerve. The attempt to • Tumor size
Management of Patients with VS 191

• Tumor extension in the IAC • Arachnoid membrane/cleavage plane


• Tumor extension in the CP angle • Adherence to neural structures
• Tumor consistency • Previous surgeries
• Tumor capsule • Previous radiotherapy/radiosurgery
• Tumor vascularization
• Cystic tumors

Fig. 5.32 Schematic


illustration of the steps of VS
removal. (a) Internal
decompression of the
extracanalicular tumor part.
(b, c) Dissection from the
cranial nerves, using the
2-hands technique.
(d) Closure of the IAC with
b
fat pieces or muscle
192 5 Vestibular Schwannomas

Fig. 5.32 (continued)

d
Management of Patients with VS 193

Intracanalicular (Extension Grade 1) VS loss, in patients with VS on the only hearing side,
Pure intracanalicular VS or VS grade T1, or those unwilling to undergo surgery. Radiosurgery
according to our tumor extension classification, could be recommended to patients whose somatic
comprised less than 10 % of all VSs operated condition prohibits open surgery or to those insist-
by the senior author [194, 198]. Some clinical ing on this type of management.
and epidemiological observations indicate that Considering the size of the tumor and its slow
these VSs may form a separate subgroup with a growth tendency, the decision when to perform
specific natural evolution. The diagnostic delay surgery should be taken by the patient. If surgery
in such schwannomas is shorter. In a previous is not scheduled, a thorough radiological follow-
publication we showed that age distribution up to detect early any change in VS’s size is war-
has an inverse linear correlation to tumor exten- ranted. Surgical removal, however, is indicated in
sion and size [145]. Patients with intrameatal patients with useful hearing and good BAEP: in
tumors were symptomatic at a more advanced order to have a chance for hearing-preserving
age than those with larger tumors (48.9 vs. 44.4 surgery, it should be done prior to its deteriora-
years). The duration of subjective deafness and tion. As pointed earlier in the text, the occurrence
dizziness were shorter but the duration of tin- of hearing loss is not related to the size of VS and
nitus was longer. The incidence of tinnitus was can be of sudden nature. Rare indications for
highest in intracanalicular tumors: 86 % in VS early surgery are severe intractable vestibular
extension grade T1, 65 in grade 2, 66 % in problems. Some small VS may cause so severe
grade T3, and 51 % in grade T4. Cranial nerve vertigo that removal of the tumor with sectioning
symptoms/signs correlated positively with the of the involved vestibular nerve/s is the only
degree of tumor extension: vestibular nerve therapeutic solution (Fig. 5.33).
signs were most pronounced in VSs grade T1. The principles of intracanalicular VS surgery
Expectedly functional hearing rate was highest were described above in the section on Removal
in intracanalicular tumors. of Intracanalicular VS. Important consideration
Intracanalicular VSs occur in several varieties is that they are a heterogeneous group of tumors
according to the extent of IAC involvement: and the surgery should always be targeted to the
• VSs limited to the medial IAC near the porus specific features of each individual case (see Case
acusticus Illustrations 5.2 and 5.3).
• VSs limited to the lateral part of the IAC near Tumor extension and size are main predic-
the fundus tors of the functional and general outcome of
• VSs involving the entire IAC surgery. Complete tumor removal is possible in
In regard to the bony changes, grade T1 tumors more than 99 % of intracanalicular schwanno-
cause local, diffuse, or no dilatation of the IAC. mas—exceptions are those cases in which the
The management options of intracanalicular tumor infiltrates the facial nerve. The facial
VSs are microsurgery, stereotactic radiosurgery, nerve anatomical preservation rate in intrac-
and follow-up or “wait-and-scan” strategy. The analicular VSs in our experience is 100 % and
last two options have been discussed comprehen- that of the cochlear nerve, 94 % [179]. Excellent
sively elsewhere in the text. In summary, initial facial function (House–Brackmann grades I
observation may be acceptable option in elderly or and II) is achieved in almost all patients. Useful
somatic unstable patients with mild nonprogres- hearing is preserved in 57 % of those patients
sive symptoms, in patients with complete hearing with good preoperative hearing level.
194 5 Vestibular Schwannomas

a b

Fig. 5.33 This 29-year-old patient presented with severe patient was unable to have normal life. Therefore, consid-
vertigo and nystagmus to the left side. Hearing on the right ering all possible management options, she decided for
side was only slightly decreased (class II according to the surgery. The tumor was removed via the RS approach;
New Hannover classification) and on the left side was nor- facial nerve function and serviceable hearing were pre-
mal. The MRI demonstrated very small VS in the lateral served. The vertigo was initially stronger but within 2
part of the IAC on the right side (a–c, arrows). The epi- weeks disappeared almost completely and in 2 months
sodes of vertigo were so frequent and severe that the resolved
Management of Patients with VS 195

Case Illustration 5.2 MRI study demonstrated an intracanalicular


Elongated VS Extension Grade T1, Normal tumor in the right IAC, extending lateral up to
IAC the fundus. The tumor had an elongated form,
This 55-year-old patient presented to our insti- and the IAC was not dilated. The patient decided
tute complaining of tinnitus and progressive for surgery, and the tumor was removed com-
hearing loss since 1 year. Her neurological pletely via the RS approach (a–d). The postop-
examination revealed no abnormalities, besides erative period was completely uneventful. At
a complete loss of hearing on the right side. discharge she had normal facial nerve function.

a b

c d

Preoperative CT and MRI revealing the grade T1 VS on Excision of the dura around the posterior wall of the
the right side: the tumor has an elongated shape and did IAC. (h, i) Exposure and removal of the intracanalicular
not cause dilatation of the IAC. (a) Bone-window CT. VS. The facial (7) and cochlear nerves (8) are preserved.
(b, c) T1-weighted sequences after contrast administra- (j) The dotted line indicated the amount of bone that had
tion. (d) CISS sequence. Intraoperative images. (e) to be drilled off additionally in order to gain access to
Dural incision along the transverse and sigmoid sinuses. the most lateral part of the tumor. The distance to the
A retractor (R) is used to elevate the cerebellar hemi- fundus has been determined by palpation with an angled
sphere, and the lateral cerebellomedullary cistern is microhook. (k) The hook points to the transverse crest
opened, releasing CSF. The lower cranial nerves are vis- of the fundus. (l) Complete tumor removal and preser-
ible. (f) Only when sufficient CSF is drained, a retractor vation of the facial and cochlear nerves (V—vestibular
is placed to hold the cerebellum without any tension. (g) nerve, from which the tumor originated)
196 5 Vestibular Schwannomas

e f

g h

i j
Management of Patients with VS 197

k l
198 5 Vestibular Schwannomas

Case Illustration 5.3 The patient was operated via the RS


VS of the Lateral IAC, Slightly Enlarged approach with the goal to remove the tumor
IAC completely and preserve the facial nerve
Sixty-year-old male patients with com- function, as well as to attempt hearing pres-
plaints of strong vertigo and slight hearing ervation. The details of surgery are pre-
loss on the left side. The audiogram and the sented on figures (d–j) and the postoperative
speech discrimination test showed preserved MRI on figures (k, l).
useful hearing (class H1 according to the The postoperative period was completely
New Hannover hearing classification). The uneventful. At discharge—14 days after
MRI study of the patient is presented on surgery—the patient had normal facial nerve
figures (a–c): a small intracanalicular VS is function and useful hearing (class H2 according
seen, located in the lateral part of the IAC. to the New Hannover hearing classification).

a b

Preoperative MRI of the patient demonstrating the VS dissection of the tumor from the cranial nerves and
located in the lateral part of the IAC: (a, b) T1-weighted removal. (i) Complete tumor removal with preservation
sequences after gadolinium administration. (c) CISS of the functions of the facial and cochlear nerves. (j) The
sequence. Intraoperative images. (d) The posterior wall IAC has been sealed off with multiple fat pieces and
of the IAC has been removed and the tumor located in fibrin glue. (k, l) Postoperative MRI performed 6 months
the lateral part of the IAC is exposed. (e–h) Stepwise after surgery: no recurrences or remnants are detectable
Management of Patients with VS 199

d e

f g

h i
200 5 Vestibular Schwannomas

j k

l
Management of Patients with VS 201

Giant (Extension Grade T5) VS schwannomas it may be extremely difficult to


Giant VSs or grade 5 VSs are defined as those identify the correct arachnoid plane, in partic-
that measure >4.0 cm or extend over the midline. ular the one to the brain stem. The pontine sur-
They always cause significant brain stem com- face veins can be used as a guide for finding
pression and typically contact and/or compress the proper dissection layer. With the 2-hands
both the trigeminal and the lower cranial nerves. technique—holding the tumor capsule with
The successful management of such tumors is one hand with a forceps and performing
especially challenging: the operation is more dissection with the other hand—inadvertent
complex, the general operative risks are higher, injuries to the neural tissue can be avoided
and the functional facial and cochlear nerves (see Case Illustrations 5.4 and 5.5)
preservation is extremely demanding. Surgery The risk of hemorrhagic complications due
with the primary goal of brain stem decompres- to venous bleeding after surgical removal of
sion and alleviation of the intracranial hyperten- large or giant VSs is higher [186, 266]. The
sion is the only treatment option in giant VSs. draining veins in the CP angle might be engorged
The opinions on the extent of surgery in the lit- and extremely fragile, with increased bleeding
erature vary: from partial or subtotal removal fol- tendency during and after surgery. In order to
lowed by observation or radiosurgery to complete detect bleeding from any open or torn bridging
removal in 1, 2 or more stages [1, 5, 21, 98, 186]. veins, we apply jugular venous compression
We believe that even in giant VSs complete surgi- twice: at the end of the procedure, while the
cal removal and preservation of facial nerve func- retractor is still in place, and after the removal
tion should be the goal. In case useful hearing is of the retractor to detect.
present before surgery, an attempt to preserve it The size of the VS is one major predicting fac-
should always be made [196]. tor for the chance of facial nerve anatomical and
Patients with giant VSs typically have severe functional preservation [26, 106, 223]. In a
hearing loss at presentation (96–100 % of all recently published series, including 50 patients
patients in different series). The trigeminal nerve with giant VSs, we found that complete tumor
is affected in 40–78 % of the patients, the lower removal was possible in all cases [196]. The
cranial nerve in 10–14 %, the facial nerve in anatomical integrity of facial nerve was preserved
0–14 %, and the abducens nerve in 4–7 % in 92 % of the patients. In 4 patients facial recon-
[4, 104, 107, 125]. Gait instability caused by the struction with hypoglossal–facial anastomosis
compression of the middle cerebellar peduncle was performed. At 12 months follow-up, the
and cerebellum is in 44–88 % of the patients and facial nerve function recovered to HB grade IV in
long-tract signs due to brain stem compression 2 of them and to HB grade III in the other 2.
in 14–25 %. The mass effect of the tumor and Overall at last follow-up, 75 % of all patients had
the frequent hydrocephalus cause intracranial excellent or good facial nerve function (defined
pressure elevation: we observed signs of intrac- as HB grade I–III). Some surgeons believe that it
ranial hypertension in 26 % of our patients with is even theoretically impossible to preserve hear-
giant VSs. ing in patients with large VS and therefore favor
The advantages offered by the RS approach the hearing-destructing translabyrinthine
are crucial for achieving optimal operative approach [23, 269]. Our experience shows though
results in giant VSs (Fig. 5.34). In such cases that even in patients with giant VSs, there is a
the CP angle neural and vascular structures are chance of hearing preservation, provided the
more vulnerable due to the long-standing com- patient has useful hearing level and normal or
pression. The underlying brain parenchyma is near-normal BAEP preoperatively. In 11 % of
frequently softened and tends to bleed dif- these patients, useful hearing could be preserved
fusely even after slight manipulations. In giant and 33 % had some hearing.
202 5 Vestibular Schwannomas

a b

c d

Fig. 5.34 Preoperative MR images (a–c) of a patient with giant VS on the right side (T1-weighted scans following
gadolinium administration). (d) Postoperative CT, performed prior to discharge 13 days after surgery
Management of Patients with VS 203

Case Illustration 5.4 and blurred vision developed. Ophthalmoscopy


Giant VS 1 revealed bilateral papilledema. MR examina-
This 41-year-old patient presented with com- tion demonstrated a very large VS on the left
plaints of progressive hearing loss since 5 side, causing severe compression and disloca-
years. During the last year his gait became tion of the brain stem (a–c). No signs of hydro-
unstable; since 5–6 months strong headache cephalus were, however, evident. The patient

a b

(a–c) MRI of the patient (T1-weighted sequences fol- superior to the bulb (double small arrows). (h) Exposure
lowing gadolinium administration), demonstrating a of the intracanalicular tumor part and identification of
giant VS—maximal diameter of 4.6 cm. Intraoperative the facial nerve (i). (j) Removal of the extracanalicular
images. (d) Initial view of the CP angle. (e) The dura tumor part. The facial nerve next to the porus of IAC is
around the posterior wall of the IAC has been incised marked with an arrowhead. (k, l) Complete tumor
and is about to be stripped off the bone with the platelet removal and anatomical preservation of the facial nerve
knife. (f) Drilling of the posterior IAC wall. (g) The (arrowheads). The abducens nerve (6) is well visible in
jugular bulb (large arrow), which was located high the depth due to the massive dislocation of the brain stem
(grade I), was partially exposed. The IAC was opened caused by the tumor. (m, n) CT scan 1 day after surgery
204 5 Vestibular Schwannomas

d e

f g

h i
Management of Patients with VS 205

j k

n
206 5 Vestibular Schwannomas

had complete hearing loss, as shown by the was preserved. No new neurological deficits
audiogram, and partial abducens nerve palsy appeared and the postoperative period was
on the left side. uneventful. He has been transferred to the nor-
He has been operated via the RS approach mal ward the day after surgery and discharged
(d–l) and the tumor was removed completely. 14 days later. At discharge he had an HB grade
The anatomical integrity of the facial nerve II facial nerve function.
Management of Patients with VS 207

Case Illustration 5.5 sided hemiparesis (grade 4/5). MRI demon-


Giant VS 2 strated a very large tumor on the left side with
The patient presented to our institute with a imaging characteristics of VS (a–d). The
history of headache, gait instability, taste dis- tumor was removed completely via the RS
turbance, and hearing loss on the left side approach (the details are presented on figures
since several years. Neurological examination e–o). After surgery the patient had complete
revealed trigeminal nerve hypesthesia (V1–3) hearing loss and HB grade III facial nerve
and hypacusis (class H3 according to the New palsy. At the first follow-up examination 6
Hannover classification) on the left side, as months after the operation, however, the func-
well as severe gait ataxia and a central right- tion had already improved to HB grade II.

a b

Preoperative bone-window MRI (a–c) and CT (d) of rator. (i) Identification of the proximal segment of the
the patient: giant VS on the left side with a maximal facial nerve (7) at the brain stem. (j) Further tumor
diameter >5 cm, extreme compression of the brain removal: internal decompression and dissection at the
stem, and dilation of the lateral and third ventricles. arachnoid plane. Longer segment of the facial nerve
(a, b) T1-weighted contrast-enhanced sequences. (c) (arrows) is now visible. (k) Identification of the lateral
T2-weighted sequence. The tumor extended up to the intracanalicular segment of the facial nerve. (l–n) The
fundus (c) but did not cause significant dilatation of facial nerve was further dissected from the tumor cap-
the IAC (d). Intraoperative images. (e) Initial view of sule: it was splayed (umbrellalike) on the anteroinfe-
the tumor in the CP angle. (f) The IAC is opened with rior tumor surface (BS brain stem). (o) Complete tumor
a diamond drill. (g) The adhesions at the inferior and removal and preservation of the facial nerve. (p, q)
superior borders of the porus acusticus internus are Early postoperative CT demonstrating the wide open-
released, which allowed better mobilization of the ing of the IAC. Note the preserved semicircular
tumor. (h) Tumor exenteration with an ultrasonic aspi- canals
208 5 Vestibular Schwannomas

c d

e f

g h
Management of Patients with VS 209

i j

k l
210 5 Vestibular Schwannomas

m n

p
o

q
Management of Patients with VS 211

Case Illustration 5.6 nerve was identified laterally in the fundus


VS with Large Petrous Bone Involvement area and could be preserved (j, k). Then the
This patient (44-year-old male) presented with CP angle tumor part was debulked and dis-
a history of a gradual hearing loss on the left sected from the cerebellum (l). The facial
side. Later on, a feeling of heaviness in the ear nerve was identified at the brain stem and fol-
appeared, and neuroimaging was performed, lowed laterally (m–o). The tumor was removed
which demonstrated a large VS on the left side completely, and the facial nerve anatomical
with massive destruction and involvement of integrity was preserved (p). The whole opened
the petrous bone (a–h). At admission he had area of the petrous bone was filled with fat
left-sided anacusis and otherwise normal neu- pieces and sealed with fibrin glue.
rological status. He was operated via the ret- The patient recovered quickly after surgery.
rosigmoid route. Initially, the dura and the thin His early postoperative CT scan is shown on
layer of bone overlying the petrous tumor figures (q–s). On the eighth postoperative day,
were removed, and this part, which reached however, a rhinoliquorrhoea appeared and
from the petrous apex to the jugular foramen treated with a lumbar drainage for 1 week. At
and compressed the lower cranial nerves and discharge he had only HB grade IV facial weak-
the jugular bulb, was debulked (i). The facial ness, which at follow-up improved to grade II.

a b

Preoperative imaging: (a, b) Bone-window CT demon- the facial nerve at the fundus and its gradual dissection.
strating the wide destruction of the petrous bone. (c–f) (k) (FN—facial nerve). (l) Debulking of the CP angle
T1-weighted MR images with contrast. (g, h) T2-CISS tumor part. (m) Identification of the facial nerve at the
images. (Intraoperative images: (i) Exposure of the brain stem and its gradual dissection from the tumor
tumor; a small bone septum (small arrow) divides the capsule (n, o). (p) Complete tumor removal. (q–s)
petrous from the CP angle tumor parts (P petrous bone, Early postoperative CT control. The opened petrous
large arrow petrous tumor part). (j) Identification of bone is filled with fat pieces
212 5 Vestibular Schwannomas

c d

e
f

g
Management of Patients with VS 213

i j

k l

m n
214 5 Vestibular Schwannomas

o p

q r

s
Management of Patients with VS 215

Tumor Adhesion/Presence of Capsule strated even with the most advanced neuroimag-
The degree of adhesion of the tumor capsule to ing techniques and cannot be predicted before
the cranial nerves is a major predictor of func- surgery (see Case Illustration 5.8). In contrast to
tional and general outcome after VS surgery. VSs meningiomas, the presence of perilesional edema
are traditionally described as being well encapsu- does not necessarily correlate with a lack of
lated. At surgery the tumor surface tends to be arachnoid plane (Fig. 5.35).
firmer and more vascular than its central regions. Keeping to the principles of VS microsurgery
Histologically, however, only an extremely thin allows achieving optimal results even in very
connective tissue layer (3–5 mm) appears to cover adherent tumors: dissection plane can almost
the tumor [121]. The most probable origin of this always be found and should be meticulously fol-
layer appears to be the perineurium of the ves- lowed. The tumor should be dissected from the
tibular nerves [205]. VSs represent a heteroge- nerve, not the nerve from the tumor! The stan-
neous group of tumors in terms of presence of dard technique of dissection by gripping the
such surgical capsule or of an arachnoid plane arachnoid sheath with two forceps and carefully
between the tumor and the neural structures. The stripping it of the capsule, however, is not appli-
treatment strategy in each case should be there- cable if the arachnoid plane does not exist. If no
fore individualized according to these character- capsule exists, the “onion-skin technique” of
istics. In case a distinct arachnoid cleavage plane tumor removal may be applied: the tumor is
is present, the dissection of the tumor is straight- removed layer by layer—from the inner to outer
forward (see Case Illustration 5.7). In a consider- one—until the outermost layer is reached and
able number of cases, however, the tumor is removed piecemeal (see Case Illustration 5.9).
densely adherent to the surrounding CP angle This modified technique prolongs the operative
structures, in particular to the facial and cochlear time but allows for cochlear and facial nerve
nerves. The extent of adhesion cannot be demon- functional preservation.

a b

Fig. 5.35 Preoperative MR images (a–d) of VS with diffuse bleeding. On the early postoperative CT (e)—1
pronounced perifocal edema. The fourth ventricle was day after surgery—only air in the CP angle was visible.
obstructed with resulting hydrocephalus. Despite the The next CT performed 3 days later to control the hydro-
radiological evidence of edema, however, the dissection cephalus (f), however, demonstrated small hematoma in
of the facial nerve did not prove to be more difficult and a the tumor bed area. The patient remained asymptomatic
good arachnoid plane was available. The adjacent cerebel- and was managed conservatively
lar parenchyma, however, was softened and prone to
216 5 Vestibular Schwannomas

c d

e f

Fig. 5.35 (continued)

In case the adhesion is so strong that the tumor remnant correlates with the risk of recur-
integrity of the nerve is at risk, the most adherent rence, as shown earlier in the text, every attempt
part of the capsule should not be removed (see should be made to remove as much tumor as
Case Illustration 5.10). Since the size of the safely possible.
Management of Patients with VS 217

Case Illustration 5.7 and MR imaging showed an intracanalicular


Intracanalicular VS with Well-Developed VS (extension grade T1) on the right side,
Capsule and Lack of Adhesions spreading to the fundus of the IAC (a, b).
This 43-year-old female presented with a his- Audiogram and speech discrimination test
tory of tinnitus, vertigo, and progressive hear- showed that she still had good hearing (class
ing loss on the right side since 12 months. CT H2 according to the New Hannover

a b

c d

7–8

(a) Preoperative MRI of the patient: intracanalicular is still intact. (e) Partial removal of the tumor. The
VS on the right side (T1-weighted sequences follow- superior vestibular nerve, from which the tumor origi-
ing gadolinium administration). (b) Intraoperative nated, is visible (VN). (f) The tumor had a well-devel-
images. (c) Initial view of the CP angle and vestibulo- oped capsule and was not adherent. It could be
cochlear nerve complex (7–8). The dura around the relatively easily dissected from the facial (FN) and
posterior IAC wall is incised (arrowheads). (d) The cochlear nerves (CN) and removed completely (g). (h)
IAC has been opened, but the dura overlying the tumor The IAC is sealed with fat pieces
218 5 Vestibular Schwannomas

e f

g h
Management of Patients with VS 219

Case Illustration 5.8 and decided for surgery, which was performed
Intracanalicular VS with Very Strong via the RS approach (c–h). The tumor was
Adhesions to the Nerves found to be very adherent to the facial and
This 54-year-old male patient was admitted cochlear nerves; its removal required pro-
with a history of hearing loss, tinnitus, and longed and meticulous dissection. Following
vertigo since several months. He had been surgery the patient had transient mild facial
diagnosed to have a very small tumor in the nerve palsy (HB grade II) that recovered com-
lateral part of the left IAC with imaging char- pletely within 3 months. Hearing was slightly
acteristics of VS (a, b). The audiogram worse than before surgery—class H3 accord-
showed that he had still useful hearing (class ing to the New Hannover classification. The
H2 according to the New Hannover vertigo resolved completely within several
classification.) The patient was informed in weeks, but the tinnitus became slightly more
detail on the alternative management options annoying.

a b

Preoperative MRI of the patient: small intracanalicular superior and inferior vestibular nerves (SVN and IVN,
VS on the left side, located in the lateral part of the IAC respectively) is seen. (f) The lateral part of the tumor is
(a) T1-weighted sequences following gadolinium admin- dissected from the facial (FN) and cochlear nerves (CN)
istration. (b) CISS sequence. Intraoperative images. and mobilized medially. (g) Despite its small size, the
(c) Initial view of the CP angle and the vestibulocochlear tumor was extremely adherent to the nerves, and its
nerve complex. The cerebellomedullary cistern has been removal required prolonged and meticulous dissection.
opened, and sufficient amount of CSF has been drained. Still, it could be removed completely, preserving the
(d, e) The IAC has been opened, and the tumor with the integrity of all neural structures (h)
220 5 Vestibular Schwannomas

c d

e f
Management of Patients with VS 221

g h
222 5 Vestibular Schwannomas

Case Illustration 5.9 the New Hannover classification). The MRI


VS Without Obvious Tumor Capsule study is presented on figures (a, b).
Fifty-three-year-old male patient was diag- The tumor was approached via the RS
nosed to have VS on the left side 6 months route. It had no capsule and was “sticking” to
prior to presentation to our institute. He suf- the neural structures (c–h). Therefore, it had
fered from strong left-sided headache, imbal- to be removed with the “onion-skin peeling
ance, and progressive hearing loss since technique”—piecemeal removal from inside
several years. At admission he had moderate to outside. The VS was removed completely,
hearing on the left side (class H3 according to and the anatomical integrity of the facial and

a b

c d

(a) Preoperative MRI of a patient with a VS in the tumor part was approached. Internal debulking has
left CP angle—extension grade T3b (T1-weighted been performed and the facial nerve identified
sequences following gadolinium administration). (b) (arrow). The VS had no capsule and appeared
Intraoperative images illustrating the steps of tumor “sticked” to the neural structures. (f) Further tumor
removal. (c) The posterior wall of IAC has been removal—mainly piecemeal—from inside to outside.
drilled off, and the intracanalicular tumor portion is (g) Last pieces of the tumor are dissected from the
exposed. (d) Initial removal of the intracanalicular facial nerve. (h) Complete tumor removal. The elec-
tumor and identification of the distal segments of trical stimulation of the facial nerve evoked good
facial and cochlear nerves. (e) Then, the CP angle response
Management of Patients with VS 223

cochlear nerves was preserved. Still, the was uneventful. At the first follow-up exami-
patient had an HB grade V facial nerve func- nation, the facial nerve function had improved
tion after surgery, and the hearing was not to HB grade II.
useful. Otherwise, the postoperative period

e f

g h
224 5 Vestibular Schwannomas

Case Illustration 5.10 intense adhesions between the tumor capsule


Incomplete Tumor Removal Due to Strong and the facial nerve in the area next to the
Adhesions porus acusticus internus were found. The
The patient presented with a history of pro- attempt to dissect this part seemed to be related
gressive right-sided hearing loss and intermit- to a very high risk of nerve injury or complete
tent tinnitus since 2 years and unsteadiness nerve disruption. Hence, a decision was taken
since 3 months. At admission he had service- not to remove this tiny tumor part. The patient
able hearing on the right side, gait ataxia, and recovered well from surgery and was dis-
horizontal spontaneous nystagmus toward the charged with an improved gait, anacusis, and
side of the tumor. The MR examination dem- an HB grade III facial nerve function on the
onstrated a large right-sided VS (grade T4a), right side. He is being followed up with yearly
causing moderate compression of the brain MR studies. Within 12 months the facial nerve
stem. function improved to an HB grade II. Up to
The patient was operated via the RS now—36 months after surgery—the remnant
approach (figures c–n). During surgery very remains unchanged.

a b

(a, b) Large VS (grade T4b) on the right side extracanalicular tumor part with CUSA. (g) Gradually
(T1-weighted sequences after gadolinium adminis- the brain stem (BS) and the facial nerve (7) in its
tration). Intraoperative images. (c) Overview of the proximal segment were exposed. PV—petrous vein.
CP angle and the VS (Tu). TS transverse sinus, SS (h) Dissection of the facial nerve. The tumor rest is
sigmoid sinus. (d) The pyramid bone has been widely held with slight tension in a superior and lateral
drilled off (arrowheads), exposing the IAC (black direction. (i, j) Further dissection of the nerve by
dots). (e) The intracanalicular tumor part has been pulling the tumor laterally. Note the splayed or
removed, and the facial nerve (7) has been dissected umbrellalike shape of the nerve (arrows). (k, l) The
free. The cochlear nerve is visible inferior to the last part to be dissected is the one close to the porus
facial nerve. BAEP at the beginning of surgery acusticus internus. (m) Due to the intense adhesions
showed increased latency and decreased amplitude between the tumor rest and the facial nerve and the
of both waves III and V. During the bone removal, impossibility to dissect the nerve without endanger-
they were suddenly completely lost. The retractor ing its integrity, this small part of the tumor was not
was removed and a pause was made, but the waves removed (double arrow). The electrical stimulation
did not recover. (f) Internal decompression of the of the facial nerve evoked good response (n)
Management of Patients with VS 225

c d

e f
226 5 Vestibular Schwannomas

g h

i j
Management of Patients with VS 227

k l

m n
228 5 Vestibular Schwannomas

Medial VSs does not show an intracanalicular component,


Medial VSs are located in the CP angle and do but at surgery tumor extension up to the fundus
not extend or extend minimally into the IAC, may be found. Therefore, regardless of the pre-
while the lateral part of the IAC is free of tumor operative imaging, the initial step is always to
[96, 248]. Although they are relatively rare—2 % open the IAC and to identify all relevant nerves
of all VSs—they deserve special attention because distal to the tumor (see Case Illustration 5.11).
the tumor may reach a considerable size before The extent of lateral tumor extension should be
diagnosis [230, 248]. Nevertheless, as shown by considered when planning the IAC opening: tai-
Strauss et al. [230] and by our own experience, the lored bone removal affords enough space for
facial and cochlear nerves are less adherent to the tumor removal and avoids unnecessary risks.
tumor than in case of tumors extending into the The functional outcome of surgery of medial
IAC. The MRI may be misleading when the lateral VSs has been shown to be better than in usual VS
tumor extension is evaluated: not uncommonly it of comparable size [230].
Management of Patients with VS 229

Case Illustration 5.11 loss of more than 90 dB in the main speech


Medial VS area. MR images demonstrated relatively large
This 45-year-old male patient presented with VS with a maximal diameter of 3.7 cm and a
a history of progressive hearing loss on the left predominant craniocaudal extension (a–d)
side since 5 years and intermittent tinnitus with a minimal growth into the IAC, and
since 3 years. His neurological examination medial VS was suspected. The caudal tumor
was normal, besides the hypacusis and a slight extension could explain the early and severe
gait ataxia. The audiogram showed a hearing hearing loss.

a b

c d

(a) Preoperative MRI demonstrating a medial VS in did not extend into the canal, as demonstrated on the
the left CP angle. The IAC is free of tumor (c, d). Note images. (g) The lower cranial nerves were than dis-
the pronounced caudal tumor extension (b). (a, b) sected free from the tumor (*, **, ***—ninth, tenth,
T1-weighted sequences following gadolinium admin- and 11th cranial nerves, respectively). Image (h) dem-
istration. (c) T2- weighted sequence. (d) T2-CISS onstrates the splayed “umbrellalike” facial nerve after
sequence. Intraoperative images illustrating the steps the complete tumor removal (BS brain stem). Despite
of tumor removal. (e) Limited opening of the IAC has this shape, the nerve reacted normally to electrical
been performed, and the facial nerve (FN) has been stimulation (i)
identified, applying electrical stimulation (f). The VS
230 5 Vestibular Schwannomas

The typical RS approach was performed, case of medial VS (e). The tumor originated
but the IAC was opened only in its medial 1/3: from the lower vestibular nerve. The facial
a limited, 2–3-mm, intracanalicular tumor nerve was seen and verified with electrical
extension was seen, proving that this was a stimulation (f). Next, the extrameatal part was

e f

g h

i
Management of Patients with VS 231

addressed: the VS was found to extend cau- later—it was displaced more cranially. The
dally below the level of the caudal cranial nerve was further dissected by slightly luxat-
nerves (g). ing the remaining tumor cranially. The lateral
Therefore, the caudal tumor part was cisternal segment of the facial nerve was
approached initially. It was debulked, and the spread like an umbrella (h). A small tumor
caudal cranial nerves, which were severely piece was strongly adherent to the nerve and—
compressed by the tumor, were dissected free. following the principle of intermittent dissec-
Then, the more cranial portion was debulked tion from different sides—the dissection
and dissected from the cerebellum and brain proceeded from lateral to medial. The tumor
stem. The facial nerve could not be seen at its was removed completely, and the preserved
usual location and was identified somewhat facial nerve responded well to stimulation (i).
232 5 Vestibular Schwannomas

Cystic VS
Cystic VS should be considered as a separate
subgroup. They have different pathological
structure and a tendency for more aggressive or
unpredictable biological behavior. Frequently a
more rapid growth rate is observed. A higher
degree of nuclear atypia and differences in the
gene expression profiles of such tumors have
been found [38, 261].
It is supposed that cyst formation is caused
by central necrosis, coalescence of microcysts,
and/or repeated intratumoral hemorrhage
[164]. Their further enlargement can be
explained by the formation of an osmotic gra- a
dient between the cyst and the solid tumor and
impairment of the blood–tumor barrier. In a
recent study, Moon et al. [153] reported to have
identified high levels of proteolytic enzymes,
such as matrix metalloproteinase, in cyst fluid
and cyst walls. These enzymes are proposed to
play an important role in the pathogenesis of
cyst formation and peritumoral adhesion in
cystic VS.
Cystic VSs comprise between 2 and 13 % of
all VSs [33, 72, 170, 194]. The criteria for cys-
tic VS are radiological and pathological: on
T1-weighted sequences, the cystic tumor parts
are hypointense, while on T2-weighted b
sequences, they are hyperintense [62]
(Fig. 5.36). The cysts do not enhance with gad-
olinium administration, while their wall
enhances—a feature that allows their differen-
tiation from arachnoid and epidermoid cysts.
Cystic tumors should be differentiated from
VS with an arachnoid pseudocyst in the periph-
ery, which is not part of the tumor. The wall of
such pseudocysts neither enhances with gado-
linium administration, nor does it have the
structure of a typical arachnoid cyst (Fig. 5.36).
Occasionally cystic changes in the brain stem
may be observed, which appear to be similar to
those developing in case of intramedullary c
tumors. The mechanism of their development
in VS is unclear. Fig. 5.36 MR images of patients with solid (a) and cys-
tic VSs (b, c). The peripherally located large cysts in cases
a and b is a CSF accumulation (arachnoid pseudocyst),
while the contrast-enhancing peripheral cyst in case c is a
real tumor cyst
Management of Patients with VS 233

Case Illustration 5.12 The patient was operated via the RS approach
Cystic VS 1 (see figures d–n), complete tumor removal was
Forty-two-year-old male patient presenting achieved, and the facial nerve was preserved.
with a history of progressive hearing loss, The postoperative period was uneventful, and
unsteadiness, and vertigo. His neurological at discharge the only new deficit was an HB
examination revealed trigeminal nerve hypes- grade IV facial nerve palsy. The patient was
thesia (V2–3), absent corneal reflex, and pro- examined again 13 months after the surgery. He
found hearing loss on the right side. The MRI had no complaints, and the facial nerve recov-
demonstrated a large cystic VS on the right ered completely (HB grade I). The follow-up
side with maximal diameter of 3.6 cm (a–c). MRI is presented on figures (o–r).

a b

Cystic VS. MR images of a patient with a large cystic VS is exposed and consecutively dissected from the tumor
on the left side (a, b) T1-weighted sequences following capsule (i–k). (l) The most adherent part of the tumor,
contrast administration. (c) CISS sequence. Note the typi- near the porus of the IAC, was removed at the end. (m)
cal contrast enhancement of the cyst wall, which differen- Complete tumor removal with preservation of the facial
tiates it from arachnoid cysts or pseudocysts. Intraoperative nerve. (n) Overview of the CP angle: BS brain stem, 7
images. (d) The IAC has been opened, and the intrac- facial nerve, 6 abducens nerve. Postoperative images
analicular tumor part has been exposed (arrows). (e) The demonstrating the completeness of tumor removal. (o)
partially cystic CP angle tumor part is well visible (arrow- Bone-window CT. Note the wide exposure of the IAC up
head). (f) Tumor debulking. (g) Dissection of the distal to its fundus. (p, q) Axial and coronal MR sequences fol-
segment of the facial nerve (white arrow) in the IAC. lowing contrast administration. The fat plug in the proxi-
(h) The proximal segment of the nerve at the brain stem mal IAC is visualized. (r) T2-weighted sequence
234 5 Vestibular Schwannomas

d e

f g
Management of Patients with VS 235

h i

j k
236 5 Vestibular Schwannomas

l m

n o

p
Management of Patients with VS 237

q r
238 5 Vestibular Schwannomas

Many VSs appear internally heterogeneous facial nerve function after surgery [272]. Most
containing interspersed regions of soft and firm large clinical series confirm this negative corre-
consistency. In case of real cystic VS, however, at lation [72, 170, 194]. Charabi et al. [35] reported
surgery fluid is seen within the cysts. Although it on the outcome of treatment of 571 VS; among
has been proposed to subdivide homogeneous them 23 were cystic. In the cystic tumor group,
and heterogeneous VSs, this subclassification has the facial nerve was accidentally transected in
no practical significance. 43 %—a much higher rate than the facial nerve
Duration of symptoms in patients with cystic transection rate for noncystic VS of similar size
VSs prior to diagnosis is shorter than in those in their experience. In a previous study of 1,000
with solid tumors. The cystic component may patients, we found that the anatomic preserva-
expand rapidly and cause brain stem compres- tion rate of the facial nerve in cystic VS was
sion and hydrocephalus. Cystic VSs tend to reduced from 93 to 88 % and of the cochlear
have a more unfavorable surgical outcome in nerve from 68 to 55 % [199].
regard to facial nerve function, hearing, and In case of very severe adhesions, a reasonable
morbidity rate [72, 198]. Nevertheless, surgery option is to leave behind a small part of the cyst
is definitely the optimal treatment option of not to destroy the nerve. In a recently evaluated
such tumors (Figs. 5.37 and 5.38). “Wait-and- series of 200 VSs [194], we performed subtotal
scan” policy is not recommended because of the tumor removal in four cases (2 %). Three of
frequently observed rapid enlargement with them had cystic VSs: in all the adhesion was so
neurological deterioration [38]. Cystic VS may tight that the attempt to dissect the nerve from
respond poorly to radiosurgery. Furthermore, the tumor capsule led to intense electromyo-
the rate of facial nerve weakness after radiosur- graphically demonstrated changes. Moreover,
gery is higher than in solid or NF-2-associated from the 3 patients (1.5 %), in whom the facial
schwannomas [169]. Close adherence to the nerve could not be preserved anatomically, 2
principles of VS microsurgery, meticulous had cystic VS.
hemostasis, and extremely careful observation Patients with cystic tumors are at higher risk
of the patients after surgery allow achieving for postoperative hemorrhagic complications. A
acceptable operative results (Fig. 5.37). common observation during surgery is that the
Surgery may be complicated by the tighter cerebellar surface in contact with the cystic
adherence of the cyst wall to neurovascular struc- schwannoma is softened and has a propensity for
tures. The solid portions of cystic VSs are fre- diffuse bleeding, probably as a consequence of
quently hypervascularized and more difficult to the proteolytic action of the cyst enzymes. In case
handle (see Case Illustration 5.12). During sur- perfect hemostasis has not been achieved, this
gery the abrupt change in the shape of the lesion, can cause delayed bleeding in the CP angle.
following the aspiration of the fluid content, can Therefore, in addition to the “standard” hemosta-
lead to rupture of the surface vessels and cause sis, we coagulate the whole softened cerebellar
excessive traction to the facial nerve. surface and cover it abundantly with Tabotamp.
The cranial nerves are typically splayed over In summary, although cystic VSs pose
the cysts, and an adequate subarachnoid dissec- specific challenges to the surgeon, strict adher-
tion plane is frequently absent. The facial nerve ence to the principles of VS removal and use of
can be markedly distorted and may be encoun- meticulous dissection technique allow to
tered unexpectedly. Cystic tumor components achieve results, comparable to that in common
are one of the most reliable predictors for the tumors.
Management of Patients with VS 239

a
b

c
d

e f

Fig. 5.37 MR images of a patient with a cystic left-sided examination 12 months after surgery, the patient had nor-
VS, presenting with complete hearing loss, vertigo, head- mal facial nerve function. The postoperative MR study is
ache, and trigeminal nerve sensory dysfunction. presented on figures (e–h). (e, f) Native T1-weighted
Preoperative images: (a) Bone-window CT, (b, c) sequence. (g) T1-weighted sequence after contrast admin-
T1-weighted images after contrast administration, and (d) istration. Note the fat plug in the area of the drilled IAC,
T2-weighted image. The tumor was removed completely visible as high intensity signal on images (e and f). (h)
and the facial nerve was preserved. At the follow-up T2-CISS image
240 5 Vestibular Schwannomas

g h

Fig. 5. 37 (continued)

Surgery of Recurrent VS 194, 266]. In the senior authors’ series, it is


Patients with tumor remnants after incomplete approximately 0.5 % of patients without NF-2
removal are followed with regular yearly MR and (0.8 % in the older series). In case a small recur-
clinical examinations. If the remnant shows a rence is detected in asymptomatic patient, initial
growth tendency, redo surgery should be per- follow-up is warranted. If the recurrence shows a
formed and the tumor should be removed com- steady growth tendency or causes neurological
pletely. The recurrence rate after VS removal symptoms, operative removal should be recom-
ranges from 0.5 to 18 % in the literature [133, mended (Figs. 5.39 and 5.40).
Management of Patients with VS 241

a b

Fig. 5.38 MRI of a patient with a large cystic VS. (a, b) T1-weighted sequences after contrast application. (c, d) CISS
sequences. (e) Intraoperative image: complete tumor removal with preservation of the facial nerve (arrow)
242 5 Vestibular Schwannomas

a b

c d

Fig. 5.39 MRI images of a 45-year-old patient, obtained irregular shape of the tumor, in particular on images (b
24 months after his initial surgery (radiologically verified and d), which is not typical for primary VSs. (a)
complete removal of a right-sided intracanalicular VS in T1-weighted MRI. (b, c) T1-weighted contrast-enhanced
another department via an RS approach). Tumor recur- sequences. (d) T2 sequence. (e) CISS sequence
rence with extrameatal extension is obvious. Note the
Management of Patients with VS 243

a b

c d

Fig. 5.40 (a–e) Preoperative MR images of a patient pre- removed completely and the facial nerve was preserved.
senting with a large recurrent VS 14 years after the first At discharge he had an HB grade IV facial palsy and no
surgery. Besides the headache that he had since several further deficits. The facial nerve function improved to HB
months, he had no further complaints. Hearing on the grade II at 12 months follow-up examination. (f–h)
right side was lost at initial surgery. The tumor was Postoperative MRI (12 months after surgery)
244 5 Vestibular Schwannomas

e f

g h

Fig. 5.40 (continued)


Management of Patients with VS 245

Case Illustration 5.13 ataxia, rendering walking—even with help—


Giant VS Operated Seven Times impossible. A ventriculoperitoneal shunt has
Previously been placed after the third surgery and was
This patient has been operated seven times in still patent. The patient had furthermore signs
different neurosurgical facilities before pre- of respiratory insufficiency but was not venti-
sentation in INI. At admission he had trigemi- lator dependent.
nal hypesthesia, abducens nerve palsy, HB Still, taking into account the size of the
grade V facial palsy, anacusis, and lower cra- tumor, the compression of the brain stem,
nial nerve dysfunction on the right side. He and the lower cranial nerve deficit, a plastic
had severe discoordination syndrome and tracheostomy was done prior to surgery. The

a b

c d

Preoperative MRI images (a–d). Postoperative CT—2 weeks after surgery (e)
246 5 Vestibular Schwannomas

e tumor could be removed completely via the


standard RS approach despite the very severe
adhesions and lack of any arachnoid plane.
The facial nerve, however, could not be
identified—probably it was destroyed
previously.
After surgery the patient received pro-
longed intensive treatment. Gradually, how-
ever, he could be weaned off the ventilator.
His initially worsened swallowing improved,
and the tracheal cannula could be removed.
The balance and body control also improved,
and at discharge he was able to walk with
support.
Management of Patients with VS 247

Preservation of functions after previous sur- (approach, extent of tumor removal), size and
gery can be very challenging. Typically adhe- location of the remnant/recurrent tumor, neuro-
sions and scar tissue obscure the normal logical status of the patient, and goal of surgery
anatomical landmarks. The initial orientation (see Case Illustration 5.14). Thus, if the patient
within the CP angle and the exposure of the presents with complete facial palsy prior to sur-
tumor are more demanding (see Case Illustration gery, an EMG should be performed and opera-
5.13). The tumor is frequently very adherent to tive reconstruction of the nerve should be
the cranial nerves, and the nerves themselves planned. Even in patients with incomplete but
may be more vulnerable. Hence, tumor removal severe facial palsy that did not recover after
should proceed only after definitive identification previous surgery, the optimal management may
of the neural structures. The facial nerve should be to remove the tumor, sacrifice the nerve
be identified initially in its healthy segment; the (which in such circumstances is typically atro-
scar/adhesion area along its course should be phic and responds poorly to electrostimulation),
approached last. The surgical strategy should be and reconstruct it with an interposition graft or
adapted to the findings in each individual case. perform a hypoglossal–facial nerve anastomo-
Factors to consider are type of previous surgery sis (see Chap. 12 for details).
248 5 Vestibular Schwannomas

Case Illustration 5.14 grade T3a VS, HB grade III facial nerve palsy,
Recurrent VS After Middle Fossa Removal and hypacusis (PTA of 50–60 dB).
This 42-year-old male patient complained The RS approach was used in the case, and
since 2007 from progressive hearing loss on the IAC was opened in the standard technique
the right side. A pure intracanalicular VS was (e–h). In the area of the fundus, however, some
diagnosed, and he has been operated at another fibrotic changes were seen, which led us to
department in the early 2008: the tumor was conclude that the facial nerve might be densely
removed partially via the MF approach. adherent or even involved by the scar tissue
Following surgery the patient had HB grade V (during the MF approach, the facial nerve lies
facial nerve palsy that later improved. The between the tumor and the surgeon, as
follow-up MRI controls, however, revealed described in the Chap. “MF Approach”). The
that the residual tumor had a growth tendency surgical strategy in case of previous surgery
and expanded into the CP angle cistern (a–d). via the MF approach should be modified,
The patient presented to our institute with a taken into consideration the expected adhe-

a b

MRI examination performed at our institute: the large view of the CP angle and the vestibulocochlear nerve
tumor remnant in the right IAC with a small extension complex, dislocated posteriorly by the tumor. (f) The
in the CP angle is seen. (a–c) T1-weighted sequences IAC has been opened, and the tumor has been partially
following gadolinium administration. (d) T2-weighted removed. (g) The most lateral tumor part is the last to
sequence. Intraoperative images. See the text for be removed. (h) Complete tumor removal with preser-
detailed description of the operative steps. (e) Initial vation of the facial and cochlear nerves
Management of Patients with VS 249

sions of the facial nerve and the tumor especially difficult to differentiate the tumor
capsule/fibrous tissue in the area of the previ- from fibrous tissue in the fundus area (g), and
ous surgery. Tumor debulking is performed we had to rely on the electrostimulation to
initially in the CP angle, and the facial and identify the nerve. The cochlear nerve was
cochlear nerves are identified medially to the found to be less involved by the scar.
tumor (f). Even in case of large VSs, the nerves Ultimately, the tumor could be completely
can be identified at the brain stem. removed, and both nerves were preserved (h).
Then, the tumor dissection and removal After the surgery the patient did not have
proceeded laterally with constant verification any new deficit, and the preoperative hearing
of the location of the facial nerve with electro- level was preserved. He was discharged on the
stimulation. In this particular case, it was tenth postoperative day.

e f

g h
250 5 Vestibular Schwannomas

Facial Nerve Preservation in VS Surgery system, as pointed by Kanzaki et al. [110], is the
The facial nerve function is a major factor deter- definition of grade 3 dysfunction. Patients cate-
mining the quality of life of patients after CP gorized in this grade may have dry eye and/or
angle surgery. Facial nerve palsy besides the cos- exposure keratitis that is not accounted for.
metic disfigurement increases the risk of ocular Furthermore, the function of the intermediate
complications, such as corneal damage, ulcer- nerve is not taken into consideration. At the
ation, and even blindness. The increased or Tokyo Consensus Meeting, a modification of the
reduced tearing (dry eye), salivation disturbances, House–Brackmann grading scale was proposed,
increased nasal secretions, and sensitivity to light claiming to allow more objective and precise
or taste abnormalities may cause significant dis- evaluation [110]. In this facial motion
comfort. The weakness of orbicularis oris and classification, grade III is subdivided into grade
buccinators muscles impairs speech and chew- IIIA and grade IIIB, and criteria for the degree of
ing. The negative effects are further exaggerated synkinesis and hemifacial spasm are added. The
by the development of synkinesis and the relative classification allows further a record of nervus
hyperactivity of the contralateral side. intermedius function (Table 5.4).
Patients with facial dysfunction experience
distress that—according to some studies—is not Predictive Factors for the Preservation of
correlated to the clinical grade of the facial the Facial Nerve
nerve paralysis. Even an incomplete facial Several factors are known to influence the ability
weakness can have devastating impact on the of the surgeon to preserve facial nerve function:
social life and psychological well-being of the tumor size, tumor type, existence of arachnoid
patients [176]. plane, intraoperative facial nerve monitoring,
previous surgery or radiosurgery, surgical
Facial Nerve Grading Systems approach, and experience of the surgical team.
The generally accepted facial nerve function
grading system is the House–Brackmann grad- Tumor Characteristics
ing scale, proposed in 1985 (Table 5.3) [95]. It Tumor size is perhaps the most important predic-
has been widely validated and allows for estima- tor of the quality of facial nerve function after
tion of facial nerve function with small interob- surgery. During tumor growth the facial nerve is
server variability. The major weakness of this progressively stretched and becomes extremely

Table 5.3 House–Brackmann grading system [95]


Grade Description Characteristics at rest Characteristics in motion
1. Normal Normal function in all muscles Normal Normal
2. Mild dysfunction Slight weakness noticeable Normal symmetry and No synkinesis, contracture, or
only on close inspection tone hemifacial spasm
3. Moderate Obvious but not disfiguring Normal symmetry and Noticeable, but not severe
dysfunction weakness, no functional tone synkinesis or contracture,
impairment hemifacial spasm, or both
4. Moderately severe Obvious weakness, disfiguring Normal symmetry and Obvious synkinesis or mass
dysfunction asymmetry; possible functional tone action, hemifacial spasm
impairment
5. Severe dysfunction Only barely perceptible motion Asymmetry w/ droop at Synkinesis, contracture, and
corner of mouth and hemifacial spasm usually
decreased nasal labial absent
fold
6. Total paralysis No motion No tone, asymmetry, no
synkinesis, contracture,
or hemifacial spasm
Management of Patients with VS 251

Table 5.4 Facial motion classification


Grade Tonus at rest Global aspect Forehead Mimic eye Mouth Hemispasm
1. Normal Normal Normal Normal Normal occlusion Normal Absent
2. Light Normal Light Light Eyelid occlusion: Very light Absent or
asymmetry asymmetry complete without asymmetry very light
effort
3. Moderate Normal or very Moderate Moderate Eyelid occlusion: Clear Present,
A (light) light asymmetry asymmetry asymmetry complete with asymmetry moderate
effort comfortable protracted lips
eye occluded
3. Moderate Normal or very Clear Clear Eyelid occlusion: Clear Present,
B (heavy) light asymmetry asymmetry asymmetry complete with asymmetry clear
effort ocular protracted lips
discomfort not occluded
4. Real Real asymmetry Real Real Eyelid occlusion: Real Present,
asymmetry asymmetry incomplete asymmetry greater
5. Heavy Greater Greater Greater Eyelid occlusion: Greater Absent
asymmetry asymmetry asymmetry roughed asymmetry
6. Total Flaccidity Greater Flaccidity No eyelid Flaccidity Absent
asymmetry occlusion
Note Crocodile tears: Taste troubles: Eye: not dry
CO, C+ T0, T+
Kanzaki et al. [110]

vulnerable. It is frequently splayed over the tive facial nerve function correlated significantly
tumor capsule and consists of multiple small to tumor stage, diameter, and tumor volume. The
bundles or has an “umbrellalike” appearance. increase of any of the extrameatal tumor diame-
Any traction or manipulation during tumor ters led to worse outcome. However, the anterior
removal may easily lead to the rupture of these and/or caudal tumor extension had more
nerve fibers even if the continuity of the bundles significant correlation than the posterior and/or
is not obviously affected. cranial extensions. VSs with significant anterior
The correlation between tumor size and facial extension could lead to more severe stretching
outcome has been proven by numerous studies and thus vulnerability of the facial nerve. This
[125, 194, 198]. The anatomical preservation rate may be the explanation for the above-mentioned
in our general series was 98.5 %, while in tumor correlations.
grades T1, T2, and T3, it was 100 %. Functional The shape of the tumor also showed significant
outcome correlated to tumor size and best results correlation to facial nerve outcome: the outcome
were achieved in VSs grade T1 and in grade T3a is poorer in polycyclic VSs compared to round or
(HB grade I in 90 and 96 %, respectively). oval ones. The characteristics of the intracanali-
Recently we analyzed the predictive value of cular tumor portion, including tumor length,
various preoperative existing factors. We evalu- tumor width, and tumor–fundus distance, as well
ated patient-related and tumor-related factors, as the bony changes and characteristics of the
such as tumor diameter (anteroposterior or paral- ICA did not have significant relationship to
lel to the petrous ridge, mediolateral or perpen- immediate facial nerve outcome. Other authors
dicular to the petrous ridge, and sagittal or vertical have also reported that the rate of good facial
diameters); cranial, caudal, anterior, and poste- nerve function does not correlate to the extent of
rior tumor extension in relation to the IAC; width IAC destruction and/or widening [265].
and length of the intracanalicular tumor portion; The extent of adherence of the tumor to the
tumor–fundus distance; and tumor shape [77]. nerve—or the presence of a good arachnoid dis-
Our analysis showed that immediate postopera- section plane—is of even greater importance.
252 5 Vestibular Schwannomas

While the chance to preserve the facial nerve in Reoperations


large VSs is related mainly to the experience of During revision surgery, intense adhesions are
the surgeon, the presence of dense adherence generally encountered between the neurovascular
may hinder complete tumor removal even in structures and the tumor remnant. Occasionally it
experienced hands. might be difficult even to identify the facial nerve.
Frequent electrostimulation of the suspicious
Patient Age areas in such cases is extremely helpful.
Another important parameter is the patients’ age.
If increased age is associated with poor facial Radiosurgery
outcomes, as reported by Grey et al. [84], a deci- The clinical experience with VSs treated opera-
sion in favor of radiosurgery for older patients tively after failed radiosurgery shows that generally
would be logical. Multiple authors, however, abundant adhesions are present and the cleavage
showed that age is not a negative predictor [41, plane to the nerve is not so obvious. Some histo-
116]. Couloigner et al. [48] evaluated patient’s logical examinations revealed no clear interface
characteristics and clinical features, such as age, between the vestibular and facial nerves (see sec-
gender, and symptom duration, and found no cor- tion “Radiosurgery”). Not infrequently, the patients
relation to postoperative facial nerve function. have been both operated and irradiated previously,
Our analysis showed that age at initial symptom which further complicates the surgery. Despite the
and surgery did not correlate with immediate difficulties encountered in case of previously oper-
postoperative facial function. Furthermore, symp- ated or irradiated patients, facial nerve preservation
tom duration before surgery and patient’s gender is realistic, provided the general microsurgical
also did not correlate with outcome. The only principles of VS removal are followed.
neurological symptoms at presentation that cor-
related significantly with worse immediate facial Intraoperative Facial Nerve Monitoring
nerve function were gait instability and preopera- In a historical study, Harner et al. [91] compared
tive facial palsy. 48 matched cases operated with and without mon-
itoring and demonstrated that this technique sub-
NF-2 stantially improved the rate of facial nerve
Patients with NF-2-associated VSs tend to have preservation, especially in patients with larger VS.
poorer facial nerve outcome after surgery [195]. Nowadays the constant monitoring of the facial
The extent of adhesion of such tumors to the nerve is an absolute prerequisite for the preserva-
nerve is more intense, and a clear histologic tion of its functions. As shown recently, the facial
cleavage plane may be missing. Furthermore, the nerve motor evoked potential monitoring is a valu-
location of the facial nerve is highly variable and able adjunct to conventional facial nerve monitor-
less predictable. It may pass between the separate ing and may predict the functional outcome [2].
tumor lobules, and hence the risk of its inadver- Facial nerve stimulation thresholds have been
tent injury is higher. shown to have prognostic significance to the
degree of postoperative function. According to
Cystic Tumors the study of Neff et al. [159], stimulus threshold
The dissection and preservation of facial nerve in of 0.05 mA or less and a response amplitude of
cystic VSs are more difficult (see section “Cystic 240 mV or greater predicted a House–Brackmann
VSs”), and its preservation rates are respectively grade 1 or 2 function with 98 % probability. Any
lower. In our experience, the anatomic preserva- of the parameters alone, however, had a much
tion rate of the facial nerve was reduced from 93 lower predictive value.
in solid VSs to 88 % in cystic tumors and of the
cochlear nerve from 68 to 55 %, respectively. Preoperative Facial Palsy
Similar correlation exists in regard to the func- Facial palsy is a rare symptom prior to surgery:
tion of the nerves. its rate was found to be 2.3 % in a series of 1,000
Management of Patients with VS 253

patients [145]. Patients who presented with facial superior. We consider this controversy as a part
nerve palsy have a higher risk of poor postopera- of VS history. It is obvious that optimal results
tive facial nerve functioning. However, preopera- are obtained if the surgeon applies the technique
tive facial nerve dysfunction is more frequent in that he best masters.
large tumors. These two factors may, therefore,
influence independently the functional outcome. Causes of Facial Nerve Injury
(Modified from Sampath et al. [200])
Experience of the Surgical Team • Direct trauma, stretch, or torsion on the nerve.
The personal experience of the surgeon is of par- This mechanism is probably the most frequent
amount importance in regard to facial nerve pres- cause of facial nerve dysfunction and occurs
ervation. Many studies have demonstrated that during its dissection from the tumor capsule.
the outcome is inferior in the initial phases of the Less severe stretching causes neuropraxia and
learning curve. Welling et al. [262] demonstrated axonotmesis, which are potentially reversible.
a learning curve of 20 patients to be necessary for • Mechanical disruption or nerve transection.
attaining acceptable standards in the surgical Risk factors are less operative experience,
removal of VS. Elsmore and Mendoza [60] found cystic VSs, as well as the attempt to dissect
that the improvements occurred mainly between the nerve from the tumor from above.
the 40th and 100th cases. Buchmann et al. [29] • Devascularization of nerve segments. Most of
presented similar numbers: 60 cases are neces- the microvascular blood supply to the facial
sary before the new team achieved results similar nerve is in the subarachnoid space and may be
to those of highly experience surgeons. Moffat damaged if the correct dissection plane is not
et al. [151] in a review of 300 patients demon- followed.
strated obvious improvement in facial nerve pres- • Compromise of facial nerve vascular supply.
ervation from the first to the sixth group of 50 The facial nerve vascular supply is variable
patients: from 88 to 98 %. Interestingly, the and is derived from three separate systems: the
authors noted that a significant improvement labyrinthine artery, the greater superficial pet-
occurred after the first 50 patients and then again rosal branch of the middle meningeal artery,
after patient 200. In our recent analysis of a series and the stylomastoid artery off the external
of 200 patients, we demonstrated that the facial carotid system. Therefore, interruption of a
nerve outcome could be improved even after an feeding artery or prolonged vasospasm can
experience of more than 3,000 cases [194]. The cause varying degree of facial nerve palsy.
improved outcome allowed us to conclude that an Topical application of nimodipine or papaver-
ultimate plateau of learning is never reached. ine has been recommended, but their efficacy
For the achievement of optimal results, the is not definitely proven.
experience of the whole team involved in the • Thermal injury due to bipolar coagulation
patients’ management is critical. In a profound close to the nerve or due to the heat generated
meta-analysis, Hastan et al. [93] showed that a while drilling.
linear relation between hospital volume and pres-
ervation ratio of facial function exists. The Prevention of Facial Nerve Palsy
authors concluded that concentration of surgical Prevention of facial nerve injury is based on the
treatment might be a consideration to optimize knowledge of its location, on its early identification
clinical outcome. according to constant anatomical landmarks, and
on the refinement of the microsurgical technique
Operative Approach of dissection and tumor manipulation. Ideally,
During the past three decades, a major topic in neuroimaging should allow to define the position
the literature has been the relation of the opera- of the facial nerve and to plan the surgery accord-
tive approach to the outcome. Numerous authors ingly. The anatomical imaging, however, does
have claimed that their operative technique is not provide reliable information on the facial
254 5 Vestibular Schwannomas

nerve course. DTI and fiber tracking is a rapidly surgery, and at least 1 year after surgery, there
evolving field that allows for visualization of the were no patients with complete facial palsy.
nerve. Its reliability has been proven, but the clin-
ical value of this information remains to be estab- Delayed Facial Nerve Palsy
lished [39, 76, 240]. Postoperative use of steroid Delayed facial nerve palsy is a term used to
medications (dexamethasone) is recommendable describe those cases in which spontaneous wors-
to minimize both cerebellar and perineurial swell- ening of facial nerve function is seen in patients
ing. Electrolyte homeostasis and appropriate with normal or near-normal function early after
intravenous hydration should be achieved. surgery. The reported incidence is highly vari-
able—between 2.3 and 29 %—due to the lack of
Outcome consensus on the precise inclusion criteria, in par-
Anatomical preservation of the facial nerve is ticular the definition of the time range [140, 201].
achieved in more than 90 % of the cases in expe- The pathogenetic mechanisms leading to delayed
rienced hands. The functional outcome correlates facial nerve palsy are poorly understood, and fac-
closely with tumor size, but in small-to-medium tors such as perineural edema, vasospasm, delayed
large tumors, excellent or good functional result immune response, viral reactivation, and nerve
is achieved in more than 80 % [56, 124, 235]. entrapment within its bone canal have been
In a series of 1,000 VSs operated by the senior implied. Respectively, various management
author in the period 1978 to 1993, the facial nerve options have been proposed: treatment with nimo-
anatomical preservation rate was 93 % [199]. The dipine, acyclovir, methylprednisolone, or dexam-
early facial nerve function was normal in 51 % of ethasone, as well as operative decompression of
them. In 45 % the function recovered within 1–12 the nerve in its labyrinthine segment. In the series
months to HB grade II in 13 %, HB grade III in of 200 patients, we observed facial palsy develop-
15 %, HB grade IV in 6 %, HB grade V in 11 %, ing 4 days to 2 weeks postoperatively in seven
and HB grade VI (no recovery) in 4 %, despite patients (3.5 %). A course of dexamethasone
nerve continuity. Of these last patients, 1.7 % treatment was performed in all patients, and the
showed no recovery. In 11 of them, hypoglossal– outcome at 1 year was favorable. The good prog-
facial nerve anastomosis was done and good nosis in case of delayed facial palsy is confirmed
facial reanimation achieved. by the experience of other authors who point that
The anatomical continuity of the nerve was recovery of facial nerve function to its original
lost in 60 cases: in 23 of them the facial nerve postoperative status nearly always occurs.
was reconstructed in the CP angle, in 16 cases
intracranial–intratemporal reconstruction, and in Intermediate Nerve Dysfunction
3 cases intracranial–extracranial transplantation Although this nerve is rarely evaluated after sur-
was performed. The facial nerve was reanimated gery and its disorders are classified as “minor,”
by a donor nerve in 18 cases (the contralateral its deficit can cause significant discomfort to the
facial nerve or the hypoglossal nerve). patient, in particular increased or reduced tearing
In a later series (2000–2003) of 200 patients, (dry eye), salivation disturbances, increased nasal
the rate of excellent facial function 2 weeks post- secretions, and taste abnormalities. The reduced
operatively increased to 62 % (vs. 51 % in the frequency of blinking in addition to the decreased
above-mentioned series) [194]. Good function tearing contributes to the severity of the dry eye
(HB grade III) had 14 % of the patients, fair func- syndrome.
tion (HB grade IV) 16 %, poor function (HB Some studies showed that nervus intermedius
grade V) 5 %, and no function (HB grade VI) abnormalities following VS surgery are more com-
3 %. At the last follow-up examination, 81 % of mon than generally anticipated. Stripf et al. [231]
patients had excellent or good facial nerve func- reported that 45 % of their patients experienced
tion (vs. 79 % in the previous series). All patients crocodile tears, 40 % had dry eyes, whereas 44 %
with complete palsy immediately after surgery complained of increased nasal secretion. In the
showed improvement after at least 1 year after series of Magliulo et al. [139], 42.3 % of the cases
Management of Patients with VS 255

Table 5.5 Gardner–Robertson Grade PTA or SRT SDS (%)


scale [73]
1 0–30 And 70–100
2 31–50 And 50–69
3 51–90 And 5–49
4 91 (ML) And 1–4
5 NR And NR
PTA pure-tone average, SRT speech reception threshold, SDS speech discrimina-
tion score, NR no response, ML maximum loss

Table 5.6 AAO-HNS Class Pure-tone threshold Speech discrimination (%)


hearing classification A £30 dB ³70
B ³30 dB, £50 dB ³50
C >50 dB ³50
D Any level <50
Note: classes A, B, and C require both the pure-tone average and speech discrimi-
nation criteria to be met

had crocodile tears and 59.6 % had altered tearing. Vestibular Schwannomas and Hearing
Irving et al. [97] evaluated a series of 224 patients: In 1954 Elliot and McKissock first reported suc-
44 % had postoperative crocodile tears, absence or cessful hearing preservation in 2 of 3 operated
significant reduction in the production of tears was patients with VSs [59]. Hearing is an important
noted in 72 %, and a taste abnormality in 48 % of determinant of quality of life, and its preservation
the patients. The overall rate of recovery of normal should be hence attempted in every patient who
tearing within 7 months was 27 %. In patients with has functional/useful hearing before surgery,
better facial nerve motor function (HB grades regardless of tumor size.
I–III), the tearing recovered in almost 50 %.
Hearing Classification
Taste Disturbance The first hearing classification was proposed by
The incidence of taste disturbance after surgery is Silverstein et al. in 1985 [218]. In 1988, Gardner
surprisingly high: between 29 and 38 % of the and Robertson [73] presented a classification
patients with normal preoperative function had similar to that of Silverstein et al. It was later
such deficit postoperatively [40, 215]. Although named the “Gardner and Robertson classification”
Watanabe et al. [259] reported that the function (Table 5.5). This system gained popularity,
recovered completely in 65 %, the extent of although it has some weaknesses: 30 dB—and
recovery is variable and unpredictable. The risk not the physiological 25 dB—is accepted as
for taste disturbance is higher with the MF upper limit of useful hearing, and the last studied
approach than with the RS approach. The study frequency is 3,000 Hz.
of Stripf et al. [231] also revealed strong tendency The AAO-HNS classification is also com-
to a higher rate of intermediate nerve-related monly applied (Table 5.6). Its drawbacks are that
symptoms following the MF approach. it separates hearing into only four categories and
The time course and degree of facial nerve is not sufficiently refined [110]. Normal and sub-
recovery depend upon the extent of its injury. In normal hearing are not separated [203].
less severe injuries, the function recovers gradually Other popular classifications are those of
within several weeks to months after surgery. In Gardner, Shelton, and House and of the Mayo
case of more severe injuries, it may take more than Clinic. Sanna et al. [203] classified hearing in steps
12 months to recover. Preoperative counseling on of 10 % for SDS and by steps of 20 dB for PTA. At
this particular aspect in all patients undergoing sur- the Tokyo Consensus Meeting, an agreement was
gery for VS is essential to adequately motivate them met that the PTA evaluation should be made at
and to reduce their psychological discomfort. 500, 1,000, 2,000, and 4,000 Hz [110]. Six hearing
256 5 Vestibular Schwannomas

Table 5.7 New Hannover Audiometry Speech discrimination score


hearing classification [197] Class Hearing (PTA) (SDS) (%)
H1 Normal hearing 0–20 dB 95–100 %
H2 Useful hearing 21–40 dB 70–95 % or better
H3 Moderate hearing 41–60 dB 40–65 % or better
H4 Poor hearing 61–80 dB 10–35 % or better
H5 No functional hearing >80 dB 0–5 % or better
PTA pure-tone average: the average of air conduction data at 1,000, 1,500, 2,000,
and 3,000 kHz

classes were defined using a diagram on which the patients had worse preoperative hearing. The
hearing result is located on the cross point of SDS studies of Lapsiwala et al. [127] and Badie et al.
(speech discrimination score) and PTA. [9] showed that a positive correlation exists
Word recognition is a more sensitive parame- between hearing loss and the increased intrac-
ter of communication ability than pure-tone analicular pressure due to tumor expansion.
thresholds. Patients with good word recognition Respectively, only the size of the intrameatal
should benefit significantly from amplification tumor component and not the total tumor size
than those with little word recognition and good were found to correlate to the hearing level.
pure-tone average [79]. Interestingly, a correlation was found between
Very important issue in regard to hearing the pressure elevation and BAEP features, such
preservation is the definition of serviceable (or as I to wave V intervals and wave V latency. The
functional or useful) hearing. This issue has authors speculated that an ischemic injury to the
been variably presented in the literature, pro- auditory apparatus or pressure to the intrameatal
hibiting any meaningful comparison between part of the cochlear nerve is the cause of hearing
different series [70, 86, 110, 182, 201]. The dysfunction. Our data, however, showed that both
“more relaxed” criterion defined useful hearing intrameatal and extrameatal tumor extension
as PTA £ 50 dB and SDS ³ 50 % [203]. influenced the hearing level, suggesting that some
According to the “strict criteria,” useful hearing additional mechanism is involved [75]. Tumor
corresponds to PTA better than 30 dB and SDS growth perpendicular to the pyramid toward the
better than 70 % in the presence of a normal brain stem correlates significantly with hearing
contralateral ear [81]. loss. A possible explanation for these findings
We favor the New Hannover classification might be that the underlying mechanisms are
[197], presented at Table 5.7. If the PTA is 40 dB stretching, rather than compressing, of the
or less and an SDS is 70 % or greater, hearing is cochlear nerve fibers. Caudal tumor growth has
defined as useful, functional, or serviceable. If similar effect on the cochlear nerve [147, 154].
hearing level is worse, balance cannot be achieved The relation of tumor size and hearing level has
between the good ear and the impaired ear, direc- been controversially discussed in the literature.
tional hearing is impossible, and hearing in noisy Although some authors could not find significant
environments becomes very bothersome. Hearing correlation, in our material both the size and vol-
level worse than 90 dB is defined as “functional ume of the VS correlated positively with the
deafness.” hearing level [75].
The most reliable predictive factors for hear-
Factors Related to the Preoperative Hearing ing preservation after surgical removal of VSs are
Level and Predictors of Hearing Preservation tumor size, preoperative hearing levels, and qual-
In a recently published study [75], we evaluated ity of the BAEP. Other features that might be rel-
the factors that correlate with preoperative hear- evant for hearing preservation are tumor location,
ing level. Patients’ sex and age at onset of clinical tumor consistency, presence of transient evoked
symptoms and at surgery were found to be impor- otoacoustic emissions and a short duration of
tant predictive parameters: younger and female hearing loss [149, 152, 174, 206].
Management of Patients with VS 257

The extent of lateral extension of the VS has ing, whatever the tumor size. Adhering to this
been reported to be an independent prognostic philosophy, our rate of anatomically preserving
factor [152]. Our analysis proved that the degree the cochlear nerve increased from 68 % in our
of intrameatal tumor growth and the lateral VSs first 1,000 cases to 75.8 % in a recent series (79
extension toward the fundus correlated with pre- and 84 % in patients with preoperative hearing)
operative hearing loss. The extensive osseous [194]. Preservation of hearing was achieved in
erosion or dilatation of the IAC is another uncer- 46 % of patients in our previous study and
tain predictor of hearing preservation. improved to 51 % in the later series. Furthermore,
The degree of tumor adhesion and surgical if at least some hearing is available before sur-
expertise are intuitively known as major predic- gery, we always attempt to preserve the cochlear
tive factors for hearing preservation but are nerve, whatever the size of the tumor. According
difficult to quantify [29, 154, 194]. With the cur- to some studies [68], microscopic involvement of
rently available neuroimaging techniques, it is still the cochlear nerve is frequent even when the sur-
impossible to determine the degree of adhesion geon sees no gross involvement. The extreme low
between tumor capsule and cranial nerves, hence rate of tumor recurrences in our series even in
the difficulty of tumor dissection. The experience case of cochlear nerve preservation is an obvious
of the surgeon is perhaps the single most impor- argument against such possibility.
tant factor determining the outcome of surgery. Hearing might be lost at any stage of VS
Younger patients, as well as those with tinni- removal. Most frequent causes are destruction of
tus as initial symptom or at surgery, have better the inner ear structures during IAC opening,
postoperative hearing. Obviously the impact of vibration-induced injury to inner ear, traction
VS on the cochlear nerve might be via two differ- injury to the myelin sheath, thermal injury or
ent pathophysiological mechanisms causing direct trauma to the cochlear nerve, and ischemia
either nerve inhibition (hearing loss) or irritation due to vessel occlusion or vasospasm [45].
(tinnitus). Detailed knowledge of the normal and patho-
logical microanatomy of the cochlear nerve and
VS Removal and Hearing Preservation inner ear structures is essential. Similar to the
Some authors suggest that hearing preservation principle of facial nerve preservation, early
surgeries should be undertaken only in carefully identification of the cochlear nerve is important.
selected cases in regard to tumor size and preop- Its constant relations to anatomical landmarks in
erative hearing level: attempt at hearing preser- the IAC and medially, at the brain stem, are uti-
vation is worthwhile only when hearing is lized: in the fundus of the IAC, it occupies the
excellent or when the tumor extends less than anteroinferior quadrant below the transverse plate
20–25 mm outside the IAC. They state that in and anterior to the vertical crest. In the CP angle
larger tumors the chances of hearing preserva- the cochlear nerve is generally located on the
tion are negligible and this consideration should anteroinferior tumor surface. More rarely it is
not influence the approach selection. Further found on the anterior middle tumor portion and at
arguments are that the attempt to preserve hear- the inferior tumor pole [202]. It joins the brain
ing prolongs operative time and may increase stem in the lateral part of the pontomedullary sul-
the risk of injury to the facial nerve and the risk cus, approximately 1–2 mm posterior to the exit
of recurrences. The concept that in VS ³ 2–3 cm site of the facial nerve [182].
the hearing-destructive translabyrinthine The cochlear nerve is extremely sensitive to
approach should be utilized has therefore sup- traction, which may destroy the nerve fibers at
porters [21, 26, 83, 269]. their weakest point at the Obersteiner–Redlich
We, as well as an ever-increasing number of zone. Therefore, excessive retraction of the cere-
surgeons worldwide, believe that cochlear nerve bellum with the self-retaining retractor should be
preservation should be attempted in all VS sur- avoided. Monitoring of the BAEP allows avoid-
geries and hearing preservation should be the ing this complication: whenever the extent of
goal in all patients who have good or useful hear- traction exceeds a certain physiological value,
258 5 Vestibular Schwannomas

the wave V latency increases or its amplitude


decreases. Actually, the retractor should be used
only to support and protect the cerebellar
hemisphere.
The integrity of the labyrinth and vestibular
aqueduct should be preserved, as described ear-
lier (see Chap. 4). During drilling of the IAC,
the local temperature increases significantly.
Continuous irrigation is required to avoid ther-
mal injury to the underlying neural structures.
Mohr et al. [152] showed that the presence of
CSF space between the tumor and the fundus
may enhance the dissection of the tumor and,
consequently, may improve the outcome. Fig. 5.41 MRI of a patient with an intracanalicular VS
Hearing preservation in VSs reaching the fun- on the right side and normal preoperative hearing. See
Fig. 5.42 for details
dus is more difficult because the attempt to
expose the entire IAC can lead to violation of
the labyrinth [86, 242]. Extensive drilling of the to cause isolated or combined component losses
posterior wall of the IAC, which is needed to and subsequent hearing losses:
expose the entire tumor, may increase the risk • Wave I: 86 % risk of deafness existed if it was
of opening the posterior semicircular canal lost permanently, and an 11 % risk existed if
[147]. As shown by Tatagiba et al. [242], the the loss was temporary. Highest risk for its
extent of labyrinthine opening correlates with loss was associated with pulling the tumor–
postoperative hearing preservation. The amount nerve complex down or medially away from
of bone removal should be therefore carefully the cochlea.
planned. • Wave III: 82 % risk of deafness existed if it
The dissection of the cochlear nerve from the was lost permanently, and a 12 % risk existed
tumor capsule is of key importance. It should if it was temporary. The wave is the most sen-
proceed from known to unknown structures and sitive warning sign.
should be performed always in the arachnoid • Wave V: 90 % risk of deafness existed if the
plane (see Chap.“RS Approach” in VSs). The loss was permanent, and a 14 % risk existed if
dissection should be alternated from different it was temporary. Loss of wave V is the sign
directions, and prolonged stretching of the tumor– most closely associated with deafness.
nerve complex in one direction has to be Importantly, earlier warning signs—disap-
avoided. pearance of wave I or III—precede wave V
The BAEP waves amplitude or latency reflect loss. The most frequent actions, during which
the functional integrity of the auditory pathway wave V disappeared, were pulling the tumor–
and are highly sensitive to any manipulation of or nerve complex back or down, nerve dissec-
even near to the auditory structures. This tech- tion, hemostasis, and plugging of the IAC.
nique provides almost real-time feedback infor- As soon as deterioration of BAEP trace
mation every 30–90 s. In a study published in appears—usually increase of latency or decrease
1997, Matthies and Samii [146] investigated the of amplitude of waves III or V—the ongoing sur-
significance of the presence or absence of ABR gical action is stopped. The cause of BAEP trace
waves I, III, and V as functional representatives change or component loss is not always obvious,
of the cochlea, the nucleus cochlearis, and the and its dynamics cannot be predicted: in some
colliculus inferior, respectively. They could show cases it recovers completely, while in others the
that certain microsurgical maneuvers were likely waves disappear completely (Figs. 5.41, 5.42,
Management of Patients with VS 259

a b

c d

Fig. 5.42 Intraoperative images of the patient from (11:12–11:17). (e, f) Initial tumor removal (further
Fig. 5.41. After the tumor has been removed, an unex- decrease of the amplitude and increase of the latency)
plained change in the BAEP trace occurred: decreased (11:18–11:21). (g, h) The tumor has been completely
amplitude and increased latency of wave V. At the end of removed, and the facial and cochlear nerves are preserved
surgery, the BAEP trace recovered completely. The steps (sudden worsening of the BAEP, in particular of the wave
of tumor removal and the corresponding BAEP wave V amplitude and latency) (11:22–11:28). (i, j) Plugging of
characteristics are demonstrated. (a, b) Dura opening the IAV with fat pieces and fibrin glue (slight improve-
(baseline wave amplitude and latency) (11:07–11:11). (c, ment BAEP) (11:28–11:32). (k, l) Dura closure (recovery
d) Drilling of the IAC (slight decrease of the amplitude) of the BAEP trace) (11:34–11:47)
260 5 Vestibular Schwannomas

e f

g h

Fig. 5.42 (continued)


Management of Patients with VS 261

i j

k l

Fig. 5.42 (continued)


262 5 Vestibular Schwannomas

wards are modified. Calcium channel blockers


(nimodipine) can be applied locally if BAEP
deterioration persists.
One highly sensitive technique is the use of
direct brain stem recording of the auditory evoked
potentials. The potentials are recorded by placing
a retractor with electrodes attached to its tip at the
cerebellomedullary junction [144]. Any move-
ment of the retractor, however, causes artifacts,
and monitoring throughout the procedure may
prove difficult.

Outcome
Hearing preservation rates reported in the liter-
ature vary from 17 to 71 % [79, 119, 189, 232,
245, 264]. This wide range of variation reflects
the use of different criteria for patient selection
and outcome evaluation, as well as the individ-
ual/institutional surgical expertise. The out-
come correlates with tumor size: hearing is
most likely to be preserved in VSs < 15 mm [79,
152]. The rate of hearing preservation in the
series of Yates [269] decreased from 72 % in
intracanalicular tumors to 34 % in tumors with
extension above 1 cm in the CP angle. Wiet
Fig. 5.43 Preoperative MRI: intracanalicular VS that
extends up to the fundus area of the IAC. See Fig. 5.44 for et al. [264] described a tendency toward wors-
details ening hearing outcome for VSs beyond 1.1 cm:
useful hearing rate dropped from 57 % for intra-
canalicular tumors to 48 % for all tumors with
5.43, 5.44, 5.45, 5.46, and 5.47). In case of quick <1.0 cm extension into the CP angle and to only
recovery, the surgery is continued. If the change 17 % in case of tumors with <1.6 cm
persists, a short break is made and the retractor is extension.
loosened or removed completely. The site and The two hearing-preserving operative
type of surgical dissection or manipulation after- approaches are the MF and the RS. If one

Fig. 5.44 Loss of the BAEP response, occurring without amplitude decreased severely and the latency increased
any obvious cause after the tumor removal. The intraop- (11:55–11:59). Therefore, assuming that a compression-
erative images and the corresponding BAEP wave charac- induced cochlear nerve dysfunction occurred, the fat was
teristics are presented. (a, b) The dura has been incised, removed (one such piece is still visible). (i, j) Further
CSF has been evacuated, and a retractor has been posi- deterioration of the BAEP trace. A Gelfoam soaked with
tioned—baseline BAEP trace (11:24–11:30). (c, d) Drill- nimodipine was placed over the cochlear nerve (assumed
ing of the posterior wall of the IAC—sudden worsening of vasospasm) (11:59–12:04). (k, l) Despite the removal of
the BAEP trace (11:30–11:37). (e, f) Complete tumor the fat pieces and application of a nimodipine, no recovery
removal—improvement of the amplitude of wave V and was seen. The IAC was plugged again—almost complete
of the latency (11:44–11:49). (g, h) During the plugging loss of all BAEP components (12:04–12:09). (m, n) Dural
of the IAC with fat pieces, the BAEP trace worsened—the closure—complete loss of the BAEP (12:08–12:20)
Management of Patients with VS 263

a b

c d
264 5 Vestibular Schwannomas

e
f

g h

Fig. 5.44 (continued)


Management of Patients with VS 265

i j

k l

Fig. 5.44 (continued)


266 5 Vestibular Schwannomas

m n

Fig. 5.44 (continued)

compares only tumors of equal size, the hearing Long-Term Outcome


preservation rates in experienced hands are simi- Durability of preserved postoperative hearing is
lar, whatever the approach. The experience of the an important issue, more often discussed in the
surgeon rather than the advantages of a particular neurootological literature. The exact incidence
technique determines the outcome. In large and pathophysiological mechanisms of late
tumors, however, the RS approach is the only hearing loss have to be yet determined. Fibrosis
hearing-preserving option. and scarring in the IAC, which gradually involve
The hearing preservation rate in our general the cochlear nerve, has been suggested as
series is above 51 %. The best overall preserva- provoking factor [252]. Histopathologic studies
tion rates were found in VSs with slight exten- of the temporal bone have demonstrated new
sion in the CP angle (grade T2), 56 %, followed bone formation within the cochlea. Progressive
by pure intracanalicular VSs (grade T1), 51 %, vascular changes have also been implicated
and VSs filling the CP cistern (grade T3), 44 %. [149]. The important conclusion from these
These rates increased in the recent 200 cases to studies is that hearing loss late after surgical
72, 60, and 56 % in VSs grade 2, grade 1, and removal of VSs does not necessarily mean
grade 3a, respectively. Normal preoperative hear- tumor recurrence.
ing provided the highest preservation scores
(£88 %), whereas good or moderate hearing pro- Hearing Preservation in Large VSs
vided a chance of preservation of one-third to Hearing preservation in patients with large VS
more than two-thirds. is very difficult to achieve but is possible, pro-
Management of Patients with VS 267

a b

Fig. 5.45 T2 vestibular schwannoma on the right side. (a, b) T1-weighted sequences after contrast administration. (c)
T2-weighted sequence. See Fig. 5.46 for details

vided the patient has good preoperative hearing 21.4 % of the patients with VSs 3 cm or larger:
level and well-preserved BAEP. Increasing in those with normal preoperative hearing
number of surgeons report on successful hear- (Gardner–Robertson class I hearing preopera-
ing preservation in such patients [52, 61, 174, tively), the rate was 30.8 %. In our experience,
178, 258]. Wanibuchi et al. [258] reported in tumors extending to and compressing the
recently hearing preservation rate of 54 % of the brain stem (grade T4a), the rate of hearing pres-
patients who had tumors larger than 20 mm. Di ervation (hearing classes I–III) after surgery was
Maio et al. [52] achieved hearing preservation in high as 29 %, while in VSs grade T4b, the rate
268 5 Vestibular Schwannomas

a b

Fig. 5.46 Intraoperative images of the steps of tumor (f) The IAC is opened up to the fundus, as proven by the
removal and the corresponding changes of the BAEP palpation with the angled microhook. (g) Further worsen-
trace. (a, b) Exposure of the CP angle (Tu tumor, 7 facial ing of the BAEP. (h) The dura of the IAC has been excised.
nerve, 8 vestibular nerve, P posterior pyramid) and inci- (i) Tumor removal. (j) Removal of the last tumor part in
sion of the dura around the posterior lip of the IAC (arrow- the fundus. (k) Complete tumor removal. (l) The BAEP is
heads). (c) Baseline BAEP trace. (d) Drilling of the discernible but worse than initially
posterior wall of the IAC. (e) Worsening of the BAEP.
Management of Patients with VS 269

h g

Fig. 5.46 (continued)


270 5 Vestibular Schwannomas

i j

Fig. 5.46 (continued)


Management of Patients with VS 271

a b

Fig. 5.47 Postoperative images of the patient from the previous figure

was 27 % [194] (Fig. 5.48). Hearing preserva- Hydrocephalus Associated with VSs
tion is possible even in patients with giant VSs. Patients with VSs are prone to develop hydro-
In a recent study [196], we found that if the cephalus, either as a consequence of tumor-
patient has useful hearing before surgery, the induced obstruction at the level of fourth
rate of hearing preservation (hearing classes ventricle and its outlets or due to malabsorption
I–III) was 33 %; useful hearing had 11 % of the of CSF. The incidence of hydrocephalus varies
patients in the series. strongly in published series—from 4 to
272 5 Vestibular Schwannomas

a b

d
c

Fig. 5.48 Large VS, operated by the senior author in the early 1990s. (a) Preoperative MR image. (b, c) Tumor expo-
sure and complete removal. (d) Normal postoperative facial nerve function. (e) Preserved functional hearing
Management of Patients with VS 273

42 %—reflecting the variable tumor size and cri- proportionally small fourth ventricle in relation
teria for hydrocephalus [78, 239, 255]. The to the lateral and third ventricles in patients with
radiological criteria for hydrocephalus that we large VSs [185]. Complete VS removal does not
apply are those described by Pirouzmand et al. always lead to resolution of the hydrocephalus,
[172]: the ratio of the maximal width of the fron- indicating that at least in some of these patients, a
tal horns to the maximal diameter of the inner malabsorptive component coexists. Hence, the
table of the cranium at the same level. If the ratio actual incidence of such “mixed hydrocephalus”
is 30–39 %, the hydrocephalus is assessed as is still not known.
mild, if the ratio is 40–45 % the hydrocephalus is The clinical manifestation of hydrocephalus is
moderate, and if it is >46 % it is severe. The dif- related to its severity and rate of development:
ferentiation of VS-associated hydrocephalus either NPH-like symptoms or symptoms/signs of
from ventricular enlargement due to other causes intracranial hypertension.
should be based on additional criteria: progres-
sion in the clinical symptoms that are not related Management of Patients with Hydrocephalus
to the tumor or progressive ventricular enlarge- The VS-associated hydrocephalus is potentially
ment [172]. reversible condition, and complete tumor
Communicating or malabsorptive hydroceph- removal frequently leads to resolution of the
alus is the more frequent type (61–87.2 % of all symptoms. This strategy allows avoiding an
patients with hydrocephalus) (Figs. 5.49 and additional invasive procedure, such as EVD or
5.50). Its precise pathogenesis is still unclear, and VP shunt placement, and shunt dependence in
factors such as protein sloughing from the tumor most patients. In patients presenting with severe
secondary to increased vascular endothelial per- hydrocephalus-related symptoms, however, the
meability, development of meningeal adhesions, hydrocephalus should be treated before tumor
arachnoiditis, or seeding of tumor cells are surgery by placing an external ventricular
believed to play an important role [145]. drainage (EVD). When the intracranial pres-
Obstructive hydrocephalus is caused by obstruc- sure is controlled, the VS can be safely removed.
tion of the CSF flow at the level of the fourth ven- In case the tumor surgery has to be postponed
tricle, its outlets, or basal cisterns (Fig. 5.51). for some reason, a VP shunt has to be inserted
Characteristic radiological findings are the dis- (see Case Illustration 5.15).
274 5 Vestibular Schwannomas

a b

c d

Fig. 5.49 MR study of a patient with severe VS-associated communicating hydrocephalus. Note the unobstructed
fourth ventricle, indicating that the cause of hydrocephalus is CSF malabsorption
Management of Patients with VS 275

Fig. 5.49 (continued) b

Fig. 5.50 Communicating hydrocephalus. (a) Patient


with VS grade T2. (b) Patient with VS grade T4a (b)
276 5 Vestibular Schwannomas

a b

Fig. 5.51 Obstructive hydrocephalus due to occlusion of the fourth ventricle (arrow)
Management of Patients with VS 277

Case Illustration 5.15 required 2 weeks of intensive therapy. Once


Large VS with Associated Hydrocephalus the acute infection was healed and the infec-
This 69-year-old patient presented with a his- tious parameters were normalized, a VP shunt
tory of progressive hearing loss and gait insta- was inserted and the patient was discharged.
bility for many years. During the last few The patient was admitted again in 3 months,
weeks, the gait worsened very rapidly and and the VS was removed completely via the
strong headache with nausea appeared. retrosigmoid approach (i–m). Although the
Neurological examination revealed profound facial nerve was preserved anatomically, early
left-sided hearing loss, decreased corneal after surgery the patient had severe facial palsy
reflex on the left side, and severe gait ataxia. (HB grade V). She had furthermore swallow-
The MRI demonstrated large VS on the left ing difficulty, which was interpreted as a
side (grade T4b) and communicating hydro- sequence of the manipulation of the lower cra-
cephalus (a–f). As an incidental finding, nial nerves during tumor removal (note the
inflammatory changes involving the mastoid extent of caudal tumor extension on figure f).
process and middle ear on the side of the During her hospital stay, however, peroral feed-
tumor were found. Considering the potential ing could be successfully initiated. The facial
risk related to the retrosigmoid craniotomy in nerve function improved rapidly, and at dis-
such circumstances, a decision was taken to charge she was able to close the eye completely.
perform the tumor removal only after healing At follow-up 12 months after the surgery, she
the mastoid infection. An EVD was inserted had normal gait and balance (she could ski
to control the intracranial hypertension, and again), very good facial nerve function (HB
the mastoiditis was treated surgically (g, h). grade II), and normal swallowing. The MR
The recovery period was protracted and study at follow-up is presented on figures n–q.
278 5 Vestibular Schwannomas

a b

c d

Preoperative CT (a, b) and MRI (c–f) of the patient been opened, and the facial nerve is identified. (k, l)
demonstrating the VS and the hydrocephalus. The Dissection of the extracanalicular part of the facial
inflammatory changes of the mastoid can be well rec- nerve by slightly pulling the tumor in lateral (k) or
ognized on both CT and MRI sequences. (a, b) Bone- superior direction (l). (m) Complete tumor removal
window CT. (c) T1-weighted MR sequence after and preservation of the integrity of the facial nerve (Tu
gadolinium administration. (d) CISS sequence. (e) tumor, 5 trigeminal nerve, 7 facial nerve, BS brain
T2-weighted sequence. (f) Coronal slice demonstrat- stem). Postoperative images of the patient demonstrat-
ing the tumor and the dilatation of the ventricular sys- ing the completeness of tumor removal. (n, o)
tem. (g, h) CT scans after the operative treatment of T1-weighted sequence after gadolinium administra-
the suppurative mastoiditis. Intraoperative images tion. Note the artifact caused by the valve of the VP
illustrating the steps of tumor removal. (i) Initial over- shunt. (p) CISS MR sequence. (q) CT scan showing
view of the CP angle and of the VS. (j) The IAC had the normal size of the ventricular system
Management of Patients with VS 279

e f

g h
h
280 5 Vestibular Schwannomas

i j

k l

m
Management of Patients with VS 281

n o

p q
282 5 Vestibular Schwannomas

Following tumor removal the patients should The postoperative follow-up of the patients
be observed closely, both radiologically and clini- should be mainly clinical because the ventriculo-
cally. If clinical deterioration occurs and the CT megaly may persist for months. As noted by
examination shows a persisting or increasing Pyrasmound et al. [172], the slow resolution of
widening of the ventricles, an EVD is inserted. the ventriculomegaly could be related to a mild
Initially the EVD, regardless if it is placed before derangement in the CSF circulation or absorp-
or after surgery, is positioned 10–12 cm H2O tion or to a change in the elastance of the brain
above the level of the external auditory canal; tissue.
once the patients’ condition is stabilized, a pro-
cess of gradual “weaning from the EVD” is initi-
ated. If no symptoms/signs of intracranial Radiosurgical Management of VS
hypertension appear, the level of the EVD is ele-
vated progressively. In case the patient becomes In 1969 Lars Leksell was the first to treat a patient
drowsy or other symptoms/signs occur, the level with VS with gamma knife radiosurgery [129,
is decreased and more CSF is allowed to drain for 175]. During the following decades, the radiation
several days. The whole period of weaning may techniques and protocols continued to evolve,
last up to 3 weeks. The drain is ultimately closed and the procedure was established as one of the
for 24 h, and if the patient tolerates it well and the main VS treatment modalities. Increasing num-
intracranial pressure does not exceed 25 cm H2O ber of patients are currently being treated with
for more than 5 min, a CT is performed and the radiation therapy—most frequently with radio-
EVD is removed. A VP shunt is inserted only in surgery (gamma knife or cyber knife) or with
case the “weaning” from the EVD fails. In case fractionated radiotherapy [7, 175]. The reported
hydrocephalus develops in a patient after surgery, tumor growth control rate in recent series is up to
it was managed similarly with a temporary EVD. 91–98 % [16, 92, 117, 118, 132, 181, 192, 267].
Recently we evaluated the management of 53 Shrinkage of the tumor is observed in 21–75 % of
patients with VS-associated hydrocephalus. In the cases, and tumor growth control (or absence
48/53 patients, the tumor was removed initially, of progressive growth) is achieved in 18–59 %.
and 42 of them (87.5 %) did not need additional The facial nerve is preserved in more than 80 %
hydrocephalus treatment. Only 3/53 patients of the patients, and the probability of preserving
required ultimately a VP shunt. Contrary to the useful hearing ranges from 33 to 74 %.
opinion of many authors [172, 239], we found These reported good results; the relatively
that the general and functional outcome in noninvasiveness and the cost-effectiveness of the
patients with primary VS removal did not corre- procedure promoted its wide spreading. According
late to the existence of hydrocephalus. Moreover, to a recent survey by the Acoustic Neuroma
the surgery was not more complex or risky. Association (2007–2008), 20 % of the patients
Important prerequisites for safe surgery are the reported treatment by radiation compared with
sufficient and adequate anesthesia and relaxation 5 % of patients in the 1998 survey [15].
of the patient, operating the patient in the semi- On the other hand, radiation therapy is not
sitting position, and drainage of sufficient CSF without hazards, in particular of cranial neuropa-
from the lateral cerebellomedullary cistern. thy, balance disturbances, headache, and hydro-
Extremely important is the prevention of brain cephalus. The reported rate of permanent facial
edema during surgery, which is frequently caused neuropathy ranges from 4 to 19 %, and the
by an excessive cerebellar retraction or interrup- trigeminal neuropathy rate is from 11 to 16 %.
tion of the venous drainage. We avoid any unnec- Notably, radiation therapy is not curative: its goal
essary opening of the basal cisterns in order to is to achieve tumor growth control. Treatment
prevent the wide distribution of blood, bone dust, failure or secondary regrowth after radiosurgery
and tumor cells, thereby reducing the risk of that necessitates additional treatment is observed
obstruction of CSF flow or its resorption. in 2–9 % of the patients [7, 16, 42, 156].
Management of Patients with VS 283

We believe that complete tumor removal is the The probable explanation for the unrespon-
mainstay of VS management: it cures the patient siveness of some VS to radiotherapy may be
and in experienced hands is related to very low related to their inherently low proliferation index
risk of morbidity. Radiosurgery is an option in and the radioresistance of the tumor bulk [131,
those patients that cannot be operated in general 270]. Treatment failure after radiosurgery/radio-
anesthesia for some somatic problem and those therapy is usually defined as progressive tumor
that are unwilling to have surgery. Chronological enlargement demonstrated on at least 2 imaging
age per se is not a contraindication for surgery. studies [173]. Patients with failed radiosurgery
Surgery of VSs, however, is very demanding and are managed either with repeated radiosurgery or
follows a steep learning curve. An experience of surgically [69, 74, 173]. Surgery is the only rea-
at least 200–300 cases is required to achieve an sonable option in case of large VSs that compress
acceptable proficiency level. Furthermore, opti- the brain stem and in patients with progressive
mal results are achieved in centers of excellence, neurological deterioration or major neurological
in which at least 50–100 patients with VS are symptoms. Tumor enlargement or development
operated annually. Contrary to that in case of of new neurological symptoms per se, however,
radiosurgery, high standards may be achieved is not an indication for immediate surgery. During
even with less experience, provided that the rec- the first year after radiosurgery, VS frequently
ommended protocols are followed. Therefore, we increases in tumor volume and then shrinks spon-
believe that in neurosurgical settings with lower taneously [175]. Such tumor enlargement may
case volume, radiosurgery should be the first line cause transient neurological worsening and
of VS management. should be differentiated from the real treatment
Radiation therapy can be safely applied only failure. New neurological deficits do not neces-
in small-to-medium-sized VSs. Recent popular sarily indicate tumor activity: they may be a
trend in the management of large VSs is to per- delayed sequence of radiation, and a trial with
form initially tumor debulking and later on radio- steroids should be undertaken. Therefore, surgery
surgery of the tumor remnant [71, 98]. The in the early phase after radiosurgery is indicated
cumulative effect of these two different types of very rarely.
tissue injury, however, has still not been ade- The goal of surgery should be total tumor
quately studied. The ultimate outcome has not removal, as it is the only option that cures the
been systematically evaluated, and data from patient [74]. Some authors advocate tumor deb-
larger series with sufficient follow-up period have ulking or internal decompression as the preferred
not yet been published. In our experience surger- management strategy for patients after radiosur-
ies in patients that have undergone radiosurgery gical failure in order to decrease the complication
after prior incomplete operative tumor removal and morbidity rates and to improve the chances
are most challenging and have worse outcome. of preservation of cranial nerves function [128,
The detailed presentation and evaluation of 173, 216]. Subtotal removal may indeed lead to
this management option is beyond the scope of better functional outcome in some cases, but it is
this book. We would refer to the multiple recent a short-term solution. It is difficult to assume that
publications on the topic. a remnant of VS that expanded despite the previ-
ous therapy will change its biological behavior
Surgical Removal of VS After Failed and stop growing. Furthermore, even subtotal
Radiosurgery tumor removal does not guarantee high rates of
The focus of this chapter will be the surgical functional preservation if performed from less
management of VSs after failed previous radia- experienced surgeons.
tion therapy. Considering the rising number of Surgery after previous radiosurgery is indi-
patients that undergo radiosurgery/radiotherapy, cated in patients with large tumors compressing
it is obvious that this issue is becoming especially the brain stem, in case of sustained tumor growth,
important. which is documented on serial MR examina-
284 5 Vestibular Schwannomas

tions, or in case of progression of symptoms, postradiational changes, causing tight adherence


such as significant cerebellar ataxia, pyramidal of the tumor capsule or lack of clear arachnoid
tract signs, or intracranial hypertension, that plane, increased vulnerability of the facial nerve,
could be definitely related to tumor enlargement and radiation-induced vasculopathy with the
and do not respond to steroid therapy. In a related tendency for delayed rupture and bleed-
recently evaluated series of 28 patients with VS ing [209, 220]. Our surgical concept and opera-
operated after failed previous radiosurgery by tive technique in such cases do not differ from
the senior author, tumor enlargement was docu- those in non-treated VSs of the same size (see
mented on serial MR examinations in 27 patients Case Illustrations 5.16 and 5.17). Important is the
[74]. Thirteen patients presented with progres- sufficient initial CSF drainage to prevent addi-
sive neurological symptoms: intracranial hyper- tional retraction injury to the cerebellum. During
tension in 5 patients, gait imbalance in 7 patients, surgery especially important is the delicate dis-
and contralateral hemiparesis in 1 patient. The section in the arachnoid plane. Although it might
indications for surgery were sustained tumor be difficult to identify it, such a plane usually
enlargement and progression of neurological exists. The 2-hand technique for tumor dissection
symptoms in 12 patients and sustained tumor is especially helpful in this regard. Every effort
enlargement in 15 patients. One patient was should be made to preserve even the small drain-
operated despite the absence of progressive ing veins. The blood vessels are often more frag-
tumor growth. He had a large VS (maximal ile, and thorough hemostasis at the end of surgery
extrameatal diameter of 4.2 cm) and signs of is mandatory.
intracranial hypertension with papilledema. The analysis of our results showed that total
Surgery in the early stage after radiosurgery was removal of VSs after failed previous radiosur-
performed only in one patient. He presented 6 gery/radiotherapy can be achieved without mor-
months following radiosurgery with rapid neuro- tality and major morbidity [74]. Total tumor
logical deterioration and a 4.7-cm-large VS that removal was achieved in all patients. The post-
severely compressed the brain stem. operative facial nerve function is indeed worse
than in untreated VSs of similar size, and the
Operative Technique risk of new neurological deficits or CSF leak
Microsurgery following previously radiosurgery tended to be higher, but the differences were not
is complicated by the development of extensive significant.
Management of Patients with VS 285

Case Illustration 5.16 ing loss, tinnitus, and slight facial nerve palsy
Surgery Following Previous Radiosurgery (HB grade II) on the left side, as well as pro-
This patient (46-year-old female) presented 4 nounced gait ataxia. She was operated via the
years after cyber knife treatment of her VS retrosigmoid approach, and the tumor was
(a–d). The follow-up MRI studies showed a removed completely (see figures m–x). The
sustained tumor growth tendency, as well as postoperative period was unremarkable, and the
progressive dilatation of the ventricular system patient was discharged 2 weeks after surgery.
(e–l). She complained of gradually worsening Her facial nerve function early after surgery
gait and headache. At presentation the patient was HB grade IV but improved to grade III at
had diminished corneal reflex, complete hear- discharge (the follow-up is just 3 months).
286 5 Vestibular Schwannomas

a b

c d

MR examination performed 1 year after radiosurgery: (TS transverse sinus, SS sigmoid sinus, P posterior
(a, b) T1-weighted sequence after gadolinium admin- pyramid, Tu tumor). (n) The tumor was covered with
istration. (c) CISS sequence. (d) T2-weighted an unusually firm capsule, which was as thick as the
sequence. MRI performed 36 months after radiosur- dura overlying the pyramid (arrows) and had to be
gery: the growth tendency of the tumor is obvious on excised with microscissors. (o) The IAC had been
all slices: (e, f) T1-weighted sequence after gadolin- widely opened (arrowheads—superior and inferior
ium administration. (g) CISS sequence. (h) walls of the IAC). (p) Partial removal of the intrac-
T2-weighted sequence. The MRI study 46 months analicular tumor part and identification of the facial
after radiosurgery demonstrated further tumor enlarge- nerve (FN). (q) Verification of the facial nerve with
ment, as well as dilatation of the lateral and third ven- electrostimulation (small arrows—petrous vein). (r)
tricles: (i, j) T1-weighted sequence after gadolinium Further removal of the intracanalicular tumor allowed
administration. (k) CISS sequence. (l) T2-weighted to expose the cochlear nerve (CN). (s) The arachnoidea
sequence. A decision for surgery was taken at this surrounding the extracanalicular tumor part (asterisks)
point. The patient was operated via the retrosigmoid was very tightly attached to neighboring structures and
approach. (m) Initial view of the CP angle and the VS required meticulous dissection (small arrows—petrous
Management of Patients with VS 287

e f

g h

vein). (t, u) Once the tumor was debulked sufficiently, the IAC was most adherent and was dissected at the
its capsule could be dissected, and the facial nerve end. Both the cisternal and the intracanalicular seg-
close to the brain stem was exposed. The nerve was ments of the facial nerve (FN) are well visible on the
dissected in the arachnoid plane further laterally; note images. (x) Complete tumor removal; the facial nerve
the slight lateral (t) and superior (u) traction of the was preserved anatomically and showed a good
tumor remnant, which allows to find and to follow the response to electrical stimulation. Through hemostasis
correct plane (the tumor is hold with a tumor forceps). and sealing of the IAC with fat pieces followed
(v, w) The tumor part immediately next to the porus of
288 5 Vestibular Schwannomas

i j

k l
Management of Patients with VS 289

m n

o p
290 5 Vestibular Schwannomas

q r

s t
Management of Patients with VS 291

u v

w x
292 5 Vestibular Schwannomas

Case Illustration 5.17 The patient was therefore reoperated in our


VS Removal Following Previous Surgery institute via the RS approach. The surgery
and Radiosurgery was complicated by the extensive thick scar
Twenty-year-old female patient presenting tissue surrounding the tumor. Interestingly,
with headache, nausea, severe gait ataxia, as the tumor core was stone hard, which was a
well as trigeminal hypesthesia, abducens surprise, considering the MRI findings (typi-
nerve palsy, complete facial nerve palsy, and cal “postradiational” hypointensity on
anacusis on the right side. She had been diag- T1-weighted images). The tumor extended
nosed approximately 2 years earlier of having into the dilated Meckel’s cave—in order to
large VS on the right side. An attempt was remove this part, the petrous apex was drilled
made to remove the VS surgically twice in her off (suprameatal extension of the retrosigmoid
city of origin; however, both surgeries had to approach). Thus, complete tumor removal
be interrupted after prolonged (>14 h) tumor was achieved, and the ventrally dislocated
removal. The VS could be removed only par- abducens nerve was decompressed. Expectedly
tially. Hearing and facial nerve function were the facial nerve was not identified. The early
completely lost. Gamma knife treatment was postoperative CT control is shown on figure f.
performed 5 months later, followed by implan- The patient had an uneventful recovery, and 2
tation of a VP shunt for hydrocephalus. Several weeks later an end-to-side facial-to-hypoglos-
months after the gamma knife treatment, hyp- sal nerve anastomosis was performed (h–j).
esthesia in all three branches of the trigeminal One year later she could close the eyes com-
nerve appeared and could not be influenced by pletely with minimal effort. There was almost
the dexamethasone therapy. The MRI follow- complete symmetry at rest with slight asym-
up examinations showed that the tumor con- metry of the mouth. Her Cushing’s syndrome
tinued to grow steadily (a–e). Her neurological recovered completely (k–n). The MRI is
condition also deteriorated, and she developed shown on figure g.
iatrogenic Cushing’s syndrome.
Management of Patients with VS 293

a b

c d

Preoperative MRI (a–e) and postoperative CT images hypoglossal nerve anastomosis was performed 2
(e) of the patient. Note the hypointense on T1 weeks after tumor removal. (h) Exposure of the facial
sequences tumor core (a–c) and the edema of cerebel- (7) and hypoglossal nerves (12). (i) Exposure of the
lum and ipsilateral temporal lobe (d, e). The CT was proximal segment of the facial nerve in the mastoid
made the day after surgery and shows moderate pneu- (7). (j) Fixation of the facial nerve stump on the par-
mocephalus and hyperdense areas surrounding the tially incised hypoglossal nerve (12) with 2 sutures.
resection cavity (probable blood accumulation). The Photos of the patient before (k, l) and 12 months after
occipital bone had not been reconstructed after the ini- reconstruction of the facial nerve (m, n). Note the ini-
tial surgeries. Despite the watertight dural closure, we tial cushingoid appearance. The patient could close the
did not make methyl methacrylate (Palacos) cranio- eyes completely with minimal effort; there was almost
plasty at this stage in order to prevent CSF leak. complete symmetry at rest with slight asymmetry of
(g) Postoperative MRI study. End-to-side facial-to- the mouth
294 5 Vestibular Schwannomas

g h
Management of Patients with VS 295

i j

k l
296 5 Vestibular Schwannomas

m n
Management of Patients with VS 297

Operative Complications: Avoidance exception (0.3 % in the series of Sanna et al.


and Management [204]). Extreme care is required in case of com-
The risk for surgery-related complications corre- bined lesions to both the trigeminal and facial
lates with certain patient-, tumor- and surgeon- nerves because this may cause major eye trophic
related factors. Although the morbidity is higher problems and corneal ulcerations.
in the elderly, as shown by some studies [161], Large tumors with major inferior extension
we believe that patient’s general physical condi- may cause lower cranial nerve deficits, which
tion—but not his age—is a more important con- usually become even more severe after surgery. If
sideration. Large tumors and NF-2-associated preoperative deficit related to these nerves is
tumors are generally related to higher morbidity absent, however, the risk of new deficits after the
rate. The experience of the surgeon and of the surgery is lower. The lower cranial nerves are
whole team involved in the management of the usually protected by an arachnoid layer and can
patient, along with tumor size, is the key factor be safely dissected. New lower nerve deficit has
determining the outcome. It has been repeatedly been reported to occur in 0.14–4.2 % of the
shown those both functional and general out- patients [198, 204, 234]. In the recent series of
comes reflect the level of the learning curve of the senior author, three patients (1.5 %) had lower
the operator [29, 194, 262]. cranial nerve disturbances in the early postopera-
When evaluating the morbidity rates reported tive period. Two of them had caudal nerve signs
in the literature, an issue of utmost importance is preoperatively [194]. Lower cranial nerve dys-
whether the outcome is correlated with tumor function can cause diminished gag reflex, dys-
size and with the completeness of tumor removal. phagia, hoarseness, and paralysis of vocal cord,
The comparisons of different surgical approaches with a threat of aspiration. Even in the presence
or modes of VS management are of little value if of gag reflex at examination, however, the coordi-
these issues, as well as the surgeons and institu- nation of the swallowing mechanism might be
tions experience with a given approach, are not dysregulated (see Chap. 10 for detailed
taken into account. discussion).
The ocular motor nerves are at risk in case of
Cranial Nerves large tumors with medial extension in the petro-
The main principle for avoiding cranial nerve- clival area. As pointed earlier, the RS approach
related complications is their early identification allows for early identification of the abducens
and avoiding their injury during surgical manipu- nerve, either in the area of Dorello’s canal or at
lations (stay in the arachnoid plane!). Importantly, its brain stem exit site. The trochlear nerve can be
patients at risk for developing such complications identified along the tentorial incisura. The
should be actively managed in the postoperative reported rates of such deficits range between 1.7
period to avoid potential problems. and 7 % [109, 198, 204]. Frequently the patients
Large tumors may cause compression and/or complain of difficulty in focusing the eyes or
dislocation of the trigeminal, trochlear, abducens, diplopia but have no obvious strabismus: such
and lower cranial nerves, which renders them complaints usually resolve in few weeks.
more susceptible to injury during surgical manip-
ulations. The trigeminal nerve is more resistant to Cerebellar Complications
compression and manipulations than the facial or Cerebellar symptoms usually improve after sur-
cochlear nerves. Patients with VS present very gery. Most patients complain of temporary rela-
rarely with trigeminal neuralgia. Removal of the tive mild unsteadiness that clears over several
tumor usually leads to improvement or resolution days to several weeks. In case of severe walking
of the pain. Nevertheless, its dissection from the difficulty, discoordination, and dysarthria the
tumor may lead to numbness in some of its recovery period is, of course, longer [198].
branches. Trigeminal neuralgia after surgical Exacerbation of the gait instability or cerebellar
removal of VS has been also reported but is an dysfunction occurs rarely after VS removal. In
298 5 Vestibular Schwannomas

the previously evaluated series of 1,000 patients and even cause the death of the patient. The inci-
with VS operated by the senior author, the cere- dence of vascular complications in large series
bellum was hemorrhagic or ischemic in seven ranges from 1 to 7 % [25, 83, 198, 204, 234].
patients. Severe permanent ataxia persisted in Hemorrhagic complications are more frequent
two of them, while the ability to walk freely than ischemic. In a recently published study, Sade
recovered in the rest. et al. evaluated a series of patients with VS that
underwent 413 procedures. Vascular complica-
Vestibular Symptoms tions were detected in 11 patients (2.7 %): hem-
Removal of a VS usually destroys completely the orrhage in 8 patients and ischemia in 3 patients.
vestibular function on the side of the lesion. The comparison of the incidence of these compli-
Exceptions are patients with small tumors when cations after the RS approach and the translaby-
one of the vestibular nerves is preserved. rinthine approach showed that it was similar.
Vestibular symptoms, such as disequilibrium, Bleeding in the CP angle is the most common
dizziness, vertigo, and imbalance, are therefore hemorrhagic complication, followed by cerebel-
frequent complaints after VS resection [138, lar bleeding. Occasionally epidural or subdural
228]. The decompensated state after surgery, hematomas occur. The hemorrhage is usually
characterized by strong vertigo, nystagmus, nau- diagnosed early after the patients’ extubation: in
sea, and vomiting, lasts usually for only up to 3 33–40 % they are detected within the first 24 h
days. Vestibular dysfunction after surgery tends [56, 198, 204]. Early recognition and active man-
to be transient, and the vast majority of the agement of such complications are prerequisite
patients recover due to the phenomenon of ves- for avoiding worse outcome. Patients with a pro-
tibular compensation. Some patients, however, longed awakening phase, those who remain
need much longer to recover—up to several drowsy or show progressive deterioration in their
weeks. The complaints are more severe after level of consciousness or worsening neurological
removal of small VS when normal or near-normal deficit, should undergo immediate CT scanning.
vestibular function was present preoperatively Delayed diagnosis, for example, when the patient
[228]. The preservation of a given vestibular is already comatose, can explain the high mor-
nerve neither prevents vestibular disturbances bidity rates after CP angle bleeding. In our expe-
nor does it promote functional recovery. Of para- rience cystic and/or uncapsulated tumors have a
mount importance for the recovery are the physi- higher risk of postoperative hematoma. Patients
cal activity and the systematic vestibular with such tumors, as well as those with large VS
training. or in case of difficult hemostasis, should be fol-
lowed especially carefully. The blood pressure
Tinnitus should be permanently monitored—initially with
Tinnitus is the second most frequent symptom in an intra-arterial sensor—and arterial hyperten-
patients with VSs and may severely affect the sion should be treated aggressively (the hypoten-
quality of life [10, 108]. Removal of the tumor sion may have also a deleterious effect!).
does not, however, influence its occurrence or Patients with a postoperative epidural hema-
characteristics. The evolution of tinnitus follow- toma have a relatively better outcome (Fig. 5.52).
ing surgery cannot be predicted. Furthermore, They are most frequently caused by a bleeding
our experience shows that the cochlear nerve sec- artery in the suboccipital area: the blood collec-
tioning does not have an effect on the tinnitus tion is in the soft tissue and secondarily extends
either. into the epidural space. We have never had hem-
orrhagic complications, such as subdural or
Vascular Complications intracerebral hemorrhages, related to the semi-
The vascular complications are perhaps the most sitting position.
serious morbidity that can occur after surgery. In some cases, small blood collections in the
They may have serious and lasting consequences tumor cavity or CP angle are detected on routine
Management of Patients with VS 299

postoperative CT scans. If they are not causing


mass effect or clinical symptoms, they are man-
aged conservatively and the blood is absorbed
within several days. Still, the possibility of grad-
ual increase of the blood volume should be con-
sidered and regular imaging follow-up should be
performed. Immediate evacuation is indicated if
the hemorrhage has a mass effect and/or the con-
dition of the patient deteriorates. The blood cloths
should be removed and a meticulous hemostasis
should be made. The cranial nerves are at higher
risks in case of large hemorrhages with absent
normal anatomical landmarks.
a Electrophysiological monitoring is advisable,
although not always available in case of emer-
gency. Frequently, the source of bleeding cannot
be identified, and a venous origin may be sus-
pected. The exposed brain surfaces are covered
with a layer of Tabotamp.
Brain stem infarcts occur usually during the
dissection of adherent tumor parts from the brain
stem when perforating vessels are interrupted
(Darrouzet). Sade et al. refer to the description of
Rhoton that recurrent perforating arteries can ini-
tially course toward the IAC but subsequently
turn medially and supply the brain stem.
Occlusion of a large artery is currently a rare
b cause of ischemic complications. Vasospasm is
another proposed but not proven mechanism
leading to infarction.
Although some surgeons state that the petrosal
veins can be sacrificed with relative safety, we
believe that the venous anatomy of the CP angle
is so variable that the consequences of such inter-
ruption cannot be predicted.

Sinus Thrombosis
This is a relatively underestimated complication,
probably due to its more subtle and late manifes-
tation. According to one study, its rate after VS
surgery may reach 4.7 % [112]. The thrombosis
may occur after injury or laceration to the sinus,
c
drying and shrinkage of the sinus if it is overex-
posed and the protective layer of bone is missing,
Fig. 5.52 Patient with an acute postoperative epidural
hematoma in the craniotomy area. (a) Preoperative MRI or after venous stasis due to its prolonged retrac-
study showing the VS. (b, c) Postoperative CT, performed tion (required in the translabyrinthine approach).
because of the prolonged awaking phase. The early detec- The symptoms and speed of clinical deterioration
tion of the hematoma allowed rapid treatment and avoid-
depend on the anatomical structure of a given
ance of any related complications
300 5 Vestibular Schwannomas

patient: dominance and size of the sinus, pres- CSF Leak


ence of collaterals and of a communicating torcu- CSF leak is one of the relatively common compli-
lar herophili [191]. cations after CP angle surgery, whatever approach
Sinus occlusion may have a milder evolution is utilized. The reported incidence in the litera-
with headache and visual deterioration beginning ture varies widely—from 2 to 30 % [24, 64, 134,
days after the surgery. Alternatively, it may cause 150, 184, 198]. Higher rates have been reported
a dramatic clinical picture, especially if concomi- in the older series. In the last decade, however,
tant hemorrhages appear. the improved techniques of dura and wound clo-
sure lead to significant reduction of the CSF leak
Pneumocephalus rate: in large current series, the rate is below
Pneumocephalus occurs relatively frequently in 5 %.
patients operated in the semi-sitting position. CSF leaks appear typically during the first 2
Symptomatic pneumocephalus is, however, rarely weeks after surgery. Delayed leak is rare but pos-
seen. The volume of trapped air tends to correlate sible, and the patients should be instructed
with the duration of surgery, the amount of evac- accordingly. Although some authors reported that
uated CSF, and the degree of brain atrophy of the the most frequent site of CSF leakage is the skin
patient. If pneumocephalus is suspected, a CT [24], in our experience this is exceptional. Key
should be made to assess its space occupying factors are the good operative technique, meticu-
effect. Patients with symptomatic tension pneu- lous attention to dura and wound closure—allow-
mocephalus and radiological evidence of more ing quick wound healing—and avoidance of
severe compression of the ventricles may require infection. Rhinorrhea is the most frequent type of
burr hole trepanation and evacuation of the air CSF leak in our experience. The communication
collection (a detailed discussion on the topic is between the cisternal space and the external
provided in the Chap. “RS Approach”). structures is usually air cells surrounding the
IAC. The extent of pneumatization of the bone
Postoperative Suboccipital Headache correlates with the risk of CSF leak. These air
The issue of postoperative suboccipital headache cells are frequently opened during the meatal
related to the RS approach and its prevention is drilling and should be thoroughly occluded at the
discussed earlier (section “Retrosigmoid end of surgery. As shown by Hoffman, the leak
Approach”). In summary, the technique of skull rate is 16 % after drilling the IAC and 0 % if the
base reconstruction with methyl methacrylate IAC is not drilled. Of course, this is a historical
reduces significantly the risk of such headache, observation, and the need to open the IAC is a
most probably by preventing scar tissue forma- basic principle of VS surgery.
tion between the dura and neck muscles: less than In case of paradoxical rhinorrhea, the CSF
1 % of the patients in our experience have persis- leak pathway is through a dural defect to the epi-
tent headache. Very strong occipital pain with dural space and then to the mastoid cells. If these
possible cranial irradiation, however, may be cells are not sealed adequately, the CSF may
caused by the formation of posttraumatic scar enter the middle ear and the Eustachian tube.
neuroma or scar tissue around the occipital Otorrhea with the RS approach is an exceptional
nerves. In such cases, a trigger point can always finding reported in the literature when the tym-
be found near the incision line. A reliable test panic membrane has been perforated while posi-
with potential therapeutic effect is the infiltration tioning the cochlear electrodes [24].
of the area around the trigger point with local Factors associated with the risk for CSF leak
anesthetics: it usually ameliorates the symptoms. are related to the surgical technique, the clinical
In case the pain subsides but later on recurs, the management, the patient’s anatomy, and the
scar should be widely excised. A more limited tumor features [134, 227].
excision of the scar or of the neuroma may lead to Surgical Technique: The factors discussed in
a recurrence. the literature are suboptimal dural and/or wound
Management of Patients with VS 301

closure, wound infection, inadequate wound 5.6 %) [227], which may reflect the more pro-
healing, increased intracranial pressure, or nounced pneumatization of the mastoid bone in
opened and not occluded air cells in the petrous males. A predisposing factor for CSF leak is the
bone or mastoid. Inadequate dura closure may increased intracranial pressure. In patients with
cause CSF leak through the wound or fluid accu- hydrocephalus, the leak rates are significantly
mulation under the skin (pseudomeningocele). higher [239]. If the CSF leak persists despite
The closure of air cells opened during the sufficient drainage via the lumbar drainage, CT
craniotomy or IAC drilling is a crucial issue. of the head has to be performed to exclude
Various materials have been applied in the attempt hydrocephalus.
to achieve a leakproof occlusion. Bone wax has Tumor Factors: The relation between tumor
been used widely in the past. The experience has size and CSF leak rate is controversially dis-
shown, however, that this material cannot provide cussed in the literature. Large tumor size is con-
reliable sealing. Furthermore, if a large amount sidered by some authors as a factor increasing the
of wax is used, it may cause foreign body reac- leak rate [25, 28], while others did not find any
tions and/or infectious complications. significant correlation [63, 125]. The evaluation
Hydroxyapatite bone cement for the reconstruc- of our series showed that highest CSF leak rate
tion of the petrous bone or of the drilled IAC [11] had patients with small intrameatal schwanno-
is also not recommendable. In a recent study, mas (class T1), which was 9.7 %, while in those
Kerr et al. [114] showed that in 7.3 % of the with large VS (grade T4a), the rate was 2.7 %.
patients, a delayed extrusion of the hydroxyapa- Similar data have been presented by Sanna et al.
tite cement used for petrous reconstruction [204]: the CSF leak rate was higher in patients
occurred. Autologous materials are utilized most with small tumors. Previous radiotherapy is con-
frequently: muscle, fat, bone dust, fascia, and sidered as a factor that significantly increases the
even soft tissue flaps. These materials are plugged risk of CSF leak [55]. In our series we could not
into the defect and fixed with fibrin glue. We con- proof that such correlation exists. Previous sur-
sider the use of fat instead of muscle pieces sealed gery, however, is a negative prognostic factor
with fibrin glue as a major breakthrough in the [55]. We also found that the CSF leak rate in such
prevention of CSF leaks. Muscle tends to shrink patients is 11.1 % compared to a 4.4 % rate in the
over the time and is reorganized to a scar. The control group of patients.
CSF leak rate in our older series, published in Prevention and Management: The operative
1996, was 9.2 %. In a later patient series, oper- technique is the key to CSF leak prevention (dis-
ated between 2001 and 2004, the rate was reduced cussed elsewhere in the text). The patients should
to 5.4 %. Since we started to apply fat in the be closely monitored in the postoperative period.
described above technique in 2004, the rate They have to be instructed to avoid activities
dropped to 2.2 % [134]. Furthermore, almost all leading to increased intracranial pressure, such as
patients with CSF leak are managed blowing the nose or pressing against the closed
conservatively. glottis, during the early postoperative period.
It has been frequently discussed which CSF leaks should be detected early and managed
approach is related to higher risks for CSF leak. adequately in order to prevent potential compli-
Many authors found no significant correlation cations, such as meningitis caused by ascending
between the approach and the leak incidence. We bacteria or tension pneumocephalus.
believe that whatever approach is used, if the dura As pointed by Luedemann et al. [134], the
and wound are closed meticulously, the risk of leak of fluid out of the nose after surgery does not
this complication can be reduced significantly. necessarily mean that it is CSF. The source can
Patient Factors: The extent of petrous bone be the fluid collected and retained in the mastoid
pneumatization correlates with the risk of CSF air cells during surgery or nasal secretion. Nasal
leak. The CSF leak rate is somewhat lower in decongestants can be given to exclude a local
female compared to male patients (3.4 % vs. nasal cause. The patient’s sensation may give a
302 5 Vestibular Schwannomas

clue: CSF has a metallic, salty, or sweet taste. In explanations: besides to the general predisposing
case a leak is suspected, we perform a provoca- factors, meningitis is found to correlate to the
tion test: the patient is asked to lower his head for presence of a CSF leak. Aseptic meningitis usu-
at least 5 min and is observed. If some fluid is ally becomes apparent 2–21 days after surgery
collected, it is tested for glucose or for beta-trace and is treated with tapering dose of steroids.
protein to assess if it is CSF. After albumin, the
beta-trace protein is the protein with highest con- Postoperative Hydrocephalus
centration in the CSF. Bachmann-Harildstad [8] Symptomatic dilatation of the ventricular system
showed recently that both beta2-transferrin and following surgery is a well-known phenomenon
beta-trace protein are reliable immunological that can cause acute deterioration of the patient.
markers for the detection of CSF traces. It has to be detected and treated early enough to
We do not favor an initial period of conserva- prevent irreversible brain damage. Surgery in
tive measures, consisting only of bed rest, head itself may hinder normal CSF dynamics and
elevation, pressure dressings, or frequent lumbar thereby potentiate an existing hydrocephalus or
punctures. If the presence of CSF leak is proven, cause the development of new hydrocephalus.
a lumbar drain is placed for at least 7 days. Obstruction at the level of fourth ventricle and its
Prophylactic broad-spectrum antibiotic is also outlets is provoked most frequently by cerebellar
given, and the CSF infectious parameters are edema or postoperative hemorrhage. Blood prod-
regularly tested. The level of the drainage is gen- ucts in the CSF can affect its absorption and
erally adjusted to produce between 150 and cause chronic hydrocephalus and NPH-like
250 ml in 24 h. If the CSF leak stops, the drain- symptoms.
age system is closed at the seventh day for 24 h Careful operative technique and meticulous
and then the provocation test is repeated. hemostasis are critical for the prevention of this
In our recently analyzed series of 420 patients complication. Cerebellar edema can be caused by
operated during the period 2001–2006, CSF leak excessive cerebellar retraction or interruption of
had 19 patients (4.5 %). The conservative man- venous drainage. In large tumors, causing long-
agement, described above, was successful in all standing neural compression, even the slightest
but 1 of the patients. There were no cases of manipulations can provoke cerebellar edema.
meningitis. The basal cisterns should be opened only as much
Indications for surgical revision are the cases as necessary for the tumor exposure. Redundant
when the leak continues despite the drainage or opening of the cisterns is to be avoided in order to
reappears after its closure. The patient is put prevent wide spread of blood, bone dust, and
supine on the operative table with rotated head. tumor cells or particles within the CSF.
The area around the IAC is examined for opened Active management is necessary in case of a
and non-occluded air cells. Frequently, the fat progressive dilatation of the ventricular system
pieces that have been inserted at the initial sur- that correlates with a prolonged awakening phase,
gery are found to be dislocated. The reason for incomplete awakening, or clinical deterioration
that can be strong CSF pulsations or inadequate of the patient (Fig. 5.53). One should not rely
distribution of the fibrin glue. The fat pieces are simply on the absolute measurements of the ven-
removed; the area is thoroughly inspected and tricular size. It is important always to compare
sealed again. the ventricles with the previous images of the
same patient and determine if a tendency for their
Meningitis enlargement exists. Patients with less severe
The reported rates of meningitis after VS surgery hydrocephalus can be initially followed. General
vary from 0 to >10 % [25, 125, 142, 143, 198]. In measures, such as head elevation and dexametha-
the recent series, it is less than 2 % with a ten- sone, are applied, and the neurological and clini-
dency for further decline. The optimized preven- cal status (blood pressure, heart rate!) are
tion of CSF leaks is one of the possible monitored closely. Radiological control with CT
Management of Patients with VS 303

a b

Fig. 5.53 Fifty-five-year-old female patient with a VS on was performed and showed extensive cerebellar edema
the left side. The MRI study demonstrated a cystic VS with causing obstruction of the fourth ventricle and correspond-
highly irregular surface and moderate compression of the ing hydrocephalus (d). An EVD was inserted immediately.
brain stem (a–c). The surgery was straightforward: the Within 10 days the swelling subsided (e), and the patient
tumor was removed completely, and all neural and vascular could be successfully weaned from the EVD. At discharge
structures were preserved. Several hours after extubation, did not have any new neurological deficits besides an HB
however, the patient became somnolent. CT examination grade II facial nerve function
304 5 Vestibular Schwannomas

is performed frequently, guided by the patient’s 2. Acioly MA, Gharabaghi A, Liebsch M, Carvalho CH,
clinical condition. Aguiar PH, Tatagiba M (2011) Quantitative parame-
ters of facial motor evoked potential during vestibular
In patients with pronounced symptoms, an schwannoma surgery predict postoperative facial
EVD is inserted. It is kept as long as necessary to nerve function. Acta Neurochir (Wien). doi:10.1007/
allow regression of the cerebellar edema and s00701-011-0995-4
recovery of the normal CSF circulation. 3. Allcutt DA, Hoffman HJ, Isla A, Becker LE,
Humphreys RP (1991) Acoustic schwannomas in
Afterwards, an attempt for weaning from the children. Neurosurgery 29(1):14–18
EVD is done, as described in the section on 4. Anderson DE, Leonetti J, Wind JJ, Cribari D, Fahey K
hydrocephalus. A VP shunt is required if the (2005) Resection of large vestibular schwannomas:
weaning fails. In a recently evaluated group of facial nerve preservation in the context of surgical
approach and patient-assessed outcome. J Neurosurg
400 patients, we found five, in whom symptom- 102(4):643–649. doi:10.3171/jns.2005.102.4.0643
atic hydrocephalus after the VS surgery devel- 5. Angeli RD, Piccirillo E, Di Trapani G, Sequino G,
oped. In all cases an EVD was inserted. Weaning Taibah A, Sanna M (2011) Enlarged translabyrinthine
from the EVD was successful in 3 cases; the approach with transapical extension in the manage-
ment of giant vestibular schwannomas: personal expe-
remaining 2 required a VP shunt. rience and review of literature. Otol Neurotol
32(1):125–131. doi:10.1097/MAO.0b013e3181ff7562
Mortality 6. Arriaga MA, Carrier D, Houston GD (1995) False-
During the last decades, the mortality rate after positive magnetic resonance imaging of small internal
auditory canal tumors: a clinical, radiologic, and
VS surgery has been dramatically reduced and pathologic correlation study. Otolaryngol Head Neck
nowadays should be an exception. In the large Surg 113(1):61–70, S0194599895001720 [pii]
series it is between 1 and 2 %. In our earlier expe- 7. Arthurs BJ, Fairbanks RK, Demakas JJ,
rience [198] the mortality rate was 1.1 %. The Lamoreaux WT, Giddings NA, Mackay AR, Cooke
BS, Elaimy AL, Lee CM (2011) A review of treat-
cause of this outcome was myocardial infarction; ment modalities for vestibular schwannoma.
acute pulmonary embolism; sinus thrombosis and Neurosurg Rev 34:265–277. doi:10.1007/s10143-
contralateral media infarction with a large edema, 011-0307-8
secondary to revision surgery for a postoperative 8. Bachmann-Harildstad G (2008) Diagnostic values of
beta-2 transferrin and beta-trace protein as markers
hemorrhage; or aspiration pneumonia due to for cerebrospinal fluid fistula. Rhinology 46(2):
severe caudal cranial nerve palsies. One patient 82–85
was reoperated for a postoperative CP angle hem- 9. Badie B, Pyle GM, Nguyen PH, Hadar EJ (2001)
orrhage but did not recover. The patients at risk Elevation of internal auditory canal pressure by ves-
tibular schwannomas. Otol Neurotol 22(5):696–700
are premorbid patients with previous surgery or 10. Baguley DM, Humphriss RL, Axon PR, Moffat DA
with severe brain stem compression, patients with (2006) The clinical characteristics of tinnitus in
very large VSs and complete caudal nerve palsy patients with vestibular schwannoma. Skull Base
after surgery, and patients prone to have postop- 16(2):49–58. doi:10.1055/s-2005-926216
11. Baird CJ, Hdeib A, Suk I, Francis HW, Holliday MJ,
erative hemorrhage (cystic or nonencapsulated Tamargo RJ, Brem H, Long DM (2007) Reduction
tumors) [198]. In premorbid patients with very of cerebrospinal fluid rhinorrhea after vestibular
large tumors causing significant brain stem com- schwannoma surgery by reconstruction of the drilled
pression, the option of performing only internal porus acusticus with hydroxyapatite bone cement.
J Neurosurg 107(2):347–351. doi:10.3171/JNS-
decompression should be considered. In the last 07/08/0347
10 years, the mortality rate was 0 %. 12. Bakkouri WE, Kania RE, Guichard JP, Lot G, Herman
P, Huy PT (2009) Conservative management of 386
cases of unilateral vestibular schwannoma: tumor
growth and consequences for treatment. J Neurosurg
References 110(4):662–669. doi:10.3171/2007.5.16836
13. Barker GF, Ojemann RG (1993) Surgical approaches
1. Abe T, Izumiyama H, Imaizumi Y, Kobayashi S, to tumors of the posterior fossa cranial nerves (exclud-
Shimazu M, Sasaki K, Matsumoto K, Kushima M ing acoustic neuromas). In: Barrow DL (ed) Surgery
(2001) Staged resection of large hypervascular ves- of the cranial nerves of the posterior fossa. American
tibular schwannomas in young adults. Skull Base Association of Neurological Surgeons, Park Ridge, pp
11(3):199–206 289–306
References 305

14. Baser ME, Makariou EV, Parry DM (2002) Predictors Dumanski JP (1999) A group of schwannomas with
of vestibular schwannoma growth in patients with interstitial deletions on 22q located outside the NF2
neurofibromatosis Type 2. J Neurosurg 96(2):217– locus shows no detectable mutations in the NF2 gene.
222. doi:10.3171/jns.2002.96.2.0217 Hum Genet 104(5):418–424
15. Bassim MK, Berliner KI, Fisher LM, Brackmann DE, 28. Bryce GE, Nedzelski JM, Rowed DW, Rappaport JM
Friedman RA (2010) Radiation therapy for the treat- (1991) Cerebrospinal fluid leaks and meningitis in
ment of vestibular schwannoma: a critical evaluation acoustic neuroma surgery. Otolaryngol Head Neck
of the state of the literature. Otol Neurotol 31(4):567– Surg 104(1):81–87
573. doi:10.1097/MAO.0b013e3181d8d3ad 29. Buchman CA, Chen DA, Flannagan P, Wilberger JE,
16. Battista RA (2009) Gamma knife radiosurgery Maroon JC (1996) The learning curve for acoustic
for vestibular schwannoma. Otolaryngol Clin North tumor surgery. Laryngoscope 106(11):1406–1411
Am 42(4):635–654. doi:10.1016/j.otc.2009.04.009, 30. Cafer S, Bayramoglu I, Uzum N, Yilmaz M, Memis
S0030-6665(09)00064-4 [pii] L, Uygur K (2008) Expression and clinical significance
17. Bederson JB, von Ammon K, Wichmann WW, of Ki-67, oestrogen and progesterone receptors in
Yasargil MG (1991) Conservative treatment of acoustic neuroma. J Laryngol Otol 122(2):125–127.
patients with acoustic tumors. Neurosurgery 28(5): doi:10.1017/S0022215107000229,
646–650; discussion 650–651 S0022215107000229 [pii]
18. Bloch DC, Oghalai JS, Jackler RK, Osofsky M, 31. Caye-Thomasen P, Borup R, Stangerup SE, Thomsen
Pitts LH (2004) The fate of the tumor remnant after J, Nielsen FC (2009) Deregulated genes in sporadic
less-than-complete acoustic neuroma resection. vestibular schwannomas. Otol Neurotol 31(2):256–
Otolaryngol Head Neck Surg 130(1):104–112, 266. doi:10.1097/MAO.0b013e3181be6478
S0194599803015985 [pii] 32. Chandler CL, Ramsden RT (1993) Acoustic schwan-
19. Bonneville F, Savatovsky J, Chiras J (2007) Imaging noma. Br J Hosp Med 49(5):335–343
of cerebellopontine angle lesions: an update. Part 2: 33. Charabi S, Mantoni M, Tos M, Thomsen J (1994)
intra-axial lesions, skull base lesions that may invade Cystic vestibular schwannomas: neuroimaging and
the CPA region, and non-enhancing extra-axial growth rate. J Laryngol Otol 108(5):375–379
lesions. Eur Radiol 17(11):2908–2920. doi:10.1007/ 34. Charabi S, Thomsen J, Mantoni M, Charabi B,
s00330-007-0680-4 Jorgensen B, Borgesen SE, Gyldensted C, Tos M
20. Brackmann DE, Bartels LJ (1980) Rare tumors of the (1995) Acoustic neuroma (vestibular schwannoma):
cerebellopontine angle. Otolaryngol Head Neck Surg growth and surgical and nonsurgical consequences of
88(5):555–559 the wait-and-see policy. Otolaryngol Head Neck Surg
21. Brackmann DE, Green JD (1992) Translabyrinthine 113(1):5–14, S0194-5998(95)70138-9 [pii]
approach for acoustic tumor removal. Otolaryngol 35. Charabi S, Tos M, Borgesen SE, Thomsen J (1994)
Clin North Am 25(2):311–329 Cystic acoustic neuromas. Results of translabyrin-
22. Brackmann DE, Green JD Jr (2008) Translabyrinthine thine surgery. Arch Otolaryngol Head Neck Surg
approach for acoustic tumor removal 1992. Neurosurg 120(12):1333–1338
Clin N Am 19(2):251–264. doi:10.1016/j.nec.2008. 36. Charabi S, Tos M, Thomsen J, Charabi B, Mantoni M
02.007, S1042-3680(08)00008-9 [pii], vi (2000) Vestibular schwannoma growth – long-term
23. Brackmann DE, Owens RM, Friedman RA, results. Acta Otolaryngol Suppl 543:7–10
Hitselberger WE, De la Cruz A, House JW, Nelson 37. Charabi S, Tos M, Thomsen J, Charabi B, Mantoni M
RA, Luxford WM, Slattery WH 3rd, Fayad JN (2000) (2000) Vestibular schwannoma growth: the continu-
Prognostic factors for hearing preservation in vestibu- ing controversy. Laryngoscope 110(10 Pt 1):
lar schwannoma surgery. Am J Otol 21(3):417–424 1720–1725. doi:10.1097/00005537-200010000-
24. Brennan JW, Rowed DW, Nedzelski JM, Chen JM 00030
(2001) Cerebrospinal fluid leak after acoustic neu- 38. Charabi S, Tos M, Thomsen J, Rygaard J, Fundova P,
roma surgery: influence of tumor size and surgical Charabi B (2000) Cystic vestibular schwannoma –
approach on incidence and response to treatment. J clinical and experimental studies. Acta Otolaryngol
Neurosurg 94(2):217–223. doi:10.3171/jns.2001.94. Suppl 543:11–13
2.0217 39. Chen DQ, Quan J, Guha A, Tymianski M, Mikulis D,
25. Briggs RJ, Fabinyi G, Kaye AH (2000) Current man- Hodaie M (2011) Three-dimensional in vivo model-
agement of acoustic neuromas: review of surgical ing of vestibular schwannomas and surrounding cra-
approaches and outcomes. J Clin Neurosci 7(6):521– nial nerves with diffusion imaging tractography.
526. doi:10.1054/jocn.2000.0728, S0967-5868(00) Neurosurgery 68(4):1077–1083. doi:10.1227/
90728-8 [pii] NEU.0b013e31820c6cbe
26. Briggs RJ, Luxford WM, Atkins JS Jr, Hitselberger 40. Chishti AH, Kim AC, Marfatia SM, Lutchman M,
WE (1994) Translabyrinthine removal of large acous- Hanspal M, Jindal H, Liu SC, Low PS, Rouleau GA,
tic neuromas. Neurosurgery 34(5):785–790; discus- Mohandas N, Chasis JA, Conboy JG, Gascard P,
sion 790–791 Takakuwa Y, Huang SC, Benz EJ Jr, Bretscher A,
27. Bruder CE, Ichimura K, Tingby O, Hirakawa K, Fehon RG, Gusella JF, Ramesh V, Solomon F,
Komatsuzaki A, Tamura A, Yuasa Y, Collins VP, Marchesi VT, Tsukita S, Hoover KB et al (1998) The
306 5 Vestibular Schwannomas

FERM domain: a unique module involved in the link- vestibular schwannoma. Otol Neurotol 30(5):681–682.
age of cytoplasmic proteins to the membrane. Trends doi:10.1097/MAO.0b013e3181859a3e
Biochem Sci 23(8):281–282, S0968000498012377 54. Doherty JK, Ongkeko W, Crawley B, Andalibi A,
[pii] Ryan AF (2008) ErbB and Nrg: potential molecular
41. Cho YD, Choi GH, Lee SP, Kim JK (2003) (1)H-MRS targets for vestibular schwannoma pharmacotherapy.
metabolic patterns for distinguishing between menin- Otol Neurotol 29(1):50–57. doi:10.1097/mao.0b013e
giomas and other brain tumors. Magn Reson Imaging 31815d4429, 00129492-200801000-00012 [pii]
21(6):663–672, S0730725X03000973 [pii] 55. Duong DH, O’Malley S, Sekhar LN, Wright DG
42. Chung WY, Pan DH, Lee CC, Wu HM, Liu KD, Yen (2000) Postoperative hydrocephalus in cranial base
YS, Guo WY, Shiau CY, Shih YH (2010) Large surgery. Skull Base Surg 10(4):197–200
vestibular schwannomas treated by Gamma Knife 56. Ebersold MJ, Harner SG, Beatty CW, Harper CM Jr,
surgery: long-term outcomes. J Neurosurg 113(Suppl): Quast LM (1992) Current results of the retrosigmoid
112–121. doi:10.3171/2010.8.GKS10954 approach to acoustic neurinoma. J Neurosurg
43. Ciric I, Zhao J, Rosenblatt S, Wiet R, O’Shaughnessy 76(6):901–909. doi:10.3171/jns.1992.76.6.0901
B (2005) Suboccipital retrosigmoid approach for 57. El-Kashlan HK, Zeitoun H, Arts HA, Hoff JT,
removal of vestibular schwannomas: facial nerve Telian SA (2000) Recurrence of acoustic neuroma
function and hearing preservation. Neurosurgery 56: after incomplete resection. Am J Otol 21(3):389–392
560–570 58. Eldridge R, Parry D (1992) Vestibular schwannoma
44. Clemis JD, Ballad WJ, Baggot PJ, Lyon ST (acoustic neuroma). Consensus development confer-
(1986) Relative frequency of inferior vestibular ence. Neurosurgery 30(6):962–964
schwannoma. Arch Otolaryngol Head Neck Surg 59. Elliott FA, McKissock KW (1954) Acoustic neuroma;
112(2):190–194 early diagnosis. Lancet 267(6850):1189–1191
45. Colletti V, Fiorino F (2003) Middle fossa versus ret- 60. Elsmore AJ, Mendoza ND (2002) The operative learn-
rosigmoid-transmeatal approach in vestibular schwan- ing curve for vestibular schwannoma excision via the
noma surgery: a prospective study. Otol Neurotol retrosigmoid approach. Br J Neurosurg 16(5):448–455
24(6):927–934 61. Fahlbusch R, Neu M, Strauss C (1998) Preservation
46. Comey CH, Jannetta PJ, Sheptak PE, Joh HD, of hearing in large acoustic neurinomas following
Burkhart LE (1995) Staged removal of acoustic removal via suboccipito-lateral approach. Acta
tumors: techniques and lessons learned from a series Neurochir (Wien) 140(8):771–777; discussion 778
of 83 patients. Neurosurgery 37(5):915–920; discus- 62. Falcioni A, Piccirillo E, Mancini F (2000) Cystic ves-
sion 920–921 tibular schwannoma. Am J Otol 21(4):595–596
47. (2001) Consensus meeting on systems for reporting 63. Falcioni M, Mulder JJ, Taibah A, De Donato G, Sanna
results in acoustic neuroma. November 7–9, 2001. M (1999) No cerebrospinal fluid leaks in translabyrin-
Tokyo, Japan. Abstracts (2001). Keio J Med 50(Suppl thine vestibular schwannoma removal: reappraisal of
4):13–77 200 consecutive patients. Am J Otol 20(5):660–666
48. Couloigner V, Gervaz E, Kalamarides M, Ferrary E, 64. Fayad JN, Schwartz MS, Slattery WH, Brackmann DE
Rey A, Sterkers O, Henin D (2003) Clinical and histo- (2007) Prevention and treatment of cerebrospinal fluid
logic parameters correlated with facial nerve function leak after translabyrinthine acoustic tumor removal.
after vestibular schwannoma surgery. Skull Base Otol Neurotol 28(3):387–390. doi:10.1097/01.
13(1):13–19. doi:10.1055/s-2003-820553 mao.0000265188.22345.d4, 00129492-200704000-
49. Dandy W (1925) An operation for the total removal of 00019 [pii]
cerebellopontine (acoustic) tumors. Surg Gynecol 65. Fernandez-Valle C, Tang Y, Ricard J, Rodenas-
Obstet 41:129–148 Ruano A, Taylor A, Hackler E, Biggerstaff J, Iacovelli
50. Deguine O, Maillard A, Bonafe A, el Adouli H H, J (2002) Paxillin binds schwannomin and regulates its
Tremoulet M, Fraysse B (1998) Pre-operative and density-dependent localization and effect on cell mor-
per-operative factors conditioning long-term facial phology. Nat Genet 31(4):354–362. doi:10.1038/
nerve function in vestibular schwannoma surgery ng930, ng930 [pii]
through translabyrinthine approach. J Laryngol Otol 66. Fitzgerald DC, Grundfast KM, Hecht DA, Mark AS
112(5):441–445 (1999) Intralabyrinthine schwannomas. Am J Otol
51. Delsanti C, Regis J (2004) Cystic vestibular schwan- 20(3):381–385
nomas. Neurochirurgie 50(2–3 Pt 2):401–406, MDOI- 67. Flint D, Fagan P, Panarese A (2005) Conservative
NCHIR-06-2004-50-2-3-C2-0028-3770-101019- management of sporadic unilateral acoustic neuro-
ART26 [pii] mas. J Laryngol Otol 119(6):424–428. doi:10.1258/
52. Di Maio S, Malebranche AD, Westerberg B, 0022215054273089
Akagami R (2011) Hearing preservation after micro- 68. Forton GE, Cremers CW, Offeciers EE (2004)
surgical resection of large vestibular schwannomas. Acoustic neuroma ingrowth in the cochlear nerve:
Neurosurgery 68(3):632–640. doi:10.1227/NEU. does it influence the clinical presentation? Ann Otol
0b013e31820777b1; discussion 640 Rhinol Laryngol 113(7):582–586
53. Dispenza F, De Stefano A (2009) Imaging of vestibular 69. Friedman RA, Brackmann DE, Hitselberger WE,
schwannoma with prevalent cystic component: cystic Schwartz MS, Iqbal Z, Berliner KI (2005) Surgical
References 307

salvage after failed irradiation for vestibular of-life outcome. Otol Neurotol. doi:10.1097/
schwannoma. Laryngoscope 115(10):1827–1832. MAO.0b013e3181b4e3c9
doi:10.1097/01.mlg.0000175063.76945.75, 83. Gormley WB, Sekhar LN, Wright DC, Kamerer D,
00005537-200510000-00022 [pii] Schessel D (1997) Acoustic neuromas: results of cur-
70. Frohlich AM, Sutherland GR (1993) Epidemiology rent surgical management. Neurosurgery 41(1):50–
and clinical features of vestibular schwannoma in 58; discussion 58–60
Manitoba, Canada. Can J Neurol Sci 20(2):126–130 84. Grey PL, Moffat DA, Palmer CR, Hardy DG, Baguley
71. Fuentes S, Arkha Y, Pech-Gourg G, Grisoli F, Dufour DM (1996) Factors which influence the facial nerve
H, Regis J (2008) Management of large vestibular outcome in vestibular schwannoma surgery. Clin
schwannomas by combined surgical resection and Otolaryngol Allied Sci 21(5):409–413
gamma knife radiosurgery. Prog Neurol Surg 21: 85. Guermazi A, Lafitte F, Miaux Y, Adem C, Bonneville
79–82. doi:10.1159/000156709, 156709 [pii] JF, Chiras J (2005) The dural tail sign – beyond men-
72. Fundova P, Charabi S, Tos M, Thomsen J (2000) ingioma. Clin Radiol 60(2):171–188. doi:10.1016/j.
Cystic vestibular schwannoma: surgical outcome. J crad.2004.01.019, S0009-9260(04)00074-1 [pii]
Laryngol Otol 114(12):935–939 86. Haberkamp TJ, Meyer GA, Fox M (1998) Surgical
73. Gardner G, Robertson JH (1988) Hearing preserva- exposure of the fundus of the internal auditory canal:
tion in unilateral acoustic neuroma surgery. Ann Otol anatomic limits of the middle fossa versus the ret-
Rhinol Laryngol 97(1):55–66 rosigmoid transcanal approach. Laryngoscope 108(8
74. Gerganov V, Giordano M, Samiii A, Samii M (2012) Pt 1):1190–1194
Surgical treatment of patients with vestibular schwan- 87. Hajioff D, Raut VV, Walsh RM, Bath AP, Bance ML,
nomas after failed previous radiosurgery. J Neurosurg Guha A, Tator CH, Rutka JA (2008) Conservative
116(4):713–720 management of vestibular schwannomas: third review
75. Gerganov V, Nouri M, Stieglitz L, Giordano M, of a 10-year prospective study. Clin Otolaryngol
Samii M, Samii A (2009) Radiological factors related 33(3):255–259. doi:10.1111/j.1749-4486.2008.
to pre-operative hearing levels in patients with ves- 01705.x, COA1705 [pii]
tibular schwannomas. J Clin Neurosci 16(8):1009– 88. Hakyemez B, Erdogan C, Bolca N, Yildirim N,
1012. doi:10.1016/j.jocn.2008.08.029, S0967-5868 Gokalp G, Parlak M (2006) Evaluation of different
(08)00486-4 [pii] cerebral mass lesions by perfusion-weighted MR
76. Gerganov VM, Giordano M, Samii M, Samii A (2011) imaging. J Magn Reson Imaging 24(4):817–824.
Diffusion tensor imaging-based fiber tracking for doi:10.1002/jmri.20707
prediction of the position of the facial nerve in rela- 89. Hanemann CO, Evans DG (2006) News on the genet-
tion to large vestibular schwannomas. J Neurosurg. ics, epidemiology, medical care and translational
doi:10.3171/2011.7.JNS11495 research of Schwannomas. J Neurol 253(12):
77. Gerganov VM, Klinge PM, Nouri M, Stieglitz L, 1533–1541. doi:10.1007/s00415-006-0347-0
Samii M, Samii A (2009) Prognostic clinical and 90. Hardy M, Crowe S (1936) Early asymptomatic acous-
radiological parameters for immediate facial nerve tic tumor. Arch Surg 32:292–301
function following vestibular schwannoma surgery. 91. Harner SG, Daube JR, Ebersold MJ, Beatty CW
Acta Neurochir (Wien) 151(6):581–587. doi:10.1007/ (1987) Improved preservation of facial nerve func-
s00701-009-0288-3; discussion 587 tion with use of electrical monitoring during removal
78. Gerganov VM, Pirayesh A, Nouri M, Hore N, of acoustic neuromas. Mayo Clin Proc 62(2):
Luedemann WO, Oi S, Samii A, Samii M (2011) 92–102
Hydrocephalus associated with vestibular schwanno- 92. Hasegawa T, Fujitani S, Katsumata S, Kida Y,
mas: management options and factors predicting the Yoshimoto M, Koike J (2005) Stereotactic radiosur-
outcome. J Neurosurg 114(5):1209–1215. gery for vestibular schwannomas: analysis of 317
doi:10.3171/2010.10.JNS1029 patients followed more than 5 years. Neurosurgery
79. Gjuric M, Rudic M (2008) What is the best tumor size 57(2):257–265, 00006123-200508000-00009 [pii];
to achieve optimal functional results in vestibular discussion 257–265
schwannoma surgery? Skull Base 18(5):317–325. 93. Hastan D, Vandenbroucke JP, van der Mey AG (2009)
doi:10.1055/s-0028-1086056 A meta-analysis of surgical treatment for vestibular
80. Gjuric M, Wigand ME, Wolf SR (2001) Enlarged schwannoma: is hospital volume related to preserva-
middle fossa vestibular schwannoma surgery: experi- tion of facial function? Otol Neurotol. doi:10.1097/
ence with 735 cases. Otol Neurotol 22(2):223–230; MAO.0b013e3181b0d04a
discussion 230–231 94. House JW, Bassim MK, Schwartz M (2008)
81. Glasscock ME 3rd, Hays JW, Minor LB, Haynes DS, False-positive magnetic resonance imaging in
Carrasco VN (1993) Preservation of hearing in sur- the diagnosis of vestibular schwannoma. Otol
gery for acoustic neuromas. J Neurosurg 78(6): Neurotol 29(8):1176–1178. doi:10.1097/MAO.
864–870. doi:10.3171/jns.1993.78.6.0864 0b013e318187e199
82. Godefroy WP, Kaptein AA, Vogel JJ, van der Mey AG 95. House JW, Brackmann DE (1985) Facial nerve grad-
(2009) Conservative treatment of vestibular schwan- ing system. Otolaryngol Head Neck Surg 93(2):
noma: a follow-up study on clinical and quality- 146–147
308 5 Vestibular Schwannomas

96. Inamasu J, Shiobara R, Kagami H, Sato S, Kawase Neurosurg 112(1):152–157. doi:10.3171/2009.3.


T, Kanzaki J (2000) Medial (intra-cisternal) acoustic JNS081053
neuromas. Acta Otolaryngol 120(5):623–626 109. Kanzaki J, Ogawa K, Tsuchihashi N, Inoue Y,
97. Irving RM, Viani L, Hardy DG, Baguley DM, Yamamoto M, Ikeda S (1991) Postoperative compli-
Moffat DA (1995) Nervus intermedius function after cations in acoustic neuroma surgery by the extended
vestibular schwannoma removal: clinical features middle cranial fossa approach. Acta Otolaryngol
and pathophysiological mechanisms. Laryngoscope Suppl 487:75–79
105(8 Pt 1):809–813. doi:10.1288/00005537- 110. Kanzaki J, Tos M, Sanna M, Moffat DA, Monsell EM,
199508000-00007 Berliner KI (2003) New and modified reporting sys-
98. Iwai Y, Yamanaka K, Ishiguro T (2003) Surgery tems from the consensus meeting on systems for
combined with radiosurgery of large acoustic neuro- reporting results in vestibular schwannoma. Otol
mas. Surg Neurol 59(4):283–289, Neurotol 24(4):642–648; discussion 648–649
S0090301903000259 [pii]; discussion 289–291 111. Kartush JM, Lundy LB (1992) Facial nerve outcome
99. Jackler R (1994) Acoustic neuroma (vestibular in acoustic neuroma surgery. Otolaryngol Clin North
schwannoma). In: Jackler RK, Brackmann DE (eds) Am 25(3):623–647
Neurotology. Mosby-Year Book, St. Louis, pp 112. Keiper GL Jr, Sherman JD, Tomsick TA, Tew JM Jr
729–785 (1999) Dural sinus thrombosis and pseudotumor
100. Jackson CG (1993) Basic surgical principles of cerebri: unexpected complications of suboccipital
neurotologic skull base surgery. Laryngoscope craniotomy and translabyrinthine craniectomy. J
103(11 Pt 2 Suppl 60):29–44 Neurosurg 91(2):192–197. doi:10.3171/jns.1999.91.
101. Jacoby LB, Jones D, Davis K, Kronn D, Short MP, 2.0192
Gusella J, MacCollin M (1997) Molecular analysis 113. Kemink JL, Langman AW, Niparko JK, Graham MD
of the NF2 tumor-suppressor gene in schwannoma- (1991) Operative management of acoustic neuro-
tosis. Am J Hum Genet 61(6):1293–1302. mas: the priority of neurologic function over com-
doi:10.1086/301633, S0002-9297(07)60230-4 [pii] plete resection. Otolaryngol Head Neck Surg
102. Jacoby LB, MacCollin M, Barone R, Ramesh V, 104(1):96–99
Gusella JF (1996) Frequency and distribution of 114. Kerr RG, Hearst MJ, Samy RN, van Loveren HR,
NF2 mutations in schwannomas. Genes Chromo- Tew JM Jr, Pensak ML, Theodosopoulos PV (2009)
somes Cancer 17(1):45–55, 10.1002/(SICI)1098- Delayed extrusion of hydroxyapatite cement after
2264(199609)17:1<45::AID-GCC7>3.0.CO;2-2 transpetrosal reconstruction. Neurosurgery
[pii]. 10.1002/(SICI)1098-2264(199609)17:1<45:: 64(3):527–531. doi:10.1227/01.NEU.0000338070.
AID-GCC7>3.0.CO;2-2 85190.3B, 00006123-200903000-00015 [pii]; dis-
103. Jacoby LB, MacCollin M, Louis DN, Mohney T, cussion 531–532
Rubio MP, Pulaski K, Trofatter JA, Kley N, Seizinger 115. Khrais T, Romano G, Sanna M (2008) Nerve origin
B, Ramesh V et al (1994) Exon scanning for muta- of vestibular schwannoma: a prospective study. J
tion of the NF2 gene in schwannomas. Hum Mol Laryngol Otol 122(2):128–131. doi:10.1017/
Genet 3(3):413–419 S0022215107001028, S0022215107001028 [pii]
104. Jain VK, Mehrotra N, Sahu RN, Behari S, Banerji D, 116. Kino T, Takeshima H, Nakao M, Nishi T,
Chhabra DK (2005) Surgery of vestibular schwan- Yamamoto K, Kimura T, Saito Y, Kochi M, Kuratsu
nomas: an institutional experience. Neurol India J, Saya H, Ushio Y (2001) Identification of the cis-
53(1):41–45; discussion 45 acting region in the NF2 gene promoter as a potential
105. James MF, Lelke JM, Maccollin M, Plotkin SR, target for mutation and methylation-dependent
Stemmer-Rachamimov AO, Ramesh V, Gusella JF silencing in schwannoma. Genes Cells 6(5):
(2008) Modeling NF2 with human arachnoidal 441–454, gtc432 [pii]
and meningioma cell culture systems: NF2 silencing 117. Kondziolka D, Lunsford LD, Flickinger JC (2000)
reflects the benign character of tumor growth. Gamma knife radiosurgery for vestibular schwanno-
Neurobiol Dis 29(2):278–292. doi:10.1016/j. mas. Neurosurg Clin N Am 11(4):651–658
nbd.2007.09.002, S0969-9961(07)00211-2 [pii] 118. Kondziolka D, Lunsford LD, McLaughlin MR,
106. Jung S, Kang SS, Kim TS, Kim HJ, Jeong SK, Kim SC, Flickinger JC (1998) Long-term outcomes after
Lee JK, Kim JH, Kim SH, Lee JH (2000) Current sur- radiosurgery for acoustic neuromas. N Engl J Med
gical results of retrosigmoid approach in extralarge 339(20):1426–1433. doi:10.1056/NEJM199811123
vestibular schwannomas. Surg Neurol 53(4):370–377, 392003
S0090301900001968 [pii]; discussion 377–378 119. Koos WT, Day JD, Matula C, Levy DI (1998)
107. Jung S, Kim SH, Kim HW, Kim TS, Kim JH, Neurotopographic considerations in the microsurgi-
Kang SS, Lee JH (1998) Prediction of facial nerve cal treatment of small acoustic neurinomas. J
displacement in extra large vestibular schwannoma. Neurosurg 88(3):506–512. doi:10.3171/jns.1998.
Acta Neurochir (Wien) 140(11):1143–1145 88.3.0506
108. Kameda K, Shono T, Hashiguchi K, Yoshida F, 120. Koos WT, Lang J (1993) Tumors of the cerebello-
Sasaki T (2010) Effect of tumor removal on tinni- pontine angle. In: Color atlas of microneurosurgery.
tus in patients with vestibular schwannoma. J Thieme Medical Publishers, New York, pp 510–608
References 309

121. Kuo TC, Jackler RK, Wong K, Blevins NH, Pitts LH up review of 11 patients. J Neurosurg 74(3):422–425.
(1997) Are acoustic neuromas encapsulated tumors? doi:10.3171/jns.1991.74.3.0422
Otolaryngol Head Neck Surg 117(6):606–609, 134. Ludemann WO, Stieglitz LH, Gerganov V, Samii A,
S0194-5998(97)70040-8 [pii] Samii M (2008) Fat implant is superior to muscle
122. Kveton JF, Tarlov EC, Drumheller G, Katcher P, implant in vestibular schwannoma surgery for
Abbott C (1989) Cochlear nerve conduction block: the prevention of cerebrospinal fluid fistulae.
an explanation for spontaneous hearing return after Neurosurgery 63(1 Suppl 1):ONS38–ONS42.
acoustic tumor surgery. Otolaryngol Head Neck doi: 10.1227/01.neu.0000335009.53122.a3 ,
Surg 100(6):594–601 00006123-200807001-00006 [pii]; discussion 42–43
123. Lallemand D, Manent J, Couvelard A, Watilliaux A, 135. Luetje C (2000) Spontaneous involution of acoustic
Siena M, Chareyre F, Lampin A, Niwa-Kawakita M, tumors. Am J Otol 21:393–398
Kalamarides M, Giovannini M (2009) Merlin 136. Luetje CM, Whittaker CK, Calloway LA, Veraga G
regulates transmembrane receptor accumulation (1983) Histological acoustic tumor involvement of
and signaling at the plasma membrane in primary the VIIth nerve and multicentric involvement of the
mouse Schwann cells and in human schwannomas. VIIIth. Laryngoscope 93:1133–1139
Oncogene 28(6):854–865. doi:10.1038/onc.2008. 137. Lutchman M, Rouleau GA (1995) The
427, onc2008427 [pii] neurofibromatosis type 2 gene product, schwan-
124. Lalwani AK, Butt FY, Jackler RK, Pitts LH, nomin, suppresses growth of NIH 3T3 cells. Cancer
Yingling CD (1994) Facial nerve outcome after Res 55(11):2270–2274
acoustic neuroma surgery: a study from the era of 138. Lynn SG, Driscoll CL, Harner SG, Beatty CW,
cranial nerve monitoring. Otolaryngol Head Neck Atkinson EJ (1999) Assessment of dysequilibrium after
Surg 111(5):561–570, S0194599894000938 [pii] acoustic neuroma removal. Am J Otol 20(4):484–494
125. Lanman TH, Brackmann DE, Hitselberger WE, 139. Magliulo G, Cordeschi S, Sepe C, de Vincentiis M
Subin B (1999) Report of 190 consecutive cases of (1998) Taste and lacrimation after acoustic neuroma
large acoustic tumors (vestibular schwannoma) surgery. Rev Laryngol Otol Rhinol (Bord) 119(3):
removed via the translabyrinthine approach. J 167–170
Neurosurg 90(4):617–623 140. Magliulo G, D’Amico R, Di Cello P (2003) Delayed
126. Lanser MJ, Sussman SA, Frazer K (1992) Epide- facial palsy after vestibular schwannoma resection:
miology, pathogenesis, and genetics of acoustic clinical data and prognosis. J Otolaryngol 32(6):
tumors. Otolaryngol Clin North Am 25(3):499–520 400–404
127. Lapsiwala SB, Pyle GM, Kaemmerle AW, Sasse FJ, 141. Malis LI (2001) Nuances in acoustic neuroma sur-
Badie B (2002) Correlation between auditory func- gery. Neurosurgery 49(2):337–341
tion and internal auditory canal pressure in patients 142. Mamikoglu B, Wiet RJ, Esquivel CR (2002)
with vestibular schwannomas. J Neurosurg Translabyrinthine approach for the management of
96(5):872–876. doi:10.3171/jns.2002.96.5.0872 large and giant vestibular schwannomas. Otol
128. Lee CC, Yen YS, Pan DH, Chung WY, Wu HM, Guo Neurotol 23(2):224–227
WY, Chen MT, Liu KD, Shih YH (2010) Delayed 143. Mass SC, Wiet RJ, Dinces E (1999) Complications
microsurgery for vestibular schwannoma after of the translabyrinthine approach for the removal of
gamma knife radiosurgery. J Neurooncol 98(2):203– acoustic neuromas. Arch Otolaryngol Head Neck
212. doi:10.1007/s11060-010-0178-9 Surg 125(7):801–804
129. Leksell L (1971) A note on the treatment of acoustic 144. Matthies C, Samii M (1997) Direct brainstem
tumours. Acta Chir Scand 137(8):763–765 recording of auditory evoked potentials during ves-
130. Lescanne E, Francois P, Bakhos D, Velut S, Robier tibular schwannoma resection: nuclear BAEP
A, Pollak A (2008) Vestibular schwannoma: dissec- recording. Technical note and preliminary results. J
tion of the tumor and arachnoidal duplication. Otol Neurosurg 86(6):1057–1062. doi:10.3171/jns.
Neurotol 29(7):989–994. doi:10.1097/MAO. 1997.86.6.1057
0b013e3181845812 145. Matthies C, Samii M (1997) Management of 1000
131. Linskey ME (2000) Stereotactic radiosurgery versus vestibular schwannomas (acoustic neuromas): clini-
stereotactic radiotherapy for patients with vestibular cal presentation. Neurosurgery 40(1):1–9; discus-
schwannoma: a Leksell Gamma Knife Society 2000 sion 9–10
debate. J Neurosurg 93(Suppl 3):90–95. doi:10.3171/ 146. Matthies C, Samii M (1997) Management of vestib-
jns.2000.93.supplement3.0090 ular schwannomas (acoustic neuromas): the value of
132. Lobato-Polo J, Kondziolka D, Zorro O, Kano H, neurophysiology for evaluation and prediction of
Flickinger JC, Lunsford LD (2009) Gamma knife auditory function in 420 cases. Neurosurgery
radiosurgery in younger patients with vestibular 40(5):919–929; discussion 929–930
schwannomas. Neurosurgery 65(2):294–300. 147. Matthies C, Samii M, Krebs S (1997) Management
doi: 10.1227/01.NEU.0000345944.14065.35 , of vestibular schwannomas (acoustic neuromas):
0006123-200908000-00010 [pii]; discussion 294–300 radiological features in 202 cases–their value for
133. Lownie SP, Drake CG (1991) Radical intracapsular diagnosis and their predictive importance. Neuro-
removal of acoustic neurinomas. Long-term follow- surgery 40(3):469–481; discussion 481–482
310 5 Vestibular Schwannomas

148. McFarland WH, Linthicum FH, Waldorf RA (1989) 160. Niemczyk K, Vaneecloo FM, Lecomte MH,
Auditory and vestibular tests (in acoustic neuroma). Lejeune JP, Lemaitre L, Skarzynski H, Vincent C,
Semin Hear 10:313–326 Dubrulle F (2000) Correlation between Ki-67 index
149. McKenna MJ, Halpin C, Ojemann RG, Nadol JB Jr, and some clinical aspects of acoustic neuromas (ves-
Montgomery WW, Levine RA, Carlisle E, Martuza tibular schwannomas). Otolaryngol Head Neck Surg
R (1992) Long-term hearing results in patients after 123(6):779–783. doi:10.1067/mhn.2000.111356,
surgical removal of acoustic tumors with hearing S0194-5998(00)41879-6 [pii]
preservation. Am J Otol 13(2):134–136 161. Oghalai JS, Buxbaum JL, Pitts LH, Jackler RK
150. Millen SJ, Meyer G (1993) Surgical management of (2003) The effect of age on acoustic neuroma sur-
CSF otorhinorrhea following retrosigmoid removal gery outcomes. Otol Neurotol 24(3):473–477
of cerebellopontine angle tumors. Am J Otol 14(6): 162. Ohata K, Tsuyuguchi N, Morino M, Takami T,
585–589 Goto T, Hakuba A, Hara M (2002) A hypothesis of
151. Moffat DA, Hardy DG, Grey PL, Baguley DM epiarachnoidal growth of vestibular schwannoma at
(1996) The operative learning curve and its effect on the cerebello-pontine angle: surgical importance. J
facial nerve outcome in vestibular schwannoma Postgrad Med 48(4):253–258; discussion 258–259
surgery. Am J Otol 17(4):643–647 163. Okamoto K, Furusawa T, Ishikawa K, Sasai K,
152. Mohr G, Sade B, Dufour JJ, Rappaport JM (2005) Tokiguchi S (2006) Focal T2 hyperintensity in the
Preservation of hearing in patients undergoing dorsal brain stem in patients with vestibular schwan-
microsurgery for vestibular schwannoma: degree of noma. AJNR Am J Neuroradiol 27(6):1307–1311,
meatal filling. J Neurosurg 102(1):1–5. doi:10.3171. 27/6/1307 [pii]
jns.2005.102.1.0001 164. Park CK, Kim DC, Park SH, Kim JE, Paek SH,
153. Moon KS, Jung S, Seo SK, Jung TY, Kim IY, Kim DG, Jung HW (2006) Microhemorrhage, a pos-
Ryu HH, Jin YH, Jin SG, Jeong YI, Kim KK, Kang sible mechanism for cyst formation in vestibular
SS (2007) Cystic vestibular schwannomas: a possi- schwannomas. J Neurosurg 105(4):576–580.
ble role of matrix metalloproteinase-2 in cyst devel- doi:10.3171/jns.2006.105.4.576
opment and unfavorable surgical outcome. J 165. Parry DM, Eldridge R, Kaiser-Kupfer MI,
Neurosurg 106(5):866–871. doi:10.3171/jns.2007. Bouzas EA, Pikus A, Patronas N (1994) Neuro-
106.5.866 fibromatosis 2 (NF2): clinical characteristics of 63
154. Moriyama T, Fukushima T, Asaoka K, Roche PH, affected individuals and clinical evidence for
Barrs DM, McElveen JT Jr (2002) Hearing preserva- heterogeneity. Am J Med Genet 52(4):450–461.
tion in acoustic neuroma surgery: importance of doi:10.1002/ajmg.1320520411
adhesion between the cochlear nerve and the tumor. 166. Patel AK, Alexander TH, Andalibi A, Ryan AF,
J Neurosurg 97(2):337–340. doi:10.3171/ Doherty JK (2008) Vestibular schwannoma
jns.2002.97.2.0337 quantitative polymerase chain reaction expression
155. Muranen T, Gronholm M, Renkema GH, Carpen O of estrogen and progesterone receptors. Laryngo
(2005) Cell cycle-dependent nucleocytoplasmic scope 118(8):1458–1463. doi:10.1097/MLG.
shuttling of the neurofibromatosis 2 tumour suppres- 0b013e318177e20b, 00005537-200808000-00024
sor merlin. Oncogene 24(7):1150–1158. doi:10.1038/ [pii]
sj.onc.1208283, 1208283 [pii] 167. Patni AH, Kartush JM (2005) Staged resection of
156. Murphy ES, Suh JH (2011) Radiotherapy for large acoustic neuromas. Otolaryngol Head Neck
vestibular schwannomas: a critical review. Surg 132(1):11–19. doi:10.1016/j.otohns.2004.09.
Int J Radiat Oncol Biol Phys 79(4):985–997. 094, S0194599804018777 [pii]
doi:10.1016/j.ijrobp.2010.10.010, S0360- 168. Pelton PD, Sherman LS, Rizvi TA, Marchionni MA,
3016(10)03428-0 [pii] Wood P, Friedman RA, Ratner N (1998) Ruffling
157. Nedzelski JM, Schessel DA, Pfleiderer A, Kassel membrane, stress fiber, cell spreading and prolifera-
EE, Rowed DW (2008) Conservative management tion abnormalities in human Schwannoma cells.
of acoustic neuromas 1992. Neurosurg Clin N Am Oncogene 17(17):2195–2209. doi:10.1038/sj.onc.
19(2):207–216. doi:10.1016/j.nec.2008.02.013, v. 1202141
S1042-3680(08)00014-4 [pii] 169. Pendl G, Ganz JC, Kitz K, Eustacchio S (1996)
158. Neff BA, Oberstien E, Lorenz M, Chaudhury AR, Acoustic neurinomas with macrocysts treated with
Welling DB, Chang LS (2006) Cyclin D(1) and D(3) Gamma Knife radiosurgery. Stereotact Funct
expression in vestibular schwannomas. Laryngo- Neurosurg 66(Suppl 1):103–111
scope 116(3):423–426. doi:10.1097/01.mlg. 170. Piccirillo E, Wiet MR, Flanagan S, Dispenza F,
0000195076.05466.6c, 00005537-200603000- Giannuzzi A, Mancini F, Sanna M (2009) Cystic
00017 [pii] vestibular schwannoma: classification, management,
159. Neff BA, Ting J, Dickinson SL, Welling DB (2005) and facial nerve outcomes. Otol Neurotol 30(6):826–
Facial nerve monitoring parameters as a predictor of 834. doi:10.1097/MAO.0b013e3181b04e18,
postoperative facial nerve outcomes after vestibular 00129492-200909000-00024 [pii]
schwannoma resection. Otol Neurotol 26(4): 171. Pillai P, Sammet S, Ammirati M (2009) Image-
728–732, 00129492-200507000-00031 [pii] guided, endoscopic-assisted drilling and exposure of
References 311

the whole length of the internal auditory canal and 183. Roche PH, Bouvier C, Chinot O, Figarella-Branger
its fundus with preservation of the integrity of the D (2008) Genesis and biology of vestibular schwan-
labyrinth using a retrosigmoid approach: a labora- nomas. Prog Neurol Surg 21:24–31. doi:10.1159/
tory investigation. Neurosurgery 65(6 Suppl):53–59. 000156556, 156556 [pii]
doi: 10.1227/01.NEU.0000343521.88537.16 , 184. Rodgers GK, Luxford WM (1993) Factors affecting
00006123-200912001-00004 [pii]; discussion 59 the development of cerebrospinal fluid leak and
172. Pirouzmand F, Tator CH, Rutka J (2001) Management meningitis after translabyrinthine acoustic tumor
of hydrocephalus associated with vestibular surgery. Laryngoscope 103(9):959–962. doi:10.1288/
schwannoma and other cerebellopontine angle 00005537-199309000-00003
tumors. Neurosurgery 48(6):1246–1253; discussion 185. Rogg JM, Ahn SH, Tung GA, Reinert SE, Noren G
1253–1254 (2005) Prevalence of hydrocephalus in 157 patients
173. Pollock BE (2006) Management of vestibular with vestibular schwannoma. Neuroradiology
schwannomas that enlarge after stereotactic radio- 47(5):344–351. doi:10.1007/s00234-005-1363-y
surgery: treatment recommendations based on a 15 186. Roland JT Jr, Fishman AJ, Golfinos JG, Cohen N,
year experience. Neurosurgery 58(2):241–248. Alexiades G, Jackman AH (2004) Cranial nerve
doi: 10.1227/01.NEU.0000194833.66593.8B , preservation in surgery for large acoustic neuromas.
00006123-200602000-00005 [pii]; discussion Skull Base 14(2):85–90. doi:10.1055/s-2004-828699;
241–248 discussion 90–91
174. Post KD, Eisenberg MB, Catalano PJ (1995) Hearing 187. Rosenberg S (2000) Natural history of vestibular
preservation in vestibular schwannoma surgery: schwannomas. Laryngoscope 110:497–508
what factors influence outcome? J Neurosurg 188. Rouleau GA, Merel P, Lutchman M, Sanson M,
83(2):191–196. doi:10.3171/jns.1995.83.2.0191 Zucman J, Marineau C, Hoang-Xuan K, Demczuk S,
175. Prasad D, Steiner M, Steiner L (2000) Gamma sur- Desmaze C, Plougastel B et al (1993) Alteration in a
gery for vestibular schwannoma. J Neurosurg new gene encoding a putative membrane-organizing
92(5):745–759. doi:10.3171/jns.2000.92.5.0745 protein causes neuro-fibromatosis type 2. Nature
176. Prasad S, Hirsch BE, Kamerer DB, Durrant J, Sekhar 363(6429):515–521. doi:10.1038/363515a0
LN (1993) Facial nerve function following cerebel- 189. Rowed DW (1999) Hearing preservation in vestibu-
lopontine angle surgery: prognostic value of intraop- lar schwannoma surgery. J Neurosurg 90(2):
erative thresholds. Am J Otol 14(4):330–333 374–376
177. Preston-Martin S, Thomas DC, Wright WE, 190. Ruttledge MH, Rouleau GA (2005) Role of the
Henderson BE (1989) Noise trauma in the aetiology neurofibromatosis type 2 gene in the development of
of acoustic neuromas in men in Los Angeles County, tumors of the nervous system. Neurosurg Focus
1978–1985. Br J Cancer 59(5):783–786 19(5):E6, 190506 [pii]
178. Raftopoulos C, Abu Serieh B, Duprez T, Docquier 191. Sade B, Mohr G, Dufour JJ (2006) Vascular
MA, Guerit JM (2005) Microsurgical results with complications of vestibular schwannoma surgery: a
large vestibular schwannomas with preservation of comparison of the suboccipital retrosigmoid and
facial and cochlear nerve function as the primary translabyrinthine approaches. J Neurosurg 105(2):
aim. Acta Neurochir (Wien) 147(7):697–706. 200–204. doi:10.3171/jns.2006.105.2.200
doi:10.1007/s00701-005-0544-0; discussion 706 192. Sakamoto GT, Borchers DJ 3rd, Xiao F, Yang HJ,
179. Rangwala R, Banine F, Borg JP, Sherman LS (2005) Chang SD, Adler JR Jr (2009) Cyberknife radiosur-
Erbin regulates mitogen-activated protein (MAP) gery for trigeminal schwannomas. Neurosurgery
kinase activation and MAP kinase-dependent inter- 64(2 Suppl):A14–18. doi:10.1227/01.NEU.
actions between Merlin and adherens junction pro- 0000341629.57676.DB, 00006123-200902001-
tein complexes in Schwann cells. J Biol Chem 00004 [pii]
280(12):11790–11797. doi:10.1074/jbc. 193. Samii A, Brinker T, Kaminsky J, Lanksch WR,
M414154200, M414154200 [pii] Samii M (2000) Navigation-guided opening of the
180. Raut VV, Walsh RM, Bath AP, Bance ML, Guha A, internal auditory canal via the retrosigmoid route for
Tator CH, Rutka JA (2004) Conservative manage- acoustic neuroma surgery: cadaveric, radiological,
ment of vestibular schwannomas – second review of and preliminary clinical study. Neurosurgery
a prospective longitudinal study. Clin Otolaryngol 47(2):382–387; discussion 388
Allied Sci 29(5):505–514. doi:10.1111/j. 194. Samii M, Gerganov V, Samii A (2006) Improved
1365-2273.2004.00852.x, COA852 [pii] preservation of hearing and facial nerve function in
181. Regis J, Roche PH, Delsanti C, Thomassin JM, vestibular schwannoma surgery via the retrosigmoid
Ouaknine M, Gabert K, Pellet W (2007) Modern approach in a series of 200 patients. J Neurosurg
management of vestibular schwannomas. Prog 105(4):527–535. doi:10.3171/jns.2006.105.4.527
Neurol Surg 20:129–141. doi:10.1159/0000100100, 195. Samii M, Gerganov V, Samii A (2008) Microsurgery
100100 [pii] management of vestibular schwannomas in
182. Rhoton AL Jr (2000) The cerebellopontine angle and neurofibromatosis type 2: indications and results.
posterior fossa cranial nerves by the retrosigmoid Prog Neurol Surg 21:169–175.
approach. Neurosurgery 47(3 Suppl):S93–S129 doi:10.1159/000156905, 156905 [pii]
312 5 Vestibular Schwannomas

196. Samii M, Gerganov VM, Samii A (2009) Functional Norrell H (eds) Neurological surgery of the ear.
outcome after complete surgical removal of giant Aesculapius, Birmingham, pp 193–197
vestibular schwannomas. J Neurosurg 112(4): 209. Schulder M, Sreepada GS, Kwartler JA, Cho ES
860–867. doi:10.3171/2009.7.JNS0989 (1999) Microsurgical removal of a vestibular
197. Samii M, Matthies C (1997) Management of 1000 schwannoma after stereotactic radiosurgery: surgical
vestibular schwannomas (acoustic neuromas): hear- and pathologic findings. Am J Otol 20(3):364–367;
ing function in 1000 tumor resections. Neurosurgery discussion 368
40(2):248–260; discussion 260–262 210. Seedat RY, Claassen AJ, Mol DA (2002) Incidence
198. Samii M, Matthies C (1997) Management of 1000 and management of acoustic neuromas in South
vestibular schwannomas (acoustic neuromas): surgi- Africa. Otol Neurotol 23(6):996–998
cal management and results with an emphasis on 211. Sekhar LN, Gormley WB, Wright DC (1996) The
complications and how to avoid them. Neurosurgery best treatment for vestibular schwannoma (acoustic
40(1):11–21; discussion 21–13 neuroma): microsurgery or radiosurgery? Am J Otol
199. Samii M, Matthies C (1997) Management of 1000 17(4):676–682; discussion 683–689
vestibular schwannomas (acoustic neuromas): the 212. Sekhar LN, Tzortzdis F (1999) Retrosigmoid
facial nerve – preservation and restitution of func- approach to the cerebellopontine angle. In: Sekhar
tion. Neurosurgery 40(4):684–694; discussion LN, de Oliveira E (eds) Cranial microsurgery.
694–685 Approaches and techniques. Thieme, New York, pp
200. Sampath P, Holliday MJ, Brem H, Niparko JK, 352–377
Long DM (1997) Facial nerve injury in acoustic neu- 213. Seol HJ, Kim CH, Park CK, Kim DG, Chung YS,
roma (vestibular schwannoma) surgery: etiology and Jung HW (2006) Optimal extent of resection in ves-
prevention. J Neurosurg 87(1):60–66. doi:10.3171/ tibular schwannoma surgery: relationship to recur-
jns.1997.87.1.0060 rence and facial nerve preservation. Neurol Med
201. Sampath P, Rhines LD, Holliday MJ, Brem H, Chir (Tokyo) 46(4):176–180; discussion 180–181.
Long DM (1998) Late-onset facial nerve degenera- JST.JSTAGE/nmc/46.176 [pii]
tion after vestibular schwannoma surgery: incidence, 214. Shao KN, Tatagiba M, Samii M (1993) Surgical
putative mechanisms, and prevention. Neurosurg management of high jugular bulb in acoustic
Focus 5(3):e6, 050306 [pii] neurinoma via retrosigmoid approach. Neurosurgery
202. Sampath P, Rini D, Long DM (2000) Microanatomical 32(1):32–36; discussion 36–37
variations in the cerebellopontine angle associated 215. Sheptak PE, Jannetta PJ (1979) The two-stage
with vestibular schwannomas (acoustic neuromas): a excision of huge acoustic neurinomas. J Neurosurg
retrospective study of 1006 consecutive cases. J 51(1):37–41. doi:10.3171/jns.1979.51.1.0037
Neurosurg 92(1):70–78. doi:10.3171/jns.2000.92. 216. Shuto T, Inomori S, Matsunaga S, Fujino H (2008)
1.0070 Microsurgery for vestibular schwannoma after
203. Sanna M, Khrais T, Russo A, Piccirillo E, Augurio A gamma knife radiosurgery. Acta Neurochir (Wien)
(2004) Hearing preservation surgery in vestibular 150(3):229–234. doi:10.1007/s00701-007-1486-5;
schwannoma: the hidden truth. Ann Otol Rhinol discussion 234
Laryngol 113(2):156–163 217. Silverstein H, McDaniel A, Norrell H, Wazen J (1985)
204. Sanna M, Taibah A, Russo A, Falcioni M, Agarwal M Conservative management of acoustic neuroma in the
(2004) Perioperative complications in acoustic elderly patient. Laryngoscope 95(7 Pt 1):766–770
neuroma (vestibular schwannoma) surgery. Otol 218. Silverstein H, McDaniel AB, Norrell H (1985)
Neurotol 25(3):379–386, 00129492-200405000- Hearing preservation after acoustic neuroma surgery
00029 [pii] using intraoperative direct eighth cranial nerve mon-
205. Sasaki T, Shono T, Hashiguchi K, Yoshida F, itoring. Am J Otol (Suppl):99–106
Suzuki SO (2009) Histological considerations of 219. Skinner A (1923) The origin of acoustic nerve
the cleavage plane for preservation of facial tumors. Br J Surg 16:440–463
and cochlear nerve functions in vestibular schwan- 220. Slattery WH 3rd (2009) Microsurgery after radiosur-
noma surgery. J Neurosurg 110(4):648–655. gery or radiotherapy for vestibular schwannomas.
doi:10.3171/2008.4.17514 Otolaryngol Clin North Am 42(4):707–715.
206. Satar B, Yetiser S, Ozkaptan Y (2003) Impact of doi:10.1016/j.otc.2009.04.007, S0030-6665(09)
tumor size on hearing outcome and facial function 00062-0 [pii]
with the middle fossa approach for acoustic neu- 221. Smouha EE, Yoo M, Mohr K, Davis RP (2005)
roma: a meta-analytic study. Acta Otolaryngol Conservative management of acoustic neuroma: a
123(4):499–505 meta-analysis and proposed treatment algorithm.
207. Saunders JE, Luxford WM, Devgan KK, Laryngoscope 115(3):450–454. doi:10.1097/01.
Fetterman BL (1995) Sudden hearing loss in acous- mlg.0000175681.52517.cf, 00005537-200503000-
tic neuroma patients. Otolaryngol Head Neck Surg 00011 [pii]
113(1):23–31, S0194-5998(95)70140-0 [pii] 222. Somers T, Casselman J, de Ceulaer G, Govaerts P,
208. Schuknecht H (1977) Pathology of vestibular Offeciers E (2001) Prognostic value of magnetic
schwannoma (acoustic neuroma). In: Silverstein H, resonance imaging findings in hearing preservation
References 313

surgery for vestibular schwannoma. Otol Neurotol 235. Sughrue ME, Yang I, Rutkowski MJ, Aranda D,
22(1):87–94 Parsa AT (2010) Preservation of facial nerve func-
223. Staecker H, Nadol JB Jr, Ojeman R, Ronner S, tion after resection of vestibular schwannoma. Br J
McKenna MJ (2000) Hearing preservation in Neurosurg 24(6):666–671. doi:10.3109/02688697.2
acoustic neuroma surgery: middle fossa versus 010.520761
retrosigmoid approach. Am J Otol 21(3):399–404 236. Sughrue ME, Yeung AH, Rutkowski MJ, Cheung SW,
224. Stangerup SE, Caye-Thomasen P, Tos M, Thomsen J Parsa AT (2011) Molecular biology of familial and
(2006) The natural history of vestibular schwan- sporadic vestibular schwannomas: implications for
noma. Otol Neurotol 27(4):547–552. doi:10.1097/01. novel therapeutics. J Neurosurg 114(2):359–366.
mao.0000217356.73463.e7, 00129492-200606000- doi:10.3171/2009.10.JNS091135
00018 [pii] 237. Sunderland S (1945) The arterial relations of the
225. Stangerup SE, Tos M, Thomsen J, Caye-Thomasen internal auditory meatus. Brain 68:23
P (2010) True incidence of vestibular schwannoma? 238. Tallan EM, Harner SG, Beatty CW, Scheithauer BW,
Neurosurgery 67(5):1335–1340. doi:10.1227/ Parisi JE (1993) Does the distribution of Schwann
NEU.0b013e3181f22660; discussion 1340 cells correlate with the observed occurrence of
226. Sterkers JM, Perre J, Viala P, Foncin JF (1987) The acoustic neuromas? Am J Otol 14(2):131–134
origin of acoustic neuromas. Acta Otolaryngol 239. Tanaka Y, Kobayashi S, Hongo K, Tada T, Sato A,
103(5–6):427–431 Takasuna H (2003) Clinical and neuroimaging char-
227. Stieglitz LH, Giordano M, Gerganov V, Raabe A, acteristics of hydrocephalus associated with vestibu-
Samii A, Samii M, Ludemann WO (2010) Petrous lar schwannoma. J Neurosurg 98(6):1188–1193.
bone pneumatization is a risk factor for cerebrospi- doi:10.3171/jns.2003.98.6.1188
nal fluid fistula following vestibular schwannoma 240. Taoka T, Hirabayashi H, Nakagawa H, Sakamoto M,
surgery. Neurosurgery 67(2 Suppl Operative): Myochin K, Hirohashi S, Iwasaki S, Sakaki T,
509–515. doi:10.1227/NEU.0b013e3181f88884, Kichikawa K (2006) Displacement of the facial
00006123-201012002-00027 [pii] nerve course by vestibular schwannoma: preopera-
228. Stieglitz LH, Samii A, Kaminsky J, Gharabaghi A, tive visualization using diffusion tensor tractogra-
Samii M, Ludemann WO (2005) Nausea and phy. J Magn Reson Imaging 24(5):1005–1010.
dizziness after vestibular schwannoma surgery: a doi:10.1002/jmri.20725
multivariate analysis of preoperative symptoms. 241. Tarlov E (1980) Total one-stage suboccipital micro-
Neurosurgery 57(5):887–890; discussion 887–890. surgical removal of acoustic neuromas of all sizes:
00006123-200511000-00009 [pii] with emphasis on arachnoid planes and on saving the
229. Stokowski RP, Cox DR (2000) Functional analysis facial nerve. Surg Clin North Am 60(3):565–591
of the neurofibromatosis type 2 protein by means of 242. Tatagiba M, Samii M, Matthies C, el Azm M M,
disease-causing point mutations. Am J Hum Genet Schonmayr R (1992) The significance for postopera-
66(3):873–891. doi:10.1086/302812, S0002-9297 tive hearing of preserving the labyrinth in acoustic
(07)64015-4 [pii] neurinoma surgery. J Neurosurg 77(5):677–684.
230. Strauss C, Bischoff B, Romstock J, Rachinger J, doi:10.3171/jns.1992.77.5.0677
Rampp S, Prell J (2008) Hearing preservation in 243. Thamburaj K, Radhakrishnan VV, Thomas B, Nair S,
medial vestibular schwannomas. J Neurosurg Menon G (2008) Intratumoral microhemorrhages on
109(1):70–76. doi:10.3171/JNS/2008/109/7/0070 T2*-weighted gradient-echo imaging helps differen-
231. Stripf T, Braun K, Gouveris H, Stripf EA, Mann WJ, tiate vestibular schwannoma from meningioma.
Amedee RG (2007) Influence of different approaches AJNR Am J Neuroradiol 29(3):552–557.
to the cerebellopontine angle on the function of the doi:10.3174/ajnr.A0887, ajnr.A0887 [pii]
intermediate nerve. J Neurosurg 107(5):927–931. 244. Theodosopoulos PV, Pensak ML (2012) Contem-
doi:10.3171/JNS-07/11/0927 porary management of acoustic neuromas. Laryngo-
232. Sughrue ME, Yang I, Aranda D, Kane AJ, Parsa AT scope 121(6):1133–1137. doi:10.1002/lary.21799
(2010) Hearing preservation rates after microsurgi- 245. Tonn JC, Schlake HP, Goldbrunner R, Milewski C,
cal resection of vestibular schwannoma. J Clin Helms J, Roosen K (2000) Acoustic neuroma sur-
Neurosci 17(9):1126–1129. doi:10.1016/j.jocn.2010. gery as an interdisciplinary approach: a neurosurgi-
01.018, S0967-5868(10)00157-8 [pii] cal series of 508 patients. J Neurol Neurosurg
233. Sughrue ME, Yang I, Aranda D, Lobo K, Pitts LH, Psychiatry 69(2):161–166
Cheung SW, Parsa AT (2010) The natural history of 246. Tos M (1998) Increasing incidence of vestibular
untreated sporadic vestibular schwannomas: a com- schwannoma: is it real and realistic? Ugeskr Laeger
prehensive review of hearing outcomes. J Neurosurg 160(45):6538
112(1):163–167. doi:10.3171/2009.4.JNS08895 247. Tos M, Charabi S, Thomsen J (1999) Incidence of
234. Sughrue ME, Yang I, Aranda D, Rutkowski MJ, vestibular schwannomas. Laryngoscope 109(5):
Fang S, Cheung SW, Parsa AT (2011) Beyond audio- 736–740
facial morbidity after vestibular schwannoma sur- 248. Tos M, Drozdziewicz D, Thomsen J (1992) Medial
gery. J Neurosurg 114(2):367–374. doi:10.3171/ acoustic neuromas. A new clinical entity. Arch
2009.10.JNS091203 Otolaryngol Head Neck Surg 118(2):127–133
314 5 Vestibular Schwannomas

249. Tos M, Thomsen J (1991) Management of acoustic neu- 261. Welling DB, Lasak JM, Akhmametyeva E,
romas. A review. Acta Otolaryngol 111(4):616–632 Ghaheri B, Chang LS (2002) cDNA microarray
250. Tran Ba Huy P, Kania R, Legac MS (2008) Vestibular analysis of vestibular schwannomas. Otol Neurotol
schwannoma (acoustic neurinoma). Natural history 23(5):736–748
and quality of life. Bull Acad Natl Med 192(9):1725– 262. Welling DB, Slater PW, Thomas RD, McGregor JM,
1737; discussion 1738–1740 Goodman JE (1999) The learning curve in vestibular
251. Trofatter JA, MacCollin MM, Rutter JL, Murrell JR, schwannoma surgery. Am J Otol 20(5):644–648
Duyao MP, Parry DM, Eldridge R, Kley N, Menon 263. Whitehouse K, Foroughi M, Shone G, Hatfield R (2009)
AG, Pulaski K et al (1993) A novel moesin-, ezrin-, Vestibular schwannomas – when should conservative
radixin-like gene is a candidate for the neurofi- management be reconsidered? Br J Neurosurg
bromatosis 2 tumor suppressor. Cell 72(5):791–800, 24(2):185–190. doi:10.3109/02688690903272634
0092-8674(93)90406-G [pii] 264. Wiet RJ, Mamikoglu B, Odom L, Hoistad DL (2001)
252. Umezu H, Aiba T, Tsuchida S, Seki Y (1996) Early Long-term results of the first 500 cases of acoustic
and late postoperative hearing preservation in neuroma surgery. Otolaryngol Head Neck Surg
patients with acoustic neuromas. Neurosurgery 124(6):645–651, S0194599801514382 [pii]
39(2):267–271; discussion 271–272 265. Yamakami I, Uchino Y, Kobayashi E, Saeki N,
253. Utermark T, Alekov A, Lerche H, Abramowski V, Yamaura A (2002) Prognostic significance of
Giovannini M, Hanemann CO (2003) Quinidine changes in the internal acoustic meatus caused by
impairs proliferation of neurofibromatosis type vestibular schwannoma. Neurol Med Chir (Tokyo)
2-deficient human malignant mesothelioma cells. 42(11):465–470; discussion 470–471
Cancer 97(8):1955–1962. doi:10.1002/cncr.11275 266. Yamakami I, Uchino Y, Kobayashi E, Yamaura A,
254. Valvassori GE, Shannon M (1991) Natural history of Oka N (2004) Removal of large acoustic neurinomas
acoustic neuromas. Skull Base Surg 1(3):165–167 (vestibular schwannomas) by the retrosigmoid
255. Wada K, Nawashiro H, Shimizu A, Shima K (2003) approach with no mortality and minimal morbidity. J
MRI analysis of hydrocephalus associated with acous- Neurol Neurosurg Psychiatry 75(3):453–458
tic neurinoma. Acta Neurochir Suppl 86:549–551 267. Yang I, Sughrue ME, Han SJ, Aranda D, Pitts LH,
256. Walsh RM, Bath AP, Bance ML, Keller A, Rutka JA Cheung SW, Parsa AT (2009) A comprehensive anal-
(2000) Consequences to hearing during the conser- ysis of hearing preservation after radiosurgery for
vative management of vestibular schwannomas. vestibular schwannoma. J Neurosurg. doi:10.3171/
Laryngoscope 110(2 Pt 1):250–255. doi:10.1097/ 2009.8.JNS0985
00005537-200002010-00012 268. Yasargil M (1996) Acoustic neuromas, vol IVb,
257. Walsh RM, Bath AP, Bance ML, Keller A, Tator CH, Microneurosurgery. Thieme, New York
Rutka JA (2000) The natural history of untreated 269. Yates PD, Jackler RK, Satar B, Pitts LH, Oghalai JS
vestibular schwannomas. Is there a role for conser- (2003) Is it worthwhile to attempt hearing preserva-
vative management? Rev Laryngol Otol Rhinol tion in larger acoustic neuromas? Otol Neurotol
(Bord) 121(1):21–26 24:460–464
258. Wanibuchi M, Fukushima T, McElveen JT Jr, 270. Yeung AH, Sughrue ME, Kane AJ, Tihan T, Cheung
Friedman AH (2009) Hearing preservation in sur- SW, Parsa AT (2009) Radiobiology of vestibular
gery for large vestibular schwannomas. J Neurosurg schwannomas: mechanisms of radioresistance and
111(4):845–854. doi:10.3171/2008.12.JNS08620 potential targets for therapeutic sensitization.
259. Watanabe K, Saito N, Taniguchi M, Kirino T, Neurosurg Focus 27(6):E2. doi:10.3171/2009.9.
Sasaki T (2003) Analysis of taste disturbance before FOCUS09185
and after surgery in patients with vestibular schwan- 271. Yoshimoto Y (2005) Systematic review of the natu-
noma. J Neurosurg 99(6):999–1003. doi:10.3171/ ral history of vestibular schwannoma. J Neurosurg
jns.2003.99.6.0999 103(1):59–63. doi:10.3171/jns.2005.103.1.0059
260. Welling DB, Guida M, Goll F, Pearl DK, 272. Zaouche S, Ionescu E, Dubreuil C, Ferber-Viart C
Glasscock ME, Pappas DG, Linthicum FH, (2005) Pre- and intraoperative predictive factors
Rogers D, Prior TW (1996) Mutational spectrum in of facial palsy in vestibular schwannoma surgery.
the neurofibromatosis type 2 gene in sporadic and Acta Otolaryngol 125(4):363–369. doi:10.1080/
familial schwannomas. Hum Genet 98(2):189–193 00016480410025216, K682N0KGGLRKMN0D [pii]
Neurofibromatosis Type 2
and Other Bilateral 6
Cerebellopontine Angle Tumors

Neurofibromatosis Type 2 NF-2 cases in patients, whose parents did not display
the disease, are mosaic [30]. If the mutation
NF-2 is a dominantly inherited tumor predispo- occurs early in embryogenesis, bilateral VSs may
sition syndrome characterized by a tendency for develop rendering the disease clinically indistin-
developing bilateral VSs and other CNS tumors guishable from nonmosaic forms. Mutation at
(meningiomas, ependymomas, astrocytomas, or a later stage of embryogenesis causes segmen-
other schwannomas). Typically the syndrome tal NF-2, characterized by unilateral VS and an
manifests itself during young adulthood—in the additional ipsilateral intracranial tumor, usually
second or third decade of life. The severity of the meningioma. This NF-2 subtype has understandably
disease ranges from mild to aggressive in regard to a very low inheritance risk [47].
age of onset, tumor types, and biological aggres-
siveness [50]. Traditionally NF-2 is subdivided
into two groups: the Wishart and the Gardner type Criteria for Diagnosis
[13]. The Wishart type is characterized by more
severe clinical presentation, earlier onset (in the Several different sets of diagnostic criteria for NF-2
late teens or early 20s), and occurrence of multiple are used currently [3, 13, 24, 49], but the modified
tumors. In addition to bilateral VSs, the patients NIH criteria and the Revised Clinical criteria are
frequently have multiple associated brain/spinal regarded as most sensitive (Tables 6.1 and 6.2).
tumors. Patients with Gardner-type NF-2 have later
onset, slower development, and less severe pre-
sentation. The incidence of associated intracranial Genetics
tumors is lower. The birth incidence of NF-2 was
estimated to vary between 1/25,000 and 1/87,410, NF-2, as well as sporadic VSs, is caused by
while the population-based disease prevalence is inactivation of the NF2 tumor suppressor gene
1/210,000 [13, 14]. Disease prevalence has now on chromosome 22q12 (see Chap. 5). NF-2 is
risen to around 1/60,000 due to earlier diagnosis caused by a single germline mutation of the
and longer survival of the patients [12]. chromosome band 22q12. The normal allele is
Recently a third subgroup—the mosaic lost due to a somatic mutation in the cells giv-
NF-2—was outlined, which is caused by a ing rise to the tumor. It has been estimated that
somatic mutation occurring during embryogen- NF-2 has the highest spontaneous mutation rate
esis [16]. Only a portion of the patients’ cells of any human genetic disorder. If a person
carry the mutation hence, and the germline DNA inherits the abnormal gene, there is a 95 %
is not affected [30, 61]. Approximately 25 % of chance that he will develop bilateral VSs.

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 315


DOI 10.1007/978-3-642-35422-9_6, © Springer-Verlag Berlin Heidelberg 2013
316 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Table 6.1 Modified NIH Main criteria Bilateral VS or


criteria for the diagnosis of Family history of NF-2 plus
NF2 [13]
1. Unilateral VS or
2. Any two of: meningioma, glioma, posterior subcapsular
lenticular opacities
Additional criteria Unilateral VS plus any two of meningioma, glioma,
neurofibroma, schwannoma, and posterior subscapular opacities
Or
Multiple meningioma (2 or more) plus
Unilateral VS or any two of glioma, neurofibroma, schwannoma,
and cataract

Table 6.2 NNFF criteria or revised clinical criteria for NF-2


Characteristics
A. Confirmed or definite 1. Bilateral vestibular schwannomas
NF-2 2. First-degree family relative with NF2 and unilateral VS at less than 30 years of
age or any two of meningioma, schwannoma, glioma, juvenile posterior
subcapsular lenticular opacities
B. Presumptive or probable 1. Unilateral vestibular schwannoma at less than 30 years of age and at least one of
NF-2 meningioma, schwannoma, glioma, juvenile lens opacity (posterior subcapsular
cataract or cortical cataract)
2. Multiple meningiomas (two or more) and unilateral vestibular schwannoma at
less than 30 years of age or at least one of schwannoma, glioma, juvenile lens
opacity (posterior subcapsular cataract or cortical cataract)
From Gutmann et al. [24]

Approximately 50 % of the patients represent Table 6.3 Clinical manifestation in NF-2 patients in the
new mutations that were not inherited. It is sup- largest published series
posed that the functional inactivation of the Clinical finding Rate (%)
NF2 gene is caused by two germline mutation Bilateral VS 71–95
(“double-hit” hypothesis). The penetrance of Spinal tumor 26–90
NF-2 is 100 %, but it has wide phenotypic vari- Intracranial meningioma 18–58
ability [16]. Practically all individuals who Peripheral nerve tumor 34
have pathogenic mutation develop the disease Skin tumors 32–68
in an average lifetime [13]. Unilateral VS 26
Café-au-lait macules 42–47
Intracranial astrocytoma 1.6–4.1
Ependymoma 2.5–6
Clinical Presentation
Nonvestibular cranial nerve 23
schwannoma
The mean age at onset of the disease is Presenile cataract 14–81
17–22 years [15, 44, 46, 54]. Clinical presenta- Cranial calcification 6
tion is highly variable and is related to tumor References: [15, 28, 44, 54]
types, size and extension, and more rarely to
the cutaneous or non-nervous-system-related Bilateral VSs are the hallmark of the disease
abnormalities. The clinical manifestation in and occur in about 95 % of adult patients with NF-2
NF-2 patients in the largest published series is (Fig. 6.1). Children with NF-2, however, often
presented on Table 6.3. present initially with nonvestibular tumors [45].
Neurofibromatosis Type 2 317

Fig. 6.1 MRI studies of patient with NF-2 and bilateral VSs. Note the characteristic multilobular structure of the VS
on the left side

The VS may reach large size before the onset of of a VS [13]. The likelihood of developing
audiological or vestibular symptoms. Compared NF-2 in young patients (<18 years) present-
to sporadic VSs, they tend to be more invasive, ing with isolated meningioma or VS is 20 and
have a higher degree of dividing cells, and are 10 %, respectively. This rate drops dramati-
frequently multifocal. A polycyclic tumor shape cally afterward and is very unlikely in patients
is more often observed. The growth rate of VSs >30 years [17].
is highly variable and remains unpredictable. It is Approximately half of all NF-2 patients develop
generally higher in younger patients, but great meningiomas, and the lifetime risk is estimated to
variations exist, both between different patients be as high as 75 % [13]—see Case Illustration 6.1.
and in the same patient over time [4, 43]. These meningiomas have a higher degree of divid-
Patients may present with cranial meningioma/s ing cells than sporadic meningiomas and are usu-
or with spinal tumor/s prior to the appearance ally fibroblastic [13].
318 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Case Illustration 6.1 recovery. Since several months, however, her


Multiple NF-2-Associated Meningiomas gait ataxia became more severe. The MRI
and Schwannoma showed significant enlargement of the right
This 21-year-old female patient presented after CP angle tumor.
multiple cranial and spinal surgeries at another At presentation the patient had right-sided
institution. She had been diagnosed of having slight facial weakness (HB grade II), bilat-
NF-2 seven years ago and has been operated eral hypacusis, more pronounced on the
for a parasagittal meningioma, craniocervi- right side (PTA of 70 dB vs. PTA of 15 dB
cal meningioma, and schwannoma of the C5 on the left side), slight weakness in both
nerve root on the right side. One year ago the arms (grade 4/5), and severe gait ataxia with
VS on the right side was partially removed. positive Romberg test. The MRI examina-
After the surgery she had transient swallowing tion (a–f) showed a large tumor in the right
difficulty and facial palsy with almost complete CP angle with characteristics compatible with

a b

c d

(a–f) MRI study of the patient with bilateral VSs and multiple supra- and infratentorial meningiomas (arrows).
(g–i) Follow-up MRI performed 12 months after the surgery
Neurofibromatosis Type 2 319

e f

g h

i
320 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

VS, extension grade T4b. The tumor com- accessible meningiomas, were removed com-
pressed severely and dislocated the brain stem. pletely. Although the facial nerve was pre-
A smaller tumor was seen in the left CP angle. served anatomically, it was very atrophic and
Several other contrast-enhancing lesions were reacted weakly when stimulated. After sur-
seen in the right CP angle, posterior cranial gery the patient had complete facial palsy
fossa, and supratentorially with imaging char- (HB grade 6/6), anacusis on the right side,
acteristics of meningiomas. Multiple spinal and dysphagia. The swallowing difficulty
extramedullary tumors (probably schwanno- proved to be transient and at discharge recov-
mas) were also visualized. ered almost completely. A tendency for
Considering the fact that the large right- improvement of the facial palsy was also
sided VS caused the current neurological noted. Two weeks later it was assessed as HB
deterioration, as well as the size of this tumor grade V. At 12 months follow-up, she could
and the absence of useful hearing on this side, close the eye and her face was symmetrical at
a decision was taken to remove it surgically. rest (HB grade III). The other tumors remained
A standard RS approach (re-craniectomy) asymptomatic and the MRI showed no growth
was performed, and the VS, as well as the tendency (g–i).
Neurofibromatosis Type 2 321

Spinal schwannomas arising from the dorsal General Management


spinal roots are very common findings—Fig. 6.2
[42]. Most of them are small and asymptomatic. A The treatment of patients with bilateral VSs is
dumbbell appearance is frequently seen (Fig. 6.2c). different from that of patients with sporadic
Radiologically these schwannomas cannot be dif- unilateral VSs. The lifelong tendency to forma-
ferentiated from neurofibromas arising from spi- tion of new CNS tumors predetermines the
nal roots in NF-1 patients [73]. Close follow-up is impossibility of definitive cure of NF-2 patients.
required because some schwannomas may enlarge Therefore, management is focused on life pro-
and cause compression of the spinal cord or may longation, preservation or rehabilitation of cra-
even compress adjacent organs (Fig. 6.2e). nial nerve function, and maintenance of quality
Peripheral schwannomas can arise from any of life [6, 63]. The treatment should be individ-
nerve; sensory nerves are, however, more fre- ualized and performed in specialized treatment
quently affected than motor nerves [13, 73]. The centers in close collaboration with the patient
clinical manifestation is caused by impaired and his family. Patient’s clinical presentation,
motor or sensory functions or by nerve irritation, attitude, and expectations, as well as the tumor
causing pain or paresthesias. Superficial periph- characteristics, should guide the decision-making
eral schwannomas present either as subcutaneous process. The tumor size and the function of the
nodules or as slightly raised well-circumscribed cochlear nerves on both sides are issues of par-
cutaneous lesions [15, 73]. Neurofibromas pre- amount importance.
senting as skin tumors are very rare. Management guidelines of NF-2-associated
The ependymomas and astrocytomas in NF-2 VSs are controversial in regard to timing and
are most commonly intramedullary spinal or type of treatment. The options are observation,
cauda equina tumors but may be also intracranial. radiosurgery, or surgery [50]. Several surgi-
Patients with NF-2, in contrast to those with cal strategies have been put forward: partial or
NF-1, rarely develop non-tumor-related neuro- total tumor removal aiming hearing preserva-
logic manifestations, such as peripheral neuropa- tion (via the retrosigmoid or the middle fossa
thy and hyporeflexia [22]. Seizures, epilepsy, and approaches), total removal of the tumor with
cognitive dysfunction are not typical. Visual placement of an auditory brain stem implant
impairment is common in the NF-2 population— (via the retrosigmoid or the translabyrinthine
up to 75 %—and is due to posterior subcapsular approaches), or decompression of the cochlear
lenticular opacities, retinal abnormalities, partic- nerve in the IAC. Some surgeons advocate sub-
ularly hamartomas, or epiretinal membrane [57]. total tumor removal in all cases. Regrowth of
Between 14 and 81 % of patients have presenile the residual tumor, however, is highly probable
cataracts, which rarely require active treatment. because of the typical young patients’ age and
The average actuarial survival from the time of the fact that VSs are typically rapidly growing in
diagnosis was estimated to be 15 years in the NF-2. Surgeries in case of recurrences are more
1970s–1980s but is constantly improving with earlier difficult and more dangerous; clear arachnoidal
diagnosis and better treatment [2, 13, 15]. According dissection plane to facial nerve is often absent.
to the study of Baser et al. [4], published in 2002, age Our treatment goal has always been total tumor
at diagnosis, intracranial meningiomas, and type of removal. The only exceptions have been made in
constitutional NF2 mutation are predictors of the order to preserve hearing function or facial nerve
relative risk of mortality. Another interesting finding integrity. Deliberate subtotal resections can be
was that patients who are treated in specialty centers performed for brain stem decompression and for
had a significantly lower risk of mortality than those hearing preservation in the last hearing ear (see
who were treated in nonspecialty centers. Case Illustration 6.2).
322 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

a b

c d

Fig. 6.2 Multiple spinal schwannomas (a, b) in a patient bilateral extraspinal component (c–e), extending in the
with NF-2 with bilateral VSs (c, arrowheads). The tumors upper thorax (e)
at the cervicothoracal junction level (arrows) have a major
Neurofibromatosis Type 2 323

Fig. 6.2 (continued)


324 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Case Illustration 6.2 and schwannomas with extracranial exten-


Subtotal Tumor Removal for Brain Stem sion, as well as multiple spinal tumors, were
Decompression and Hearing Preservation seen as well. Obviously, the clinical deterio-
in the Last Hearing Ear ration correlated mostly to the growth of the
This 19-year-old patient presented with NF-2 left-sided VS. Therefore, a decision was
and multiple intracranial and spinal tumors. taken to remove this tumor in an attempt to
He has been operated three times previously: prolong the life expectancy of the patient,
a large VS on the right side and a cervical according to his wish and the wish of his
schwannoma have been removed and a VP family. A tracheostomy was performed, and
shunt has been inserted. During the last then the tumor was removed completely via
2 years, he was followed with regular MRI the RS approach. The facial nerve was com-
examinations (a–d). At presentation he was pletely destroyed by the tumor, even in its
in poor general condition and was unable to petrous segment. All other structures could
walk independently. He had oculomotor dis- be preserved. The postoperative CT scans (j)
turbances, bilateral trigeminal hypoesthesia, showed small blood collections in the tumor
facial weakness (HB grade III) on the right bed area, which were clinically irrelevant and
side, complete bilateral hearing loss, lower gradually resorbed spontaneously (k). The
cranial nerve dysfunction, and quadriparetic patient had no new neurological deficits
syndrome. The MRI examination demon- besides the facial nerve palsy and was trans-
strated significant enlargement of the left- ferred to a rehabilitation center in a stable
sided VS when compared to the previous condition with improvement of gait and
images (e–i). Multiple cranial meningiomas quadriparesis.
Neurofibromatosis Type 2 325

a b

c d

(a–d) MRI of the head and spine, performed 10 months (j, k) Postoperative CT scans (day 1 after surgery):
prior to admission. (e–i) MRI examination of the patient, small blood collection in the tumor bed area is visible,
performed at our institute. Almost all tumors showed a which had no clinical significance and was managed
growth tendency. Note the significant enlargement of conservatively
the left-sided VS when compared to the initial images
326 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

e f

g h
Neurofibromatosis Type 2 327

j k
328 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Fig. 6.3 Very large bilateral VSs, extreme compression of the brain stem: the only management option is surgical
removal of both tumors

Large VSs Smaller VS, Useful Hearing

Symptomatic large tumors have to be removed Some surgeons advocate a more conservative
surgically in order to decompress the brain stem approach in such cases in order to preserve hear-
(Figs. 6.3, 6.4, and 6.5). An attempt to pre- ing. Surgery is postponed as long as possible: the
serve available hearing should always be made patients are followed with serial MRIs, and sub-
(Fig. 6.4). Alternatively, the tumor may be total intracapsular resection is done if the VS
removed partially, and the remaining part may be enlarges [49]. Surgery on the better hearing ear is
subjected to radiosurgery. Drawback of this lat- avoided, regardless of tumor size. With this man-
ter management concept, however, is the added agement strategy, surgery is performed only
morbidity risk of the two procedures. Therefore, when the tumors have reached a considerable
we do not recommend it. size, which significantly worsens the outcome.
Neurofibromatosis Type 2 329

a b

Preoperative audiogram

c R L
Frequency in kHz
01.5 0.25 0.5 1 1.5 2 3 4 5 8 10 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10

–10 –10
0 0
10 10
20 20
Hearing loss in dB HL

30 30
40 40
50 50
60 60
70 70
80 80
90 90
100 100
110 110

Speech discrimination in %
0 20 40 60 80 100 0 20 40 60 80 100
0 0

10 10

20 20
Sound pressure level in dB

30 30

40 40

50 50

60 60

70 70

Fig. 6.4 Patient with NF-2 and very large bilateral VSs. He follow-up examination was HB grade II on both sides. (a, b)
was operated in 1982: both tumors were removed and the Preoperative CT scans. (c) Preoperative audiogram. (d)
hearing was preserved. The facial nerve function at 12 months’ Postoperative CT scan. (e) Postoperative audiogram
330 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Postoperative audiogram

18 months after bilateral removal


e R Frequency in kHz
L

01.5 0.25 0.5 1 1.5 2 3 4 5 8 10 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10


–10 –10
0 0
10 10
Hearing loss in dB HL

20 20
30 30
40 40
50 50
60 60
70 70
80 80
90 90
100 100
110 110

Speech discrimination in %
0 20 40 60 80 100 0 20 40 60 80 100
0 0

10 10

20 20
Sound pressure level in dB

30 30

40 40

50 50

60 60

70 70

Fig. 6.4 (continued)


Neurofibromatosis Type 2 331

a b

c d

Fig. 6.5 Removal of a giant VS on the nonhearing side. The management decision was to remove the large tumor
Preoperative CT (a) and MRI (b–e) of a patient with NF-2. operatively and to follow the smaller one. The tumor
A giant VS on the left side and a smaller one on the right could be removed completely via the RS approach, and
side are visible. At presentation hearing was completely the facial nerve could be preserved. (f) Postoperative CT
lost on the left and was still functional on the right side.
332 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

e f

Fig. 6.5 (continued)

We accept an initial observational period in Surgery on the Only Hearing Side


selected cases: in older medically unstable
patients, in those with poor surgical risks, or in Management of VSs on the only hearing side (or
those who refuse surgery. We, as well as other “in the single ear”) is especially challenging. In
experienced surgeons, favor early proactive case of large tumors with compressive effect, sur-
treatment of NF-2 patients, based on the obser- gery is without alternatives. In smaller tumors,
vation that VSs can be removed and hearing however, the consideration is whether to treat
preserved [6, 63, 66, 67]. Our goal has always them actively early, accepting the risk of complete
been to preserve useful hearing for as long as hearing loss, or to observe the patient until hear-
possible. If the chances for useful hearing pres- ing deteriorates. With the later strategy, the patient
ervation are realistic, our recommendation is may enjoy hearing for a certain time. The draw-
for early surgery. Tumor extension, hearing back, however, is that sooner or later the hearing
level, and BAEP features, which are the main will be completely lost. Ultimately, the patient
predictors of successful hearing preservation, should take the decision on the type of treatment.
determine which side should be operated ini- We recommend early surgical treatment in
tially. The side with best chances for hearing those patients that have realistic chances for
preservation—smaller tumor and better hear- hearing preservation: useful hearing and good
ing level—is treated first. In case the VSs are quality of the preoperative BAEP. The goal of
of similar size, the side with better hearing surgery is to decompress the IAC and to remove
level is operated initially (Fig. 6.6). If the hear- the tumor. The extent of tumor removal, however,
ing and the tumor size are similar on both sides, is determined intraoperatively. As shown ear-
our decision is based on the preoperative BAEP. lier, BAEP monitoring is a highly sensitive test
The side with better BAEP quality is operated for the function of auditory pathways. Surgery
first because the chances of successful hearing proceeds as far as the BAEP remains stable. In
preservation are higher (Fig. 6.7). case that even slight microsurgical actions are
Neurofibromatosis Type 2 333

Fig. 6.6 Patient with relatively small VSs and good bilateral hearing. The tumor on the left side (better hearing level)
was removed initially

followed by severe deterioration of the auditory These tumors are generally very slow growing.
evoked responses, partial resection is performed. Only symptomatic tumors or those showing a
Applying this strategy, we could preserve hearing steady growth tendency should be treated (see
in 15 patients. Follow-up examinations indicated Case Illustration 6.2).
that preserved hearing remained functional for Radiosurgery is another treatment option.
periods of up to 15 years. If the BAEP is of low Some studies, however, showed that the outcomes
quality before surgery, with loss of waves or con- are not as good as in individuals with sporadic
siderable interval prolongation, this feedback is unilateral VS (see Case Illustration 6.3). The
not available. In such case, we would not recom- long-term tumor control rate ranges between 60
mend early surgery. and 81 % [41, 55, 60]. In a study of Rowe et al.
If the patient decides against surgery, the [60], 20 % of all patients required further treat-
tumor should be followed and removed when ment 8 years after radiosurgery, and in 30 % there
hearing is lost or the tumor progression leads was some concern about tumor control. Hearing
to serious neurological symptoms. The res- preservation in the published series is approxi-
toration of hearing with an auditory implant mately 33–43 %, although late deterioration is
should be planned and performed at the same observed in up to 40 % of the patients [41, 55,
surgery or in several weeks. Bilateral deafness 60]. As pointed by Brackmann et al. [6], hearing
of NF-2 patients is often inevitable, and an preservation rate and facial nerve function after
ABI implantation is the only option to provide radiosurgery are not superior to those follow-
some hearing sensation. ing microsurgery by experienced surgeons. The
Approximately 40 % of NF-2-associated VSs trigeminal nerve dysfunction rate is, however,
are multilobular, and the nerves and vessels may higher. Radiosurgery is therefore not the optimal
pass between the tumor lobules. The tumor can primary therapy in NF-2. It is best reserved for
infiltrate the fibers of individual nerves, which is those patients who have particularly aggressive
seldom the case in unilateral tumors. The man- tumors, those with medical contraindications for
agement of non-CP angle tumors associated with microsurgery, patients who refuse surgery, or for
NF-2 follows the general neurosurgical principles. the elderly [2].
334 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

01.5 0.25 0.5 1 1.5 2 3 4 5 8 10


a b–10
0
10
20
30
40
50
60
70
80
90
100
110

c 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10 d


–10
0
10
20
30
40
50
60
70
80
90
100
110

Fig. 6.7 This is the first patient with successfully second tumor was also removed, and the hearing and
removed by the senior author bilateral VSs and normal facial nerve function were preserved. (a) Preoperative CT.
postoperative facial nerve function and preserved useful (b, c) Preoperative audiogram (b, right side; c, left side).
hearing after surgery, done. She has been operated in (d) Postoperative photo of the patient, demonstrating the
1981. Initially the larger tumor on the side with worse normal facial nerve function on both sides. (e, f) Post-
hearing—the left side—was removed. In 3 months the operative audiogram
Neurofibromatosis Type 2 335

e 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10 f 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10

–10 –10
0 0
10 10
20 20
30 30
40 40
50 50
60 60
70 70
80 80
90 90
100 100
110 110

Fig. 6.7 (continued)


336 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Case Illustration 6.3 first time to the senior author, he had no hear-
Initial Follow-Up, Radiosurgery, and ing on both sides, bilateral trigeminal hypesthe-
Microsurgery sia, severe gait ataxia, and weakness of the right
This 18-year-old male patient was diagnosed extremities, as well as intracranial hypertension.
of having NF-2 with bilateral VSs in 1996 (a). The MRI study demonstrated that both VSs
Initially he was followed radiologically; the MRI have enlarged further (c, d). The brain stem was
examination in 1 year showed clear enlargement severely compressed, and the ventricles were
of both tumors (b). He had profound hearing loss dilated. A new intra-axial tumor was also visible
on the left side and still useful hearing on the right in the low medulla oblongata. The left-sided VS
side. Gamma knife treatment was performed in was removed initially; in 3 months the second
1997. One year later, when he presented for the VS was also removed successfully.

a b

c d

(a–d) Bilateral VSs that have been initially observed, then treated with radiosurgery, and then removed operatively.
See the text for details
Neurofibromatosis Type 2 337

Personal Series schwannomas) originated from the facial nerve


or no cleavage plane between the cranial nerves
Over a period of approximately 40 years, the and the tumor could be found (see Case Illustration
senior author has treated surgically 175 patients 6.4). In 205 cases (90 %) the facial nerve could
with NF-2. The total number of VS surgeries in be separated from the VS and its continuity pre-
these patients was 228. Complete tumor removal served. The overall rate of hearing preservation
rate was 85 %, while in 15 % deliberately subto- in the series was 35 %. If only patients with pre-
tal removal was performed for brain stem decom- served functional preoperative hearing are
pression and hearing preservation in the only included, however, the rate was 65 %. Bilateral
hearing ear. Facial nerve preservation was usu- hearing after surgery had 23 % of the patients.
ally possible unless the schwannoma (or multiple See Case Illustrations 6.5 and 6.6.
338 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Case Illustration 6.4 a


Siblings with Bilateral VSs
These two siblings with NF-2 presented to
the senior author in the early 1990s. Their
father had been operated for bilateral VSs
5 years earlier. The boy was 17 years old,
and although his VSs were of similar size,
hearing level was better on the left side
(a–c). The girl was 13 years old at presen-
tation. Her hearing was better on the side
of the larger VS (d–f). Both siblings were
admitted together and operated the same
day (g, h). See the text to the figures for
details.

Left side
May 1992 June 1992 June 1993
Preoperative Postoperative Postoperative
Frequency in kHz
b 01.5 0.25 0.5 11.5 2 3 4 5 8 10 01.5 0.25 0.5 11.5 2 3 4 5 8 10 01.5 0.25 0.5 11.5 2 3 4 5 810
–10 –10 –10
Hearing loss in dB HL

0 0 0
10 10 10
20 20 20
30 30 30
40 40 40
50 50 50
60 60 60
70 70 70
80 80 80
90 90 90
100 100 100
110 110 110
Speech discrimination in %
0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100
0 0 0
Sound pressure level in dB

10 10 10
20 20 20
30 30 30
40 40 40
50 50 50
60 60 60
70 70 70

Preoperative MRI demonstrating bilateral VSs in a 13-year-old sister was operated initially on the left side,
17-year-old boy (a). The tumor on the left side was and later on the right-sided VS was removed (d). Hearing
removed initially, and hearing could be preserved (b). could be preserved on both sides (e, f). (g, h) Postoperative
One year later the right-sided tumor was removed, the photos of the siblings, approximately 3 months follow-
facial nerve was preserved, but hearing was lost (c). The ing last surgery: normal bilateral facial nerve function
Neurofibromatosis Type 2 339

Right side
May 1993 May 1993
Preoperative Postoperative
Frequency in kHz
c 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10

–10 –10
0 0
10 10
20 20
Hearing loss in dB HL

30 30
40 40
50 50
60 60
70 70
80 80
90 90
100 100
110 110

d
340 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Left side
May 1992 June 1992 June 1993
Preoperative Postoperative Postoperative
Frequency in kHz
e 01.5 0.25 0.5 11.5 2 3 4 5 8 10 01.5 0.25 0.5 11.5 2 3 4 5 810 01.5 0.25 0.5 11.5 2 3 4 5 8 10
Hearing loss in dB HL

–10 –10 –10


0 0 0
10 10 10
20 20 20
30 30 30
40 40 40
50 50 50
60 60 60
70 70 70
80 80 80
90 90 90
100 100 100
110 110 110

Speech discrimination in %
0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100
0 0 0
Sound pressure level in dB

10 10 10
20 20 20
30 30 30
40 40 40
50 50 50
60 60 60
70 70 70
Neurofibromatosis Type 2 341

Right side
May 1993 May 1993
Preoperative Postoperative
Frequency in kHz
f 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10 01.5 0.25 0.5 1 1.5 2 3 4 5 8 10

–10 –10
0 0
10 10
Hearing loss in dB HL

20 20
30 30
40 40
50 50
60 60
70 70
80 80
90 90
100 100
110 110

Speech discrimination in %
0 20 40 60 80 100 0 20 40 60 80 100
0 0

10 10
Sound pressure level in dB

20 20

30 30

40 40

50 50

60 60

70 70
342 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

h
Neurofibromatosis Type 2 343

Table 6.4 Modified from Yohay [73]


Features NF-1 NF-2
Inheritance Autosomal dominant Autosomal dominant
Incidence 1:3,500–4,000 1:25,000–40,000
Chromosome abnormality 17q11 22q12
Gene product Neurofibromin Merlin
Typical clinical presentation/age Six or more café-au-lait macules/infancy Hearing loss or vestibular dysfunction/
or early childhood young adults
Nerve sheath tumors Neurofibroma, plexiform neurofibroma Schwannoma
Intracranial tumors Gliomas (optic gliomas, other low-grade Bilateral vestibular schwannomas,
astrocytomas/gliomas), ependymomas, meningiomas
meningiomas, and/or PNETs
Spinal tumors Nodular plexiform neurofibromas Schwannomas, ependymomas,
astrocytoma
Cognitive Learning disability, AHDH Normal
Other tumors Chronic myeloid leukemia, None
pheochromocytoma
Ophthalmologic findings Lisch nodules, congenital glaucoma Posterior subcapsular lenticular
opacities, presenile cataracts, retinal
hamartomas, or epiretinal membrane

Differential Diagnosis The neurological symptoms in patients with


Neurofibromatosis Type 1 NF-1 are caused by brain, spine, or peripheral
NF-2 is distinctly different disease from neuro- nerves tumors; epilepsy; macrocephaly; hydro-
fibromatosis type 1 (NF-1). NF-1 or the “von cephalus; meningoceles; and/or peripheral neu-
Recklinghausen disease” is an autosomal-domi- ropathy [72]. Typical finding is multiple peripheral
nant disorder and results from mutation of a tumor neurofibromas: either cutaneous neurofibromas,
suppressor gene mapped to chromosome 17: the subcutaneous neurofibromas, nodular plexiform
NF1 gene [32]. It occurs with an incidence of 1 neurofibromas, or diffuse plexiform neurofibromas.
per 3,500–4,000 live births. The diagnostic cri- Plexiform neurofibromas may undergo malig-
teria for NF1, formulated by the NIH Consensus nant transformation. A higher risk for developing
Development Conference on NF, include six intracranial tumors is observed in NF-1 patients,
or more café-au-lait macules, two or more compared to the general population. The tumor
neurofibromas of any type or 1 or more plexiform types, associated with NF-1 and NF-2, however,
neurofibroma, two or more Lisch nodules, distinct are different. NF-1 patients tend to have most fre-
osseous lesions, or first degree relative with NF-1. quently gliomas, ependymomas, meningiomas,
When two or more of these signs are present in and/or PNETs. Gliomas are typically low grade and
one patient, the diagnosis NF-1 is accepted. involve the optic pathways, hypothalamus, cerebel-
Patients with NF-1 have specific clinical pre- lum, brain stem, and spinal cord. Optic pathway
sentation, which is completely different from that gliomas generally have a more indolent natural
seen in case of NF-2 (see Table 6.4). Cutaneous, history and are less aggressive than optic gliomas
ophthalmological, musculoskeletal, and/or neuro- in patients without NF-1 [68]. They involve fre-
logical symptoms occur in varying combinations. quently the anterior visual pathways and occur in
The cutaneous and ocular manifestations— 11–19 % of NF-1 patients. Vascular complications,
typically café-au-lait macules, axillary freckling, such as carotid or middle cerebral artery occlusion,
and Lisch nodules—are the most common [72]. can occur [32]. NF-1 has a significant effect on
Ninety-nine percent of the patients have café-au-lait learning and cognition and causes frequently psy-
macules. chiatric disorders.
344 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Schwannomatosis MRI of the whole neuraxis, should be performed


Schwannomatosis is a recently defined entity, (NIH Consensus conference). All patients with
characterized by multiple spinal, peripheral nerve NF-2 and their families (relatives at risk) should
or subcutaneous schwannomas in the absence of have access to genetic testing because early, at
VSs [39, 64]. Schwannomatosis is segmental or best presymptomatic, diagnosis improves clinical
localized to a certain body part, such as a single outcome.
limb, several contiguous segments of the spine, Presently, prenatal diagnosis of pregnancies is
or one side of the body, in approximately 30 % of possible: the DNA of fetal cells obtained by
patients. Most schwannomas are caused by amniocentesis (15–18 weeks of gestation) or
somatic NF2 gene inactivation; less frequently chorionic villous sampling (10–12 weeks of ges-
mutations in the INI1/SMARCB1 gene are tation) can be examined [13]. Such test is recom-
responsible [25]. The disease is rarely familial. mended for those at 50 % risk for developing
Patients with schwannomatosis frequently pres- NF-2. A prerequisite for the test is the identification
ent with intractable pain rather than cranial nerve of the disease-causing allele of an affected family
deficits. Other intracranial tumors are not typical member or the establishing of a linkage in the
for this condition. family.
The offspring of patients with NF-2 should be
Multiple Meningiomas (See Chap. 7) followed and examined at regular intervals for
earliest possible detection of NF-2. Annual oph-
Follow-Up and Screening of NF-2 Patients thalmological examinations should start soon
Patients with known NF-2 should be followed after birth, while the annual neurological exami-
closely. In case no new symptoms/signs appear, nations should start at 7 or 10 years of age
annual audiological and ophthalmological exam- [18, 51]. Audiogram should be performed twice
inations, as well as MRI of the brain, are manda- per year. MRI studies of the brain are recom-
tory. Once the diagnosis of NF-2 is made, relatives mended annually or at 2 years interval starting at
who are at risk should be screened for the dis- 10–12 years of age. Spine MRI has to be per-
ease: a thorough dermatological, ophthalmologi- formed in case signs or symptoms suggestive of
cal, and neurological examinations, as well as myelopathy or radiculopathy appear.
Neurofibromatosis Type 2 345

Case Illustration 6.5 quickly and had only a slight facial weakness
Bilateral NF-2-Associated VSs (HB grade II)—figures d, e. He decided for
The patient presented with a history of hearing initial follow-up of the right-sided VS. Two
loss on the left side and dizziness since more years later hearing remained stable and the
than 2 years. Gradually his gait became unsta- tumor showed no major growth tendency.
ble and headache appeared. At admission he Afterward, however, the right-sided VS began
had anacusis on the left side and slightly to grow quickly, the patients’ condition dete-
reduced but useful hearing on the right side, riorated again, and hearing worsened.
trigeminal hypesthesia on the left side (V1–3), Furthermore, he complained of worsened gait
decreased gag reflex, and gait ataxia. MRI and very annoying tinnitus. At presentation he
revealed giant VS on the left side, with a maxi- had bilateral anacusis and gait ataxia, prohib-
mal diameter of approximately 5 cm, causing iting independent walking. The MRI study,
severe compression of the brain stem (a–c). No shown on figures f–i, showed that the tumor
signs of hydrocephalus were, however, visual- on the right side had reached a large size and
ized. The tumor on the right side was consider- caused compression of the brain stem. The
ably smaller—extension grade 3a. patient was therefore operated again: com-
The left-sided VS had to be removed at the plete removal of the tumor was achieved, and
first step for obvious reasons: giant size, severe the facial nerve was preserved. Figures j, k
compressive effect, no hearing on this side. show the CT 1 day after surgery with moder-
The surgery was extremely difficult: all neigh- ate pneumocephalus. The whole postoperative
boring neural structures were very vulnerable period was smooth, and the patient was dis-
due to the long-standing severe compression. charged with a slight (HB grade II facial weak-
The facial nerve was atrophic and splayed in ness on the right side) and improved balance.
an umbrella-like fashion over the anterior Implantation of an ABI was discussed with
tumor surface. Still, its anatomical integrity the family but was not plausible because the
could be preserved and complete tumor patient did not fulfill the criteria outlined pre-
removal achieved. The patient recovered viously in text.
346 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

a b

T1-weighted contrast-enhanced (a, b) and CISS (c) MRI the patient prior to discharge. Slight weakness of the
images of the patient, demonstrating a small VS on the inferior facial branch on the left side is noted (HB grade
right side and a giant one on the left side. The degree of II). MRI images before (f–i) the second surgery: the
brain stem compression and deformation of the fourth enlargement of the right-sided tumor is obvious. Note
ventricle, as well as the large intracanalicular tumor part, the fat plug in the left IAC and a small contrast-enhanc-
are obvious. (d) MRI, performed before discharge, ing structure lateral to it, which is suspicious for local
confirming the complete removal of the left VS. The recurrence. (f) T1-weighted native MRI. (g, h)
right-sided VS is visualized, as well as the fat in the left T1-weighted MRI after contrast administration. (i) CISS
IAC. Fat has characteristic signal on T1 and T2 and sequence. (j, k) CT 1 day after surgery with moderate
should not be interpreted as tumor residual. (e) Photo of pneumocephalus
Neurofibromatosis Type 2 347

d e

f g

h i
348 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

j k
Neurofibromatosis Type 2 349

Case Illustration 6.6 removed completely, and both the facial and ves-
Removal of Both VSs During the Same tibular nerves were preserved anatomically (e).
Hospital Stay and Hearing Preservation The patient recovered quickly and had only a
This patient suffered from a brain stem stroke slight facial weakness (HB grade II). The man-
at the age of 15 years. He recovered partially agement options of the tumor on the only hear-
and still had some phonation difficulty, right- ing side were discussed with the patient and
sided hearing loss, and hemiparesis. At the his family. Our recommendation was to per-
age of 24 years, he started complaining of form the surgery in 3–6 months—after the
vertigo and tinnitus on the left side. MRI was recovery from the initial surgery—because of
performed that showed bilateral CP angle the good chances of hearing preservation (good
tumors. At presentation he was 25 years old hearing level, good BAEP). The patient and his
and complained of hearing loss on the right family, however, insisted on having early sur-
side, swallowing difficulty, and vertigo. The gery during the same hospital stay. Taking into
audiogram revealed non-useful hearing on account the excellent postoperative condition
the right side (hearing loss of more than of the patient and the fact that he came from a
80 dB in the main speech area) and normal very distant country to our department, we
hearing on the left side (hearing loss of decided to perform the second surgery 2 weeks
15 dB). MRI showed bilateral VSs (a–d): on after the first one. Notably, the BAEP were
the right side the tumor was larger (grade IV stable throughout the surgery, which allowed
B, approximately 3.5 cm in its maximal us to proceed with tumor removal. The VS was
diameter) and polycyclic; the left tumor was removed completely and the cranial nerves
grade III B (reaching but not compressing the were preserved (e). Postoperative audiogram
brain stem). demonstrated preservation of the preoperative
A decision was taken to remove initially the hearing level on the left side (PTA of 15 dB)
larger tumor on the side with non-useful (h). Until discharge the patient’s facial weak-
hearing—the right-sided VS. The tumor was ness improved to HB grade II.
350 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

a b

c d

e f

Preoperative MRI demonstrating the bilateral VSs. (a–c) following the second surgery. Complete tumor removal
T1-weighted MRI with contrast enhancement (axial on both sides. The fat used to plug the IAC is well seen.
and sagittal). (c) CISS sequence. (e) CT of the head 1 Preoperative (g) and postoperative (h) audiograms—
day after the initial surgery. (f) MRI performed 3 days left ear
Neurofibromatosis Type 2 351

h
352 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Other Bilateral CP Angle Tumors extensive skull base or intra-axial tumors may
extend into both CP angles but per definition
Bilateral CP angle tumors are usually seen in should not be regarded as “bilateral CP angle
patients with NF-2 and are typically vestibu- tumors” (Fig. 6.10).
lar or facial nerves schwannomas or—infre- The diagnosis may be straightforward in the
quently—meningiomas [1]. Other bilateral CP presence of NF-2 disease or if the typical MR
angle lesions have been rarely reported: bilateral characteristics of schwannomas are seen. The
choroid plexus papillomas [8], bilateral endo- presence of multiple intracranial or extracranial
lymphatic sac tumors [29], bilateral primary tumors or of a primary focus, as well as rapid
central nervous system lymphoma [27], bilat- clinical deterioration, should arise the suspicion
eral arachnoid cysts [70], or bilateral metasta- of metastatic disease. In some cases, however,
ses (Fig. 6.8) [5, 10, 33, 56]. Infrequently fourth clinical examination and neuroradiological
ventricle tumors may extend into both CP angles images do not allow to ascertain preoperatively
via the foramina of Luschka (Fig. 6.9). Some the tumor type.

Fig. 6.8 Metastatic lesions in both cerebellar hemispheres with extension in the CP angles in a patient with a primary
pulmonary adenocarcinoma
Other Bilateral CP Angle Tumors 353

Fig. 6.9 MR images of a 62-year-old male patient with a The tumor was removed completely via a median suboc-
subependymoma of the fourth ventricle, which extends cipital craniotomy
into both CP angles (arrows) via the foramina of Luschka.
354 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Fig. 6.10 Extensive skull base meningioma with involvement of both CP angles
Other Bilateral CP Angle Tumors 355

Case Illustration 6.7 The standard RS approach was used and the
Bilateral Osteomas tumor, which had the features of an osteoma,
A 30-year-old female patient presented to our was seen attached to the anterosuperior lip of
department with a history of increasing ver- the IAC (e, f). It was anterior and superior to
tigo and tinnitus on the left side. Her neuro- the seventh-eighth nerve bundle, compressing
logical examination revealed no deficits. The and dislocating them posteriorly, although not
audiogram showed hearing loss of 25 dB in adherent to the lesion. The osteoma was
the main speech area on the left side and nor- removed completely with a diamant micro-
mal hearing on the right side. HRCT with drill, and the nerves were decompressed.
bone window showed hyperdense lesions with Histopathological examination proved the
bone density and irregular margins close to diagnosis of an osteoma.
both IAC (a, b). MRI showed a lesion in the The patient recovered rapidly from surgery,
area of the porus of the IAC, but the image and at discharge the vertigo and tinnitus were
characteristics were unspecific (c, d). completely resolved (g). Hearing even
Considering the size of the lesion and the pro- improved with a mean of 20 dB in the main
gressive nature of the symptoms, a decision speech area (h, i). Considering the small size
was made to remove the left-sided tumor, to of the contralateral lesion and the lack of any
verify it pathologically, and to decompress the related symptoms, we recommended regular
vestibulocochlear nerve. CT scan follow-up.
356 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

a c

b
d

(a, b) Preoperative bone window CT showing the the left porus acusticus internus. (f) The osteoma has
lesions attached to both IAC (arrowheads). The left been completely removed. The seventh-eighth nerves
tumor has a maximal diameter of 1.2 cm and the right have been decompressed. Postoperative bone window
one, 0.3 cm. The diagnosis of osteomas relies entirely CT. (g) Preoperative and (h) postoperative audio-
on CT scanning, which demonstrates well-demarcated, grams—improvement of hearing following the
dense bone outgrowth. (c, d) MR imaging: the lesion cochlear nerve decompression (Reprinted from
in the area of the porus has unspecific characteristics. Gerganov et al. [21]. Copyright © 2012, with permis-
(e, f) Intraoperative images. (e) Osteoma attached to sion from Wolters Kluwer Health)
Other Bilateral CP Angle Tumors 357

e f

g
358 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

h
Other Bilateral CP Angle Tumors 359

Case Illustration 6.8 left side, no further neurological deficits devel-


Melanoma Metastases oped. The final histopathological examination
This 46-year-old woman presented with a his- confirmed the lesion to be a lightly pigmented
tory of a Clark Level IV cutaneous malignant epithelioid malignant melanoma.
melanoma of the right calf and involvement of Surgery on the right side was postponed in
the inguinal lymphatic nodes. Her primary favor of palliative radiotherapy of the entire
tumor and the metastasis have been removed. neural axis. The patient decided, however, to
Subsequently alpha-interferon therapy was continue with the interferon therapy and to
instituted. One year later she began to com- undergo adjunctive chemotherapy instead of
plain of dizziness and vomiting and later on of radiation treatment. Follow-up MR imaging
acute hearing loss and tinnitus in the left ear. showed that the intracranial and spinal lesions
CT and MR examinations were performed, remained constant in size. Twenty-four months
showing bilateral enhancing intra- and extra- later the patient presented again with progres-
canalicular lesions (a–e). sive hearing loss on the right side. MRI demon-
Neurological examination revealed hearing strated slight enlargement of both intracanalicular
loss on the left side (PTA of 50 dB). No other tumors. The right tumor was approached via the
somatic metastases were detected. MR study retrosigmoid route, and a similar tumor in the
of the spine, however, showed further tumors: right IAC was detected. It could be removed
at the level of T-12 and L-3 spinal nerve roots. completely because a good dissection plane to
The differential diagnosis was therefore NF-2 the cranial nerves was found (g–m). The post-
and melanoma metastases. operative MR images are presented at figures n,
A decision for surgery was taken with the o. After surgery the patient had normal facial
goal of obtaining histopathological diagnosis nerve function and preserved useful hearing.
and removing the tumor. The left-sided lesion The histopathological diagnosis was lightly pig-
was operated initially due to the hearing loss mented epithelioid malignant melanoma.
on this side. At surgery a black-colored tumor, Summary: The presence of a primary malig-
slightly protruding from the IAC, was seen. nant melanoma with regional metastases, the
The IAC was widely opened thus exposing the patient’s relatively advanced age for an initial
whole tumor, which infiltrated the facial and diagnosis of NF-2, and the absence of family
cochlear nerves. Frozen section showed that history of NF-2 suggested that the intracranial
the lesion was a malignant melanoma. Taking lesions could be metastases. The clinical
into account this diagnosis and the extensive course, however, did not give us clues to the
infiltration of the facial nerve, partial tumor diagnosis. The finding of bilateral contrast-
resection was performed (e, f). enhancing intracanalicular lesions, along with
The postoperative period was uneventful. two spinal nerve root lesions, argued for the
Besides the complete loss of hearing on the diagnosis of NF-2.
360 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

a b

c d

Bilateral contrast-enhancing lesion in the IAC; the left (g–m) Intraoperative images. (g) Initial view of the CP
one has a maximal diameter of 1.2 cm and the right angle and of the tumor (tu). (h–j) Exposure of the
one, 1.3 cm. (a, b) T1-weighted sequence after gado- intracanalicular tumor part. (k, l) Dissection from the
linium administration. (c, d) CISS sequence. (e, f) facial and vestibulocochlear nerves. (m) Complete
MRI after the partial resection of the left-sided tumor tumor removal. (n, o) MRI after the complete removal
(T1-weighted sequence after gadolinium administration). of the right-sided tumor
Other Bilateral CP Angle Tumors 361

e f

g h
362 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

i j

k l
Other Bilateral CP Angle Tumors 363

n o
364 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

Hearing Rehabilitation in patients who develop VS in the only hearing


ear, the other having been deaf from birth [58].
A central auditory prosthesis that bypasses the
cochlea and the eighth nerve is the only option Prosthesis
for restoration of some hearing sensations in The device used initially by House and
patients with bilateral loss of integrity and con- Hitselberger was a ball electrode ABI [52].
ductivity of the cochlear nerve. Auditory brain Multichannel devices replaced this early proto-
stem implants (ABI) were designed to stimulate type single-channel device in 1992. Currently
directly the second-order neurons and bypass several multichannel devices, sharing similar
both the cochlea and cochlear nerve. The cochlear basic constructional principles, are available [9,
nucleus complex is stimulated directly by placing 26, 71]. The device consists of an implanted com-
the ABI electrode into the lateral recess of the ponent and an external component. The implanted
fourth ventricle. ABI was applied for the first receiver–stimulator is attached to an electrode
time in 1979 by House and Hitselberger in the array; the external component consists of a micro-
House Ear Clinic in a patient with NF-2 after the phone, speech processor, and a transcutaneous
removal of her second VS [7, 52]. It has been transmitter coil (Figs. 6.11 and 6.12).
estimated that up to year 2008, more than 600 Sound is transformed to an analog electrical
patients worldwide have received the ABI, most signal by a microphone. The electrical signal is
of whom with NF-2 [9]. modified by a speech processor afterward and
delivered to the set of electrodes (electrode array
Indications composed of 21 platinum disks) mounted on a
Implantation of the ABI is only the first step of a silicone and Dacron mesh carrier. These elec-
complex auditory rehabilitation program. The trodes stimulate the cochlear nucleus and evoke
ultimate success is determined to a certain extent responses that can be interpreted by the brain as
by the capability and motivation of the patient to sound [31].
complete this rehabilitation. Therefore, strict cri-
teria for patient selection have been introduced in Technique
regard to age, diagnosis, cognitive level, and gen- Accurate placement of the prosthesis is essential to
eral condition. achieve good functional outcome and avoid com-
The criteria accepted in Hannover [36] are: plications. The target for placement of the ABI
• Diagnosis of NF-2 with bilateral retrocochlear electrode array is the cochlear nucleus complex,
deafness caused by tumor growth or surgical composed of the dorsal and ventral cochlear nuclei.
tumor removal These nuclei cannot be directly visualized on the
• Absence of actual signs of serious disease brain stem surface and must be located using sev-
progression eral anatomical landmarks. In case of large VSs
• Age at least 18 years causing significant compression of the brain stem,
• Stable psychosocial condition their identification may be challenging. The ABI
• Realistic preoperative expectations is implanted either via the RS approach or via the
• Acceptable lip-reading ability translabyrinthine route [9, 40]. The RS approach
• Motivation to take part in the postimplantation has the advantage of providing wide exposure of
rehabilitation program the CP angle and allowing for removal of any
The indications for ABI implantation have additional existing tumors, as frequently observed
been expanded to include non-NF-2 patients with in NF-2 (Figs. 6.13 and 6.14). From a retrosig-
bilateral profound hearing loss and impossibility moid perspective, the entry of the cochlear nerve
of cochlear implantation, such as ossification or into the brain stem is obscured by the cerebellar
major cochlear malformations, aplasia, or avul- flocculus and covered by the choroid plexus [9].
sion of the cochlear nerves [9, 23]. A recently The plexus protrudes from the foramen of
published report describes the application of ABI Luschka, which is found between the roots of the
Other Bilateral CP Angle Tumors 365

a c

b d

Fig. 6.11 One of the first ABIs implanted by the senior tumor. (b) Skin incision. (c) Initial view of the tumor. (d)
author. (a) Initial CT demonstrating both VSs. The tumor The schwannomas have been removed completely. (e, f)
on the left side was removed initially. The implantation of Implantation of the external device. (g) Postoperative CT,
the ABI was planned after the removal of the right-sided which shows the correct position of the electrode
366 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

e f

Fig. 6.11 (continued)


Other Bilateral CP Angle Tumors 367

a b

Fig. 6.12 Implantation of a magnetless ABI. (a) Planning (c) View after the reconstruction of the bone with methyl
of the skin incision and site of device implantation. methacrylate and fixation of the device
(b) Insertion of the electrode after the tumor removal.
368 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

located immediately adjacent to the dorsal cochlear


nucleus and the posterior tip of the ventral cochlear
nucleus [36]. Telemetry and electrical BAEP are
used to optimize the electrode position [9]. The
receiver/stimulator is placed in a bony well, drilled
in the temporoparietal area—superior and anterior
to the craniotomy—and fixed.
Initially, the ABI was placed in patients with
bilateral VSs at the second VS surgery. Some
authors advocate device placement during sur-
gery for the first tumor, even though the patient
has useful hearing in the opposite ear. The theo-
retical advantage of early implantation is that it
allows the patient to learn to use the device before
complete loss of hearing occurs in both ears.
Furthermore, if the initial device fails to provide
Fig. 6.13 MRI of an NF-2 patient with a right-sided VS. useful auditory sensation, it can be implanted on
The larger left-sided VS has been removed earlier (note the second side after tumor removal. The benefit
the fat plug in the right IAC area). Implantation of an ABI of this approach, however, remains unproven.
was planned after the removal of the right-sided tumor
Complications
The specific complications, related to ABI place-
facial and glossopharyngeal nerves. With a slight ment, include electrode migration, non-auditory
caudal-to-cranial retraction of the cerebellum, these side effects, and CSF leaks. Placement of the
structures can be seen. In case a stump of the electrode array deep into the lateral recess can
cochlear nerve remains after VS removal, it may be prevent its migration, which usually occurs as a
a useful landmark for the lateral recess of the fourth result of unstable positioning or changes of the
ventricle. The lateral recess can be palpated and brain stem after VS removal.
opened with a microhook. If adhesions are found, If the electrode is positioned too low, the
they should be dissected. The observation of CSF glossopharyngeal nerve can be stimulated, result-
egress through the foramen of Luschka during a ing in a sense of tingling or constriction in the
Valsalva maneuver is another helpful clue. The use throat. If it is placed too high, the facial nerve
of an angled endoscope may be helpful to control could be stimulated and cause facial twitching.
the placement of the ABI electrode [20]. These non-auditory side effects can be avoided or
The placement of the electrode completely at least reduced by switching reference elec-
within the lateral recess provides optimal contact trodes, increasing the duration of the stimulus
to the nuclei. Furthermore, this aids in the stabili- pulse or turning off the more medial or lateral
zation of the electrode [19, 65]. The array is thus electrodes. Previously turned off electrodes can

Fig. 6.14 Removal of a right-sided VS and implantation the last part of the tumor from the facial nerve. (e) The posi-
of an ABI—intraoperative images. (a) Initial view of the tion of the retractor is changed, and the cerebellum is
tumor from an RS perspective. (b) The IAC has been retracted slightly in a caudal-to-cranial direction, thus
opened, and the intracanalicular part of the tumor has been exposing the flocculus (double arrows) and the choroid
removed, exposing the facial nerve. (c) Tumor debulking plexus (single arrow). (f) A microhook is used to open the
with the ultrasonic aspirator. (d) Dissection and removal of lateral recess. (g) The electrode of the ABI has been placed
Other Bilateral CP Angle Tumors 369

a b

c d

e f
370 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

g the impact of brain stem compression or distor-


tion caused by large VSs, and the presence of
other tumors, common in NF-2 patients. Another
unsolved problem is related to the more complex
processing of afferent signals at the level of
second-order auditory pathway neurons [35].
The performance of patients with ABIs is
comparable to that of patients with earlier single-
channel cochlear implants. Most patients acquire
environmental-sound awareness and understand-
ing of closed-set words, consonants, and vowels.
Ebinger et al. [11] report that 85 % of patients
undergoing implantation receive auditory sensa-
tions. Combined with lip-reading cues, 93 % of
patients demonstrate improved sentence under-
standing. Open-set speech recognition, however,
Fig. 6.14 (continued) remains an exception.
In our series (Samii and T. Lenarz), more than
50 % of the patients developed lip-reading
be reactivated later because with time some adap- enhancement in the first 3 months after device
tation occurs and the severity of the non-auditory activation [36]. Optimal lip-reading enhancement
sensations often decreases [36]. was mostly observed 6 months after device acti-
CSF leak along the electrode lead is avoided vation. Regarding the speech tracking and
by accurately sealing the dura opening with fat Freiburger numbers test, the plateau effect was
pieces and fibrin glue. less prominent. Sound recognition and speech
perception generally continue to improve for up
Rehabilitation to 8 years following implantation. oto
The device is activated usually 4–8 weeks after Lenarz et al. [35] reported on the patients’
implantation. Rehabilitation program starts after assessment of their hearing and communication
the first fitting with a basic training period of about abilities with the help of ABI. All patients profited
2 weeks consisting of intensive speech and hearing from the ABI: due to the increased environmental
therapy program every day [35, 36]. Thereafter, awareness, certain dangers could be avoided and
refitting and reevaluation of the auditory perfor- the patients’ self-confidence increased. Most of
mance is performed initially at 3-month intervals them distinguished not merely speech from envi-
and then at 6-month intervals for 3 years, provided ronmental sounds but could perceive difference
the patient’s hearing condition remains stable. between various voice qualities and differentiate
environmental sounds.
Results
Patients should be informed and comprehend that New Developments
the ABI does not provide a normal quality of Penetrating Electrode ABI
sound. The level of performance and the quality The cochlear nucleus has a highly complex struc-
of sound from the ABI are variable and do not ture and contains several distinct tonotopic repre-
reach the level obtained by the latest multichannel sentations composed of functionally different cell
cochlear implants. The reasons for these are not types. They are organized perpendicular to the
completely elucidated yet. Major contributing surface [48]. Hence, the two-dimensional elec-
factors are the unfavorable tonotopic organization trode array, which is designed for surface stimu-
of the cochlear nuclei, the distance from the sur- lation, makes poor connection with all of these
face of the fourth ventricle to the cochlear nuclei, structural layers [31, 62].
References 371

A new generation of ABI with penetrating the implant location. Open-set speech perception
microelectrodes—the penetrating electrode ABI without lip-reading cues could not be achieved.
(or PABI)—was developed to achieve a better
alignment along the tonotopic gradient of the Cochlear Implants
cochlear nucleus [19, 53]. The PABI uses 8 or 10 Cochlear implantation may be an alternative in
penetrating microelectrodes in conjunction with case the cochlear nerve is preserved anatomically
a separate array of 10 or 12 surface electrodes. and has maintained a positive electric stimulation
A recent review of the initial experience with ten response. As pointed by Roehm et al. [59], they
patients showed, however, that a major difficulty bypass the external auditory canal, ossicles, and
is the reliable placement of the penetrating elec- inner-ear hair cells and stimulate directly the
trodes into cochlear nucleus. Due to the individ- cochlear nerve within the cochlea. Some literature
ual anatomic variability and the tumor-induced data indicate that such cochlear implant patients
anatomical distortions, only 22 of 90 penetrating performed better than auditory brain stem implant
electrodes produced auditory sensations. patients and might even achieve open-set speech
The speech perception level of the patients could communication [69]. In the recently published
not be improved [53]. Still, the study demonstrated study [59], the authors found that cochlear implants
that the procedure is safe and that with the penetrat- offer superior auditory outcomes in patients with
ing microelectrodes auditory sensations are achieved retained cochlear nerves and positive results on
at lower levels of electrical stimulation than with promontory stimulation testing. The experience is
surface electrodes; the pitch range was increased. though still very limited, and any conclusions
should be evaluated critically.
Auditory Midbrain Implant
The limited success of the ABI in NF-2 patients
may be associated with tumor-induced alterations
at the level of the cochlear nucleus. Stimulation at a References
more proximal to the damage level may be a rea-
1. Baldwin D, King TT, Chevretton E, Morrison AW
sonable alternative [34]. The inferior colliculus, in (1991) Bilateral cerebellopontine angle tumors in
particular the inferior colliculus central nucleus, is neurofibromatosis type 2. J Neurosurg 74(6):910–915.
a promising site for a central auditory prosthesis in doi:10.3171/jns.1991.74.6.0910
2. Baser ME, Evans DGR, Gutmann DH (2003) Neuro-
NF-2 patients because it provides access to neural
fibromatosis 2. Curr Opin Neurol 16(1):27–33.
projections necessary for speech perception [34, 38]. doi:10.1097/01.wco.0000053583.70044.ab
The auditory midbrain implant (AMI) is a new 3. Baser ME, Friedman JM, Joe H, Shenton A, Wallace
hearing prosthesis designed for stimulation of AJ, Ramsden RT, Evans DG (2011) Empirical develop-
ment of improved diagnostic criteria for
this nucleus. An initial clinical trial included five
neurofibromatosis 2. Genet Med 13(6):576–581.
patients, in which a single shank AMI array (20 doi:10.1097/GIM.0b013e318211faa9
electrodes) was implanted. A modified lateral 4. Baser ME, Makariou EV, Parry DM (2002) Predictors
suboccipital approach was developed to ensure of vestibular schwannoma growth in patients with
neurofibromatosis type 2. J Neurosurg 96(2):217–222.
safe removal of large VS and to provide adequate
doi:10.3171/jns.2002.96.2.0217
exposure of the inferior colliculus for ideal AMI 5. Bono P, Bartoloni Saint Omer F (1994) A rare case of
implantation in one stage [62]. bilateral metastasis in the cerebellopontine angle.
The preliminary results have been presented Pathologica 86(3):324–328
6. Brackmann DE, Fayad JN, Slattery WH 3rd,
by Lim et al. [37]. They were interpreted as
Friedman RA, Day JD, Hitselberger WE, Owens RM
encouraging in terms of the safety and function- (2001) Early proactive management of vestibular
ality of the implant. Similar to the PABI, a major schwannomas in neurofibromatosis type 2. Neuro-
problem remains the optimal position of the elec- surgery 49(2):274–280; discussion 280–283
7. Brackmann DE, Hitselberger WE, Nelson RA, Moore J,
trodes. Although all patients obtained improve-
Waring MD, Portillo F, Shannon RV, Telischi FF (1993)
ments in hearing capabilities, the performance Auditory brainstem implant: I. Issues in surgical implan-
varied dramatically across patients depending on tation. Otolaryngol Head Neck Surg 108(6):624–633
372 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

8. Casar Borota O, Jacobsen EA, Scheie D (2006) 22. Gijtenbeek JM, Gabreels-Festen AA, Lammens M,
Bilateral atypical choroid plexus papillomas in cere- Zwarts MJ, van Engelen BG (2001) Mononeuropathy
bellopontine angles mimicking neurofibromatosis 2. multiplex as the initial manifestation of
Acta Neuropathol 111(5):500–502. doi:10.1007/ neurofibromatosis type 2. Neurology 56(12):
s00401-006-0067-7 1766–1768
9. Colletti V, Shannon RV, Carner M, Veronese S, 23. Grayeli AB, Bouccara D, Kalamarides M, Ambert-
Colletti L (2009) Progress in restoration of hearing Dahan E, Coudert C, Cyna-Gorse F, Sollmann WP,
with the auditory brainstem implant. Prog Brain Res Rey A, Sterkers O (2003) Auditory brainstem implant
175:333–345. doi:10.1016/S0079-6123(09)17523-4, in bilateral and completely ossified cochleae. Otol
S0079-6123(09)17523-4 [pii] Neurotol 24(1):79–82
10. Delerue O, Destee A, Devos P (1991) Bilateral 24. Gutmann DH, Aylsworth A, Carey JC, Korf B, Marks
metastases in the cerebellopontine angle. J Neurol J, Pyeritz RE, Rubenstein A, Viskochil D (1997) The
Neurosurg Psychiatry 54(6):562–563 diagnostic evaluation and multidisciplinary manage-
11. Ebinger K, Otto S, Arcaroli J, Staller S, Arndt P ment of neurofibromatosis 1 and neurofibromatosis 2.
(2000) Multichannel auditory brainstem implant: US JAMA 278(1):51–57
clinical trial results. J Laryngol Otol Suppl 27:50–53 25. Hanemann CO, Evans DG (2006) News on the genet-
12. Evans DG (2009) Neurofibromatosis 2 [Bilateral ics, epidemiology, medical care and translational
acoustic neurofibromatosis, central neurofibromatosis, research of Schwannomas. J Neurol 253(12):1533–
NF2, neurofibromatosis type II]. Genet Med 1541. doi:10.1007/s00415-006-0347-0
11(9):599–610. doi:10.1097/GIM.0b013e3181ac9a27 26. Jackson KB, Mark G, Helms J, Mueller J, Behr R
13. Evans DG (2009) Neurofibromatosis type 2 (NF2): a (2002) An auditory brainstem implant system. Am J
clinical and molecular review. Orphanet J Rare Dis 4:16. Audiol 11(2):128–133
doi:10.1186/1750-1172-4-16, 1750-1172-4-16 [pii] 27. Jaiswal AK, Chandra PS (2001) Cerebellopontine
14. Evans DG, Baser ME, O’Reilly B, Rowe J, Gleeson M, angle meningioma with acquired chiari and syringo-
Saeed S, King A, Huson SM, Kerr R, Thomas N, myelia: neuroimage. Neurol India 49(3):323
Irving R, MacFarlane R, Ferner R, McLeod R, Moffat 28. Kanter WR, Eldridge R, Fabricant R, Allen JC,
D, Ramsden R (2005) Management of the patient Koerber T (1980) Central neurofibromatosis with
and family with neurofibromatosis 2: a consensus bilateral acoustic neuroma: genetic, clinical and bio-
conference statement. Br J Neurosurg 19(1):5–12. chemical distinctions from peripheral
doi:10.1080/02688690500081206, neurofibromatosis. Neurology 30(8):851–859
M877P25502X236M6 [pii] 29. Kempermann G, Neumann HP, Scheremet R, Volk B,
15. Evans DG, Huson SM, Donnai D, Neary W, Blair V, Mann W, Gilsbach J, Laszig R (1996) Deafness due to
Newton V, Harris R (1992) A clinical study of type 2 bilateral endolymphatic sac tumours in a case of von
neurofibromatosis. Q J Med 84(304):603–618 Hippel-Lindau syndrome. J Neurol Neurosurg
16. Evans DG, Huson SM, Donnai D, Neary W, Blair V, Psychiatry 61(3):318–320
Teare D, Newton V, Strachan T, Ramsden R, Harris R 30. Kluwe L, Mautner V, Heinrich B, Dezube R,
(1992) A genetic study of type 2 neurofibromatosis in Jacoby LB, Friedrich RE, MacCollin M (2003)
the United Kingdom. I. Prevalence, mutation rate, Molecular study of frequency of mosaicism in
fitness, and confirmation of maternal transmission neurofibromatosis 2 patients with bilateral vestibular
effect on severity. J Med Genet 29(12):841–846 schwannomas. J Med Genet 40(2):109–114
17. Evans DG, Ramsden RT, Gokhale C, Bowers N, 31. Kuchta J, Otto SR, Shannon RV, Hitselberger WE,
Huson SM, Wallace A (2007) Should NF2 mutation Brackmann DE (2004) The multichannel auditory
screening be undertaken in patients with an appar- brainstem implant: how many electrodes make
ently isolated vestibular schwannoma? Clin Genet sense? J Neurosurg 100(1):16–23. doi:10.3171/jns.
71(4):354–358. doi:10.1111/j.1399-0004.2007. 2004.100.1.0016
00778.x, CGE778 [pii] 32. Lee MJ, Stephenson DA (2007) Recent developments
18. Evans DG, Sainio M, Baser ME (2000) Neuro- in neurofibromatosis type 1. Curr Opin Neurol
fibromatosis type 2. J Med Genet 37(12):897–904 20(2):135–141.doi:10.1097/WCO.0b013e3280895da8,
19. Fayad JN, Otto SR, Brackmann DE (2006) Auditory 00019052-200704000-00004 [pii]
brainstem implants: surgical aspects. Adv Otorhino- 33. Lee WT, Weber PC (2005) Melanoma metastasis mas-
laryngol 64:144–153. doi:10.1159/000094649, 94649 querading as bilateral acoustic neuromas. Otolaryngol
[pii] Head Neck Surg 132(3):505–506. doi:10.1016/j.
20. Friedland DR, Wackym PA (1999) Evaluation of sur- otohns.2004.05.004, S0194599804005546 [pii]
gical approaches to endoscopic auditory brainstem 34. Lenarz M, Lim HH, Lenarz T, Reich U, Marquardt N,
implantation. Laryngoscope 109(2 Pt 1):175–180 Klingberg MN, Paasche G, Reuter G, Stan AC (2007)
21. Gerganov VM, Samii A, Paterno V, Stan AC, Samii M Auditory midbrain implant: histomorphologic effects
(2008) Bilateral osteomas arising from the internal of long-term implantation and electric stimulation
auditory canal: case report. Neurosurgery 62(2): of a new deep brain stimulation array. Otol
E528–E529 Neurotol 28(8):1045–1052. doi:10.1097/MAO.
References 373

0b013e318159e74f, 00129492-200712000-00013 47. Mohyuddin A, Neary WJ, Wallace A, Wu CL,


[pii] Purcell S, Reid H, Ramsden RT, Read A, Black G,
35. Lenarz M, Matthies C, Lesinski-Schiedat A, Frohne C, Evans DG (2002) Molecular genetic analysis of the
Rost U, Illg A, Battmer RD, Samii M, Lenarz T (2002) NF2 gene in young patients with unilateral vestibular
Auditory brainstem implant part II: subjective assess- schwannomas. J Med Genet 39(5):315–322
ment of functional outcome. Otol Neurotol 23(5): 48. Moore JK, Osen KK (1979) The cochlear nuclei in
694–697 man. Am J Anat 154(3):393–418. doi:10.1002/
36. Lenarz T, Moshrefi M, Matthies C, Frohne C, aja.1001540306
Lesinski-Schiedat A, Illg A, Rost U, Battmer RD, 49. (1994) National Institutes of Health Consensus
Samii M (2001) Auditory brainstem implant: part I. Development Conference Statement on Acoustic
Auditory performance and its evolution over time. Neuroma, December 11–13, 1991. The Consensus
Otol Neurotol 22(6):823–833 Development Panel. Arch Neurol 51(2):201–207
37. Lim HH, Lenarz M, Lenarz T (2009) Auditory 50. Neff BA, Welling DB (2005) Current concepts in
midbrain implant: a review. Trends Amplif 13(3): the evaluation and treatment of neurofibromatosis
149–180. doi:10.1177/1084713809348372, 13/3/149 type II. Otolaryngol Clin North Am 38(4):671–684.
[pii] doi:10.1016/j.otc.2005.01.002, ix. S0030-6665(05)
38. Lim HH, Lenarz T, Joseph G, Battmer RD, Samii A, 00003-4 [pii]
Samii M, Patrick JF, Lenarz M (2007) Electrical stim- 51. Neff BA, Welling DB, Akhmametyeva E, Chang LS
ulation of the midbrain for hearing restoration: insight (2006) The molecular biology of vestibular schwan-
into the functional organization of the human central nomas: dissecting the pathogenic process at the
auditory system. J Neurosci 27(49):13541–13551. molecular level. Otol Neurotol 27(2):197–208.
d o i : 1 0 . 1 5 2 3 / J N E U RO S C I . 3 1 2 3 - 0 7 . 2 0 0 7 , doi:10.1097/01.mao.0000180484.24242.54,
27/49/13541 [pii] 00129492-200602000-00011 [pii]
39. MacCollin M, Woodfin W, Kronn D, Short MP (1996) 52. Otto SR, House WF, Brackmann DE, Hitselberger WE,
Schwannomatosis: a clinical and pathologic study. Nelson RA (1990) Auditory brain stem implant: effect
Neurology 46(4):1072–1079 of tumor size and preoperative hearing level on func-
40. Maini S, Cohen MA, Hollow R, Briggs R (2009) tion. Ann Otol Rhinol Laryngol 99(10 Pt 1):789–790
Update on long-term results with auditory brainstem 53. Otto SR, Shannon RV, Wilkinson EP, Hitselberger WE,
implants in NF2 patients. Cochlear Implants Int McCreery DB, Moore JK, Brackmann DE (2008)
10(Suppl 1):33–37. doi:10.1002/cii.383 Audiologic outcomes with the penetrating electrode
41. Mathieu D, Kondziolka D, Flickinger JC, Niranjan A, auditory brainstem implant. Otol Neurotol 29(8):1147–
Williamson R, Martin JJ, Lunsford LD (2007) 1154. doi:10.1097/MAO.0b013e31818becb4
Stereotactic radiosurgery for vestibular schwannomas 54. Parry DM, Eldridge R, Kaiser-Kupfer MI, Bouzas EA,
in patients with neurofibromatosis type 2: an analysis Pikus A, Patronas N (1994) Neurofibromatosis 2
of tumor control, complications, and hearing preser- (NF2): clinical characteristics of 63 affected individu-
vation rates. Neurosurgery 60(3):460–468. als and clinical evidence for heterogeneity. Am J Med
doi:10.1227/01.NEU.0000255340.26027.53; discus- Genet 52(4):450–461. doi:10.1002/ajmg.1320520411
sion 468–470. 00006123-200703000-00005 [pii] 55. Phi JH, Kim DG, Chung HT, Lee J, Paek SH, Jung HW
42. Mautner VF (1996) Spinal tumors in patients with (2009) Radiosurgical treatment of vestibular schwan-
neurofibromatosis type 2: MR imaging study of fre- nomas in patients with neurofibromatosis type 2:
quency, multiplicity and variety. AJR Am J Roentgenol tumor control and hearing preservation. Cancer
166(5):1231 115(2):390–398. doi:10.1002/cncr.24036
43. Mautner VF, Baser ME, Thakkar SD, Feigen UM, 56. Preuss M, Stein M, Huegens-Penzel M,
Friedman JM, Kluwe L (2002) Vestibular schwan- Kuchelmeister K, Nestler U (2008) Metastatic
noma growth in patients with neurofibromatosis type tumours mimicking vestibular schwannoma. Acta
2: a longitudinal study. J Neurosurg 96(2):223–228. Neurochir (Wien) 150(9):915–919. doi:10.1007/
doi:10.3171/jns.2002.96.2.0223 s00701-008-0007-5
44. Mautner VF, Lindenau M, Baser ME, Hazim W, 57. Ragge NK (1993) Clinical and genetic patterns of
Tatagiba M, Haase W, Samii M, Wais R, Pulst SM neurofibromatosis 1 and 2. Br J Ophthalmol 77(10):
(1996) The neuroimaging and clinical spectrum of 662–672
neurofibromatosis 2. Neurosurgery 38(5):880–885; 58. Ramsden R, Khwaja S, Green K, O’Driscoll M,
discussion 885–886 Mawman D (2005) Vestibular schwannoma in the
45. Mautner VF, Tatagiba M, Guthoff R, Samii M, only hearing ear: cochlear implant or auditory brain-
Pulst SM (1993) Neurofibromatosis 2 in the pediatric stem implant? Otol Neurotol 26(2):261–264,
age group. Neurosurgery 33(1):92–96 00129492-200503000-00023 [pii]
46. Moffat DA, Quaranta N, Baguley DM, Hardy DG, 59. Roehm PC, Mallen-St Clair J, Jethanamest D,
Chang P (2003) Management strategies in Golfinos JG, Shapiro W, Waltzman S, Roland JT
neurofibromatosis type 2. Eur Arch Otorhinolaryngol (2011) Auditory rehabilitation of patients with
260(1):12–18. doi:10.1007/s00405-002-0503-9 neurofibromatosis type 2 by using cochlear implants.
374 6 Neurofibromatosis Type 2 and Other Bilateral Cerebellopontine Angle Tumors

J Neurosurg 115(4):827–834. doi:10.3171/2011.5. 67. Slattery WH 3rd, Fisher LM, Hitselberger W,


JNS101929 Friedman RA, Brackmann DE (2007) Hearing preser-
60. Rowe JG, Radatz M, Walton L, Kemeny AA (2002) vation surgery for neurofibromatosis type 2-related
Stereotactic radiosurgery for type 2 neurofibromatosis vestibular schwannoma in pediatric patients. J
acoustic neuromas: patient selection and tumour size. Neurosurg 106(4 Suppl):255–260. doi:10.3171/
Stereotact Funct Neurosurg 79(2):107–116. ped.2007.106.4.255
doi:10.1159/000070106, 70106 [pii] 68. Thiagalingam S, Flaherty M, Billson F, North K
61. Ruggieri M (2001) Mosaic (segmental) neurofibro- (2004) Neurofibromatosis type 1 and optic pathway
matosis type 1 (NF1) and type 2 (NF2): no longer gliomas: follow-up of 54 patients. Ophthalmology
neurofibromatosis type 5 (NF5). Am J Med Genet 111(3):568–577. doi:10.1016/j.ophtha.2003.06.008,
101(2):178–180. doi:10.1002/ajmg.1332 S0161-6420(03)01420-9 [pii]
62. Samii A, Lenarz M, Majdani O, Lim HH, Samii M, 69. Tran Ba Huy P, Kania R, Frachet B, Poncet C,
Lenarz T (2007) Auditory midbrain implant: a com- Legac MS (2009) Auditory rehabilitation with
bined approach for vestibular schwannoma surgery cochlear implantation in patients with neurofibro-
and device implantation. Otol Neurotol 28(1):31–38. matosis type 2. Acta Otolaryngol 129(9):971–975.
doi:10.1097/01.mao.0000247819.16325.7d, doi:10.1080/00016480802510202, 905661602 [pii]
00129492-200701000-00006 [pii] 70. Ucar T, Akyuz M, Kazan S, Tuncer R (2000) Bilateral
63. Samii M, Gerganov V, Samii A (2008) Microsurgery cerebellopontine angle arachnoid cysts: case report.
management of vestibular schwannomas in neuro- Neurosurgery 47(4):966–968
fibromatosis type 2: indications and results. Prog Neurol 71. Vincent C, Zini C, Gandolfi A, Triglia JM, Pellet W,
Surg 21:169–175. doi:10.1159/000156905, 156905 [pii] Truy E, Fischer G, Maurizi M, Meglio M, Lejeune JP,
64. Seppala MT, Sainio MA, Haltia MJ, Kinnunen JJ, Setala Vaneecloo FM (2002) Results of the MXM Digisonic
KH, Jaaskelainen JE (1998) Multiple schwannomas: auditory brainstem implant clinical trials in Europe.
schwannomatosis or neurofibromatosis type 2? J Otol Neurotol 23(1):56–60
Neurosurg 89(1):36–41. doi:10.3171/jns.1998.89.1.0036 72. Ward BA, Gutmann DH (2005) Neurofibromatosis 1:
65. Shannon RV, Fayad J, Moore J, Lo WW, Otto S, from lab bench to clinic. Pediatr Neurol 32(4):221–
Nelson RA, O’Leary M (1993) Auditory brainstem 228. doi:10.1016/j.pediatrneurol.2004.11.002, S0887-
implant: II. Postsurgical issues and performance. 8994(04)00555-7 [pii]
Otolaryngol Head Neck Surg 108(6):634–642 73. Yohay K (2006) Neurofibromatosis types 1 and
66. Slattery WH 3rd, Brackmann DE, Hitselberger W 2. Neurologist 12(2):86–93. doi:10.1097/01.nrl.
(1998) Hearing preservation in neurofibromatosis 0000195830.22432.a5, 00127893-200603000-00003
type 2. Am J Otol 19(5):638–643 [pii]
Meningiomas
7

Cerebellopontine Angle Meningiomas (CP angle), 21 % (petroclival), 11 % (foramen


magnum), and 10 % (tentorial meningiomas).
C. Rokitansky was the first to describe a petrous
ridge meningioma in 1855 at autopsy [81]. Later
on, Virchow described a tumor originating from Pathogenesis and Pathology
the posterior lip of the meatus acusticus, which
he called “psammoma” [100]. Sir Charles Balance CP meningiomas are believed to arise from arach-
in London performed the first successful com- noid cap cell clusters located around the IAC,
plete removal of a CP angle tumor on November lower border of superior petrosal sinus, lateral
19, 1894. According to the original description, it border of the inferior petrosal sinus, around the
was most probably a meningioma: firm, well- cranial nerve foramina, and in the region of the
encapsulated tumor with wide attachment to the geniculate ganglion [65]. The variable attach-
dura of the petrous bone. The term “meningioma” ment sites and direction of growth determine the
was coined in 1922 by Harvey Cushing to high heterogeneity of CP angle meningiomas in
describe a benign neoplasm of the brain menin- terms of clinical presentation and operative
ges [19]. In 1938, Cushing and Eisenhardt challenge.
described their experience with the surgical man- Inactivation of both NF2 gene alleles is
agement of seven patients with meningiomas regarded essential for meningioma formation in
“simulating acoustic neuromas” [20]. both sporadic and in NF-2-associated menin-
giomas. The inactivation is caused most fre-
quently by biallelic losses of markers from the
Epidemiology chromosomal region (22q12.2) encompassing
the NF2 gene (40–70 % of meningiomas), muta-
Meningiomas are the second most frequent tumor tions in the NF2 gene (30–60 % of sporadic men-
of the CP angle after VSs. They comprise 6–15 % ingiomas), calpain-mediated proteolysis, or
of all CP angle tumors and 40–42 % of all poste- aberrant methylation in the 5¢ region of the NF2
rior cranial fossa meningiomas [10, 16, 74, 90]. gene [22, 24, 52]. As described in the chapter on
Their epidemiological and biological features are NF-2, the NF2 gene product functions as a reces-
similar to those of meningiomas with other loca- sive tumor suppressor. This protein is believed to
tions: they are generally slow-growing benign be crucial in regulating leptomeningeal prolifera-
lesions that occur more frequently in women in tion. Although inactivation of both copies of the
the fifth to sixth decade of their life. NF2 gene is necessary for initiation of menin-
The senior author’s experience comprises more gioma formation, its role in malignant menin-
than 1,000 posterior fossa meningiomas: 59 % gioma progression is debated [94]. Some studies

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 375


DOI 10.1007/978-3-642-35422-9_7, © Springer-Verlag Berlin Heidelberg 2013
376 7 Meningiomas

have shown that NF-2 mutations occur with simi- progression of meningiomas, similar to that of
lar frequencies in WHO grade I, WHO grade II, gliomas, is regarded as a continuum of increasing
and WHO grade III meningiomas [103]. What atypia and mitotic activity. Anaplastic (WHO
alternative mechanism is responsible for the ori- grade III) meningiomas exhibit considerably more
gin of meningiomas that do not carry NF-2 muta- malignant histologic features, compared to atypi-
tions and for atypical and anaplastic meningiomas cal ones [78].
remains unclear. Some genetic aberrations, such In the future, meningioma grading will be cer-
as deletion of the INK4a-ARF locus or altera- tainly refined by inclusion of molecular and
tions of the protein 4.1B family, which belongs to genetic criteria, such as presence of genetic alter-
the same protein superfamily as merlin, might ations on chromosomes 1p, 10q, 14q, and 9p, and
play a role [94]. telomerase activation [94].
CP angle meningiomas are typically globular Peritumoral edema, which is encountered in a
dura-based tumors; very rarely the carpet-like or considerable number of patients, is not a feature
“en plaque” form is encountered. With progres- pertinent only to atypical or anaplastic menin-
sive growth, the overlying meninges are pushed giomas and occurs also in benign WHO grade I
by the tumor, thus creating an arachnoidal mar- tumors. The extent of edema, however, tends to
gin, which serves as a natural cleavage plane dur- correlate with histological tumor grade. More
ing surgery. severe edema is seen in case of tumor transgres-
The current World Health Organization (WHO) sion of the arachnoid plane or brain invasion.
2000 classification distinguishes between WHO Tumor size has also been linked to the develop-
grade I, atypical WHO grade II, and anaplastic ment of edema, but our experience does not
(malignant) WHO grade III meningioma [53]. confirm that completely. Although the precise
Within each grade, however, a wide variability of pathogenesis of edema is not known, important
tumor biological behavior exists, in terms of role seems to play the extravasation of edemo-
growth rate, adhesion, or infiltration of adjacent genic cytokines, particularly of VEGF (vascular
neurovascular structures; brain invasion; and risk endothelial growth factor), which causes vasogenic
of recurrence (Fig. 7.1). Generally, meningiomas edema and neoangiogenesis into brain paren-
are two types: encapsulated and infiltrating. chyma [64].
Encapsulated meningiomas may be removed
without or with minimal morbidity. Some menin-
giomas, however, tend to infiltrate the pial cover- Classification
ing and even neural tissue. Microcystic, atypical,
and, especially, anaplastic meningiomas tend to Meningiomas, arising from the dura of the poste-
have more extensive and irregular tumor–brain rior surface of the pyramid lateral to the trigemi-
interface [55]. Pial infiltration is not restricted to nal nerve, are defined as “CP angle meningiomas”
the aforementioned tumor categories: it may occur and those arising medial to the trigeminal nerve
even in WHO grade I meningiomas. Brain inva- as “petroclival meningiomas” [86]. We consider
sion does not necessarily indicate malignant those meningiomas that arise from adjacent areas
tumor, in particular in the absence of frank histo- but have their main bulk in the CP angle also as
logical anaplasia [52, 78]. Grading of menin- CP angle meningiomas. These tumors are further
giomas is based on histopathological criteria, divided into five subgroups (Figs. 7.2, 7.3, 7.4,
which do not account for these individual differ- 7.5, 7.6, and 7.7): premeatal, centered at the IAC,
ences [43]. Criteria for atypical (WHO grade II) suprameatal, inframeatal, and retromeatal
meningiomas are increased mitotic activity or [66, 67]. Although the main tumor occupies the
three or more of the following features: hypercel- CP angle cistern, any of these meningiomas can
lularity, small cell transformation, macronucleoli, grow secondarily into the IAC. Meningiomas,
uninterrupted patternless growth, and foci of arising within the IAC, though very rare, comprise
spontaneous or geographic necrosis [79]. Malignant a separate subgroup. Accurate determination of
Cerebellopontine Angle Meningiomas 377

a b

Fig. 7.1 Large sphenopetroclival meningioma in a demonstrated the extent of tumor removal and brain stem
38-year-old patient. Considering the extension of the decompression. Second surgery—removal of the extra-
tumor (a), the degree of brain stem compression, and the cavernous tumor part via a frontotemporal craniotomy—
neurological status (absence of visual symptoms), a deci- was scheduled 6 months later. In 5 months, new MRI was
sion was taken for tumor removal in two steps. Initially, performed: unexpectedly, substantial growth of the
the infratentorial tumor part was removed via the retrosig- infratentorial part of the tumor was visualized (c). The
moid suprameatal approach. The pathological examina- patient is to be operated within 1 week and is planned for
tion revealed that it is a transitional meningioma (WHO postoperative radiotherapy
grade 1); Ki 67 was 2 %. The early postoperative MRI (b)
378 7 Meningiomas

a b

Fig. 7.2 Premeatal meningiomas. (a) T1-weighted MR sequence after contrast administration. (b) CISS sequence

a b

Fig. 7.3 Retromeatal meningiomas. (a) T1-weighted MR sequence after contrast administration. (b) CISS sequence

a b

Fig. 7.4 Suprameatal meningiomas. (a) T1-weighted MR sequence after contrast administration. (b) CISS sequence
Cerebellopontine Angle Meningiomas 379

a b

Fig. 7.5 Inframeatal meningiomas. (a) T1-weighted MR sequence after contrast administration. (b) CISS sequence

a b

Fig. 7.6 Meningiomas centered at the IAC. (a) T1-weighted MR sequence after contrast administration. (b) CISS
sequence

a b

Fig. 7.7 Petroclival meningiomas. (a, b) T1-weighted MR sequences after contrast administration
380 7 Meningiomas

the site of origin, however, is possible only in The extension patterns of larger CP angle
small tumors. Large tumors may spread more meningiomas can be generally classified as:
widely and occupy several adjacent areas. The • Retromeatal meningiomas:
site of origin in such cases might be inferred from – Extension into the IAC
the location of the so-called tumor matrix or dural – No extension into the IAC
attachment. Occasionally it is marked by a small • Premeatal meningiomas
hyperostosis. In most patients, the blood supply – Medial extension to petrous apex
to the tumor comes primarily through this attach- – Medial and superior extension
ment site. • Extension into the Meckel’s cave
Pure clival meningiomas are rare; they origi- • No extension into the Meckel’s cave
nate from the lower, middle, or upper clivus and – Medial and inferior extension
are regarded as a separate subtype of the petro- • Extension into the jugular foramen
clival group (Fig. 7.8). • No extension into the jugular foramen
This classification has surgical and prognostic • Extension to the foramen magnum
significance: the more medially located tumors • No extension to the foramen magnum
are related to greater surgical complexity and – Extension into the IAC
poorer outcome. As presented in a previously – No extension into the IAC
published evaluation of the senior author’s expe- • Large meningiomas located pre- and ret-
rience with CP angle meningiomas [66], a romeatal with all extension patterns described
specific pattern of dislocation of the seventh- above
eighth cranial nerves is observed in each of the
subgroups:
Premeatal meningiomas (originating between Neuroimaging
the IAC and the trigeminal nerve):
• seventh and eighth nerves are displaced most CP meningiomas display more variable features
frequently posteriorly (45 %) and inferiorly than VSs in regard to tumor type and grade, ori-
(43 %); rarely—superiorly (10 %). gin, location, and extension pattern. Preoperative
• fifth nerve is displaced primarily superiorly imaging, besides the information on tumor type
(59 %), anteriorly (26 %), or inferiorly and extension, should provide information on its
(14 %). relations to the important surrounding neurovas-
Meningiomas centered at the IAC: cular and bone structures, which is essential for
• seventh and eighth nerves are surrounded the surgical planning. The neuroradiological
by the tumor in 29 %; displaced superiorly examination should include at least thin-slice
or inferiorly (25 % each). Rarely they are bone-window CT, native MRI, and MRI follow-
displaced posteriorly (12 %) or anteriorly ing contrast administration. In case the surgery is
(8 %). supposed to be performed in the semi-sitting
Suprameatal meningiomas: position, dynamic X-ray of the cervical spine is
• seventh and eighth nerves are displaced inferi- also mandatory.
orly in 78 % and anteriorly in 17 %. On CT, meningiomas appear generally as
Inframeatal meningiomas: well-circumscribed, encapsulated tumors. Most
• seventh and eighth nerves are displaced supe- demonstrate some degree of hyperdensity with
riorly in 69 % and anterior in 19 %. respect to surrounding gray matter. Meningiomas
Retromeatal meningiomas (originate between are strongly and uniformly enhancing tumors.
the IAC and sigmoid sinus): Up to 20–25 % may show evidence of intratu-
• seventh and eighth nerves are displaced ante- moral calcification, occurring in a punctate, rim-
riorly in 62 % and inferiorly in 25 %. like, or nodular pattern or even as dense
Cerebellopontine Angle Meningiomas 381

a b

Fig. 7.8 Clival meningioma, arising from the middle third facial, and lower cranial nerves are obvious. (d) Complete
of clivus. (a, b) Preoperative MR images. (c) Initial view tumor removal with coagulation of its attachment at the
of the tumor, via a retrosigmoid perspective. The compres- clivus. (e) Postoperative CT scan (Reprinted from:
sion and dislocation of the trigeminal, vestibulocochlear/ Nakamura and Samii [70])
382 7 Meningiomas

e calcification. Calcifications appear hypointense


on all sequences. Contrast enhancement is usu-
ally intense and homogeneous. Areas of central
necrosis, which are caused by rapid tumor growth,
are hypointense on T1 and hyperintense on T2
sequences. The extent of meningioma growth
into IAC is best evaluated on axial and/or coronal
T2-weighted and CISS sequences: the presence/
absence of high signal, corresponding to CSF,
lateral to the tumor indicates its lateral extension.
Notably, radiological information may be mis-
leading, and the actual relations may become
obvious only at surgery.
CP meningiomas can be usually reliably dif-
ferentiated from VSs on preoperative images
[13, 105]. Meningiomas have broad contact to
the petrous bone or tentorium; most are centered
away from the IAC; the angle between the tumor
and the pyramid is obtuse; the IAC is not wid-
Fig 7.8 (continued) ened (Fig. 7.11). Although secondary invasion
of the IAC might be observed in 10–22 %, pri-
calcification of the entire tumor (Fig. 7.9). Bone marily meningiomas of the IAC are very rare
alterations, best visible on high-resolution bone- [83, 101]. In contrast, VSs are centered at the
window CT series, may be caused by actual widened IAC, extend almost always into the
tumor infiltration or erosion from direct pres- IAC, form an acute angle with the posterior sur-
sure or are reactive bony changes. These bone face of the pyramid, and cause different patterns
changes contribute to the discrimination of men- of bone changes. On T2-weighted sequence,
ingiomas from other tumors (Fig. 7.10). VSs meningiomas have higher intensity than VSs.
may cause dilatation of the IAC and adjacent The dural “tail” sign, although not pathogno-
bone erosion but do not produce hyperostosis monic, is strongly suggestive of meningioma
and almost never calcify. Chordomas and chon- (Fig. 7.12). This dural reaction is due to actual
drosarcomas display different patterns of bone tumor extension (wedge-shaped tumor “tongue”)
destruction. but can also be caused by nonneoplastic reac-
MRI is currently regarded as the most sensi- tive change, tissue proliferation, or hypervascu-
tive modality for detection and characterization larity. Other tumors that may have a dural “tail”
of meningiomas. The ability to obtain images and are dura-based metastases, lymphomas, sarcoi-
visualize the tumor and its surroundings in mul- dosis, solitary fibrous tumors, and hemangio-
tiple planes is important for operative planning. pericytomas [102].
On T1-weighted MRI studies, meningiomas are The possibility to predict accurately tumor type
typically isointense to slightly hypointense with and grade, tumor consistency (firm or soft menin-
respect to brain [109]. On T2-weighted MRI giomas), tumor vascularization, and tumor relation
studies, they demonstrate greater signal variabil- to the arachnoid (clear arachnoid plane or invasion)
ity: more than half are isointense, 35–40 % are would greatly benefit the decision-making process.
hyperintense, and less than 10 % are hypointense. It would allow for more precise preoperative plan-
Signal heterogeneity on T2-weighted sequences, ning and patient counseling. Although some radio-
which is not uncommon, is due to cystic tumor logical factors have been shown to have correlation
parts, necrosis, previous hemorrhages, or areas of to the meningiomas’ biological behavior, current
Cerebellopontine Angle Meningiomas 383

a b

c d

Fig. 7.9 Petroclival meningioma, causing reactive bony hyperostosis in the area anterior to the IAC (arrow). (a) Bone-
window CT scan. (b, c) T1-weighted MR images after contrast administration. (d) FLAIR MR image

neuroimaging modalities provide inconclusive arachnoid plane (See Case Illustration 7.1) [15, 32,
data. Malignant meningiomas tend to have more 92, 96]. Atypical and anaplastic meningiomas that
frequently increased signal intensity on both T1- invade the pia often provoke considerable cerebral
and T2-weighted MR scans than do benign ones. edema [54, 96]. Some studies showed that edema
The presence of irregular tumor projections into is more common in association with meningiomas
the brain surface, the so-called mushrooming, and having increased MIB-1 labeling indices [40].
the presence of indistinct margins are further Edema appears hypointense on T1-weighted
factors that might rise the suspicion of malignancy images and hyperintense on T2-weighted sequences
[2] (Fig. 7.13). Peritumoral edema tends to corre- and is associated with peripheral (or rim) enhance-
late well but not always to the loss of a patent ment on fast FLAIR sequences [73].
384 7 Meningiomas

a b

e
f

Fig. 7.10 CT (a–c) and MR images (d–h) of an unusual nal, and sagittal bone-window CT. (d) T1-weighted native
petroclival meningioma with involvement of the cavern- and (e, f) contrast-enhanced MR sequences. (g, h)
ous sinus and retrosellar growth. The dense calcification T2-weighted sequences
of the tumor is the clue to its diagnosis. (a–c) Axial, coro-
Cerebellopontine Angle Meningiomas 385

g h

Fig 7.10 (continued)

a b

Fig. 7.11 Meningioma with


secondary intracanalicular extension.
(a, b) bone-window CT, demonstrat-
ing the IAC on the left and right
sides. Both sides should be compared
always to exclude—as in this
case—an anatomically wider medial
portion of the canal. (c, e) MRI
examination: T1-weighted native
(c), T1-weighted with contrast
administration (d), and T2-CISS
(e) MR sequences. The extent of
intracanalicular tumor growth is best
appreciated on the CISS sequence,
showing both the tumor in the
medial part of the canal and the
CSF lateral to it
386 7 Meningiomas

c d

Fig 7.11 (continued)

Fig. 7.12 The dura “tail” sign (arrows), although not


pathognomonic, is strongly suggestive of meningioma
Cerebellopontine Angle Meningiomas 387

a b

c d

Fig. 7.13 MRI study of a 22-year-old patient with a giant patient was operated at another facility, but only partial
CP angle meningioma (a–c) T1-weighted sequence after removal was feasible due to its high vascularization and
contrast administration. (d) T2-weighted sequence. Note absence of arachnoid dissection plane. The patient is
the mushrooming effect: irregular tumor projections into scheduled for surgical treatment via the RS approach at
the brain surface and the intense peritumoral edema. The the INI
388 7 Meningiomas

Case Illustration 7.1 possibility was discussed with the patient, and
Petroclival Meningioma with Perifocal a decision was taken that if the tumor would
Edema turn out to be infiltrative, only a subtotal resec-
Sixty-five-year-old female patient with recur- tion would be performed. The tumor was
rent headache, progressive gait instability, exposed via the RS approach and partially
hemifacial hypesthesia, and decreased hearing resected. It was indeed infiltrative in regard to
on the left side. The MRI showed a large sphe- cranial nerves and brain stem. Multiple ves-
nopetroclival meningioma on the left side with sels were identified passing through the men-
supratentorial extension and involvement of ingioma and supplying ultimately the brain
posterior cavernous sinus (a–f). Two fea- stem. An attempt to proceed with tumor dis-
tures—the unclear border to surrounding section would inevitably cause severe iatro-
structures and the perifocal edema—indicated genic damage. Hence, once decompression of
a possible infiltration of the brain stem. This the brain stem was achieved, no attempt was

a b

c d

Preoperative MRI of an infiltrative sphenopetroclival sequence. Note the brain stem edema, which is best
meningioma. (a–d) T1-weighted sequence after con- visualized on the T2-weighted and FLAIR sequences,
trast administration. (e) T-2 sequence. (f) FLAIR and the unclear border to surrounding structures
Cerebellopontine Angle Meningiomas 389

e f

made to dissect the tumor capsule. MRI was recommended in 6 months: in case
Histopathological diagnosis was meningothe- the remnant would have growth tendency,
lial meningioma (WHO grade I). Follow-up fractionated radiotherapy would be necessary.
390 7 Meningiomas

Generally, T1-weighted signal characteris- Clinical Presentation


tics do not correlate with either histopathologi-
cal findings or tumor consistency [28]. Higher The clinical presentation of patients with CP
signal on T2-weighted images may indicate a angle meningiomas depends mainly on the size,
softer tumor consistency. Lower signal suggests site of origin, and extension of the tumor. They
harder tumor consistency; it is associated with cannot be distinguished from other CP angle
calcifications and fibrous tissue [28, 107]. The tumors based on their clinical presentation
very important from surgical standpoint cleav- alone. Some clinical clues, however, should
age plane or distinct interface between tumor alert the clinician that the lesion may be other
and adjacent brain may be visualized as a CSF than VS [56].
“cleft” around the tumor periphery. It has typi- Neurological symptomatology is due to:
cal CSF signal characteristics: hypointensity • Involvement of the cranial nerves and dura
on T1-weighted images and hyperintensity on • Mass effect on the cerebellum
T2-weighted images. • Brain stem compression
DWI might prove helpful in the differentiation • Intracranial hypertension, caused by the tumor
of the pathological grading of meningiomas: mass or due to obstructive hydrocephalus
some data indicate that atypical or malignant The onset of clinical symptoms is usually
meningiomas tend to have lower ADCs (apparent insidious, and the mean duration of symptoms
diffusion coefficient) than the benign ones [30]. before hospital admission is 12–31 months
MR spectroscopy finding of a combined elevation [10, 56]. At the time of diagnosis, the most fre-
of glutamate/glutamine and presence of alanine at quent symptoms, observed in 50–78 % of the
1.5 ppm are specific for meningiomas [18]. cases, are otologic: impaired hearing (41–68 %),
MR angiography and MR venography demon- dizziness (20–31 %), and tinnitus (12–22 %)
strate the degree of tumor vascularity and allow [10, 21, 58, 91, 98, 101]. Hearing loss is of sud-
assessing the patency and dominance of major den onset in 9–14 %. Unspecific headache as ini-
venous sinuses. In our experience, digital subtrac- tial complaint is estimated to appear in 15–24 %.
tion angiography (DSA) as a diagnostic and/or At presentation, trigeminal nerve sensory
planning tool is required in exceptional cases, such deficit (facial numbness or a decreased corneal
as giant tumors with wide extension and involve- reflex) is seen in 13–49 % of the patients. Up to
ment of major vessels. Vascular supply to CP angle 14 % may present with trigeminal neuralgia.
meningiomas is from branches of the ascending Two percent of all cases of trigeminal neuralgia
pharyngeal artery, occipital artery, vertebral artery, are due to tumor compression at the root entry
AICA (subarcuate artery), and middle meningeal zone of the trigeminal nerve, and the tumors
artery. Large tumors may be supplied additionally causing such compression most frequently are
by direct branches of the AICA and the superior meningiomas (see Case Illustration 7.2). Hear-
cerebellar artery. Preoperative embolization of ing loss has 63–83 % of all patients [9, 14, 101];
tumor-feeding arteries may be helpful in highly this rate is lower than in patients with VSs,
vascular meningiomas. Ideally, it renders the men- where it is found in the majority of the cases.
ingioma smaller, softer, and less bloody, which, Preoperative facial nerve paresis is a rare sign,
ultimately, allows for safer removal. Most menin- observed in 6–8 % of the patients [66, 101].
giomas, however, do not need to be embolized Lower cranial nerve deficits can occur in case of
because adequate blood control can be obtained caudal tumor extension and are seen in 6–21 %
intraoperatively. of all patients [10].
Cerebellopontine Angle Meningiomas 391

Case Illustration 7.2 could be successfully removed without endan-


Small Premeatal Meningioma Causing gering the surrounding cranial nerves. In order
Trigeminal Neuralgia to decrease the risk of recurrences, the hyper-
The patient suffered from symptomatic trigem- ostotic bone and the suprameatal tubercle were
inal neuralgia for approximately 18 months. drilled off. Fat pieces were used to prevent
Neurological examination revealed hypesthesia occurrence of CSF leak. Histopathological
in all branches of the fifth nerve and decreased finding was a psammomatous meningioma
corneal reflex on the left side. She was operated WHO grade I. After the surgery, the patient
via the standard RS approach. In this case, the did not have more neuralgic attacks; however,
petrous vein had to be coagulated in order to the hypesthesia persisted longer and started to
expose the tumor sufficiently. The meningioma resolve only after 6–7 months.

a b

(a–c) Small meningioma arising from the posterior hyperostosis in the area of tumor attachment. (a) Bone-
petrous dura, immediately lateral to the trigeminal window CT scan. (b) T1-weighted MR sequence.
nerve. The compressive effect of the tumor is well vis- (c) CISS sequence
ible, especially on the CISS sequence (c). Note the
392 7 Meningiomas

Cerebellar dysfunction, mainly gait ataxia, is Management


found in 25–52 % of all patients with CP angle
meningiomas. Symptoms and signs secondary to Perioperative Management
brain stem compression with obstructive hydro- Patients with extensive perifocal edema receive
cephalus occur in late stages of the disease. Signs dexamethasone for 3 days prior to surgery (12–
of increased intracranial pressure have 16–29 %, 24 mg/day). Beneficial effect—decrease in capil-
and hydrocephalus have 20–31 % of the patients lary permeability—is manifested within the first
in different series [51, 56]. In case of very large hours after its administration [93]. Dexamethasone
or giant meningiomas, these symptoms are invari- after the surgery is given to all patients in similar
ably seen. dosage—12–24 mg/day—for at least 3 days.
The origin and location of meningiomas Prophylaxis with broad-spectrum antibiotic (usu-
determine their clinical presentation. Premeatal ally cefazolin) is administered as a single preop-
and retromeatal meningiomas have distinctive erative dose and readministered every 6 h during
clinical development and symptomatology the procedure. Prophylactic antibiotics are dis-
[66, 86, 89, 90]. Premeatal tumors are diag- continued within 24 h. If the risk of infection is
nosed earlier and consequently have smaller higher, for example, due to local or systemic rea-
size. Their clinical presentation is related to sons, prolonged application or more potent anti-
cranial nerve dysfunction (such as hearing loss, biotics may be given.
trigeminal neuralgia, or hypesthesia) and facial The risk of venous thromboembolism in patients
and/or abducens nerve palsy. Retromeatal men- operated for intracranial meningiomas is elevated
ingiomas are generally larger; the predominant due to the existing neurological deficits, the related
symptoms are due to cerebellar compression. immobility of patients and/or the prolonged immo-
Meningiomas, originating above the IAC, most bilization during or after surgery. Furthermore,
often present with facial pain or other sensory release of procoagulants and fibrinolysis-inhibiting
symptoms before developing eighth cranial factors by the tumor may play a role in the devel-
nerve symptoms. The clinical presentation of opment of this procoagulant state [88]. All patients
meningiomas, originating at or near to the IAC, wear antithrombotic (compression) stockings dur-
is similar to that of patients with VSs: hearing ing the postoperative hospital stay. Intermittent
loss, tinnitus, and disequilibrium. Patients with compression devices are utilized in high-risk
large meningiomas that occupy the whole CP patients or in those with contraindications for hep-
angle present with a combination of these arin prophylaxis. All patients receive subcutane-
symptoms. ous low molecular weight heparin prophylaxis
since the day after surgery, provided the postopera-
Clinical Examinations tive CT shows no signs of intracranial hemorrhage.
All patients should undergo hearing examination, Following surgery, patients are mobilized early;
including audiogram and evaluation of speech passive range-of-motion exercises are initiated
discrimination. Neuro-ophthalmologic examina- early in those with neurological deficits.
tion is performed in patients with oculomotor
nerve deficits. In case of more caudal tumor Management
growth and preoperative swallowing or speech The treatment options of CP angle meningiomas
deficits, an ENT examination with direct laryn- are observation, surgery, radiotherapy/radiosur-
goscopy allows estimating the function of the gery, or combination thereof. The decision in
lower cranial nerves. Tumors extending supraten- each case should be taken individually, consid-
torially might compress the pituitary stalk or ering patient’s biological age, expectations, and
extend into the sella. In such cases, pituitary axis activities; his general and neurological status; as
evaluation and—if needed—endocrinologic well as tumor size and extension. Meningiomas
treatment preoperatively are indicated. are increasingly detected early, during their
Cerebellopontine Angle Meningiomas 393

“preclinical” stage due to the wide availability are related to higher risks of neurological morbid-
of MRI facilities. Furthermore, MRI is a reliable ity or operative complications, such as facial nerve
method to monitor tumor growth. Initial follow- palsy, hearing loss, and CSF leak. Moreover, with
up and surgery only in case the meningioma these techniques, a higher risk of venous-related
shows growth potential or new symptoms appear complications, such as interruption or thrombosis
are frequently recommended. On the other hand, of major draining veins or sinuses, exists. The
earlier active management is completely justified transverse and sigmoid sinuses may be injured dur-
because the outcome of surgery is related to ing their exposure or may desiccate if skeletonized
tumor size and is optimal in neurologically intact completely, which increases the risk of thrombosis
patients. We recommend an initial period of clin- with consequent parenchymal swelling or hemor-
ical and radiological observation to determine rhage. Even their prolonged retraction with the
whether the symptoms are progressive and/or the dura can cause partial or complete interruption of
tumor is enlarging in elderly or somatic unstable venous flow, leading to venous congestion or
patients with small tumors and mild stable symp- thrombosis.
toms or in those unwilling to undergo surgery. Another important issue is the possibility to
Repeat scans should be performed in 6-month preserve hearing. Some authors suggest that hear-
intervals or—if new symptoms appear—earlier. ing preservation should be attempted only in ret-
The optimal treatment of CP meningiomas, romeatal meningiomas [89]. Hearing destructing
which potentially leads to cure of the patient, is approaches through the pyramid have been pro-
their complete operative tumor removal. This posed for patients with severe hearing deficit or
goal, however, should not be strived at the expense premeatal meningiomas. Our experience has
of new neurological dysfunction and/or deterio- shown, however, that in CP angle meningiomas,
ration of the patient’s quality of life. contrary to VSs, hearing loss is usually due to
Different surgical routes are used to approach compression of the cochlear nerve. Following
CP angle meningiomas: retrosigmoid, extended tumor removal, hearing may improve even in large
retrosigmoid, presigmoid, translabyrinthine, tumors. Such improvement has been observed and
transcochlear, transpetrosal, far lateral, modified reported also by other authors [59, 66, 99].
far lateral, as well as routes through the middle Having extensive experience with all skull-
cranial fossa [6, 11, 21, 65, 73, 91, 98]. The gen- base techniques, the senior author’s current con-
eral concepts that are important for the successful cept is that simpler and safer approaches are the
removal of most meningiomas, however, are simi- prerequisite for improving outcome and reducing
lar: adequate exposure, interruption of the blood morbidity. The retrosigmoid suboccipital approach
supply along the dural attachment, internal decom- (RS approach) offers an excellent panoramic visu-
pression, and cautious dissection of the tumor cap- alization of the whole CP angle, wide exposure of
sule from brain stem and cranial nerves in the the tumor, whatever its size, and increased safety
arachnoid plane. Due to the progress in neuroanes- when working in the vicinity of the brain stem. It
thesia, critical care, and microsurgery, as well as is related to a very low procedure-related morbid-
the introduction of modern skull-base approaches, ity rate and allows hearing preservation. In case of
the morbidity and mortality related to the surgical pure CP meningiomas, the RS approach is our
treatment of meningiomas decreased considerably favorite technique. In CP meningiomas with
over the last decades. medial extension toward the petroclival area and/
In the early ages of the skull base surgery, an or the Meckel’s cave, we utilize the retrosigmoid
extensive bony exposure allowing complete visual- suprameatal approach (Samii’s technique), intro-
ization of the meningioma and all adjacent neural duced by the senior author in 1982.
and vascular structures was considered as a major All CP angle meningiomas, regardless of their
prerequisite for complete and safe tumor removal. location, can display infiltrative or adhesive char-
The approaches, providing such exposure, however, acteristics. The lack of arachnoid plane, however,
394 7 Meningiomas

a b

Fig. 7.14 MR images of two patients with large CP angle ing tumor removal in both cases. The dilatation of the
meningiomas, causing hydrocephalus. The hydrocepha- ventricular system, however, persisted for more than 6
lus-related signs/symptoms resolved completely follow- months

becomes critical in case of large premeatal or pet- which can be attributed to further dilatation of the
roclival meningiomas. The attempt to dissect ventricles, an EVD is placed. Once the patients’
their capsule may damage the pial vascular sup- condition stabilizes, the weaning program is initi-
ply or cause direct damage to the brain stem. In ated, and a decision on the necessity of a VP shunt
such cases, subtotal tumor removal is justified in is taken according to its success.
order to avoid major neurological morbidity.
General Operative Technique
Management of Hydrocephalus Similar to the technique used in case of VSs, the
in Patients with CP Angle Meningiomas meningioma is removed by alternating internal
Large meningiomas of the CP angle may occlude debulking and dissecting the capsule from sur-
the outlets of the fourth ventricle and cause rounding structures in the arachnoid plane. Initial
obstructive hydrocephalus (Fig. 7.14). In contrast internal decompression or debulking of the tumor
to VSs, meningiomas typically do not cause is performed with an ultrasonic surgical aspirator,
impairment of the CSF absorption. Once the suction, or platelet knife. Thereafter, the tumor
obstruction is removed, the normal flow of the capsule is dissected in the arachnoid plane from
CSF is reestablished and the hydrocephalus is the neural and vascular structures.
resolved. In our practice, VP shunting is very
rarely needed after radical tumor removal. Retrosigmoid Approach
In patients presenting with severe hydrocepha- The main steps of the RS approach in case of CP
lus-related symptoms, an external ventricular drain- angle meningiomas are similar to those in case of
age (EVD) is inserted. Following the tumor surgery, VSs and are described in detail in the chapter on
stepwise weaning from the EVD is performed, as RS approach. They will be therefore only sys-
described in the section on VS-related hydrocepha- temized here:
lus. A VP shunt is inserted only in case the weaning • The patient is in semi-sitting position.
is unsuccessful. In patients with less severe preop- • Continuous neurophysiological monitoring is
erative hydrocephalus symptoms, the tumor is performed throughout the surgery: from the
removed primarily, and the patient is monitored patient positioning to the skin closure. SSEP,
closely. In case neurological deterioration appears, facial nerve EMG, BAEP, and recently facial
Cerebellopontine Angle Meningiomas 395

nerve motor-evoked potential are monitored • The lateral cerebellomedullary cistern is then
routinely, while monitoring of the oculomotor opened, and sufficient CSF is allowed to
and of the lower cranial nerves is performed if egress.
needed—according to the particular tumor • Every attempt should be made to preserve the
extension and clinical presentation. During integrity of petrous or other main draining veins.
tumor removal, nerve stimulation is used to The wide variability of the venous system of the
identify the facial nerve. CP angle does not allow predicting the potential
• The head is flexed and rotated approximately consequences of their interruption [44].
30° to the involved side. Importantly, occlu- Individual tumor characteristics, such as size
sion of venous jugular outflow or hyperflexion and extension, determine the subsequent steps:
of the cervical spine should be avoided. During • Generally, the meningioma is removed by
patient positioning, SSEP are continuously alternating internal debulking and dissecting
monitored and provide feedback information the capsule from the surrounding structures.
on the spinal cord function. • The cranial nerves are identified near their
• The approach should be planned and per- entrance or exit in bone or dura and at the
formed so that damage to venous sinuses is brain stem. Their dissection is performed by
avoided. Venous bleeding should be immedi- carefully stripping off the arachnoid from the
ately controlled by bipolar coagulation or with tumor with one hand. Simultaneously, a slight
hemostatic sponges in order to avoid air traction is done with the second hand.
entrapment. • Dissection is started only after sufficient inter-
• Avoidance of clinically significant air embo- nal decompression has been achieved. It is
lism relies on continuous anesthesiological performed always in the arachnoid plane.
monitoring (end-tidal carbon dioxide and pre- Once sufficient internal decompression has
cordial Doppler echocardiography) and care- been achieved at one of the CP angle levels,
ful operative technique (preventing of the surgeon moves to the next one.
embolism and immediate control if it occurs). • Only the feeding arteries or major bleeding
Good coordination between the anesthesiolo- vessels are coagulated during tumor removal.
gist and the surgeon is essential for avoiding The continuous irrigation of the operative
air embolism-related morbidity. The surgeon field performed by the assistant obviates the
should act systematically to detect and obliter- need for more frequent coagulation. Coagu-
ate the source of air entry at the first sign of lation is avoided in the vicinity of the cranial
venous air embolism. nerves.
• A slightly curved skin incision 2.5–3.5 cm • Targeted opening of the IAC, depending on
medial to the mastoid process is made. the degree of intracanalicular tumor extension
• The craniectomy should expose just the edges and surgical goal, is performed. Violation to
of the sigmoid and transverse sinuses. the inner ear structures should be avoided.
Caudally, the floor of the posterior fossa • The dural origin of the tumor should be excised
should be reached to allow unrestricted access or—if excision is not possible—coagulated
to the lateral cerebellomedullary cistern. The meticulously. High-speed drill is used to
mediolateral diameter of the opening is remove any bone hyperostosis, which may
3.5–4.5 cm. contain viable tumor cells.
• Excessive traction to the mastoid emissary • Since the dural tail probably indicates tumor
vein/veins may cause sinus laceration. The extension (at least in about 2/3 of the cases), it
vein should be skeletonized with a diamond should be resected, if possible, to reduce the
drill until it is free of any bony encasement risk of tumor recurrence.
and can be safely coagulated. • The drilled area of the pyramid as well as any
• The dura is incised in a curvilinear manner opened mastoid air cells is sealed with multi-
1.5–2 mm medial to the sigmoid and inferior ple fat pieces and fibrin glue.
to the transverse sinus. • Watertight dura closure.
396 7 Meningiomas

a b

d
c

Fig. 7.15 Lateral retromeatal meningioma. Note the Axial, coronal, and sagittal T1-weighted MR images after
broad base to the posterior petrous bone and to the tento- contrast application. (d) CISS sequence—the facial and
rium. The tumor is adjacent to the dominant sigmoid vestibulocochlear nerves are well visible both in their cis-
sinus, which has to be considered during surgery. (a–c) ternal and intracanalicular parts

Operative Technique is straightforward (Fig. 7.15). Important issue


in Different CP Angle Meningiomas to consider is the possibility of sigmoid sinus
Retromeatal meningiomas arise from the poste- involvement, especially on the side of the domi-
rior petrous bone dura between the IAC and the nant sinus. In case the tumor invades the wall of
sigmoid sinus. According to the senior authors’ a dominant sinus, complete removal should be
experience, most of them originate posterior attempted only if its safe reconstruction is pos-
and inferior to the IAC dorsal to the jugular sible. If a large part of the sinus is involved,
foramen. The size and exact site of origin of the small layer of the tumor may be left over the
tumor determine their relation to the seventh- sinus.
eighth nerve complex and the operative com- MRI, especially CISS and T2-weighted
plexity. Small more lateral meningiomas are sequences, may provide information on the
located away from the nerves, and their removal relation of these nerves to the tumor. Often, a
Cerebellopontine Angle Meningiomas 397

a b

Fig. 7.16 Retromeatal meningioma. (a, b) T1-weighted strong contrast enhancement are obvious. (c) On
MR image after gadolinium application of a meningioma, T2-weighted sequences, the arachnoid layer, separating
arising posterior to the IAC. The broad base to the dura, the tumor from the seven to eighth nerve complex, can be
the small dural tail sign (medial), and the homogeneous appreciated

well-defined arachnoid layer separates them during tumor debulking should be avoided not to
(Fig. 7.16). injure the cranial nerves. Notably, patients with
Large or more medially located meningiomas large or giant meningiomas, which caused pro-
have more intimate relationship to the seventh nounced cerebellar compression over long
and eighth cranial nerves and usually displace period, should be very carefully monitored after
them anteriorly (63 %) or inferiorly (25 %). The surgery: hematomas tend to accumulate in tumor
nerves are encountered in later stages of tumor bed or in cerebellar parenchyma, despite how
removal, which has to be kept in mind during thorough the hemostasis had been (see Case
surgery. Penetration of the anterior capsule Illustration 7.3).
398 7 Meningiomas

Case Illustration 7.3 be achieved. The facial and cochlear nerves


Large Retromeatal Meningioma were dislocated anteriorly. An arachnoid layer
Seventy-year-old male patient presented with separated them from the tumor capsule, which
a history of increasing gait instability and allowed for their safe dissection and structural
decreased hearing on the right side since more preservation. The tumor originated from the
than a year. Later on, constant nausea and dura between the IAC and the jugular fora-
frequent vomiting appeared. He was treated men. In order to prevent recurrences, the dura
for Ménière’s disease for several months; in the area was excised and the underlying
only when the therapy turned out to be com- bone was drilled off with the diamond tip.
pletely futile, MRI examination was per- The patient did not have new neurological
formed. At presentation, he had very severe deficits after surgery; the facial nerve function
gait and truncal ataxia, so that he had to use was completely normal. Routine follow-up CT
wheelchair. His cranial nerve function was performed the next day revealed an intracerebel-
normal besides the profound right-sided hear- lar hematoma on the right side (g). Considering
ing loss. MRI showed a very large retromeatal its relatively small size, a decision was taken for
meningioma with broad attachment to the conservative treatment. Follow-up CT scans
posterior petrosal surface and tentorium (a, demonstrated the gradual resorption of the blood
b). The tumor caused pronounced compres- (h, i). The histopathological diagnosis was
sion of the cerebellum; however, no signs of WHO grade I fibrous meningioma. At dis-
edema were seen. charge, the gait was significantly improved. At 2
The patient was operated via the RS years follow-up, the patient recovered com-
approach (c–f), and complete removal of the pletely. The late follow-up MRI examination
tumor, which was highly vascularized, could showed no signs of tumor recurrence (j–l).

a b

(a, b) Large retromeatal meningioma with a broad base neurovascular structures in the CP angle have been pre-
to the posterior pyramid surface and the tentorium. The served. * fifth cranial nerve, ** seventh-eighth nerves,
cerebellum is compressed, but the fourth ventricle is not *** lower cranial nerves. (g–h) Routine follow-up CT
occluded. (c, d) The images present the steps of tumor revealed an intracerebellar hematoma on the right side.
removal. (c) Initial exposure in the CP angle. (d) Internal Considering its relatively small size, a decision was
debulking with CUSA and suction. (e) Dissection from taken for conservative treatment. Follow-up CT scans
the adjacent structures. (f) Electrical stimulation of the demonstrated the gradual resorption of the blood (i). (j,
facial nerve to verify its function after complete tumor k, l) T1-weighted MR imaging, performed 24 months
removal (the arrow points at the electrode probe). All later: no signs of residual tumor or recurrence
Cerebellopontine Angle Meningiomas 399

c d

e f

g h
400 7 Meningiomas

i j

k l
Cerebellopontine Angle Meningiomas 401

Premeatal and Inframeatal Meningiomas and trigeminal nerve, the second level between
Premeatal meningiomas are located medial to the trigeminal and seventh-eighth nerves, the third
IAC and most frequently displace the facial and level between seventh-eighth nerves and lower
cochlear nerves posteriorly and inferiorly (in 45 cranial nerves, and the lowest level, defined by the
and 43 %, respectively) [66]. Some premeatal lower cranial nerves and foramen magnum. The
meningiomas expand more medially toward the tumor is approached initially via the most
petroclival junction and may extend into the expanded level, for example, the level with the
Meckel’s cave or even supratentorially. Other widest distance between the respective structures
tumors extend caudally toward the jugular fora- (see Case Illustration 7.4). Resection of the petrous
men and foramen magnum or laterally along the apex (the suprameatal extension of the retrosig-
posterior petrous surface. These meningiomas moid approach—see the section “Petroclival
may invade the IAC (Fig. 7.17) or remain entirely Meningiomas”) may be necessary to achieve
in the CP angle cistern. sufficient exposure of the tumor attachment
Similar to the technique used in petroclival (Fig. 7.18). Inframeatal meningiomas extending
meningiomas, the surgeon has to approach the to the foramen jugulare or foramen magnum level
tumor between the cranial nerves via one of the can be removed via a similar retrosigmiod craniec-
CP angle levels, as viewed from a retrosigmoid tomy with/without additional C1 hemilaminec-
perspective: the upper level between tentorium tomy/laminectomy [85]; see Case Illustration 7.5.
402 7 Meningiomas

a b

c d

f
e

Fig. 7.17 Premeatal meningioma with limited growth enhanced (b, c). T2-weighted (d). T2 CISS (e). (f) MR
into the IAC: only the medial part of the canal is involved. cisternography. The seven to eighth nerve complex is dis-
T1-weighted native MRI (a). T1-weighted contrast- located laterally
Cerebellopontine Angle Meningiomas 403

a b

c d

Fig. 7.18 The complete removal of this premeatal men- (c)—from the brain stem to the Meckel’s cave. (d) Early
ingioma (a, b) required resection of the suprameatal postoperative CT control. (e) The patient at 6-month fol-
tubercle. Note the wide exposure of the trigeminal nerve low-up examination
404 7 Meningiomas

Case Illustration 7.4 nerve could be preserved. Targeted opening of


Premeatal Meningioma with Perifocal the IAC was performed to remove the intrac-
Edema analicular tumor part, while preserving the
This 55-year-old male patient was operated 5 facial and cochlear nerves. During the whole
years earlier in another neurosurgical facility surgery, the BAEP remained stable. The men-
for a CP angle meningioma. The tumor was ingioma was very adherent to the brain stem
removed partially, and the rest was followed but—despite the radiologically visualized
with regular MRI examinations. A growth extensive peritumoral edema—did not invade
tendency was demonstrated, and 3 years later, the pia. Thus, complete removal of the tumor
the tumor was treated with gamma knife. Over with preservation of all neurovascular struc-
the following 2 years, further tumor growth tures was achieved. Histopathologically the
was evident. At presentation to our depart- tumor was WHO grade I meningothelioma-
ment, he had a hypesthesia on the right side of tous meningioma.
the face (V1–V3), right-sided anacusis, and The CT examination performed several
severe gait difficulty. MRI demonstrated a hours after surgery because the patient was
relatively large tumor with largest diameter of drowsy showed blood collection in the tumor
3.5 cm with imaging characteristics, compati- bed area and severe compression of the
ble with those of a meningioma. The tumor fourth ventricle (c). The dilation of the lat-
was located premeatally, had a small exten- eral and third ventricles was more pro-
sion into the IAC, and caused significant com- nounced when compared to the preoperative
pression of the brain stem, as well as distortion CT scans, and a decision was taken in favor
of the fourth ventricle (a, b). T2 sequences of placing an EVD (d, e). Immediately there-
demonstrated an intense peritumoral edema, after, the patient regained his consciousness
involving both the brain stem and cerebellum. completely. Over the following days, he
Although the ventricles were slightly dilated, made rapid recovery and was successfully
the patient had no symptoms/signs of intracra- weaned from the EVD, which was removed
nial hypertension. on the eighth postoperative day. At discharge,
The meningioma was very vascularized his gait was much more stable, but he had an
and had its attachment lateral and inferior to HB grade IV facial palsy. The patient was
the Meckel’s cave. Medially it extended up to seen 6 months later: his gait had recovered
the oculomotor nerve. In order to remove the almost completely, and the facial nerve func-
part in the Meckel’s cave, the petrous apex tion was HB grade III. MRI at 6 months
(the suprameatal tubercle) was drilled off. The demonstrated no pathological finding (f).
tumor capsule was found to be strongly adher- Twelve months after the operation, he had
ent to the trigeminal nerve fascicles. Still, it normal gait and very good facial nerve
could be dissected, and the integrity of the function (HB grade II).
Cerebellopontine Angle Meningiomas 405

a b

c d

T1-weighted contrast-enhanced (a) and T2- weighted tumor bed area and almost complete occlusion of the
MR images of the tumor (b). Note the extensive peri- fourth ventricle. (d) Dilatation of the lateral ventricles;
focal edema on the T2 sequence. (c) CT performed (e) CT after the insertion of the EVD. (f) MRI per-
after the surgery, demonstrating blood collection in the formed 6 months later: no pathological findings
406 7 Meningiomas

e f
Cerebellopontine Angle Meningiomas 407

Case Illustration 7.5 he had severe gait ataxia, urine incontinence,


Inframeatal Meningioma and complained of strong headache.
This 70-year-old male patient has been diag- Audiometry revealed nonfunctional hearing
nosed to have a CP angle tumor 3 years prior on the side of the tumor. Considering the
to presentation to our institute. He decided size of the lesion and the progressive wors-
against “early” surgery in favor of a wait- ening of his condition, a decision for opera-
and-see strategy. During the last several tive treatment was taken. Due to the presence
months, however, he felt progressive gait of hydrocephalus and the clinical signs of
instability and headache. MR examination intracranial hypertension, external ventricu-
demonstrated significant tumor growth with lar drainage was inserted prior to tumor sur-
obstructive hydrocephalus (a–f). At admission, gery. The tumor was approached via the

a b

e f

Preoperative MRI, demonstrating a large premeatal meningioma, causing obstructive hydrocephalus. (a–c)
T1-weighted contrast-enhanced images. (d–f) CISS sequences
408 7 Meningiomas

standard RS craniotomy, decompressed hence, the dura in the area was intensely
internally, and dissected from the surround- coagulated.
ing structure. It encased the vertebral artery After surgery, the patient had swallowing
and dislocated the lower cranial nerves cau- difficulty and developed aspiration pneumonia,
dally. Small tumor portions entered the jugu- which had to be treated with antibiotics over a
lar foramen and the IAC. Following removal period of 2 weeks. Gradually his condition sta-
of the CP angle tumor part, the IAC was bilized, and a tendency toward improvement of
opened, and the intracanalicular extension the lower nerve function was noted. The EVD
was also resected. Then, the part in the jugu- was removed on the 12th postoperative day.
lar canal was approached, and complete Later on, the nasogastric tube was removed, and
removal with preservation of all neurovascu- peroral feeding was started. At discharge, he
lar structures was achieved. The tumor was fully mobilized: the gait was considerably
matrix was inferior and medial to the IAC; better when compared to the preoperative level.
Cerebellopontine Angle Meningiomas 409

Meningiomas Involving the IAC IAC (retromeatal) in 11 % [66]. It is still unclear


Meningiomas may arise primary or extend sec- why some tumors tend to extend into the canal,
ondary into the IAC [65, 83, 101]. It has been while others remain purely cisternal (Fig. 7.19).
estimated that 14–20 % of all CP angle menin- Approximately 10 % of all CP angle menin-
giomas have an intracanalicular part [10, 83, 101]. giomas arise from the dura of the IAC and gradu-
The extracanalicular tumor part in the senior ally extend toward the CP angle [10, 90]. They
authors’ series was found to be located anterior to are believed to originate from intrameatal arach-
the IAC (premeatal) in 38 %, superior to the noid cell clusters [65]. In a widely cited study,
IAC (suprameatal) in 22 %, inferior to the Nager and Masica [65] described such arachnoid
IAC (inframeatal) in 29 %, and posterior to the clusters not only along the dural sinuses and in

a b

d
c

Fig. 7.19 The variable extension patterns of CP angle intracanalicular extension and dilatation of the IAC. (e, f)
meningiomas in regard to the IAC are illustrated on these The intracanalicular part of this tumor extends laterally up
images. (a, b) Meningioma centered at the IAC without to the fundus of the IAC. (g, h) Petroclival meningioma
any intracanalicular growth. (c, d) Meningioma with with a limited intracanalicular extension
410 7 Meningiomas

e f

g h

Fig 7.19 (continued )

the area of Meckel’s cave but also in the IAC, in hearing (Hannover classes H1–H2), however, had
the jugular foramen, along the greater petrosal 54 % of all patients. Tinnitus had 42 %, vertigo
nerve, and in the vicinity of the geniculate had 35 %, ataxia had 22 %, trigeminal neuralgia
ganglion. had 17 %, headache had 17 %, and facial nerve
In the seniors’ author series, the mean diame- palsy had 8 %. Grey et al. reported that 45 % of
ter of meningiomas extending into the IAC was their patients with gross intrameatal tumor exten-
28 and 34 mm in male and female patients, sions were deaf preoperatively, compared with
respectively [83]. The average symptom duration 5 % of patients in whom the IAM was not
prior to diagnosis was 48 months. At presenta- involved.
tion, the most frequent neurological finding was Neuroimaging reveals the typical characteris-
related to cochlear nerve dysfunction: some tics of CP angle meningiomas and shows the
degree of hearing loss had 72 %. Functional extent of IAC involvement and the changes of
Cerebellopontine Angle Meningiomas 411

surrounding bone structures. Such meningiomas wall of the canal should be opened sufficiently—
are difficult to differentiate from VSs due to generally approximately 180° of its circumfer-
their similar MR and CT signal intensities and ence—in order to provide an unobstructed view
degree of contrast enhancement. Important of the tumor. In case of meningioma arising from
clues, if available, are the dural tail sign, the en the dura of the IAC, complete excision of the
plaque growth, and the bone hyperostosis, typi- dura in that area should be attempted. See Case
cal for meningiomas. Meningiomas, further- Illustration 7.6.
more, rarely cause dilatation of the IAC. In the In the series, the IAC was opened to achieve
seniors’ author series, dural tail was evident in tumor removal in 16 patients, while in 19
21 %, petrous bone hyperostosis in 13 %, and patients, the intracanalicular tumor portion could
bone erosion in 20 % of all meningiomas with be removed completely without opening of the
IAC extension. En plaque appearance had 6 % of canal. Radical removal of all affected bone adja-
the tumors [83]. cent to the tumor site of origin is recommended
The necessity and the extent of IAC opening, to prevent recurrences in cases of severe preop-
as in case of VS, are determined by the extension erative facial and cochlear nerve dysfunction. In
of the tumor in the canal, its adherence to neural such patients, surgical radicality may necessitate
structures, and the preoperative cranial nerve even complete resection of the infiltrated nerve
function. In patients with normal preoperative and its primary reconstruction in the CP angle
cranial nerve function, every attempt should be (see section “Facial Nerve Reconstruction”).
made to avoid new deficits. The attempt to dis- The decision to sacrifice a normally functioning
sect very adherent tumors from the nerves could nerve should be taken on an individual basis.
cause neurological morbidity. Following this Factors to consider are the patients wish, the
concept, in 2/20 patients with meningiomas orig- possibility to achieve radical surgery, and the
inating from the IAC, the canal was left intact, tumor grade.
while in the remaining 18/20 patients, it was Generally, meningiomas arising from the dura
opened to achieve more complete resection or to of the IAC are infiltrative or, at least, are very
decompress the neurovascular bundle [83]. The adhesive, whereas the degree of secondary intra-
latter option is used to gain functional time in canalicular tumor extension does not affect clini-
case the cranial nerves are infiltrated. cal outcome so strong. In our series, 83 % of the
Targeted opening of the posterior wall of the patients with HB grade I or II preoperative facial
IAC is performed with the high-speed drill function had similar function immediately after
(Fig. 7.20). Meningiomas with limited intrac- surgery. Twenty-five percent of the patients
analicular extension can be safely removed by maintained normal hearing after surgery, two
drilling off just few millimeters of the posterior patients improved from preoperative hearing
lip of the IAC. Wide opening of the IAC is class H2 to normal hearing, and 32 % of those
required to remove meningiomas extending more with preoperative class H2 retained hearing at
laterally. Thin-slice bone-window CT provides the same level. Interestingly, hearing recovered
the information necessary for the precise plan- in 1 of the 11 preoperatively deaf patients.
ning of this step. Important issues to consider are Complete tumor resection (defined as Simpson
the degree of bone pneumatization and the loca- grades I and II) could be achieved in 86 % of the
tion of the jugular bulb and of the inner ear struc- cases. Facial nerve function was significantly
tures. The dura behind the IAC is incised better in case the meningioma involved the IAC
circumferentially, stripped off, and excised. secondarily. The extent of resection and hearing
Large-sized diamond burr (4–5 mm in diameter) outcome, however, did not correlate significantly
is used initially and changed to smaller in the to tumor origin [83]. At long-term follow-up,
depth. Bone is drilled off layer by layer in 1-mm four patients had recurrences (mean follow-
increments, and the last bone layer or lamina is up—60 months). Two of them had atypical
removed with a small platelet knife. The posterior meningiomas.
412 7 Meningiomas

a b

c
d

Fig. 7.20 Small premeatal meningioma with intracanali- (c) The IAc has been opened, and the tumor is mobilized.
cular extension. (a) MR image. (b) Initial view of the CP (d) Complete tumor removal. All essential structures have
angle tumor part ventral to the seventh-eighth nerves. been preserved
Cerebellopontine Angle Meningiomas 413

Case Illustration 7.6 rological examination was normal besides the


Premeatal Meningioma with Intracanali- hypacusis (PTA of 50 dB). MRI revealed a CP
cular Extension angle meningioma with lateral extension up to
The patient (32-year-old female) presented the fundus of IAC. The patient was operated
with a history of decreased hearing and ver- via the RS approach. Once CSF was drained
tigo since approximately 6 months. Her neu- from the lateral cerebellomedullary cistern

a b

(a–c) Preoperative MRI of the patient: (a, b) (f) The dura around the posterior lip of the IAC had
T1-weighted sequences after contrast administration. been coagulated. The dotted line depicts the area of the
(c) T2-weighted sequence. The tumor extends laterally petrous bone to be drilled off (arrowhead seventh-
up to the fundus area, as demonstrated on the T2 eighth nerve complex). (g) The posterior wall of the
sequence: no CSF is seen lateral to the tumor. (d) Initial IAC has been removed widely, the dura was excised,
view of the CP angle, demonstrating the tumor and its and the intracanalicular meningioma part was exposed
relations (Tu tumor, P posterior pyramid, R retractor, C and gradually removed (h). (i) The tumor has been
cerebellar hemisphere). (e) Removal of the retro- and removed completely, and the neural and vascular struc-
inframeatal tumor parts (arrow—lower cranial nerves). tures were preserved (arrow—lower cranial nerves)
414 7 Meningiomas

d e

f g

h i
Cerebellopontine Angle Meningiomas 415

and the retractor was placed, the tumor, which was widely removed with the high-speed drill
covered the seventh-eighth nerve complex with diamond burrs. The intracanalicular
and—partially—the lower cranial nerves, was tumor part, which was located posterior and
seen. The meningioma originated from the inferior to the nerve complex, was then
dura anterior to the porus and extended like a removed completely. The integrity of the
carpet premeatally, inframeatally down to the nerves, as well as their vasculature, was pre-
jugular foramen, suprameatally, and slightly served. After thorough hemostasis, the IAC
retromeatally. Its retro- and inframeatal parts was sealed off with fat pieces and fibrin glue.
were dissected from the lower cranial nerves Following surgery, the patient did not have
and removed. A large tumor part was seen to new neurological deficits and recovered
extend along the seventh-eighth nerves into quickly. The audiogram showed an improve-
the IAC—its dissection, however, was post- ment of the hearing (PTA of 40 dB), which
poned until the canal was widely opened. The was stable and confirmed at follow-up exami-
dura behind the IAC was coagulated, incised nations. Histopathological examination
circumferentially around the porus of the showed that this was a case of lymphoplasma-
canal, and excised. Then, the posterior wall cyte-rich WHO grade I meningioma.
416 7 Meningiomas

Intracanalicular Meningiomas Meningiomas rarely cause widening of the IAC


Meningiomas confined to the IAC or “intrac- but induce more frequently petrous bone or IAC
analicular meningiomas” are extremely rare hyperostosis.
[8, 49, 69]. Their clinical and neuroradiological The relationship of the tumor to the seventh
features are very similar to those of VSs, and in and eighth cranial nerves is less reliably predict-
most cases, the correct diagnosis is made only able in meningiomas than in VSs. The direction
at surgery. The initial tumor manifestation is of nerve displacement is related to the site of
usually decreased hearing. At presentation, the tumor origin. Another fact to consider is that
majority of patients have vestibulocochlear meningiomas tend more frequently to be severe
nerve disturbances. In the series of the senior adherent or to infiltrate these nerves [7, 69].
author, including eight patients with intrac- Asaoka et al. [7] pointed that the adhesions
analicular meningiomas, tinnitus had 88 %, between the meningioma and facial nerve are
hearing loss of variable degree had 75 %, and much more difficult to separate, than in VS, even
dizziness and vertigo had 63 % [69]. In contrast in case of smaller tumors.
to VSs, meningiomas can cause facial nerve The operative strategy and technique used in
symptoms, including facial paresis or hemifa- surgeries of intracanalicular meningiomas do not
cial spasm [7, 69]. differ from those in case of meningiomas involv-
Preoperative differentiation between these ing secondarily the IAC (see Case Illustration
meningiomas and VSs is difficult because they 7.7). The goal is complete tumor removal and
have similar neuroradiological characteristics. functional preservation of all cranial nerves. In
Still, some findings could help their differentia- our series, complete tumor removal was achieved
tion. Meningiomas and VSs have similar signal in all patients without major morbidity. Facial
intensities; on T2-weighted images, however, and cochlear nerves preserved anatomically in all
VSs tend to show higher signal intensity than cases; one patient had postoperative facial weak-
meningiomas. The visualization of intratumoral ness that resolved completely. In 71 % of the
calcification, the type of bone changes, the gen- patients, hearing was preserved. Interestingly, in
eral shape of the tumor, and/or the presence of three of the seven patients with tinnitus at presen-
“dural tail” may be further helpful clues, tation, the symptom resolved, in three patients it
although these findings are nonspecific [7, 46]. improved, and only in one it persisted.
Cerebellopontine Angle Meningiomas 417

Case Illustration 7.7 however, was not dilated. All management


Intracanalicular Meningioma options were discussed with the patient, but he
This 47-year-old patient presented with a his- decided for surgery.
tory of right-sided hearing loss since more The CP angle was exposed via an RS route. In
than 10 years. In the last months, he suffered the area of the porus of the IAC, a small protrud-
from annoying tinnitus and headache on the ing tumor part was seen, which appeared like a
right side. His neurological examination was meningioma. Frozen section confirmed this sus-
normal besides the right-sided hypacusis. CT picion. The strategy of its removal was similar to
and MRI demonstrated a pure intracanalicular the one applied in case of intracanalicular VSs:
tumorous lesion with imaging characteristics, wide opening of the IAC up to its fundus, preser-
compatible with those of VS (a–f). The IAC, vation of the inner ear structures, excision of the

a b c

(a, b) Bone-window CT of the patient: normal IAC on after contrast application. (e, f) T2-weighted sequences.
both sides. (c–f) MRI clearly demonstrated the pure (g, h) MRI preformed 12 months after surgery: no
intracanalicular tumor. (c, d) T1-weighted sequences remnant or recurrent tumor is detectable
418 7 Meningiomas

g h

dura of IAC, identification of the cranial nerves, waves III and V could be recognized but had still
and stepwise dissection of the meningioma from long latency and low amplitude.
surrounding structures. The meningioma, includ- The patient had no new neurological deficits
ing the dura from which it originated, was after surgery, and the hearing level remained at
removed completely, and the anatomical integ- the same nonfunctional level. Histopathological
rity of the facial and cochlear nerves was pre- diagnosis was psammomatous meningioma
served. The severely impaired BAEP showed WHO grade I. Follow-up MRI (g, h) confirmed
slight improvement after tumor removal: the the completeness of tumor removal.
Cerebellopontine Angle Meningiomas 419

Intraosseous Meningiomas and outer cortical layers of the skull [1]. The MRI
Extradural meningiomas comprise 1–2 % of all findings of intraosseous are similar to those of
meningiomas [25]. Primary intraosseous are intradural meningiomas: they are typically
those extradural meningiomas that arise within hypointense on T1-weighted images and hyper-
the bone [80]. Intraosseous meningiomas of the intense on T2-weighted images with intense
skull base are extremely rare. Most are osteoblas- homogeneous enhancement after contrast appli-
tic or mixed osteoblastic–osteolytic lesions, while cation. Although the classical dural tail sign is
purely lytic lesions are the least common not seen, contrast enhancement of the underlying
[1, 50, 82]. Such tumors may be purely intraosseous dura may be noted [5]. The differential diagnosis
without evidence of dural invasion or may have of intraosseous meningiomas includes metasta-
additional soft tissue component [82]. In order to sis, epidermoid tumor, multiple myeloma, eosino-
classify a tumor with soft tissue component as a philic granuloma, fibrous dysplasia, chordoma,
primary extradural meningioma, its origin and chondrosarcoma, brown tumor, skull dermoid,
center should be located outside the dura [48]. Of giant cell tumor, hemangiopericytoma, and
note, clear-cut criteria still do not exist [25]. hemangioma [82].
Skull-base intraosseous meningiomas are gen- Treatment of choice of such meningiomas is
erally slow-growing, painless tumors. Due to their wide surgical excision. In the skull base,
their location, they cause cranial nerve deficits however, this is rarely possible, and maximal
and/or cosmetic deformation. On CT, the osteo- tumor removal with decompression of vital neu-
blastic intraosseous meningioma-induced hyper- ral structures is performed (see Case Illustration
ostosis is demonstrated as focally thickened 7.8). Tumor remnants are followed and in case of
hyperdense lesion. The osteolytic lesions cause biological activity are subjected to radiation
thinning, expansion, and interruption of the inner therapy.
420 7 Meningiomas

Case Illustration 7.8 nerve dysfunction with hoarseness and dys-


Intraosseal Meningioma phagia, weakness of the sternocleidomastoid
Twenty-seven-year-old male patient pre- muscle, and hemitongue atrophy on the right
sented with a history of diplopia for more side. MRI showed a contrast-enhancing
than 9 years. In the last 3 years, progressive lesion of the medial petrous bone, extending
swallowing difficulties, vertigo, and inter- medially to the clivus (a–d). The tumor had
mittent headache developed. At presenta- a large soft tissue component in the lower
tion, the patient had trigeminal hypesthesia CP angle, causing compression of the lower
(V2), abducens nerve palsy, lower cranial pons and of the medulla. On CT, the lesion

a b

c d

Preoperative MR images of a patient with an intraos- tures was not extirpated in order to avoid major
seal petrous meningioma. (a) T2-weighted. (b–d) deficits. (f) MR venography: occlusion of the lateral
T1-weighted sequences after contrast administration. transverse and of the sigmoid sinuses on the side of
(e) Postoperative MRI. A small part of the menin- surgery; patent contralateral sinus
gioma that was most adherent to surrounding struc-
Cerebellopontine Angle Meningiomas 421

e f

had a mixed osteoblastic and osteolytic The patient suffered an epileptic fit shortly
appearance. after extubation, and an early cranial CT scan
The patient was operated via a transmastoid was performed. Thrombosis of the sigmoid
presigmoid approach. Frozen section examina- and of the lateral segment of transverse
tion suggested a WHO grade I meningioma. The sinuses was suspected and confirmed on the
tumor was partially hard and fibrous with ossified MR venography (f). Obviously the contralat-
parts. Initially the intraosseal tumor part was eral sinus was patent because no signs of
removed; the dura was found to be infiltrated and venous infarction or edema were detected.
was opened to inspect the CP angle cistern; how- Appropriate therapy was initiated, and the
ever, no signs of dural penetration were found. patient recovered completely. Following sur-
Considering the extension of the lesion and the gery, his hearing level remained unchanged,
dense adherence to surrounding structures, com- but a facial palsy (HB grade IV) developed.
plete tumor removal and preservation of the nor- At discharge, the facial nerve function
mal facial nerve function and hearing was deemed improved to HB grade III, and the patient
impossible. Moreover, the petrous carotid was noted an improvement in swallowing.
encased by the tumor. The most densely adherent Histopathological diagnosis was WHO grade
tumor parts were, therefore, not removed (e). I sclerosing meningioma.
422 7 Meningiomas

Postoperative Management The outcome of surgery depends mainly upon


The patients should emerge smoothly from anes- the location and consistency of the tumor and to a
thesia with minimal agitation, and coughing lesser extent upon its size [10, 90]. In premeatal
should be prevented. Invasive arterial blood pres- meningiomas, the preservation of facial and audi-
sure monitoring is performed for at least 24 h: tory function is less likely. Two further important
major elevation of blood pressure should be factors are the extent of skull-base involvement
avoided, especially during the early postopera- and the adherence of the tumor to brain stem, cra-
tive period. In case the patient is expected to have nial nerves, and/or vessels [98]. In the large recent
lower cranial nerve deficit after surgery, he series, the rate of total removal is above 80 %,
should be extubated only when fully awake and and the mortality is below 5 %.
cooperating (see Chap. 10). Patients that remain Nakamura et al. [66] evaluated 421 patients
intubated (preferably nasally) should be mini- with CP meningiomas operated by the senior
mally sedated, which allows estimation of their author. They found that the radicality of tumor
level of consciousness and early detection of removal as well as the functional and general out-
possible intracranial complications. If deeper come correlated with the topographic location of
sedation is required, the medication should be these tumors. The overall rate of complete
decreased at regular intervals (e.g., every 2 h), removal (Simpson grades 1 and 2) was 86 %.
and the patient should be examined. CT is per- Considering the tumor subtypes, highest rate was
formed routinely in all patients the day after achieved in suprameatal meningiomas (90 %), in
surgery. retromeatal meningiomas (89 %), and in those
We perform steroid treatment (dexametha- involving the IAM (88 %). The rate was lowest in
sone) for at least 3 days after surgery. Steroids are premeatal (83 %) and in inframeatal tumors
known to inhibit vasogenic edema, to reduce (78 %). The outcome was worse in cases that
cytokines expression by the tumor cell, and to arose in or at the IAC [66].
inhibit the edemogenic response of the brain In the series, good facial nerve function
microvascular. Furthermore, they stabilize the (House–Brackmann grades 1 or 2) was observed
endothelial cell membranes [62]. in 89 % of the patients. The rate of preserved nor-
Early MRI should be made to document the mal facial nerve function was highest in ret-
extent of tumor removal. In case of incomplete romeatal meningiomas (90 %) and lowest in
resection, follow-up MRI studies are performed premeatal meningiomas (76 %). The rate of pres-
3–6 months after surgery, and radiosurgery/radio- ervation of functional hearing could be increased
therapy or follow-up of the residual tumor is rec- from 88 % (earlier experience [58]) to 91 % in the
ommended accordingly. recent series [66]. The rate of hearing preservation
was highest in patients in the suprameatal and ret-
Outcome romeatal subgroups (100 and 73 %, respectively)
The postoperative cranial nerve outcome is better and lowest in those in the premeatal subgroup
in meningiomas than in VS because meningiomas (43 %). Postoperative hearing improvement
generally do not invade but rather compress the occurred in 8 % of our patients. In 2 % even recov-
seventh and eighth cranial nerves. Removal of ery from preoperative deafness was observed [66].
the tumor can even lead to improvement of cra- Interestingly, none of the seven patients present-
nial nerve function [58]. Currently, facial nerve ing with sudden hearing loss demonstrated post-
function can be preserved in the majority of operative improvement. Bassiouni et al. [10]
patients [10, 11, 66, 101, 106]. Anatomic preser- reported on the positive effect of surgery on tinni-
vation of the seventh and eighth cranial nerves tus. They found that tinnitus was a major com-
was reported in 94 %, and functional preserva- plaint in 18 % of the patients preoperatively but
tion of the seventh and eighth cranial nerves was was ameliorated in all of them postoperatively.
reported in 86 and 77 % of CP angle menin- Lower cranial nerves, however, are more vulnera-
giomas, respectively [57, 66]. ble, and more aggressive dissection or manipulation
Cerebellopontine Angle Meningiomas 423

of the arachnoid layer might cause new postopera- between 3.2 and 23 % among the different series
tive deficits [97]. but is usually less than 10 % [6, 36, 91, 98, 101].
Meningiomas are increasingly frequently The extent of tumor removal is well known to cor-
diagnosed in elderly patients. Our opinion is that relate to the risk of recurrence. Total tumor
surgery is the most appropriate management removal, however, does not preclude recurrences
option even in this age group, provided that the [6, 98].
patient is in good general condition. Although the If the recurrence shows a growth tendency or
growth rate of meningiomas in elderly is lower becomes symptomatic, surgery should be consid-
when compared with younger patients, most of ered as the primary management option. Complete
them have a certain growth tendency [68]. In tumor removal should be the goal, if safely pos-
medically unstable patients, radiotherapy (frac- sible. Generally, the chances to remove the tumor
tionated radiotherapy or radiosurgery) is an completely without causing new major deficits
acceptable alternative. Incidentally detected are lower than in case of primary surgery. In
small tumors or tumors causing minimal symp- recurrent meningiomas, the surgery-related mor-
toms may be followed with regular radiological bidity risk is higher. An anatomical dissection
and clinical examinations. The Senior authors’ plane is usually absent; the tumor tends to be
experience encompasses 25 operated patients more adherent to the brain and to encase nerves
with CP angle meningiomas aged 70 or more. and vessels. Radiotherapy is an alternative man-
The outcome was related mostly to the preopera- agement option of large recurrences that are not
tive neurological and clinical condition of the considered amenable to operative removal. Small
patient. Complete resection (Simpson grade 1 recurrent tumors can be followed and—in case of
and/or 2) was achieved in 84 % of all cases, and further growth—may be treated with radiosur-
the general and functional outcome was similar gery or fractionated radiotherapy [31, 95].
to that in younger patients. Elderly patients were
more prone to have lung infections. Furthermore, Atypical and Anaplastic Meningiomas
the CSF leak rate was higher than in younger Atypical and anaplastic meningiomas comprise
ones. Generally, the length of hospital stay was approximately 5 % from all meningiomas and
longer in elderly patients, but the difference was have a slight male predominance [104]. They
not significant [68]. are approximately 2 % of posterior fossa
and 18–31 % of all skull-base meningiomas.
Recurrence Characteristic for this small subgroup is the
CP angle meningiomas have higher tendency more aggressive biological behavior with a ten-
toward recurrence if compared with VSs and may dency for early recurrence even after complete
recur even late after surgery [10, 90, 91, 98, 101]. tumor removal [35, 42, 45].
Patients should be therefore regularly followed, These tumors are believed to develop de novo
both clinically and radiologically. Radiologic or progress from benign to atypical or anaplastic
follow-up interval for MRI is set between 3 variants. Tumor progression, however, is rarely
months and 1 year for malignant and benign observed in skull-base meningiomas [3, 41].
meningiomas, respectively. Recurrent menin- Benign, atypical, and anaplastic meningiomas
giomas, especially those after radiotherapy, may cannot be reliably differentiated radiologically.
show a milder contrast enhancement than the pri- Some features, such as marked edema, heteroge-
mary lesion, probably due to increased fibrosis in neous appearance, irregular or nodular cerebral
older tumors. surface, mushrooming on the outer edge of the
The likelihood of recurrence correlates with lesion, extensive bone destruction, and absence
meningioma grade, extent of resection (complete of calcification, are more frequently seen in atyp-
vs. incomplete), and—according to some studies— ical and anaplastic meningiomas but are
to the loss of progesterone receptor immunostaining nonspecific [41, 54, 108]. Recent studies indi-
[39, 77]. For WHO grade I meningiomas, it varies cated that the diffusion-weighted MRI, perfusion
424 7 Meningiomas

MRI, and magnetic resonance spectroscopy Multiple Meningiomas


findings might be used to distinguish menin- Patients with NF-2, in addition to the characteris-
gioma subtypes, but the results have been incon- tic bilateral VSs, often have other tumors, such as
clusive [63]. cranial nerve schwannomas, spinal schwanno-
Recurrence rates of atypical and anaplastic mas, and meningiomas [60] (see section
meningiomas are very high, irrespective of their “Neurofibromatosis Type 2”). Meningiomas, fre-
management mode. The recurrence rate is high quently multiple, are the second most frequent
even after radical tumor removal: depending on tumor type in NF-2. They occur in 38–58 % of
the length of follow-up in different series, it is these patients [29, 34, 76].
between 50 and 95 % [55]. The 5-year local The term “multiple meningiomas” denotes the
tumor control rate following radiosurgery in the clinical entity, in which the patient has two or
study of Stafford et al. [95] was 68 % in atypical more independently situated meningiomas, not
and 0 % in anaplastic meningiomas, treated with necessarily of the same pathologic subtype
radiosurgery. In another study, the 3-year survival [12, 26]. Patients with multiple meningiomas
rate associated with linear accelerator-based may not have (or still not have) other features of
radiosurgery of atypical and anaplastic menin- NF-2, such as multiple schwannomas. The
giomas was 24 and 14 months, respectively [38]. reported incidence of patients harboring multiple
Best results are achieved with a multimodality meningiomas not related to NF-2 varies: 1–12 %
management, including radical surgery and in surgical series, 8–16 % in autopsy studies of
adjunctive radiation therapy either with fraction- meningiomas, and 1–9 % in radiological series
ated radiotherapy, stereotactic radiosurgery, or [4, 27, 34]. As noted by Goutagny and Kalamarides
proton beam irradiation [63, 71, 75]. The primary [34], however, NF-2 patients are difficult to seg-
treatment of such tumors should be their com- regate from other multiple meningioma patients,
plete operative removal, including the infiltrated and multiple meningiomas as an entity largely
bone and/or soft tissue. Every patient with ana- overlap with NF-2. In the population study of
plastic meningioma, regardless of the degree of Antinheimo et al. [4] in the absence of classic
resection, should receive adjuvant fractionated NF-2, 4 % of all patients with meningiomas had
radiotherapy soon after resection. Management multiple meningiomas. Somatic mosaicism, fre-
of atypical meningiomas is related to the extent quently seen in NF-2, is the proposed explanation
of their resection: follow-up is recommended in for developing of multiple meningiomas.
case of complete resection, while early adjuvant Meningiomatosis is a less clearly defined term; it
fractionated radiotherapy is indicated in subto- is used either to denote all patients with multiple
tally resected cases. Modha et al. [63] recom- meningiomas or only those with more pro-
mend postoperative fractionated radiotherapy nounced clinical syndrome.
even in case of complete removal of atypical In some patients, especially in children,
meningioma if the tumor was found to invade the meningiomas may be the first sign of NF-2
brain. In both subtypes, radiosurgery may be con- [84]. Patients with multiple meningiomas
sidered as an alternative to fractionated radiother- should be hence investigated to exclude NF-2:
apy in case of a focal or nodular remnant. detailed family history analysis, dermatologi-
Radical surgery is the primary management cal, ENT and ophthalmological examinations,
option for recurrences, provided the tumor is as well as MRI of the whole neuraxis should be
resectable without very high operative risks. performed.
Fractionated radiotherapy should be performed Meningiomas in NF-2 patients develop less
in patients who have not received it before. frequently in the skull base and, respectively, in
Currently, effective chemotherapy is not avail- the CP angle [4]. Their management poses
able, and its role in the management of atypical significant problems. Major issue is the lifelong
and anaplastic meningiomas remains unclear propensity for development of new neoplasms
[45, 63]. and the respective impossibility for definitive
Cerebellopontine Angle Meningiomas 425

healing. Treatment is focused on life prolonga- structures and—if possible—to remove the tumor
tion, preservation of cranial nerve function, and completely. Preservation of neural function has a
maintenance of quality of life. Symptomatic priority: in case the tumor infiltrates or is very
meningiomas, as well as progressively growing adherent to a cranial nerve, no attempt should be
tumors, are best treated surgically. The goal is to made to remove it completely. See: Case
achieve decompression of essential neural Illustrations 7.9 and 7.10.
426 7 Meningiomas

Case Illustration 7.9 and the anterior and the middle cranial arteries
Multiple Meningiomas and extended toward the tentorial incisura,
Fifty-year-old female patient presented with compressing the right oculomotor nerve. The
MRI findings of an extensive meningioma of patient had visual field defect and severe
the anterior and middle cranial fossae (right > visual loss (visual acuity on the right side, 0.3;
left) with engulfing of both optic nerves and left side, 0.9), as well as partial oculomotor
the optic chiasm (a–c). The parasellar tumor nerve palsy. She was operated via a frontolat-
on the right side engulfed the carotid artery eral (pterional) craniotomy, and complete

(a–c) Preoperative MRI (T1-weighted sequence after (g–i) Bone-window CT scan and MRI, demonstrating
contrast administration), demonstrating the size and the petroclival meningioma, involving the cavernous
extension of the tumor. (d–f) Postoperative MRI: com- sinus. A hyperostotic/calcified part is seen on the
plete removal of the skull-base meningioma. Note the bone-window CT and on some MR sequences
convexity meningioma on the left side. Two more con- (arrows). (j–l) MR images after the removal of the
vexity meningiomas of similar size were also detected. right-sided petroclival meningioma
Cerebellopontine Angle Meningiomas 427

removal of the tumor was achieved with pres- via the RS route. It compressed the trigemi-
ervation of all neurovascular structures (d–f). nal, the seventh-eighth, and the lower cranial
The patient presented 3 years later with nerves. The abducens nerve was completely
complaints of progressive hearing loss on the engulfed by the tumor. Initial removal was
right side. Neurological examination revealed started along the lower cranial nerves until the
trigeminal hypesthesia (V1,2,3) and hearing basilar artery was reached and dissected free
loss (still functional hearing). Visual acuity on from the tumor. Then, the space between the
the right side was 3/10 and on the left side was trigeminal and the seventh-eighth nerves was
9/10. The MR examination demonstrated that used to remove the meningioma further. The
the known convexity meningiomas were abducens nerve was followed from its exit
unchanged; however, a new tumor in the right zone to the Dorello’s canal. Despite the
petroclival space, reaching the IAC and com- adherence to the cranial nerves and basilar
pressing the seventh-eighth cranial nerves, artery perforators, the tumor mass could be
was detected (g–i). The tumor was approached removed, and all neurovascular structures

b
428 7 Meningiomas

were preserved. The tumor matrix in the gothelial meningioma, WHO grade I. The last
petrous apex area was thoroughly coagulated. postoperative MR control is presented on
Histopathological tumor type was menin- figures (j–l).

c
Cerebellopontine Angle Meningiomas 429

d e

f
430 7 Meningiomas

g h

i
Cerebellopontine Angle Meningiomas 431

j k

l
432 7 Meningiomas

Case Illustration 7.10 decided to remove the larger tumor via the
Two Tentorial Meningiomas Removed via infratentorial supracerebellar approach and to
One Approach attempt removal of the smaller tumor via the
The patient presented with headache and tran- same route. The craniotomy was approxi-
sient double vision. The MRI examination mately 4 × 2.5 cm, extending from the sigmoid
demonstrated two separate tumors with imag- sinus to 1 cm to the midline. Both tumors
ing characteristics compatible with tentorial could be adequately accessed via this route
meningiomas: a small one arising from the and removed without any complications.
free tentorial edge in the area of the tentorial Histological diagnosis was WHO grade 1
incisura and a larger one, arising from the meningioma. The patient recovered quickly
middle tentorial part. The larger meningioma from surgery and was discharged without any
had both a supra- and infratentorial extension. neurological deficits.
Considering the location of the tumors, it was
Cerebellopontine Angle Meningiomas 433

a b

c d

e f
434 7 Meningiomas

g h

i j

(a–f) Preoperative MRI study of the patient with 2 ten- Initially this tumor part was removed; then the attach-
torial meningiomas. (a–e) T1-weighted sequence after ments of the tumor to the falx were incised, which
contrast application. (f) T2-CISS sequence. (g–j) allowed us to mobilize the remaining supratentorial
Intraoperative images. (g) The craniotomy extends from tumor part. This part was carefully pulled down and
the sigmoid sinus (SS) laterally to approximately 1 cm removed completely. A retractor was then placed, and
from the midline. The edge of the transverse sinus (TS) the second meningioma (arrow) could be visualized
is exposed. The dura is incised along the transverse from a more lateral perspective (i). The trochlear nerve
sinus, which facilitates its later watertight closure, and was compressed and adherent to the anterior tumor
is slightly retracted with 2 sutures. The infratentorial surface (j). Nevertheless, the tumor could be com-
part of the large tentorial meningioma is visible (g, h). pletely removed, and the structures were preserved
Petroclival Meningiomas 435

Hormonal Therapy and Chemotherapy Meckel’s cave. The location and the intimate rela-
of Meningiomas tion of these tumors to important neural and vascu-
Patients with meningiomas of any histological lar structures determine the complexity of their
grade that recur or regrow after surgery and radi- surgery and the high risk of postoperative morbid-
ation therapy, as well as high-grade tumors, are ity [115]. Most challenging are meningiomas with
potential candidates for hormonal or chemother- extensive supratentorial parasellar extension and
apy. Up to know, however, the role of chemother- involvement of the cavernous sinus, sella turcica,
apy is very restricted because the currently and sphenoid sinus.
available chemotherapeutic agents have minimal Petroclival meningiomas show various exten-
activity against meningiomas [17, 61]. Hormonal sion patterns. In the senior authors’ series, purely
therapies are also largely ineffective in patients infratentorial were just 5 % of all petroclival
within this subset of meningiomas. Studies with meningiomas; 15 % of them extended to the level
estrogen inhibitors did not show any efficacy, of tentorial notch, and 80 % had supratentorial
which probably is due to the low concentration of extension [115]. Medial tumor growth causing
estrogen receptors: they are present at low levels brain stem compression was seen in 92 % of all
in approximately 10 % of meningiomas [33]. operated cases. Thirty-three percent of menin-
Progesterone and androgen and their receptors giomas with anteromedial extension reached the
are present in approximately two-thirds of men- clivus, and 67 % extended up to the parasellar
ingiomas [47, 87] . Still, studies with the applica- space. Extension into the cavernous sinus was
tion of their antagonists failed to demonstrate any observed in 48 % of the cases: petroclival tumors
treatment effect [37]. As Norden et al. [72] point, involved the posterior part of the sinus
the lack of efficacy may be explained in part by (Fig. 7.21b), while the sphenopetroclival menin-
the loss of progesterone expression in menin- giomas occupied the whole sinus (Fig. 7.21c).
giomas with increased proliferation index and The latter involve in addition the anterior cavern-
higher histological grade. Inhibition of the growth ous sinus, sella turcica, and/or sphenoid sinus.
hormone receptors or their direct blockade and Lateral growth is defined in relation to the IAC:
the use of somatostatin agonist have been also 31 % of the tumors remained medial to IAC (pre-
utilized, but the effect was minimal [23]. meatal), and 69 % had more lateral extension
Potential molecular targets for targeted ther- (retromeatal). Anterolateral growth leads to inva-
apy in the future may be the various growth fac- sion of the Meckel’s cave (Fig. 7.21d).
tors and their receptors that are overexpressed Occasionally the tumor originates inside or at the
in some meningiomas: the platelet-derived mouth of Meckel’s cave. Such tumors may grow
growth factor, the epidermal growth factor, and further in the middle fossa between the two leaves
the vascular endothelial growth factor, as well of tentorium.
as some steps of the signal transduction path- Fifty-five percent of all tumors extended infe-
ways [72]. rior to the level of IAC, while 24 % reached the
jugular foramen, and 21 % reached the foramen
magnum. Three percent of the latter extended
Petroclival Meningiomas even down to the C1–C2 level. Very large and
giant meningiomas (approximately 15 % of all
Petroclival meningiomas account for 3–10 % of all patients) extend from the foramen magnum level
posterior fossa meningiomas and for approximately to the supratentorial region.
2 % of all meningiomas [112, 114, 116, 119, 138].
They arise in the area of the petroclival junction
[150]. Included in the group are meningiomas General Features
located medial to the trigeminal nerve (Fig. 7.21a):
originating from the clivus, from the petroclival The clinical presentation of petroclival menin-
junction, from the petrous apex, or from the giomas is often of insidious onset; patients tend to
436 7 Meningiomas

a b

c d

Fig. 7.21 MRI studies of various petroclival menin- of the posterior cavernous sinus. (c) Sphenopetroclival
giomas. (a) Petroclival meningioma: the tumor originates meningioma with a large suprameatal part. The whole
medial to the trigeminal nerve (arrow). (b) Petroclival cavernous sinus is involved by the tumor. (d) Petroclival
meningioma with supratentorial extension and involvement meningioma with extension into the Meckel’s cave

have a longer history of symptoms prior to diag- palsies. Typical for meningiomas with this loca-
nosis than those with CP angle meningiomas: on tion is the more severe involvement of the trigem-
average 30–60 months [112, 114, 136, 144, 153]. inal nerve. The most frequent neurological deficit
The clinical picture is determined by the size, in our series was trigeminal nerve dysfunction in
location, and extension of the tumor and is quite 43 % of the patients, hearing loss in 43 %, ataxia
variable. It does not, however, always correlate in 30 %, and abducens nerve palsy in 23 %.
with tumor size: some large tumors may cause Oculomotor and trochlear nerve symptoms had
relatively mild symptoms. Gait ataxia, trigeminal 3 % each. Understandably, patients with spheno-
nerve sensory loss, and headache are the most fre- petroclival meningiomas and petroclival menin-
quent initial complaints. Most common present- giomas extending into the cavernous sinus have
ing signs/symptoms are related to cranial nerve more frequently extraocular motor dysfunction.
Petroclival Meningiomas 437

Later symptoms are gait ataxia, dysphagia, characterized by progressive growth and neuro-
and long-tract signs [114]. Due to the frequent logical deterioration [116, 157]. An attempt for
compression of the brain stem, long-tract signs surgical removal is therefore justified. Van
occur more frequently in petroclival than in CP Havenbergh et al. [157] studied the natural history
angle meningiomas: motor deficits have been of 21 patients who were followed conservatively
reported in 15–57 % and sensory deficits in by the senior author for a minimum of 4 years.
15–20 % of the patients with petroclival tumors Radiological growth was demonstrated in 76 % of
[112, 114, 136, 152, 159]. the tumors, and in 63 % of them, neurologic dete-
rioration occurred.
The management of petroclival meningiomas
Management should be individualized, taking into account the
patient’s biological age, general medical condi-
A detailed radiological examination of the tumor, tion, neurological symptoms and expectations, as
including high-resolution thin-slice bone-window well as the tumor size and type. Treatment goal
CT, native and contrast-enhanced MRI, and MR should be complete resection of these benign
angio- and venography, should be performed tumors, which is the only potentially curative
before surgery. Hearing evaluation is essential in option. In case of large symptomatic tumors,
all patients. In extensive tumors with para- or there are no alternatives to surgery. An inciden-
suprasellar extension, the hormonal status should tally found small tumor in an older or medically
be evaluated with attention to the thyroid and cor- unstable patient, however, can be initially fol-
tisol levels. Ophthalmologic examinations should lowed clinically and radiologically. Patients with
be also performed. In case of more caudal tumor contraindications for surgery or those refusing
extension or existing swallowing dysfunction, operative treatment may be treated with primary
direct endoscopic examination of the vocal cords stereotactic radiosurgery or fractionated radio-
and swallowing test are necessary. therapy [129, 141, 155, 158].
DSA is generally substituted for by MR angio- The extent of tumor removal is the most
and venography. Vascular supply to petroclival important predictor of recurrences, and every
meningiomas is from arterial branches of the effort should be made to remove the tumor com-
internal carotid artery at the siphon and from the pletely at initial surgery. Still, even after complete
external carotid artery via the ascending pharyn- tumor removal, the recurrence rates can be as
geal and middle meningeal arteries [126, 142]. high as 4–26 % [114, 118, 132, 133, 152]. In one
These meningiomas displace typically the basilar large study of 150 patients with petroclival men-
artery laterally away from the side of the tumor. ingiomas, the recurrence-free survival rate after
Rarely, the artery may be displaced dorsally or complete tumor removal was 100 % at 3 years,
ventrally—depending on the exact tumor origin. but declined to 93 % at 7 years and to 85 % at 12
In case the artery is encased within the tumor, no years [138]. Preservation of neurological func-
displacement is visualized. Preoperative embo- tions and quality of life after surgery has, there-
lization of the feeding vessels is rarely needed fore, the highest priority. Moreover, small
with the exception of very large hypervascular residuals are rarely biologically active, and their
tumors. progression course is often benign. In the study
Petroclival meningiomas are located at the cen- of Natarajan et al. [138], further growth of the
ter of the skull base, adjacent to multiple cranial remnant was observed in 5 % of the patients with
nerves and vessels, which render their surgical incomplete resection, compared to 4 % recur-
treatment with acceptable mortality and morbidity rence rate after complete tumor removal. Leaving
rates extremely difficult. These tumors are a small part of the tumor or of its capsule is con-
regarded as one of the most formidable challenges sequently reasonable if the attempt to remove it
in skull-base surgery. On the other hand, their completely would cause injury to essential neural
natural history is unpredictable and frequently and vascular structures.
438 7 Meningiomas

This consideration is especially important in risk of morbidity is higher, for example, in neuro-
case of small petroclival meningiomas (<3 cm): surgical settings with less experience, stereotac-
their complete resection is potentially curable but tic radiosurgery, which leads to tumor control in
should not cause any major morbidity [139] (see 91 % in 5 and 86 % at 10 years [123], may be the
Fig. 7.22 and Case Illustration 7.11). In case the better option.

a b

c d

Fig. 7.22 Small petrous apex meningioma (53-year-old the suprameatal tubercle, complete removal of the tumor
female patient, no preoperative deficits). (a) T1-weighted and coagulation of its dural attachment (arrow trochlear
MR image following contrast administration. (b) Exposure nerve, double arrows dura attachment, TS transverse
of the tumor via the retrosigmoid approach (Tu tumor, t sinus, SS sigmoid sinus). (f) Postoperative MRI. The
tentorium, SMT suprameatal tubercle). (c–e) Resection of patient had no new neurological deficits
Petroclival Meningiomas 439

e f

Fig. 7.22 (continued)


440 7 Meningiomas

Case Illustration 7.11 upper 2/3 of the clivus and medially reached
Small Petroclival Meningioma the basilar artery.
Fifty-seven-year-old male patient presented to Due to the progressive nature of the
our outpatient department with complaints of abducens palsy, caused presumably by direct
increasing double vision to the right side for 3 tumor compression, a decision was taken for
months. Neurological examination revealed operative management via the RS route. At
right-sided abducens nerve palsy. The MRI surgery, a large loop of the AICA was seen that
study demonstrated a right-sided petroclival obstructed the approach to the trigeminal nerve
meningioma, which caused slight brain stem and to the tumor. The artery was extensively
compression (a–c). The tumor occupied the dissected from the arachnoidea and displaced

a b

(a–c) Small petroclival meningioma. Preoperative nerve complex, * tumor, arrow abducens nerve, BA
T1-weighted MR images after contrast administration. basilar artery). Note the preserved petrous vein (arrow-
(d–i) Intraoperative images, illustrating the consecutive head). (j) Follow-up MRI 6 months after the surgery
steps of tumor removal. See the text for descriptions. (5 (T1-weighted axial sequence after contrast
trigeminal nerve, 7–8 facial and vestibulocochlear administration)
Petroclival Meningiomas 441

caudally, thereby opening enough working from the abducens nerve up to its
space between the trigeminal and the seventh- dural entry point (g, h). The meningioma
eighth cranial nerves (d). At the beginning, the was removed completely, and its matrix,
abducens nerve was identified—it was actu- located in the area of Dorello’s canal, was
ally encased by the meningioma (e). The distal coagulated (i).
part of the nerve at the Dorello’s canal and the The patient had no new deficits after sur-
basilar artery were also identified (f). The gery. Histopathological diagnosis was menin-
trigeminal nerve was slightly elevated by the gothelial meningioma WHO grade I. At
tumor, which had a small extension into the 3-month follow-up examination, he reported a
Meckel’s cave. Once the relations of the men- significant improvement of the double vision,
ingioma to all important surrounding struc- and at 6 months, the function of the abducens
tures were revealed, internal debulking was nerve was completely normal. The follow-up
initiated, followed by dissection of the capsule MRI is presented on figure j.

d e
5
5

*
*

7-8 7-8

f g

5
BA

BA

7-8 7-8
442 7 Meningiomas

h 5 i

7-8

j
Petroclival Meningiomas 443

Surgical Management petrosal sinus is exposed. Its opening and the


The history of petroclival meningioma surgery incision of tentorium to the tentorial incisura pro-
reflects to a large extent the evolution of skull- vide access to the petroclival region to the mid-
base surgery [113, 147]. Complete visualization clival level. The surgeon’s field of view is crossed
of the tumor with all surrounding neural and vas- only by the fourth cranial nerve medially and the
cular structures was regarded initially as a pre- fifth nerve laterally.
requisite for more radical and safer tumor The modifications of lateral cranial base
resection. Different extensive cranial base approaches—the presigmoid, retrolabyrinthine,
approaches were elaborated. Malis [134] and translabyrinthine, and transcochlear ones—differ
Mayberg and Symon [136] described the com- mainly in the amount of temporal bone removal
bined supra-infra-transtentorial approach with [121, 127, 137, 140, 148] . Their main advantages
ligation of the transverse sigmoid sinus. Hakuba are the shorter distance to tumor and surrounding
[124] first described in the neurosurgical litera- neurovascular structures; improved visualization,
ture the surgical technique and results of the especially of the anterior aspect of the brain stem;
transpetrosal–transtentorial route. In 1988, he and less brain retraction [121]. The presigmoid
described his experience using the transpetrosal approach combines a supra- and infratentorial
approach for surgery of clivus meningiomas exposure with less amount of presigmoid petrous
[125]. The combined retrosigmoid–subtemporal– bone resection. With the retrolabyrinthine
transtentorial approach with or without ligation approach, the bone anterior to the sigmoid sinus
of the sigmoid sinus was introduced later on and posterior to the semicircular canals is drilled
[112, 143, 154]. Complete or partial removal of off. The semicircular canals are skeletonized but
the petrous bone, which is the main anatomical not opened, and hearing is preserved. The trans-
obstacle to good exposure of the petroclival area, labyrinthine approach implies a complete mas-
allows exposing widely the area of interest. toidectomy and labyrinthectomy, which increases,
Transpetrosal techniques are directed either respectively, the area of exposure. The transco-
through the surface of the temporal bone (supe- chlear approach includes further resection of the
rior approaches) or through the mastoid process whole labyrinth and requires transposition of the
(lateral approaches) [110, 121, 156]. The senior facial nerve, thereby allowing better visualization
author introduced the intradural subtemporal of the structures anterior to the IAC. The trans-
approach with resection of the petrous apex. The petrosal approach with complete petrosectomy
epidural subtemporal approach was developed by was intended for large tumors in patients with
Kawase et al. and applied initially for accessing complete hearing loss. Other more extensive and
the lower basilar artery and then for petroclival rarely applied approaches to the clival region
and sphenopetroclival tumors [128, 131, 132]. include the extended transbasal approach, the
The technique includes resection of the anterior midline approaches (transsphenoidal, transoral,
petrous bone medial to the IAC, behind the hori- and transcervical), the infratemporal approach,
zontal segment of the petrous carotid artery and the facial degloving approach, and the anterior
lateral to the trigeminal nerve in Meckel’s cave. transethmoid epidural petrous apex resection
Thus, the dura mater down to the level of inferior [120, 122, 130].
444 7 Meningiomas

Case Illustration 7.12 supratentorial part (a). At that time, the senior
Presigmoid Approach author favored the presigmoid approach with-
This is a case of a large petroclival meningioma, out ligation of sigmoid sinus for such tumors
causing severe compression of the brain stem, (b). The tumor could be removed completely,
operated in the late 1980s. The tumor extended and all cranial nerve functions, including func-
into the dilated Meckel’s cave and had a large tional hearing (c–e), were preserved.
Petroclival Meningiomas 445

a b

(a–e) Intraoperative images. (a) Preoperative MRI structures (5 trigeminal nerve, 7–8 facial and vestibu-
demonstrating the size and extension pattern of this locochlear nerve complex, BS brain stem).
meningioma. (b) The approach consists of a temporal (e) Watertight reconstruction of the dura. Note the
and retrosigmoid suboccipital craniotomies plus addi- intact transverse and sigmoid sinuses. (f) The postop-
tional mastoid and partial pyramid resection (SS sig- erative bone-window CT demonstrates the extent of
moid sinus, TS transverse sinus, T temporal lobe), (c) bone resection. (g) Audiogram, showing that preserved
Two retractors (R) are used to elevate the temporal hearing (single arrowhead hearing before surgery,
lobe and the cerebellar hemisphere thus affording a double arrowhead hearing level after tumor removal).
wide exposure of the tumor (Tu). (d) Complete tumor (h) The patient 1 month after surgery
removal and preservation of all neurovascular
446 7 Meningiomas

e f

g
0.125 0.25 0.5 1 1.5 2 3 4 6 8 12
-10
-0
10
20
30
40
50
60
70
80
90
100
110
120
Petroclival Meningiomas 447

Growing experience with these extended therefore, only if the expected long-term mor-
approaches showed, however, that they are bidity is minimal and the quality of life is pre-
related to unacceptably high approach-related served [135, 151, 159].
mortality, mainly high rate of facial nerve palsy, The senior author’s experience encompasses
hearing loss, and CSF leaks. Another main dis- more than 200 surgically treated patients with
advantage is the risk of injury to the highly petroclival meningiomas. Initially, he used the
variable venous structures in the area [114, 117, frontotemporal or subtemporal approaches with
149, 151]. The interruption or even prolonged resection of the tentorium and petrous apex
stretching of the vein of Labbé, tentorial sinuses, (1970s). Later on, he introduced the combined
or any of the major draining veins may have infratentorial supratentorial presigmoid approach
unpredictable and devastating consequences (see Case Illustration 7.12). The aforementioned
(Fig. 7.23). On the other hand, as pointed out risks of venous injury or other morbidity were
earlier, even radiologically confirmed complete considered unacceptably high and forced him to
resection does not always prevent recurrences. change the operative concept and to abandon this
Complete resection should be attempted, approach (see Case Illustration 7.13).
448 7 Meningiomas

a b

c d

Fig. 7.23 Venous phase of a DSA, showing the great variability of the venous system, in particular the vein of Labbé
Petroclival Meningiomas 449

Case Illustration 7.13 old and had a large right petroclival menin-
Massive Venous Infarction Following gioma with supratentorial extension and
Presigmoid Transtentorial Approach growth into the Meckel’s cave (a, b).
This case presents a key experience that ulti- Considerable compression of the brain stem
mately led to change of the senior authors’ was evident on the MR images. The presig-
concept of operative management of petro- moid transtentorial approach was used to
clival meningiomas. The patient was 33 years remove the tumor completely. Despite the

a b

(a, b) Preoperative MRI, demonstrating the size and rioration appeared; CT performed 1 week. (d) 4 weeks.
growth pattern of the tumor. (c) CT performed 2 days (e) After surgery
after surgery when the first signs of neurological dete-
450 7 Meningiomas

d e

adhesion of part of the tumor capsule to the massive venous infarction with intracerebral
neural structures, they could be preserved. All hemorrhage in the temporal lobe, presumably
vascular channels were also intact at the end due to thrombosis of the vein of Labbè. The
of surgery. The patient awoke rapidly and was patient was re-intubated, and appropriate treat-
extubated. In the first 24 h, she did not have ment was initiated. Over the following 4
any deficits. weeks, she recovered completely and at dis-
On the second postoperative day, she lost charge had no neurological deficits. The CTs,
consciousness and developed left-sided central shown on figures c–e, demonstrate the pro-
hemiplegia and facial palsy. CT demonstrated gressive recovery from the venous infarction.
Petroclival Meningiomas 451

Tumors that are confined to the petroclival of the tentorium provides access to meningiomas
area are removed via the simple RS approach. with supratentorial extension that are limited to
Large meningiomas have already displaced the the middle cranial fossa and do not involve the
brain stem, so that the RS approach provides ade- Meckel’s cave or the cavernous sinus. If addi-
quate access even to the contralateral side or tional exposure is required, however, the petrous
supratentorially (Figs. 7.24 and 7.25). Resection apex may be resected. In 1982, the senior author

a b

Fig. 7.24 Petroclival meningioma, operated in the 1980s superiorly. * suprameatal tubercle. (c) Complete tumor
by MS via the retrosigmoid suprameatal approach. (a) CT removal. Note the extent of removal of the suprameatal
of the tumor. (b) Initial view of the tumor, dislocating the tubercle and the wide exposure of the trigeminal nerve
seventh-eighth nerves laterally and the trigeminal nerve into the Meckel’s cave
452 7 Meningiomas

introduced the technique of the intradural resec- Currently, this is our favored approach in petro-
tion of the petrous apex via the retrosigmoid clival meningiomas. Endoscopic inspection is
route—the so-called retrosigmoid suprameatal increasingly being used to inspect hidden areas
approach (RSMA) or Samii approach (Fig. 7.24). for tumor remnants (see Case Illustration 7.17).

a b

c d

Fig. 7.25 Giant petroclival meningioma with very large (c, d). The early postoperative MRI control image is
supratentorial part (a, b), removed via the simple retrosig- shown on figures e, f
moid approach with additional resection of the tentorium
Petroclival Meningiomas 453

e f

Fig. 7.25 (continued)

Thereby, the amount of bone resection or the achieved in 3.4 % of those meningiomas with
need for retraction of neural structures to visual- cavernous sinus infiltration [147].
ize a given area can be reduced.
In particularly large tumors extending into the Retrosigmoid Suprameatal (Samii)
suprasellar and parasellar areas that engulf the Approach: Technique
optic nerve and carotid artery, we prefer operat- The retrosigmoid suprameatal approach (RSMA)
ing in stages using two simple approaches [146] consists of the simple RS craniotomy and addi-
(Figs. 7.26 and 7.27). Initially, the infratentorial tional intradural resection of the petrous apex
tumor part in CP angle, clivus, and/or Meckel’s [145, 149]. The drawbacks related to the alterna-
cave is removed via a retrosigmoid suprameatal tive approaches to petrous apex, such as exten-
approach, and the brain stem is decompressed. sive petrous bone resection or retraction of the
Thus, the risk of severe neurological deteriora- temporal lobe with the associated risks of dam-
tion is prevented. The remaining supratentorial age to neural and vascular structures, are thus
component is removed via the frontotemporal avoided. Initially the tumor bulk is removed via
approach at a second stage. Planned two-staged one of the CP angle levels: the upper level
meningioma removal was performed in 17 % of between the tentorium and the trigeminal nerve,
the recently operated patients. The two-stage sur- the second level between the trigeminal and the
gery affords more radical tumor removal and facial-cochlear nerves, the third level between
allows adequate decompression of the optic the facial-cochlear nerves and the lower nerves,
nerve. Furthermore, it is associated with less and/or the lowest level, defined by the lower cra-
approach-related morbidity than the alternative nial nerves and foramen magnum. Although ana-
extended cranial base approaches. Removal of tomically these spaces may be relatively narrow,
the cavernous sinus component of such tumors the meningioma usually widens some of them
has never been the goal in patients with intact and provides adequate working corridor. Tumor
oculomotor function in order to prevent func- removal should start first at the most expanded
tional deficits. Therefore, total resection was level with the widest distance between the
454 7 Meningiomas

respective structures. Initial internal decompres- Once the accessible tumor portion is removed,
sion is performed using an ultrasonic surgical the bone located above and anterior to the IAC—
aspirator at the appropriate levels. In petroclival the suprameatal tubercle—is drilled off, provid-
meningiomas, as in almost all other CP angle ing access to the petroclival area and Meckel’s
tumors, the principle that the dissection of tumor cave (see Case Illustrations 7.14 and 7.15). The
capsule should be performed only after sufficient dura in the area is incised circumferentially and
internal decompression is achieved must be excised. Bone removal is performed with high-
always followed. speed drill with diamond spurs of decreasing

a b

c d

Fig. 7.26 Two-staged removal: retrosigmoid suprameatal and partially encased the ICA, was resected at a second sur-
and frontotemporal approach. This meningioma (a, b) was gery via the frontotemporal approach (e, f). The cavernous
removed in two stages: at the first stage, the CP angle part sinus tumor part was not removed but at follow-up showed
was resected, and the brain stem was decompressed (c, d). no growth tendency (Reprinted from: Samii et al. [146].
The supratentorial part, which compressed the optic nerve Copyright © 2010, with permission from Springer-Verlag)
Petroclival Meningiomas 455

e f

Fig. 7.26 (continued)

size, similar to the technique used to open the lateral perspective, such as the one provided by
IAC. Generally, the amount of bone resection the Kawase approach, the nerve is hidden by the
depends on the individual anatomical character- tumor and is visualized at a later stage, which
istics of the petrous bone and the tumor exten- increases the risk of its iatrogenic injury. The
sion. It has been estimated that drilling of the RSMA provides access to Meckel’s cave, the pet-
suprameatal tubercle increases the exposure up to roclival area, and the middle fossa including the
13.0 mm [117]. Opening the Meckel’s cave and posterior cavernous sinus. Following tumor
mobilizing the trigeminal nerve increase the removal and thorough hemostasis, the drilled
working angle further. Structures at risk during area of the petrous apex is sealed with multiple
the petrous apex resection are the petrosal seg- pieces of fat tissue and fibrin glue.
ment of the internal carotid artery anterolaterally, Notably, 33 % of all petroclival meningiomas
the superior petrosal sinus, and the superior semi- in our series presented with signs of peritumoral
circular canal laterally. The exposure of the ros- edema, which—as discussed previously—fre-
tral tumor portion can be further enlarged by quently indicates invasion of the pial sheet [115].
incision and retraction of the tentorium. Care Such tumors should be operated with utmost
should be taken to avoid injury to the trochlear care: in case a dissection plane does not exist,
nerve: it should be identified prior to the incision small part of the tumor or tumor layer might be
of tentorium. left to preserve the microvasculature and/or to
The abducens nerve is located typically within avoid direct injury to the brain stem or cranial
the tumor or is stretched anteriorly. With the nerves (10–20 % of all cases). Importantly, peri-
RSMA, the abducens nerve can be identified tumoral edema seen on the MRI does not always
early at its brain stem exit zone and followed dur- correlate to infiltrative or invasive tumor growth:
ing tumor removal up to Dorello’s canal, thus this issue can be determined only at surgery
reducing the risk of its inadvertent injury. From a (Figs. 7.28 and 7.29).
456 7 Meningiomas

a b

c d

Fig. 7.27 This 41-year-old man presented with a history images are shown on figures f, g. One week later, the sec-
of progressive visual disturbance, episodic headache, ond stage was performed: the remaining tumor was
chronic fatigue, and polyuria. The MRI (a–c) showed a removed completely via a right frontolateral craniotomy
large meningioma of the upper clivus, extending retro- (h, i). Immediate postoperative MRI (j, k) and photo of
and suprasellar. Initially, the lower tumor part was the patient (l). The patient had minimal functional impair-
removed via a retrosigmoid approach (d, e), and the brain ment of the right oculomotor nerve, which improved sub-
stem was decompressed. The immediate postoperative sequently later on
Petroclival Meningiomas 457

e f

Fig. 7.27 (continued)


458 7 Meningiomas

j k

Fig. 7.27 (continued)


Petroclival Meningiomas 459

a b

c d

Fig. 7.28 The presence of brain stem and/or cerebellar Nevertheless, a good arachnoid dissection plane could be
edema does not necessarily mean that the meningioma identified, and the tumor was removed completely without
invades the pia mater. On this figure, we present a patient any injury to the neural and vascular structures. The
with a petroclival meningioma, causing extensive brain patient did not have any new neurological deficits after
stem edema (a, b) that is best seen on the T2-weighted surgery. The postoperative CT examination, performed 10
sequences (c, d) and on the FLAIR sequence (e). days after surgery, is shown on figure f
460 7 Meningiomas

e f

Fig. 7.28 (continued)


Petroclival Meningiomas 461

a b

Fig. 7.29 Patient with a large petroclival meningioma, and the infratentorial tumor part could be removed com-
extending into the Meckel’s cave and posterior cavernous pletely via a retrosigmoid suprameatal approach. All cra-
sinus. The patient presented with signs of intracranial nial nerves were preserved. The early postoperative CT
pressure due to obstructive hydrocephalus and was stupor- (g) shows minimal blood collection in the tumor bed and
ous, and an EVD was immediately placed. The CT and opened fourth ventricle. (h) CT at discharge. The patient
MRI images at admission are presented on figures a–f. was discharged 14 days after surgery in a very good con-
Note the extensive perifocal edema and the occlusion of dition without neurological deficits
the fourth ventricle. The patient was operated 1 day later,
462 7 Meningiomas

e f

g h

Fig. 7.29 (continued)


Petroclival Meningiomas 463

Case Illustration 7.14 pyramid surface and extension deep into the
Retrosigmoid Suprameatal Approach 1 IAC and into the Meckel’s cave (a–d).
This 48-year-old female patient reported on The patient was operated via the retrosig-
an episode of hearing loss on the right side moid suprameatal approach—all essential
that occurred 7 years earlier. Hearing then steps are illustrated on figures e–v. The men-
recovered completely. Two months later, ingioma, which was found to originate from
however, she had a further similar episode. the dura medial to the trigeminal nerve, could
MRI of the head was performed and demon- be removed completely, and all neural and
strated a large CP angle tumor. At admission, vascular structures were preserved. After sur-
the patient did not have neurological deficits gery, the patient had an incomplete abducens
besides slight imbalance and numbness of the nerve palsy and facial nerve palsy HB grade
right half of the face. Audiogram surprisingly IV on the right side, which showed a gradual
revealed normal hearing levels on both sides. tendency to improve and recovered completely
MRI examination demonstrated a relatively within 12 months.
large tumor with broad base to the posterior
464 7 Meningiomas

a b

T1-weighted contrast-enhanced MR images of the removal of the most medial tumor part allows visual-
meningioma (a, b). T2-weighted sequences demon- ization even of the oculomotor nerve (3). (n) Further
strating clearly the extent of intracanalicular tumor opening of the IAC, up to its fundus. The main intra-
growth (c, d). Initial view of the tumor, as seen via canalicular tumor bulk was anterior to the seventh-
the RS exposure (e). The transverse and sigmoid eighth nerve complex. (o) The most lateral tumor part
sinuses are the limits of the craniotomy. CSF has was removed with an angled microhook with a tech-
been drained, and once the tension decreased nique similar to the one used in VS. (p) The abducens
sufficiently, the cerebellum was slightly retracted. nerve (6) was also dissected from the capsule—from
(R retractor, T tentorium, p posterior pyramid, Tu its exit zone to the Dorello’s canal. Tumor origin site
tumor). (f) Gradual debulking of the tumor above or matrix was found to be located medial to the IAC,
the seventh-eighth nerve complex. (g) The retro- lateral to the trigeminal nerve (*). (q) Drilling off the
suprameatal tumor part has been removed. The suprameatal tubercle allowed for access and removal
planned extent of opening of the IAC is indicated of the tumor from the Meckel’s cave (two arrow-
with dotted dark line. (h) The posterior wall of the heads). (r) Removal of the blood cloths in the opera-
IAC has been drilled off, and a part of the IAC is seen tive area and final hemostasis (BA basilar artery). (s)
in the middle. (i) In contrast to VS, the drilling is con- Final view of the CPA, demonstrating the complete
tinued above the IAC, thereby exposing the hidden tumor removal and preservation of all neurovascular
premeatal tumor part. (j) The area of exposure gained structures (LCN lower cranial nerves). (t) The drilled
by the additional drilling of the suprameatal tubercle area of the petrous bone is sealed off with fat pieces
is demarcated with a white dotted line. (k, l) The and fibrin glue. (u) Dural closure—2 or 3 sutures are
trigeminal (5) and the trochlear (4) cranial nerves placed to adapt the dura, and then an interrupted
were dissected from the tumor capsule. (m) The suture is made to achieve a watertight closure (v)
Petroclival Meningiomas 465

e f

Tu

Tu
P

7-8

g h
T

Tu

7-8
466 7 Meningiomas

i j

Tu

IAC
7-8

k l

BS
Petroclival Meningiomas 467

m n

o p

6
468 7 Meningiomas

q r

BA

s t

7-8

LCN
Petroclival Meningiomas 469

u v
470 7 Meningiomas

Case Illustration 7.15 decompression and resection of the menin-


Retrosigmoid Suprameatal Approach 2 gioma were started. The facial nerve was
Fifty-four-year-old male patient presenting identified by electrostimulation: it was dislo-
with very mild symptoms: slight hearing loss cated caudally. Therefore, the upper tumor
on the right side (35 dB). The preoperative part was further removed, which required
MRI is shown on figures a–f. Considering resection of the suprameatal tubercle. When
the fact that the major tumor part was the tumor was sufficiently decompressed
infratentorial and caused severe compression internally, indicated by the appearing brain
of the brain stem, the retrosigmoid approach stem pulsations, careful dissection of its cap-
was selected. All vessels and cranial nerves sule from the neural structures was per-
were hidden by the huge tumor and could formed. The supratentorial tumor part could
not be identified initially. Careful internal be removed through the tentorial incisura.

a b

c d

(a–f) Preoperative MRI of a large petroclival menin- after contrast administration. (b) T2 sequence. (c, f)
gioma. The tumor extends into the IAC and partially TOFF sequence. (g–i) MRI control study 12 months
into the jugular foramen (d, f) and has a large supraten- after surgery
torial portion. (a, d, e) T1-weighted MR sequences
Petroclival Meningiomas 471

The patient recovered rapidly from surgery. leak occurred at the third postoperative day
He had no new neurological deficits; the hear- but was successfully treated with a lumbar
ing was preserved but nonfunctional. CSF drainage for 1 week.

g h

i
472 7 Meningiomas

Outcome trigeminal, facial, and oculomotor nerves.


Early postoperative MRI examination is war- Meningiomas with major inferior growth toward
ranted to verify the amount of tumor resection. the jugular foramen and foramen magnum (see
This information is essential as a basis for further Case Illustration 7.16) are associated with worse
patient management. The main reasons for subto- postoperative outcome due to the involvement of
tal removal of petroclival meningiomas are the lower cranial nerves and the related neurological
following: lack of dissection plane and/or en dysfunction [115]. Best outcome, in respect to
plaque tumor growth with infiltration of cranial radicality of tumor removal and avoidance of
nerves or brain stem; fibrous tumor type with neurological morbidity, can be achieved during
hard consistency; encasement or infiltration of the first operation. Previous surgery or radiother-
major vessels (in particular of the perforating apy, besides obliterating the arachnoid planes,
arteries); and cavernous sinus invasion [115, 133]. renders the neural and vascular structures more
In case of incomplete resection, follow-up MRI vulnerable to iatrogenic injury.
studies are performed 3–6 months after surgery, In the seniors’ author series, the preoperative
and if the residual tumor demonstrates definitive abducens palsy improved or resolved completely
growth tendency, radiosurgery/fractionated radio- in 29 % of the patients. The rate of recovery of
therapy is recommended. preoperative oculomotor or trochlear palsy was
The outcome of surgery depends mainly upon much lower. The most frequent new postopera-
the location and consistency of the tumor and to a tive extraocular muscle impairment was double
lesser extent upon its size. As noted by different vision due to trochlear nerve (7 %), abducens
authors, despite the modern cranial base nerve (3 %), or oculomotor nerve (3 %) dysfunc-
approaches, refined microsurgical techniques, tion [115]. CSF leak that required surgical repair
and reliable neurophysiological monitoring, the occurred in one patient. In one patient, hydro-
surgical morbidity may still be high and is com- cephalus developed on the tenth postoperative
monly related to cranial nerve deficits and pul- day, and a VP shunt was implanted. The periop-
monary embolisms [111, 114, 118, 152]. The erative mortality in the group was 0 %.
nerves at highest risk for iatrogenic injury, espe- The management of recurrences is discussed
cially in meningiomas extending supratentorially, in the appropriate section of the chapter on CP
are the trochlear nerve, followed by the cochlear, angle meningiomas.
Petroclival Meningiomas 473

Case Illustration 7.16 years. The neurological examination at admis-


Large Petroclival Meningioma with Extension sion did not reveal any abnormalities, besides
into the Jugular Foramen a mild hearing deficit—PTA of 30 dB. The
The patient (48-year-old female) presented to MRI and CT are shown on figures a–e.
our outpatient department with a history of She was operated via the RS approach. The
progressive hearing loss and sporadic swal- meningioma was located medial to the
lowing difficulties and double vision since 3 seventh-eighth nerves and the lower cranial

a b

c d

(a–e) T1-weighted MRI after gadolinium administra- bellar hemisphere, 5 trigeminal nerve, 7–8 facial and
tion, showing the meningioma in the left CP angle. The vestibulocochlear nerve complex, LCN lower cranial
typical intense contrast enhancement is obvious. The nerves, PV petrous vein, R retractor. (m, n) The patient
tumor extends inferior to the level of the foramen mag- had relatively large pneumocephalus, visualized on the
num and has a supratentorial extension in the cavern- early postoperative CT examination (m CT scout scan
ous sinus. Small tumor extension in the jugular foramen of the head, n CT axial scan). (o–q) Postoperative
is well seen on figures c and d. (f–l) Intraoperative T1-weighted MR images (after contrast administra-
images of different stages of tumor removal (see the tion) showing complete removal of the CP angle tumor
description in the text). Tu tumor, P pyramid, C cere- part and the tumor remnant in cavernous sinus
474 7 Meningiomas

nerves and extended from the tentorium to the nerve was engulfed by the tumor and dislo-
base of the PCF, caudally to the lower cranial cated, so that it could be visualized through
nerves (f). The tumor had relatively firm con- the level between the seventh and eighth and
sistency, was well vascularized, and was the lower cranial nerves (j). Despite the strong
strongly bleeding. It was initially debulked via bleeding and its consistency, the tumor could
the most expanded level—the one between the be further removed until the abducens, the
seventh-eighth nerves and the lower cranial trigeminal, and the seventh-eighth nerves were
nerves (g). The neural structures were not dissected free. The tumor part in the jugular
infiltrated, and the lower cranial nerves could foramen could be removed with angled micro-
be gradually dissected from the tumor. Then instruments. The attachment of the tumor,
the removal proceeded via the upper level— which was in the area of the jugular foramen,
above the seventh and eighth nerves (h–i). The was coagulated (k). All neural and vascular
petrous vein determined the upper limit of the structures, including the petrous vein, were
level in this particular case. The abducens preserved (l).

e f

g h
Petroclival Meningiomas 475

The postoperative course was uneventful. examination was done later and proved the
Although relatively large pneumocephalus was complete removal of the CP angle tumor part
seen on the early postoperative CT (m, n), the (o–q). At discharge, the only deficit she had
patient had no major related symptoms besides was a worsened hearing on the left side. A rec-
some headache and nausea. He was mobilized ommendation was given to follow the cavern-
and transferred to a normal ward in 48 h. MRI ous tumor portion with an MRI in 6 months.

i j

k l
476 7 Meningiomas

m n

p q
Petroclival Meningiomas 477

Case Illustration 7.17 tumor removal: a small remnant along the


Endoscope-Assisted Removal of Petroclival abducens nerve was identified that was not
Meningiomas visualized with the microscope (l–q). In this
Fifty-six-year-old female patient presented case, the remnant was removed with endo-
with progressive headache, trigeminal nerve scopic guidance.
sensory loss (V1,2), absent corneal reflex, and The postoperative period was completely
hypacusis. The MRI findings are demonstrated uneventful. A slight abducens palsy appeared
on figures a, b. The patient was operated via early after surgery but until discharge resolved
the RS approach (see figures c–s). Endoscopic completely. Hearing level was similar to the
control was used to verify the completeness of preoperative.
478 7 Meningiomas

a b

c d

(a) Preoperative MR images demonstrating the loca- opening of the IAC. (j, k) Presumed complete tumor
tion, size, and extension of the tumor (T1-weighted removal. The basilar artery (BA), the abducens nerve
sequences after gadolinium administration). (b) (6), and the preserved petrous vein (arrowhead) are
Intraoperative images. (c) Standard RS craniotomy well seen. (l) Endoscopic inspection demonstrating
was performed exposing the edges of the transverse the whole course of the trigeminal nerve. (m) The
and sigmoid sinuses. The dura was incised along the endoscope is advanced above the trigeminal nerve: the
sinuses. (d) Elevation of the cerebellar hemisphere and petrous apex area (PA), oculomotor nerve (3), and
draining of CSF from the lateral cerebellomedullary brain stem (B) are visualized. No residual tumor is
cistern. (e) The retractor (R) is placed to support the seen in this area. (n) The tumor in the area of the IAC
hemisphere, and the tumor (Tu), lying medially to the (double small arrows) has been also removed. (o)
fifth and seventh-eighth cranial nerves, is exposed (T Then, the endoscope was directed to the lower level:
tentorium). (f) Tumor debulking performed initially between the seventh-eighth and the lower cranial
via the level determined by the tentorium and the nerves (LCN). (p) A small remnant surrounding the
trigeminal nerve. (g, h) Further tumor removal via the distal part of the abducens nerve (6) in the area of
second level—between the fifth and the seventh-eighth Dorello’s canal was visualized and removed under
nerves. (i) Removal of the small intracanalicular tumor direct endoscopic control (q). (r) Fat plug in the drilled
part: in this particular case, there was no need of wide area of the petrous bone. (s) Watertight dural closure
Petroclival Meningiomas 479

e f

g h
480 7 Meningiomas

i j

k l

m
Petroclival Meningiomas 481

o p

q r

s
482 7 Meningiomas

Foramen Magnum Meningiomas posterior and anterior meningeal arteries, as well


as from meningeal branches of the ascending
Meningiomas are the most common benign pharyngeal and occipital arteries.
tumors of the foramen magnum. Foramen mag-
num meningiomas account for 1.8–3.2 % of all
meningiomas and for 6–7 % of all posterior fossa General Features
meningiomas [160, 161, 166, 172]. They arise
primarily within the foramen magnum area or The clinical presentation of foramen magnum
involve it secondarily. Most frequently—in meningiomas is nonspecific, and the mean dura-
52–4 % of the cases—the tumor occupies the tion of symptoms prior to diagnosis is usually
anterolateral part of the foramen. Anteriorly more than 2 years. Frequent initial complaints are
located are 4–32 % of the tumors, posterolateral occipital headache and upper cervical pain, which
are 10–20 %, and posterior are 5–7 % [161, 165, may be exacerbated by neck movements or
167, 168]. Valsalva maneuvers. Meningiomas that extend to
The foramen magnum is a component of the the foramen magnum secondarily with a major
occipitocervical junction—a complex biome- intracranial portion cause brain stem, cerebellar,
chanical entity of ligaments, joints, and muscles and lower cranial nerve signs and symptoms.
connecting the atlas, axis, and occipital bone High cervical meningiomas cause typically
[176]. Its limits are the following: lower third of symptoms and signs related to the upper cervical
the clivus and upper margin of the C2 vertebral spinal cord and roots. More extensive tumors
body anteriorly, jugular tubercle down to the may present with a mixed clinical picture [165].
upper margin of the C2 lamina laterally, and ante- The typical “foramen magnum syndrome” is
rior edge of the squamous occipital bone to the rarely seen in its full form: patients present gen-
spinous process of C2 posteriorly [169]. Relevant erally with varying degrees of sensory and motor
neural structures in the area are the following: loss. Long-tract signs with spastic gait are fre-
lower cranial nerves (9–12th), cerebellar tonsils, quent findings. Atrophy of the intrinsic hand
inferior vermis, fourth ventricle, caudal aspect of muscles may be found on the side of the lesion,
brain stem, rostral aspect of spinal cord, and probably as a result of vascular changes (com-
upper cervical nerve roots (C1 and C2) [165]. pression of the anterior spinal artery or venous
The most important vascular structures in the obstruction) or syringohydromyelia. In later
area are: the vertebral arteries (VA), the posterior stages, the attempt to avoid pain by holding the
inferior cerebellar arteries (PICAs), the anterior head in fixed position leads to a clinical picture
and posterior spinal arteries, the meningeal resembling torticollis.
branches, and the veins and dural sinuses at the
occipitocervical junction. Detailed knowledge of
the microanatomical relations between these Management
structures and of their physiological significance
is of enormous importance for planning and per- Preoperative Examinations
forming safe surgeries. Both the normal anatomy The preoperative neuroradiological examinations
and the possible variations should be considered. are similar to those in other CP angle menin-
An important landmark in the foramen magnum giomas. The stability of the occipitocervical
area is the dentate ligament, whose sectioning junction should be studied on dynamic X-rays
increases the exposure to the area anterior to the (flexion and extension) of cervical spine. The
spinal cord. This ligament is attached to the pia vascular anatomy, which is surgically relevant, is
mater between the dorsal and ventral rootlets appreciated on MR angiography and MR venog-
medially and to the dura mater laterally [175]. raphy. Encasement and narrowing of the verte-
The dural blood supply to foramen magnum bral artery suggest that the adventitia of the artery
meningiomas is usually from branches of the may be invaded. Encasement of the artery is
Foramen Magnum Meningiomas 483

found intraoperatively in 40–48 % of cases in following: wide exposure and even transposition
different series. Superselective embolization of of the extradural vertebral artery, exposure of the
arterial feeders may be useful in highly vascular transverse process of C1 and of the atlantooc-
meningiomas. The function of the lower cranial cipital joint, and varying degrees of occipital
nerves should be examined with direct endo- condyle removal. The occipital condyle may be
scopic examination of the vocal cords and swal- resected partially or entirely and may be com-
lowing test. bined with removal of the lateral mass of the
atlas lateral to the dural entrance of vertebral
Principles of Surgery artery, partial mastoidectomy, or even sectioning
Surgery in the foramen magnum area can cause of the sigmoid sinus [163, 164, 171–173]. Wide
significant functional impairment and may be exposure of the tumor and its dural attachment is
related to a prolonged recovery period. The deci- thus achieved.
sion on the optimal management should be taken The benefits of such extensive approaches,
together with the patients and their families. however, have to be carefully weighted against
Important factors to consider are the following: the risk of higher morbidity, related to vascular
patients’ expectations, psychological and social complications, injury to cranial nerves, prolonged
needs, neurological status, and tumor size, exten- surgery, or CSF leak. Another drawback is the
sion, and relation to adjacent anatomical struc- potential risk of craniocervical instability that
tures. The optimal management is complete may require an additional fusion procedure, espe-
tumor removal with preservation of neurological cially in case of resection of more than 75 % of
functions and avoidance of craniocervical insta- the condyle [162, 180, 181].
bility. Radiotherapy/radiosurgery may be consid- The experience of the senior author with
ered in patients with small tumors and severe almost all these approaches showed that in nearly
somatic diseases, precluding prolonged surgery all foramen magnum meningiomas, such wide
in general anesthesia or in those unwilling sur- exposure is not required [177, 179]. Notably, the
gery. In incidentally discovered small tumors, the wide bone removal does not provide any addi-
option of initial observation period has to be dis- tional advantages for the difficult operative steps,
cussed with the patient. such as dissection of the tumor from the brain
Posterior-located meningiomas are approached stem, lower cranial nerves, or vessels. Exceptions
via a medial suboccipital craniectomy and—if are the rare strictly anterior located very small
necessary—a C1/C2 laminectomy. The approach meningiomas: resection of the posterior third of
affords bilateral access to approximately 120° of the condyle may be needed to expose them ade-
the foramen magnum [174]. Anterior, anterolateral, quately. The lateral suboccipital approach, how-
and posterolateral meningiomas are approached ever, is sufficient in case of larger anterior foramen
via posterolateral approaches, which differ in the magnum meningiomas (Figs. 7.30 and 7.31). The
degree of bone removal. Our favorite lateral sub- rate of permanent surgical morbidity in series
occipital approach consists of a retrosigmoid when the lateral suboccipital approach was used
craniectomy and a C1 hemilaminectomy/lamine- ranges from 0 to 8 %, and the mortality rate ranges
ctomy [178, 179]. More extensive approaches from 0 to 6.1 %, respectively [161, 170, 179]. In
have been developed to provide wider surgical case a transcondylar approach was used, the sur-
exposure and to enhance the feeling of safety to gical morbidity rate is 0–60 % and the mortality
the surgeon. Their common features are the rate is 0–29 % [160, 166, 169, 172].
484 7 Meningiomas

a b

c d

Fig. 7.30 Anterior foramen magnum meningioma, oper- bral artery and complete tumor removal (e). The CT scans
ated by MS more than 20 years ago. (a, b) Preoperative (f, g) demonstrate the size of the craniotomy and the open-
MR images. (c) View of the tumor after sectioning of the ing of the foramen magnum: the exposure was sufficient
dentate ligament. The spinal part of the accessory nerve to remove the tumor without undue risks
and the C1 root is well visible. (d) Exposure of the verte-
Foramen Magnum Meningiomas 485

e f

Fig. 7.30 (continued)


486 7 Meningiomas

a b

c d

Fig. 7.31 Large meningioma of the lower clivus extend- approach. (a, b) Preoperative MR images. (c, d) Post-
ing to the foramen magnum area. Complete tumor removal operative images: complete tumor removal was achieved,
could be safely achieved via the lateral suboccipital and all neurological functions were preserved
Foramen Magnum Meningiomas 487

Lateral Suboccipital Approach to the superior extension of bone opening should


Foramen Magnum Meningiomas reach the sinus (Fig. 7.32).
The approach is based on the retrosigmoid cran- • Depending on the tumor extension, the C1
iotomy, as described in the corresponding chap- lamina and even the C2 lamina have to be
ter. Here we will focus only on the differences removed laterally enough. Dissection or trans-
relevant for surgery of foramen magnum menin- position of the vertebral artery in order to gain
giomas (see Case Illustrations 7.18 and 7.19): more lateral exposure is unnecessary because
• The approach is performed on the side of the opening of the dura is always medial to
dominant tumor growth. the artery.
• A slightly curved skin incision medial to the • The dura is incised in a curvilinear manner
mastoid process is made, which has to be just 1.5–2 mm medial to the sigmoid sinus,
extended caudally. and the incision is continued in the midline
• The suboccipital muscles and fasciae are cut down to C1 or C2. At the spinal level, stay
in line with the skin incision, and the C1 arch sutures are placed on both sides and fixed to
on the side of the tumor is exposed. the muscles in order to open the dura.
• The vertebral artery is identified and followed
to its entry point in the dura but is not Tumor Removal
mobilized. The Tumor relation to the major neurovascular
• Retrosigmoid craniectomy is performed structures—vertebral arteries, posterior spinal
using bone rongeurs and high-speed drill, arteries and PICA, Cl nerve root, and lower cra-
exposing 1–2 mm from the sigmoid sinus. nial nerves—cannot be predicted before surgery.
The foramen magnum is opened widely. Their early identification helps to avoid iatro-
Usually, in foramen magnum meningiomas, genic injury but is possible only in smaller men-
there is no need to expose the transverse ingiomas. Tumor removal starts always with
sinus. Exceptions are meningiomas with a careful internal debulking, alternated with dissec-
large CP angle portion; in such cases, the tion of the capsule in the arachnoid plane. The

a b

Fig. 7.32 Extensive posterior fossa meningioma located of the lower part of the tumor in the area of foramen mag-
in the left CP angle extending caudally below the level of num (Tu tumor, BS brain stem, 7–8 facial and vestibuloco-
foramen magnum. (a, b) Preoperative MR images. (c–e) chlear nerve complex, LCN lower cranial nerves, VA
Intraoperative images. (c) Initial exposure of the tumor. vertebral artery). (f, g) Postoperative MR study, demon-
(d) Removal of the CP angle tumor portion. (e) Removal strating the complete tumor removal
488 7 Meningiomas

Fig. 7.32 (continued)


c

e
Foramen Magnum Meningiomas 489

f g

Fig. 7.32 (continued)

anterior exposure is increased by dividing the with the outcome and the possibility for complete
dentate ligaments or even sectioning of the first removal: tumor type (encapsulated or en plaque),
cervical nerve root distal to its connections with invasion of the pia of the brain stem/spinal cord
the accessory nerve. Meningiomas located medial vs. of the cranial nerves or presence of a good
to the glossopharyngeal, vagus, and accessory arachnoid plane, encasement of major artery, and
nerves are accessed via the spaces between the previous surgery or radiotherapy [179]. Complete
nerve fascicles (Figs. 7.33 and 7.34). These removal could be achieved in just 7 % of en
spaces are frequently expanded by the tumor plaque meningiomas: in 86 % of them subtotal
itself. More rostral tumor parts may be accessed removal, and in 7 %, only partial removal was
via the space between the lower margin of the safely possible. In encapsulated meningiomas,
vestibulocochlear/ facial nerves and the upper complete removal could be achieved in a much
roots of the glossopharyngeal nerve. Large men- higher rate—88 %. See Case Illustrations 7.18
ingiomas usually cause posterior and contralat- and 7.19.
eral displacement of the brain stem and upper Best results are achieved at initial surgery:
cervical spine, creating sufficient surgical corri- complete removal was possible in 76 % of all pri-
dor for their safe resection (Figs. 7.33 and 7.34). mary surgeries and in just 46 % of the reopera-
During tumor removal, care should be taken not tions. The encasement of a major artery was
to rupture the contralateral vertebral artery, which another independent factor for the completeness
is usually identified at a later stage (Fig. 7.35). of tumor removal. In case such encasement
Radical excision of the dura in the area is impos- existed, complete tumor removal was feasible in
sible; thorough coagulation of the tumor matrix/ 33 %, while if it was absent, the rate increased to
attachment is therefore recommended. 90 %. These parameters, especially en plaque
tumor type, surgery of recurrence, or after previ-
Outcome ous radiotherapy, were found to correlate also
The experience of the senior author encompasses with the complication rate. The complication rate
more than 130 foramen magnum meningiomas. increased from 14 % in encapsulated menin-
Several parameters have been found to correlate giomas to 33 % in the en plaque type.
490 7 Meningiomas

a b

Fig. 7.33 Large foramen magnum meningioma (a–c), removal, the PICA is dissected free. (f) Complete tumor
removed completely via the lateral suboccipital approach. removal; all neurovascular structures are intact (BS brain
(d) initial tumor (Tu) exposure. (e) Following partial stem)
Foramen Magnum Meningiomas 491

d e

Fig. 7.33 (continued)


492 7 Meningiomas

a c

b d

Fig. 7.34 Patient with a large foramen magnum menin- (Tu). The cerebellum (C) is slightly elevated; the lower
gioma, operated via the lateral suboccipital approach. The cranial nerves (LCN) are spread over the tumors’ surface;
tumor was located predominantly medial to the lower cra- the facial-vestibulocochlear nerve complex is not visible.
nial nerves; the facial-vestibulocochlear nerve complex (d) Complete tumor removal and preservation of all neu-
were dislocated medially. Tumor removal was accom- ral structures. (7–8 facial and vestibulocochlear nerve
plished between the fascicles of the lower cranial nerves. complex, BS brain stem). (e) Postoperative CT, demon-
(a, b) Preoperative MRI study. (c) Initial view of the tumor strating the extent of bone removal
Foramen Magnum Meningiomas 493

e Patients with small remnants should be observed


closely, both clinically and radiologically, and
treated only if the remnant does show a growth ten-
dency. Considering the mode of treatment of such
remnants, very important issue is their location: in
case the tumor contacts the brain stem, the risk of
radiation injury and severe neurological deficits is
very high. Therefore, an attempt should be made to
remove the tumor surgically. Radiotherapy (radio-
surgery or fractionated radiotherapy) may be rec-
ommended only if the tumor is separated from the
radiosensitive neural structures or as ultima ratio.
Recurrence rate after complete removal
(Simpson grade 1) is low. Still, meningiomas
may recur even late after surgery. Follow-up with
regular MRI examinations is warranted at regular
intervals. In case of symptomatic or growing recur-
rences, active management is indicated. The pre-
Fig. 7.34 (continued)
ferred option is again operative. Radiotherapy
(radiosurgery or fractionated radiotherapy) has to
Although it is still impossible to predict the bio- be considered as an adjunctive therapy, especially
logical activity of small tumor remnants, most of in case of early recurrences. Primary radiotherapy
them remain rather inactive. We do not recom- of the recurrence is an alternative option in elderly
mend early adjuvant radiotherapy or radiosurgery. somatically unstable patients.
494 7 Meningiomas

a b

c d

Fig. 7.35 Example of a foramen magnum meningioma is barely visible. (d) Elevation of the cerebellum and expo-
encompassing but not infiltrating the vertebral artery. (a, sure of the tumor. (e) Complete tumor removal. (f) The
b) Preoperative MR images. (c) Dura incision. The tumor patient 1 month later did not have neurological deficits
Foramen Magnum Meningiomas 495

Fig. 7.35 (continued)


496 7 Meningiomas

Case Illustration 7.18 of the neck, which appeared 1 year earlier and
Foramen Magnum Meningioma 1 gradually disappeared. The only neurological
This 42-year-old female patient presented to deficits at examination were an ipsilateral
our outpatient department complaining of tongue atrophy and gait ataxia. MRI study
headache and vertigo since 6 months. The ini- showed a relatively large tumor in the lower
tial complaint was strong pain in the right part CP angle with extension caudally to the

a b

c d

(a, b) Preoperative T1-weighted axial and sagittal ramus spinalis of the 11th nerve); some nerve fibers
postcontrast MRI images showing an enhancing mass were engulfed by the tumor (small arrows), and the
in the foramen magnum area (arrowhead tumor exten- 10th and 12th nerve could not be visualized. (h) A
sion in the jugular foramen). (c, d) T2-weighted MRI large calcification—appr.1 cm (asterisk)—was
images demonstrating an arachnoid plane between the removed from the area of the jugular foramen. (i) With
tumor and the brain stem. (e) RS craniotomy, exposing further tumor removal, the vertebral artery (VA) could
the edges of the transverse and sigmoid sinuses. The be identified. (j) The tumor from the JF could be
foramen magnum was also opened. The dura was removed with an angled hook and platelet knife (small
incised along the lower half of the sigmoid sinus (dot- arrows), without the need of extending the exposure.
ted line), approximately 2 mm medial to its border. (f, (k) The tumor has been removed, and the vertebral
g) Initial view of the tumor, located inferior to the artery was freed. (l) Complete tumor removal. The
jugular foramen (arrow ninth nerve, arrowheads preserved lower cranial nerves are well seen
Foramen Magnum Meningiomas 497

foramen magnum level (a–d). The tumor parts (e). At surgery, the tumor turned out to
caused severe compression and dislocation of be moderately firm and well vascularized. The
the brain stem and had imaging characteris- 9th–12th cranial nerves were tightly attached
tics, compatible with meningioma. The typi- to the tumor and dislocated (f, g). The menin-
cal dural tail sign was well visible. The tumor gioma engulfed the ipsilateral vertebral artery
had its bulk in the lower CP angle and fora- and extended into both the jugular foramen
men magnum area but extended also into the and the hypoglossal canal. Following internal
jugular foramen (b-arrowhead). decompression, its capsule could be dissected
The patient was operated via an RS from the surrounding structures. The tumor
approach with opening of the foramen mag- part in the area of JF was strongly calcified.
num to gain access to the most caudal tumor A large calcified tumor part (h) had to be

e f

g h
498 7 Meningiomas

removed, which allowed further exposure of excised. Pathological examination of the spec-
the JF and enabled us to remove the tumor imen showed that the tumor was a secretory
portions, extending into the JF and into the WHO grade I meningioma. After surgery, a
hypoglossal canal (i, j). Complete tumor transient lower cranial nerve deficit was regis-
removal could be thus achieved (k, l). The tered that recovered almost completely until
meningioma was found to originate from the discharge. The hypoglossal palsy was, how-
dura between the JF and the hypoglossal canal. ever, more pronounced and showed minimal
This part of the dura was coagulated and recovery.

i j

k l
Foramen Magnum Meningiomas 499

Case Illustration 7.19 meningioma in the area of foramen magnum,


Foramen Magnum Meningioma 2 engulfing the ipsilateral vertebral artery, are
Sixty-five-year-old female patient complain- presented on figures a–c.
ing of increasing occipital headache and gait The patient was operated via the lateral sub-
instability since 3 months and slight swallow- occipital approach (RS craniotomy, opening of
ing difficulty since several weeks. Her neuro- foramen magnum, and C1 laminectomy). The
logical examination revealed gait ataxia, slight vertebral artery was identified and followed to
sensorineural hearing deficit on both sides, its entry point into the dura but was not mobi-
and decreased gag reflex on the right side. The lized. The consecutive steps of the surgery are
MR images, which demonstrated a large shown on figures d–i. Initially sufficient CSF

a b

(a–c) Preoperative T1-weighted MRI scans showing C cerebellum). (e) The tumor has been partially
the foramen magnum meningioma, extending caudal to removed, and the accessory nerve was identified (11).
the C1 level. (d) Retrosigmoid craniotomy with open- (f, g) Gradually the ipsilateral vertebral artery (VA) was
ing of the foramen magnum and C1 laminectomy has exposed. (h, i) Complete removal of the tumor with
been performed, exposing the tumor (Tu), which preservation of all neurovascular structures (BS brain
extended caudal to the level of C1 root (C1). (D dura, stem)
500 7 Meningiomas

was allowed to egress, and tumor debulking was achieved with preservation of all neuro-
was performed. Gradually, the lower cranial vascular structures. Then, the tumor matrix
nerves were identified, and after sufficient was extensively coagulated.
internal decompression, the tumor capsule was No new neurological deficits developed after
carefully dissected from the cranial nerves and surgery. The swallowing deficit was slightly
brain stem. The vertebral artery was engulfed more pronounced in the first days but resolved
by the meningioma over a length of 2–2.5 cm until discharge. Histopathological diagnosis
but could be completely freed. Complete tumor was WHO grade I secretory meningioma.

d e
C

11
D
Tu
D
Tu

C1

f g

Tu Tu

VA
VA

h i
C

BS

VA 11
References 501

References canal: a diagnostic and surgical challenge. Skull Base


Surg 9(2):87–94
15. Carvalho GA, Matthies C, Tatagiba M, Eghbal R,
Cerebellopontine Angle Meningiomas Samii M (2000) Impact of computed tomographic and
magnetic resonance imaging findings on surgical out-
1. Agrawal V, Ludwig N, Agrawal A, Bulsara KR (2007) come in petroclival meningiomas. Neurosurgery
Intraosseous intracranial meningioma. AJNR Am J 47(6):1287–1294; discussion 1294–1295
Neuroradiol 28(2):314–315, 28/2/314 [pii] 16. Castellano F, Ruggiero G (1953) Meningiomas of the
2. Akeyson EW, McCutcheon IE (1996) Management of posterior fossa. Acta Radiol Suppl 104:1–177
benign and aggressive intracranial meningiomas. 17. Chamberlain MC, Blumenthal DT (2004) Intracranial
Oncology (Williston Park) 10(5):747–756; discussion meningiomas: diagnosis and treatment. Expert Rev
756–759 Neurother 4(4):641–648. doi:10.1586/14737175.4.4.641
3. Al-Mefty O, Kadri PA, Pravdenkova S, Sawyer JR, 18. Cho YD, Choi GH, Lee SP, Kim JK (2003) (1)
Stangeby C, Husain M (2004) Malignant progression H-MRS metabolic patterns for distinguishing
in meningioma: documentation of a series and analy- between meningiomas and other brain tumors. Magn
sis of cytogenetic findings. J Neurosurg 101(2):210– Reson Imaging 21(6):663–672, S0730725X03000973
218. doi:10.3171/jns.2004.101.2.0210 [pii]
4. Antinheimo J, Sankila R, Carpen O, Pukkala E, Sainio 19. Cushing H (1922) The meningiomas (dural endothe-
M, Jaaskelainen J (2000) Population-based analysis liomas): Their source and favoured seats of origin.
of sporadic and type 2 neurofibromatosis-associated Brain 45:282–316
meningiomas and schwannomas. Neurology 54(1): 20. Cushing H, Eisenhardt L (1938) Meningiomas: Their
71–76 classification, regional behavior, life history and sur-
5. Arana E, Diaz C, Latorre FF, Menor F, Revert A, gical end results. Charles C Thomas, Springfield
Beltran A, Navarro M (1996) Primary intraosseous 21. De Monte F, Al-Mefty O (1993) Neoplasms and the
meningiomas. Acta Radiol 37(6):937–942 cranial nerves of the posterior fossa. In: Barrow DL
6. Arriaga M, Shelton C, Nassif P, Brackmann DE (ed) Surgery of the cranial nerves of the posterior
(1992) Selection of surgical approaches for menin- fossa. American Association of Neurological Surgeons,
giomas affecting the temporal bone. Otolaryngol Park Ridge, pp 253–274
Head Neck Surg 107(6 Pt 1):738–744 22. Dezamis E, Sanson M (2003) The molecular genetics
7. Asaoka K, Barrs DM, Sampson JH, McElveen JT Jr, of meningiomas and genotypic/phenotypic correla-
Tucci DL, Fukushima T (2002) Intracanalicular men- tions. Rev Neurol (Paris) 159(8–9):727–738, MDOI-
ingioma mimicking vestibular schwannoma. AJNR RN-09-2003-159-8-9-0035-3787-101019-ART1 [pii]
Am J Neuroradiol 23(9):1493–1496 23. Drake WM, Grossman AB, Hutson RK (2005) Effect
8. Bacciu A, Piazza P, Di Lella F, Sanna M (2007) of treatment with pegvisomant on meningioma growth
Intracanalicular meningioma: clinical features, radio- in vivo. Eur J Endocrinol 152(1):161–162
logic findings, and surgical management. Otol Neurotol 24. Drummond KJ, Zhu JJ, Black PM (2004)
28(3):391–399. doi:10.1097/MAO.0b013e31803261b4 Meningiomas: updating basic science, management,
9. Baguley DM, Beynon GJ, Grey PL, Hardy DG, Moffat and outcome. Neurologist 10(3):113–130, 00127893-
DA (1997) Audio-vestibular findings in meningioma 200405000-00001 [pii]
of the cerebello-pontine angle: a retrospective review. 25. Elder JB, Atkinson R, Zee CS, Chen TC (2007)
J Laryngol Otol 111(11):1022–1026 Primary intraosseous meningioma. Neurosurg Focus
10. Bassiouni H, Hunold A, Asgari S (2004) Meningiomas 23(4):E13. doi:10.3171/FOC-07/10/E13
of the posterior petrous bone: functional outcome 26. Eljamel MS (1990) Multiple meningiomas. J Neurosurg
after microsurgery. J Neurosurg 100:1014–1024 72(5):834–835
11. Batra PS, Dutra JC, Wiet RJ (2002) Auditory and 27. Eljamel MS, Foy PM (1989) Multiple meningiomas
facial nerve function following surgery for cerebello- and their relation to neurofibromatosis. Review of the
pontine angle meningiomas. Arch Otolaryngol Head literature and report of seven cases. Surg Neurol
Neck Surg 128(4):369–374, ooa00138 [pii] 32(2):131–136
12. Bonnal J, Born JD, Tremoulet M (1979) Multiple intrac- 28. Elster AD, Challa VR, Gilbert TH, Richardson DN,
ranial meningiomas (author’s transl). Neurochirurgie Contento JC (1989) Meningiomas: MR and
25(2):78–83 histopathologic features. Radiology 170(3 Pt 1):
13. Bonneville F, Savatovsky J, Chiras J (2007) Imaging 857–862
of cerebellopontine angle lesions: an update. Part 2: 29. Evans DG, Huson SM, Donnai D, Neary W, Blair V,
intra-axial lesions, skull base lesions that may invade Newton V, Harris R (1992) A clinical study of type 2
the CPA region, and non-enhancing extra-axial neurofibromatosis. Q J Med 84(304):603–618
lesions. Eur Radiol 17(11):2908–2920. doi:10.1007/ 30. Filippi CG, Edgar MA, Ulug AM, Prowda JC, Heier
s00330-007-0680-4 LA, Zimmerman RD (2001) Appearance of menin-
14. Carney AS, Ward V, Malluci CL, O’Donoghue GM, giomas on diffusion-weighted images: correlating
Robertson I, Baldwin DL, Maw AR, Coakham HB diffusion constants with histopathologic findings.
(1999) Meningiomas involving the internal auditory AJNR Am J Neuroradiol 22(1):65–72
502 7 Meningiomas

31. Flannery TJ, Kano H, Lunsford LD, Sirin S, Tormenti 44. Koerbel A, Gharabaghi A, Safavi-Abbasi S, Samii A,
M, Niranjan A, Flickinger JC, Kondziolka D (2010) Ebner FH, Samii M, Tatagiba M (2009) Venous com-
Long-term control of petroclival meningiomas plications following petrosal vein sectioning in sur-
through radiosurgery. J Neurosurg 112(5):957–964. gery of petrous apex meningiomas. Eur J Surg Oncol
doi:10.3171/2009.8.JNS09695 35(7):773–779. doi:10.1016/j.ejso.2008.02.015,
32. Go KG, Kamman RL, Wilmink JT, Mooyaart EL S0748-7983(08)00085-1 [pii]
(1994) A study on peritumoral brain edema around 45. Krayenbuhl N, Pravdenkova S, Al-Mefty O (2007) De
meningiomas by MRI and contrast CT. Acta Neurochir novo versus transformed atypical and anaplastic
Suppl (Wien) 60:365–368 meningiomas: comparisons of clinical course, cytoge-
33. Goodwin JW, Crowley J, Eyre HJ, Stafford B, Jaeckle netics, cytokinetics, and outcome. Neurosurgery
KA, Townsend JJ (1993) A phase II evaluation of 61(3):495–503. doi:10.1227/01.NEU.0000290895.
tamoxifen in unresectable or refractory meningiomas: 92695.22, 00006123-200709000-00009 [pii]; discus-
a Southwest Oncology Group study. J Neurooncol sion 503–504
15(1):75–77 46. Lalwani AK (1992) Meningiomas, epidermoids, and
34. Goutagny S, Kalamarides M (2010) Meningiomas other nonacoustic tumors of the cerebellopontine
and neurofibromatosis. J Neurooncol 99(3):341–347. angle. Otolaryngol Clin North Am 25(3):707–728
doi:10.1007/s11060-010-0339-x 47. Lamszus K (2004) Meningioma pathology, genetics,
35. Goyal LK, Suh JH, Mohan DS, Prayson RA, Lee J, and biology. J Neuropathol Exp Neurol 63(4):
Barnett GH (2000) Local control and overall survival 275–286
in atypical meningioma: a retrospective study. Int J 48. Lang FF, Macdonald OK, Fuller GN, DeMonte F
Radiat Oncol Biol Phys 46(1):57–61, S0360- (2000) Primary extradural meningiomas: a report on
3016(99)00349-1 [pii] nine cases and review of the literature from the era of
36. Grey PL, Baguley DM, Moffat DA, Hardy DG, computerized tomography scanning. J Neurosurg
Beynon GJ (1996) Audiovestibular results after sur- 93(6):940–950. doi:10.3171/jns.2000.93.6.0940
gery for cerebellopontine angle meningiomas. Am J 49. Laudadio P, Canani FB, Cunsolo E (2004) Meningioma
Otol 17(4):634–638 of the internal auditory canal. Acta Otolaryngol
37. Grunberg SM, Weiss MH, Russell CA, Spitz IM, 124(10):1231–1234
Ahmadi J, Sadun A, Sitruk-Ware R (2006) Long-term 50. Levin M, Wertheim SE, Klein E, Lowry J (1995)
administration of mifepristone (RU486): clinical Unusual lytic intraosseous meningioma. J Neuroimaging
tolerance during extended treatment of meningioma. 5(4):247–248
Cancer Invest 24(8):727–733. doi:10.1080/ 51. Liu JK, Gottfried ON, Couldwell WT (2003) Surgical
07357900601062339, K6271K7768127257 [pii] management of posterior petrous meningiomas.
38. Hakim R, Alexander E 3rd, Loeffler JS, Shrieve DC, Neurosurg Focus 14(6):e7, 140607 [pii]
Wen P, Fallon MP, Stieg PE, Black PM (1998) Results 52. Lomas J, Bello MJ, Alonso ME, Gonzalez-Gomez P,
of linear accelerator-based radiosurgery for intracra- Arjona D, Kusak ME, de Campos JM, Sarasa JL, Rey
nial meningiomas. Neurosurgery 42(3):446–453; dis- JA (2002) Loss of chromosome 22 and absence of
cussion 453–454 NF2 gene mutation in a case of multiple meningiomas.
39. Hsu DW, Efird JT, Hedley-Whyte ET (1997) Hum Pathol 33(3):375–378, S004681770215324X
Progesterone and estrogen receptors in meningiomas: [pii]
prognostic considerations. J Neurosurg 86(1):113– 53. Louis DNSB, Budka H, von Deimling A, Kepes JJ
120. doi:10.3171/jns.1997.86.1.0113 (2000) Meningiomas. In: Kleihues PCW (ed)
40. Ide M, Jimbo M, Yamamoto M, Umebara Y, Hagiwara Pathology and Genetics of Tumours of the Nervous
S, Kubo O (1996) MIB-1 staining index and peritu- System: World Health Organization Classification of
moral brain edema of meningiomas. Cancer 78(1):133– Tumours. IARC Press, Lyon, pp 176–184
143, 10.1002/(SICI)1097-0142(19960701)78:1<133:: 54. Mahmood A, Caccamo DV, Tomecek FJ, Malik GM
AID-CNCR19>3.0.CO;2-0 [pii] 10.1002/(SICI)1097- (1993) Atypical and malignant meningiomas: a clini-
0142(19960701)78:1<133::AID-CNCR19>3.0.CO;2-0 copathological review. Neurosurgery 33(6):955–963
41. Jaaskelainen J, Haltia M, Servo A (1986) Atypical and 55. Maier H, Ofner D, Hittmair A, Kitz K, Budka H (1992)
anaplastic meningiomas: radiology, surgery, radiother- Classic, atypical, and anaplastic meningioma: three his-
apy, and outcome. Surg Neurol 25(3):233–242 topathological subtypes of clinical relevance. J Neurosurg
42. Katz TS, Amdur RJ, Yachnis AT, Mendenhall WM, 77(4):616–623. doi:10.3171/jns.1992.77.4.0616
Morris CG (2005) Pushing the limits of radiotherapy 56. Mallucci CL, Ward V, Carney AS, O’Donoghue GM,
for atypical and malignant meningioma. Am J Clin Robertson I (1999) Clinical features and outcomes in
Oncol 28(1):70–74, 00000421-200502000-00013 patients with non-acoustic cerebellopontine angle
[pii] tumours. J Neurol Neurosurg Psychiatry 66(6):
43. Kleihues P, Louis DN, Scheithauer BW, Rorke LB, 768–771
Reifenberger G, Burger PC, Cavenee WK (2002) The 57. Maniglia AJ, Fenstermaker RA, Ratcheson RA (1989)
WHO classification of tumors of the nervous system. Preservation of hearing in the surgical removal of
J Neuropathol Exp Neurol 61(3):215–225; discussion cerebellopontine angle tumors. Otolaryngol Clin
226–229 North Am 22(1):211–232
References 503

58. Matthies C, Carvalho G, Tatagiba M, Lima M, Samii 72. Norden AD, Drappatz J, Wen PY (2009) Advances in
M (1996) Meningiomas of the cerebellopontine angle. meningioma therapy. Curr Neurol Neurosci Rep
Acta Neurochir Suppl 65:86–91 9(3):231–240
59. Maurer PK, Okawara SH (1988) Restoration of hear- 73. Oguz KK, Cila A (2003) Rim enhancement of menin-
ing after removal of cerebellopontine angle menin- giomas on fast FLAIR imaging. Neuroradiology
gioma: diagnostic and therapeutic implications. 45(2):78–81. doi:10.1007/s00234-002-0914-8
Neurosurgery 22(3):573–575 74. Ojemann R (1990) Meningiomas. Neurosurg Clin N
60. Mautner VF, Lindenau M, Baser ME, Hazim W, Am 1(1):181–197
Tatagiba M, Haase W, Samii M, Wais R, Pulst SM 75. Palma L, Celli P, Franco C, Cervoni L, Cantore G
(1996) The neuroimaging and clinical spectrum of (1997) Long-term prognosis for atypical and malig-
neurofibromatosis 2. Neurosurgery 38(5):880–885; nant meningiomas: a study of 71 surgical cases.
discussion 885–886 Neurosurg Focus 2(4):e3, 020403 [pii]
61. McMullen KP, Stieber VW (2004) Meningioma: cur- 76. Parry DM, Eldridge R, Kaiser-Kupfer MI, Bouzas EA,
rent treatment options and future directions. Curr Pikus A, Patronas N (1994) Neurofibromatosis 2 (NF2):
Treat Options Oncol 5(6):499–509 clinical characteristics of 63 affected individuals and
62. Merrill MJ, Oldfield EH (2005) A reassessment of clinical evidence for heterogeneity. Am J Med Genet
vascular endothelial growth factor in central nervous 52(4):450–461. doi:10.1002/ajmg.1320520411
system pathology. J Neurosurg 103(5):853–868. 77. Perry A, Cai DX, Scheithauer BW, Swanson PE,
doi:10.3171/jns.2005.103.5.0853 Lohse CM, Newsham IF, Weaver A, Gutmann DH
63. Modha A, Gutin PH (2005) Diagnosis and treatment (2000) Merlin, DAL-1, and progesterone receptor
of atypical and anaplastic meningiomas: a review. expression in clinicopathologic subsets of menin-
Neurosurgery 57(3):538–550, 00006123-200509000- gioma: a correlative immunohistochemical study of
00017 [pii]; discussion 538–550 175 cases. J Neuropathol Exp Neurol 59(10):872–879
64. Mukhopadhyay D, Carrabba G, Guha A (2009) 78. Perry A, Scheithauer BW, Stafford SL, Lohse CM,
Meningiomas and Brain Edema. In: Pamir N BP, Wollan PC (1999) “Malignancy” in meningiomas:
Fahlbusch R (eds) Meningiomas: a comprehensive a clinicopathologic study of 116 patients, with grad-
text. Saunders Elsevier, Philadelphia, pp 135–145 ing implications. Cancer 85(9):2046–2056, 10.1002/
65. Nager GT, Masica DN (1970) Meningiomas of the (SICI)1097-0142(19990501)85:9<2046::AID-
cerebello-pontine angle and their relation to the tem- CNCR23>3.0.CO;2-M [pii]
poral bone. Laryngoscope 80(6):863–895 79. Perry A, Stafford SL, Scheithauer BW, Suman VJ, Lohse
66. Nakamura M, Roser F, Dormiani M, Matthies C, CM (1997) Meningioma grading: an analysis of histo-
Vorkapic P, Samii M (2005) Facial and cochlear nerve logic parameters. Am J Surg Pathol 21(12):1455–1465
function after surgery of cerebellopontine angle menin- 80. Politi M, Romeike BF, Papanagiotou P, Nabhan A,
giomas. Neurosurgery 57(1):77–90; discussion 77–90 Struffert T, Feiden W, Reith W (2005) Intraosseous
67. Nakamura M, Roser F, Dormiani M, Samii M, hemangioma of the skull with dural tail sign: radio-
Matthies C (2005) Intraoperative auditory brainstem logic features with pathologic correlation. AJNR Am
responses in patients with cerebellopontine angle J Neuroradiol 26(8):2049–2052, 26/8/2049 [pii]
meningiomas involving the inner auditory canal: anal- 81. Rokitansky C (1856) Lehrbuch der pathologischen
ysis of the predictive value of the responses. Anatomie. Wilhelm Braumüller, Wien
J Neurosurg 102(4):637–642. doi:10.3171/jns.2005. 82. Rosahl SK, Mirzayan MJ, Samii M (2004) Osteolytic
102.4.0637 intra-osseous meningiomas: illustrated review. Acta
68. Nakamura M, Roser F, Dormiani M, Vorkapic P, Neurochir (Wien) 146(11):1245–1249. doi:10.1007/
Samii M (2005) Surgical treatment of cerebellopon- s00701-004-0380-7
tine angle meningiomas in elderly patients. Acta 83. Roser F, Nakamura M, Dormiani M, Matthies C,
Neurochir (Wien) 147(6):603–609. doi:10.1007/ Vorkapic P, Samii M (2005) Meningiomas of the cer-
s00701-005-0517-3; discussion 609–610 ebellopontine angle with extension into the internal
69. Nakamura M, Roser F, Mirzai S, Matthies C, Vorkapic auditory canal. J Neurosurg 102(1):17–23.
P, Samii M (2004) Meningiomas of the internal audi- doi:10.3171.jns.2005.102.1.0017
tory canal. Neurosurgery 55(1):119–127; discussion 84. Ruggieri M, Iannetti P, Polizzi A, La Mantia I,
127–128 Spalice A, Giliberto O, Platania N, Gabriele AL,
70. Nakamura M, Samii M (2003) Surgical management Albanese V, Pavone L (2005) Earliest clinical mani-
of a meningioma in the retrosellar region. Acta festations and natural history of neurofibromatosis
Neurochir (Wien) 145(3):215–219 type 2 (NF2) in childhood: a study of 24
71. Noel G, Bollet MA, Calugaru V, Feuvret L, Haie- patients. Neuropediatrics 36(1):21–34. doi:10.1055/
Meder C, Dhermain F, Ferrand R, Boisserie G, s-2005-837581
Beaudre A, Mazeron JJ, Habrand JL (2005) Functional 85. Samii M, Gerganov VM (2008) Surgery of extra-
outcome of patients with benign meningioma treated axial tumors of the cerebral base. Neurosurgery
by 3D conformal irradiation with a combination of 62(6 Suppl 3):1153–1166. doi:10.1227/01.
photons and protons. Int J Radiat Oncol Biol Phys neu.0000333782.19682.76, 00006123-200806001-
62(5):1412–1422, S0360-3016(04)03155-4 [pii] 00024 [pii]; discussion 1166–1168
504 7 Meningiomas

86. Samii MAM, Ammirati M (1991) Cerebellopontine 101. Voss NF, Vrionis FD, Heilman CB, Robertson JH
angle meningiomas (Posterior pyramid menin- (2000) Meningiomas of the cerebellopontine angle.
giomas). In: Al-Mefty O (ed) Meningiomas. Raven, Surg Neurol 53(5):439–446, S0090-3019(00)00195-6
New York, pp 503–515 [pii]; discussion 446–447
87. Sanson M, Cornu P (2000) Biology of meningiomas. 102. Wallace EW (2004) The dural tail sign. Radiology
Acta Neurochir (Wien) 142(5):493–505 233(1):56–57. doi:10.1148/radiol.2331021332,
88. Sawaya RE, Ligon BL (1994) Thromboembolic 233/1/56 [pii]
complications associated with brain tumors. 103. Wellenreuther R, Kraus JA, Lenartz D, Menon AG,
J Neurooncol 22(2):173–181 Schramm J, Louis DN, Ramesh V, Gusella JF,
89. Schaller B, Heilbronner R, Pfaltz CR, Probst RR, Wiestler OD, von Deimling A (1995) Analysis of the
Gratzl O (1995) Preoperative and postoperative neurofibromatosis 2 gene reveals molecular variants
auditory and facial nerve function in cerebellopon- of meningioma. Am J Pathol 146(4):827–832
tine angle meningiomas. Otolaryngol Head 104. Wiemels J, Wrensch M, Claus EB (2010) Epidemiology
Neck Surg 112(2):228–234, S0194-5998(95)70241-5 and etiology of meningioma. J Neurooncol 99(3):307–
[pii] 314. doi:10.1007/s11060-010-0386-3
90. Schaller B, Merlo A, Gratzl O, Probst R (1999) 105. Wilms G, Plets C, Goossens L, Goffin J, Vanwambeke
Premeatal and retromeatal cerebellopontine angle K (1992) The radiological differentiation of acoustic
meningioma. Two distinct clinical entities. Acta neuroma and meningioma occurring together in the
Neurochir (Wien) 141(5):465–471 cerebellopontine angle. Neurosurgery 30(3):443–
91. Sekhar LN, Jannetta PJ (1984) Cerebellopontine 445; discussion 445–446
angle meningiomas. Microsurgical excision and fol- 106. Wu ZB, Yu CJ, Guan SS (2005) Posterior petrous
low-up results. J Neurosurg 60(3):500–505. meningiomas: 82 cases. J Neurosurg 102(2):284–
doi:10.3171/jns.1984.60.3.0500 289. doi:10.3171/jns.2005.102.2.0284
92. Sekhar LN, Swamy NK, Jaiswal V, Rubinstein E, 107. Yamaguchi N, Kawase T, Sagoh M, Ohira T, Shiga
Hirsch WE Jr, Wright DC (1994) Surgical excision H, Toya S (1997) Prediction of consistency of men-
of meningiomas involving the clivus: preoperative ingiomas with preoperative magnetic resonance
and intraoperative features as predictors of postop- imaging. Surg Neurol 48(6):579–583, S0090-
erative functional deterioration. J Neurosurg 3019(96)00439-9 [pii]
81(6):860–868. doi:10.3171/jns.1994.81.6.0860 108. Younis GA, Sawaya R, DeMonte F, Hess KR,
93. Shapiro WR, Hiesiger EM, Cooney GA, Basler GA, Albrecht S, Bruner JM (1995) Aggressive meningeal
Lipschutz LE, Posner JB (1990) Temporal effects of tumors: review of a series. J Neurosurg 82(1):17–27.
dexamethasone on blood-to-brain and blood-to- doi:10.3171/jns.1995.82.1.0017
tumor transport of 14C-alpha-aminoisobutyric acid 109. Zamani AA (2000) Cerebellopontine angle tumors:
in rat C6 glioma. J Neurooncol 8(3):197–204 role of magnetic resonance imaging. Top Magn
94. Simon MBJ, Hartmann C (2007) Molecular genetics Reson Imaging 11(2):98–107
of meningiomas: from basic research to potential
clinical applications. Neurosurgery 60:787–798
95. Stafford SLPB, Foote RL et al (2001) Meningioma
radiosurgery: tumor control, outcomes, and complica-
tions among 190 consecutive patients. Neurosurgery Petroclival Meningiomas
49:1029–1038
96. Tamiya T, Ono Y, Matsumoto K, Ohmoto T (2001) 110. Abdel Aziz KM, Sanan A, van Loveren HR, Tew JM
Peritumoral brain edema in intracranial menin- Jr, Keller JT, Pensak ML (2000) Petroclival
giomas: effects of radiological and histological fac- meningiomas: predictive parameters for transpetro-
tors. Neurosurgery 49(5):1046–1051; discussion sal approaches. Neurosurgery 47(1):139–150;
1051–1052 discussion 150–152
97. Tekkok IH, Suzer T, Erbengi A (1992) Non-acoustic 111. Al-Mefty O, Ayoubi S, Smith RR (1991) The petro-
tumors of the cerebellopontine angle. Neurosurg sal approach: indications, technique, and results.
Rev 15(2):117–123 Acta Neurochir Suppl (Wien) 53:166–170
98. Thomas NW, King TT (1996) Meningiomas of the 112. Al-Mefty O, Fox JL, Smith RR (1988) Petrosal
cerebellopontine angle. A report of 41 cases. Br J approach for petroclival meningiomas. Neurosurgery
Neurosurg 10(1):59–68 22(3):510–517
99. Vellutini EA, Cruz OL, Velasco OP, Miniti A, 113. Bambakidis NC, Kakarla UK, Kim LJ, Nakaji P,
Almeida GM (1991) Reversible hearing loss from Porter RW, Daspit CP, Spetzler RF (2007) Evolution
cerebellopontine angle tumors. Neurosurgery of surgical approaches in the treatment of petroclival
28(2):310–312; discussion 312–313 meningiomas: a retrospective review. Neurosurgery
100. Virchow R (1863) Die krankhaften Geschwülste: 61(5 Suppl 2):202–209. doi:10.1227/01.
Dreissig Vorlesungen gehalten während des neu.0000303218.61230.39; discussion 209–211
Wintersemesters 1862–1863 an der Universität zu 114. Bricolo AP, Turazzi S, Talacchi A, Cristofori L
Berlin. A. Hirschwald, Berlin (1992) Microsurgical removal of petroclival menin-
References 505

giomas: a report of 33 patients. Neurosurgery of large petroclival and cavernous sinus menin-
31(5):813–828; discussion 828 giomas. Surg Neurol 56(5):308–314, S0090-
115. Carvalho GA, Matthies C, Tatagiba M, Eghbal R, 3019(01)00622-X [pii]
Samii M (2000) Impact of computed tomographic 130. Janecka IP, Sen CN, Sekhar LN, Arriaga M (1990)
and magnetic resonance imaging findings on surgical Facial translocation: a new approach to the cranial
outcome in petroclival meningiomas. Neurosurgery base. Otolaryngol Head Neck Surg 103(3):413–419
47(6):1287–1294; discussion 1294–1295 131. Kawase T, Shiobara R, Toya S (1991) Anterior trans-
116. Castellano F, Ruggiero G (1953) Meningiomas of petrosal-transtentorial approach for sphenopetro-
the posterior fossa. Acta Radiol Suppl 104:1–177 clival meningiomas: surgical method and results in
117. Chanda A, Nanda A (2006) Retrosigmoid intradural 10 patients. Neurosurgery 28(6):869–875; discus-
suprameatal approach: advantages and disadvan- sion 875–876
tages from an anatomical perspective. Neurosurgery 132. Kawase T, Shiobara R, Toya S (1994) Middle fossa
59(1 Suppl 1):ONS1–ONS6. doi:10.1227/01. transpetrosal-transtentorial approaches for petroclival
NEU.0000220673.79877.30; discussion ONS1–ONS6 meningiomas. Selective pyramid resection and radi-
118. Couldwell WT, Fukushima T, Giannotta SL, Weiss cality. Acta Neurochir (Wien) 129(3–4):113–120
MH (1996) Petroclival meningiomas: surgical expe- 133. Little KM, Friedman AH, Sampson JH, Wanibuchi
rience in 109 cases. J Neurosurg 84(1):20–28. M, Fukushima T (2005) Surgical management of
doi:10.3171/jns.1996.84.1.0020 petroclival meningiomas: defining resection goals
119. Cushing H, Eisenberg L (1938) Meningiomas: Their based on risk of neurological morbidity and tumor
classification, regional behavior, life history and sur- recurrence rates in 137 patients. Neurosurgery
gical end results. Charles C Thomas, Springfield 56(3):546–559; discussion 546–559
120. Decker RE, Malis LI (1970) Surgical approaches to 134. Malis L (1991) The petrosal approach. Clin
midline lesions at the base of the skull: a review. Mt Neurosurg 37:528–540
Sinai J Med 37(2):84–102 135. Mathiesen T, Gerlich A, Kihlstrom L, Svensson M,
121. Erkmen K, Pravdenkova S, Al-Mefty O (2005) Bagger-Sjoback D (2008) Effects of using combined
Surgical management of petroclival meningiomas: transpetrosal surgical approaches to treat petroclival
factors determining the choice of approach. meningiomas. Neurosurgery 62(6 Suppl 3):1213–
Neurosurg Focus 19(2):E7, 190207 [pii] 1223. doi:10.1227/01.neu.0000333787.06221.e5
122. Fisch U (1978) Infratemporal fossa approach to 136. Mayberg MR, Symon L (1986) Meningiomas of the
tumours of the temporal bone and base of the skull. clivus and apical petrous bone. Report of 35 cases.
J Laryngol Otol 92(11):949–967 J Neurosurg 65(2):160–167. doi:10.3171/jns.1986.
123. Flannery TJ, Kano H, Lunsford LD, Sirin S, Tormenti 65.2.0160
M, Niranjan A, Flickinger JC, Kondziolka D (2010) 137. Morrison AW, King TT (1973) Experiences with a
Long-term control of petroclival meningiomas translabyrinthine-transtentorial approach to the cer-
through radiosurgery. J Neurosurg 112(5):957–964. ebellopontine angle. Technical note. J Neurosurg
doi:10.3171/2009.8.JNS09695 38(3):382–390. doi:10.3171/jns.1973.38.3.0382
124. Hakuba A (1978) Total removal of cerebellopontine 138. Natarajan SK, Sekhar LN, Schessel D, Morita A
angle tumors with a combined transpetrosal-tran- (2007) Petroclival meningiomas: multimodality
stentorial approach (author’s transl). No Shinkei treatment and outcomes at long-term follow-up.
Geka 6(4):347–354 Neurosurgery 60(6):965–979. doi:10.1227/01.
125. Hakuba A, Nishimura S, Jang BJ (1988) A combined NEU.0000255472.52882.D6; discussion 979–981
retroauricular and preauricular transpetrosal-tran- 139. Ramina R, Neto MC, Fernandes YB, Silva EB,
stentorial approach to clivus meningiomas. Surg Mattei TA, Aguiar PH (2008) Surgical removal of
Neurol 30(2):108–116 small petroclival meningiomas. Acta Neurochir
126. Hakuba A, Nishimura S, Tanaka K, Kishi H, (Wien) 150(5):431–438. doi:10.1007/s00701-007-
Nakamura T (1977) Clivus meningioma: six cases of 1403-y; discussion 438–439
total removal. Neurol Med Chir (Tokyo) 17(1 Pt 1): 140. Rhoton AL Jr (2000) The temporal bone and tran-
63–77 stemporal approaches. Neurosurgery 47(3
127. House WF, De la Cruz A, Hitselberger WE (1978) Suppl):S211–S265
Surgery of the skull base: transcochlear approach to 141. Roche PH, Pellet W, Fuentes S, Thomassin JM, Regis
the petrous apex and clivus. Otolaryngology J (2003) Gamma knife radiosurgical management of
86(5):ORL-770–ORL-779 petroclival meningiomas results and indications. Acta
128. Ichimura S, Kawase T, Onozuka S, Yoshida K, Ohira Neurochir (Wien) 145(10):883–888. doi:10.1007/
T (2008) Four subtypes of petroclival meningiomas: s00701-003-0123-1; discussion 888
differences in symptoms and operative findings 142. Salamon GM, Combalbert A, Raybaud C, Gonzalez
using the anterior transpetrosal approach. Acta J (1971) An angiographic study of meningiomas of
Neurochir (Wien) 150(7):637–645. doi:10.1007/ the posterior fossa. J Neurosurg 35(6):731–741.
s00701-008-1586-x doi:10.3171/jns.1971.35.6.0731
129. Iwai Y, Yamanaka K, Nakajima H (2001) Two- 143. Samii M, Ammirati M (1988) The combined supra-
staged gamma knife radiosurgery for the treatment infratentorial pre-sigmoid sinus avenue to the
506 7 Meningiomas

petro-clival region Surgical technique and clinical 157. Van Havenbergh T, Carvalho G, Tatagiba M, Plets C,
applications. Acta Neurochir (Wien) 95(1–2): Samii M (2003) Natural history of petroclival menin-
6–12 giomas. Neurosurgery 52(1):55–62; discussion 62–64
144. Samii M, Ammirati M, Mahran A, Bini W, Sepehrnia 158. Zachenhofer I, Wolfsberger S, Aichholzer M,
A (1989) Surgery of petroclival meningiomas: report Bertalanffy A, Roessler K, Kitz K, Knosp E (2006)
of 24 cases. Neurosurgery 24(1):12–17 Gamma-knife radiosurgery for cranial base menin-
145. Samii M, Carvalho GA, Tatagiba M, Matthies C giomas: experience of tumor control, clinical course,
(1997) Surgical management of meningiomas origi- and morbidity in a follow-up of more than 8 years.
nating in Meckel’s cave. Neurosurgery 41(4):767– Neurosurgery 58(1):28–36, 00006123-200601000-
774; discussion 774–775 00003 [pii]; discussion 28–36
146. Samii M, Gerganov V, Giordano M, Samii A (2010) 159. Zentner J, Meyer B, Vieweg U, Herberhold C,
Two step approach for surgical removal of petro- Schramm J (1997) Petroclival meningiomas: is radi-
clival meningiomas with large supratentorial exten- cal resection always the best option? J Neurol
sion. Neurosurg Rev 34(2):173–179. doi:10.1007/ Neurosurg Psychiatry 62(4):341–345
s10143-010-0299-9
147. Samii M, Gerganov VM (2008) Surgery of extra-
axial tumors of the cerebral base. Neurosurgery 62(6
Suppl 3):1153–1166. doi:10.1227/01.
Foramen Magnum Meningiomas
neu.0000333782.19682.76, 00006123-200806001-
00024 [pii]; discussion 1166–1168 160. Arnautovic KI, Al-Mefty O, Husain M (2000)
148. Samii M, Tatagiba M (1992) Experience with 36 sur- Ventral foramen magnum meningiomas. J Neurosurg
gical cases of petroclival meningiomas. Acta 92(1 Suppl):71–80
Neurochir (Wien) 118(1–2):27–32 161. Bassiouni H, Ntoukas V, Asgari S, Sandalcioglu EI,
149. Samii M, Tatagiba M, Carvalho GA (2000) Stolke D, Seifert V (2006) Foramen magnum menin-
Retrosigmoid intradural suprameatal approach to giomas: clinical outcome after microsurgical resec-
Meckel’s cave and the middle fossa: surgical tech- tion via a posterolateral suboccipital retrocondylar
nique and outcome. J Neurosurg 92(2):235–241. approach. Neurosurgery 59(6):1177–1185.
doi:10.3171/jns.2000.92.2.0235 doi:10.1227/01.NEU.0000245629.77968.37; discus-
150. Samii M, Ammirati M (1991) Cerebellopontine sion 1185–1187
angle meningiomas (Posterior pyramid menin- 162. Bejjani GK, Sekhar LN, Riedel CJ (2000)
giomas). In: Al-Mefty O (ed) Meningiomas. Raven, Occipitocervical fusion following the extreme lateral
New York, pp 503–515 transcondylar approach. Surg Neurol 54(2):109–115,
151. Seifert V (2010) Clinical management of petroclival S0090-3019(00)00255-X [pii]; discussion 115–106
meningiomas and the eternal quest for preservation 163. Bertalanffy H, Seeger W (1991) The dorsolateral,
of quality of life: personal experiences over a period suboccipital, transcondylar approach to the lower
of 20 years. Acta Neurochir (Wien) 152(7):1099– clivus and anterior portion of the craniocervical
1116. doi:10.1007/s00701-010-0633-6 junction. Neurosurgery 29(6):815–821
152. Sekhar LN, Wright DC, Richardson R, Monacci W 164. Borba LA, de Oliveira JG, Giudicissi-Filho M, Colli
(1996) Petroclival and foramen magnum menin- BO (2009) Surgical management of foramen mag-
giomas: surgical approaches and pitfalls. J Neurooncol num meningiomas. Neurosurg Rev 32(1):49–58.
29(3):249–259 doi:10.1007/s10143-008-0161-5; discussion 59–60
153. Spallone A, Makhmudov UB, Mukhamedjanov DJ, 165. Boulton MR, Cusimano MD (2003) Foramen mag-
Tcherekajev VA (1999) Petroclival meningioma. An num meningiomas: concepts, classifications, and
attempt to define the role of skull base approaches in nuances. Neurosurg Focus 14(6):e10, 140610 [pii]
their surgical management. Surg Neurol 51(4):412– 166. Bruneau M, George B (2008) Foramen magnum
419, S0090-3019(98)00100-1 [pii]; discussion meningiomas: detailed surgical approaches and
419–420 technical aspects at Lariboisiere Hospital and review
154. Spetzler RF, Daspit CP, Pappas CT (1992) The com- of the literature. Neurosurg Rev 31(1):19–32.
bined supra- and infratentorial approach for lesions doi:10.1007/s10143-007-0097-1; discussion 32–33
of the petrous and clival regions: experience with 46 167. David CA, Spetzler RF (1997) Foramen magnum
cases. J Neurosurg 76(4):588–599. doi:10.3171/ meningiomas. Clin Neurosurg 44:467–489
jns.1992.76.4.0588 168. George B (1991) Meningiomas of the foramen mag-
155. Stafford SL, Pollock BE, Foote RL, Link MJ, num. In: Schmidek HH (ed) Meningiomas and Their
Gorman DA, Schomberg PJ, Leavitt JA (2001) Surgical Management. WB Saunders, Philadelphia,
Meningioma radiosurgery: tumor control, outcomes, pp 459–470
and complications among 190 consecutive patients. 169. George B, Lot G, Boissonnet H (1997) Meningioma
Neurosurgery 49:1029–1038 of the foramen magnum: a series of 40 cases. Surg
156. Tedeschi H, Rhoton AL Jr (1994) Lateral approaches Neurol 47(4):371–379, S0090-3019(96)00204-2 [pii]
to the petroclival region. Surg Neurol 41(3):180– 170. Goel A, Desai K, Muzumdar D (2001) Surgery on
216, 0090-3019(94)90123-6 [pii] anterior foramen magnum meningiomas using a con-
References 507

ventional posterior suboccipital approach: a report 176. Rhoton AL Jr (2000) The foramen magnum.
on an experience with 17 cases. Neurosurgery Neurosurgery 47(3 Suppl):S155–S193
49(1):102–106; discussion 106–107 177. Samii M, Gerganov VM (2008) Surgery of extra-
171. Heros RC (1986) Lateral suboccipital approach for axial tumors of the cerebral base. Neurosurgery 62(6
vertebral and vertebrobasilar artery lesions. Suppl 3):1153–1166. doi:10.1227/01.neu.
J Neurosurg 64(4):559–562. doi:10.3171/jns.1986. 0000333782.19682.76; discussion 1166–1168
64.4.0559 178. Samii M, Gerganov V (2008) Surgery of extraaxial
172. Margalit NS, Lesser JB, Singer M, Sen C (2005) tumors of the skull base. Neurosurgery 62(6):
Lateral approach to anterolateral tumors at the fora- 1153–1168
men magnum: factors determining surgical proce- 179. Samii M, Klekamp J, Carvalho G (1996) Surgical
dure. Neurosurgery 56(2 Suppl):324–336; discussion results for meningiomas of the craniocervical junc-
324–336 tion. Neurosurgery 39(6):1086–1094; discussion
173. Menezes AH (2008) Surgical approaches: postoper- 1094–1095
ative care and complications “posterolateral-far 180. Shin H, Barrenechea IJ, Lesser J, Sen C, Perin
lateral transcondylar approach to the ventral fora- NI (2006) Occipitocervical fusion after resection of
men magnum and upper cervical spinal canal”. craniovertebral junction tumors. J Neurosurg
Childs Nerv Syst 24(10):1203–1207. doi:10.1007/ Spine 4(2):137–144. doi:10.3171/spi.2006.4.2.137
s00381-008-0597-5 181. Vishteh AG, Crawford NR, Melton MS, Spetzler RF,
174. Menezes AH, Traynelis VC, Gantz BJ (1994) Sonntag VK, Dickman CA (1999) Stability of the
Surgical approaches to the craniovertebral junction. craniovertebral junction after unilateral occipital
Clin Neurosurg 41:187–203 condyle resection: a biomechanical study. J Neurosurg
175. Rhoton AL Jr (1994) Meningiomas of the cerebel- 90(1 Suppl):91–98
lopontine angle and foramen magnum. Neurosurg
Clin N Am 5(2):349–377
Cerebellopontine Angle
Epidermoids 8

General Features Neuroimaging

Epidermoids or epidermoid cysts are congenital Radiologically, epidermoids are well-delineated


lesions that arise from ectodermal cells, mis- cystic lesions that have irregular lobulated or
placed during an aberrant closure of the neural scalloped margins. On CT, they are hypodense;
tube [25, 30]. According to the most popular rim or mural calcification is rarely observed
theory, the epithelial cell rests are transplanted (Fig. 8.3) [9, 22].
to the CP angle by the laterally migrating otic On T1-weighted MR images, epidermoids
capsule or by the developing neurovasculature. are characteristically heterogeneous lesions
Epidermoids are the most common intracranial with low signal intensity, usually isointense to
embryonal tumor and are most frequently located CSF (Fig. 8.3). T2-weighted MR images typi-
in the CP angle, in which they comprise 4–7 % cally demonstrate a nonhomogeneous lesion
of all lesions [3, 10, 29]. They are the third most with high signal intensity [11, 13, 31]. The
common CP angle tumor [15, 25, 29]. intensity of the signal on MRI is related to the
The epidermoid cyst wall is composed of cyst contents: in case cholesterol predominates,
stratified squamous epithelium, while the central the cysts are hyperintense on T1-weighted
core contains solid keratin protein—a pearly images and hypointense on T2-weighted images.
blistering material. The tumors are hence The main differential diagnosis of epidermoids
referred to as “pearly tumors.” They grow from is arachnoid cysts. Their radiologic characteris-
the slow desquamation of the stratified kerati- tics on routine CT and MRI are similar [11].
nized epithelium, which lines the cyst and spread Arachnoid cysts have characteristically less
along pathways of least resistance into the pos- mass effect and are more homogeneous on
terior fossa cisterns [15, 23]. Epidermoids cause T2-weighted images. In contrast to arachnoid
gradual displacement of neighboring neural cysts, epidermoids cause no adjacent bony reac-
structures and encase cranial nerves and vessels tion on bone-window CT [6]. Fluid-attenuated
(Fig. 8.1). Over the time, dense adhesions to cra- inversion recovery imaging (FLAIR), diffusion-
nial nerves or to brain stem may develop. weighted imaging, and CISS spin-echo MR
Extension into the Meckels’ cave, IAC, petrous images are more accurate imaging modalities
bone, or jugular foramen is occasionally seen that allow for definitive differentiation of these
(Figs. 8.2 and 8.3). entities [13]. On the FLAIR sequence, epider-
CP angle epidermoids, if untreated, tend to moid cysts can be easily differentiated from
extend through the tentorial incisura into the mid- arachnoid cysts because the former show mixed
dle cranial fossa toward the contralateral CP angle iso- to hypersignal intensities but with poor
or toward the foramen magnum (Table 8.1). demarcation, while the signal of the latter is

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 509


DOI 10.1007/978-3-642-35422-9_8, © Springer-Verlag Berlin Heidelberg 2013
510 8 Cerebellopontine Angle Epidermoids

a b

c
d

Fig. 8.1 CT and MRI images of a large CP epidermoid, extending into the prepontine cisterns and even to the
contralateral CP angle
Management 511

Clinical Presentation
a
The very slow growth of CP epidermoids causes
insidious and protracted course of symptoms and
signs. The typical patient is usually in the third to
fifth decades of life and has a long-standing
history of tinnitus and hearing loss [18, 25].
Symptoms are caused by compression of neuro-
vascular structures or by irritation due to the
adhesion of cysts’ capsule. The most common
symptoms are hearing impairment and trigeminal
neuralgia. Other common signs/symptoms are
headache, cerebellar and pyramidal signs, facial
weakness, hemifacial spasm, glossopharyngeal
neuralgia, lower cranial nerve dysfunction, and
b double vision [3, 5, 9, 10, 24]. Vestibular symp-
toms are rarely seen. Epidermoids cause trigemi-
nal neuralgia and facial nerve signs more
frequently than other CP angle tumor [25].
Patients may present with classical trigeminal
neuralgia or with constant neuralgic facial pain
[14]. The cranial nerves hyperactive dysfunction
Fig. 8.2 CISS MR sequence, clearly demonstrating the syndrome may be caused by several mechanisms
epidermoid in the IAC and a smaller part protruding into or combination thereof: direct compressive effect,
the CP angle “strangle” of the nerve caused by the growing
tumor with consequent reduction of its blood
suppressed, like the signal of CSF [16]. The supply, pushing the nerve against a pulsating
presence of residual tumor is also well visual- blood vessel, or local irritation from cholesterol
ized on these later sequences [27]. oozing [9, 14, 17, 25]. Cyst rupture—although
MR cisternography, by means of heavily rare—may cause recurrent episodes of aseptic
T2-weighted 3D sequence, demonstrates the epi- meningitis and lead to hydrocephalus [1, 7].
dermoid cyst signal hypointense to CSF, reveals
the lobulated margins of the tumor, and clearly
depicts the anatomical relation to neighboring Management
nerves and vessels, allowing precise surgical
planning [16, 21]. DWI offers a finding specific The treatment of choice of CP epidermoids is
for extra-axial epidermoid cysts by showing a surgical, and the preferred approach is the ret-
very high signal. Restricted ADC compared to rosigmoid [14, 15, 20, 25, 28, 30]—see Case
CSF, almost comparable to that of the brain, and Illustrations 8.1, 8.2, 8.3, and 8.4. The approach
T2 shine-through effect both play an important allows for removal of epidermoids of any
role in the high signal intensity of epidermoid size, even those extending beyond the CP angle
cyst at DWI (Figs. 8.3 and 8.4) [4, 16]. MR spec- [12, 25, 30]. We discourage from utilization of
troscopy might be helpful, as it shows only ele- translabyrinthine or any other hearing destructive
vated lactate peaks. approach because after tumor removal, hearing
Epidermoids are nonenhancing lesions. In can improve or even recover.
case an enhancing component is visualized, a For detailed description of the RS approach,
squamous carcinoma arising from an epidermoid please refer to the appropriate chapter. The cra-
should be suspected [19]. nial nerves are usually engulfed by the tumor
512 8 Cerebellopontine Angle Epidermoids

and usually not significantly dislocated. Due to levels—see Case Illustration 8.3. A lateral-to-
the long-standing compression, they might medial direction of removal is followed, which
have altered morphology and be very vulnera- allows for earlier identification of the cranial
ble, in particular the cochlear nerve. Large epi- nerves near their bony or dural entrance. The
dermoids with considerable extension are tumor is removed completely in one region
removed consequently at each of the CP angle before moving to the next one, thereby

a b

c d

Fig. 8.3 CT and MRI of a patient with a CP angle epider- bone-window scan b. Note the erosion of the left petrous
moid tumor with extension into the Meckels’ cave and apex (b). (d) T1-weighted sequence after gadolinium
erosion of the petrous apex. (a, b) Native (a) and bone- administration. (e) FLAIR sequence. (f, g) T2-CISS
window (b, c) CT scans. The hyperdense signal on the sequences. (h) ADC. (i) Diffusion MRI. (j–l) Proton den-
native CT is due to multiple calcifications, as seen on the sity sequences
Management 513

e f

g h

Fig. 8.3 (continued)


514 8 Cerebellopontine Angle Epidermoids

i j

k l

Fig. 8.3 (continued)

minimizing the risk of overlooking a remnant. microscope control. When a smaller tumor part
The powerful ultrasonic aspiration can easily remains, it is hold with one hand and the suc-
cause injury to neural structures and is not rec- tion with the other hand. Thus, inadvertent tear
ommendable. Simple suction, tumor forceps, or suction-induced injury of the cranial nerves
microdissector, and platelet knife are usually can be avoided. Very important consideration
sufficient. A helpful maneuver to avoid iatro- in epidermoids is that major vessels may be
genic injury is to decrease the suction power embedded in the tumor mass, in contrast to VS,
while introducing or removing the suction tip in which the large vessels are on the tumor
and increase it, while working under direct capsule.
Management 515

Table 8.1 Tumor location and extension (n = 60) access to a narrow deep space, as well as the lack
Location/extension of CP angle of appropriate instruments, prohibits its safe
epidermoids Rate (%) removal. Therefore, very large epidermoids with
CP angle alone 37 considerable supratentorial part have to be
CP angle + transtentorial extension 8 removed in two steps (Figs. 8.6, 8.7, and 8.8).
CP angle + parasellar extension 5 Tumors that involve the contralateral CP angle
CP angle + middle fossa extension 10 are also removed in two separate steps. The
CP angle + foramen magnum extension 18
symptomatic side is approached initially. Notably,
CP angle + transtentorial and foramen 17
magnum
the time between the two procedures should be
Tumor reaches midline 55 sufficient to allow recovery or improvement of
Contralateral CP angle 26 potential neurological deficits, especially of the
Modified from Samii et al. [25] lower cranial nerve dysfunction. The contralat-
eral tumor is approached more conservatively to
avoid bilateral cranial nerve deficits.
The removal of tumor capsule is performed During surgery, spillage of the epidermoid
only when sufficient debulking has been achieved, contents should be avoided in order to prevent
similar to the case of all other CP angle tumors. aseptic meningitis or later recurrences.
Capsule dissection is performed under optimal Furthermore, communicating hydrocephalus can
visualization and illumination with the two-hand develop in a delayed fashion due to leakage of
technique. In order to prevent additional neuro- cyst contents into the operative field [2]. Following
logical deficits, small remnants of the capsule tumor removal and hemostasis, the surgical field
that are firmly attached to important structures should be thoroughly irrigated to ensure that all
should not be removed. particles are eliminated.
The endoscope is an extremely useful tool in
case of CP angle epidermoids and should be
always available—see Case Illustration 8.3 and Outcome
8.4. We use straight and angled scopes—usually
30° and exceptionally 75° (Karl Storz GmbH). It The rate of radical removal in different published
is used after the microscopically visible tumor series ranges from 38 to 75 % [20, 24–26, 28].
part has been removed to inspect for hidden tumor The senior authors’ experience encompasses
remnants. All corners of the CP angle should be more than 110 CP angle epidermoids, removed
systematically inspected. Any remaining tumor via the RS approach. Complete removal was
can be removed either under microscopic or achieved in 75 % of the cases. Surgical morbidity
endoscopic control. was related mainly to cranial nerve dysfunction.
Epidermoids located purely in the petrous Possible complication after surgery is aseptic
bone are exceptionally rare. Their removal meningitis, which is reported to vary between 2.5
requires different approaches, depending on the and 40 % [25]. Its occurrence might be prevented
exact extension of the tumor (Fig. 8.5). by careful operative technique and application of
Large epidermoids, extending into adjacent dexamethasone, 16–24 mg/day, for 1–2 weeks
cranial base spaces, pose specific management after surgery.
problems. Tumors with transtentorial extension
or growth toward the foramen magnum might be
removed via the RS approach with the additional Recurrences
use of angled endoscopes. The additional inci-
sion of the tentorium and use of endoscope, how- Epidermoids tend to recur even decades after pri-
ever, does not always allow complete removal of mary surgery. In subtotally resected epidermoids,
the remaining portion. Actually, tumor rests are recurrences can develop even after 20 or 30 years.
frequently well visualized, but the restricted Close follow-up of such patients is therefore
516 8 Cerebellopontine Angle Epidermoids

mandatory. The radiological detection and differ- possible residual or recurrent tumor. Considering
entiation of small recurrent epidermoids from the very slow growth rate of small remnants,
CSF may be difficult [24]. Their diagnosis is observation might be the most adequate initial
based on specific MR sequences: the fast fluid- strategy. Repeated operative removal is indicated
attenuated inversion recovery (fast-FLAIR) MR in case the recurrence becomes symptomatic or
imaging and the echo-planar diffusion-weighted reaches considerable size and a steady growth
imaging [8]. Dechambre et al. [8] demonstrated tendency is observed. Such surgeries are more
the usefulness of DWI for postoperative follow- difficult and risky due to the development of scar
up as it allows confirmation of the presence of a tissue and adhesions.

a b

c d

Fig. 8.4 Postoperative imaging of the patient from Fig. 8.3—(a) Native CT scan. (b) T1-weighted native sequence.
(c) T1-weighted sequence after gadolinium. (d) FLAIR sequence. (e) T2-CISS sequence. (f) ADC. (g) Diffusion MRI
Management 517

e f

Fig. 8.4 (continued)


518 8 Cerebellopontine Angle Epidermoids

a b

c d

Fig. 8.5 Petrous bone epidermoid. The patient had been left side. (a, b) Bone-window CT scans, demonstrating
operated for the first time 12 years earlier in another facil- the bone remodeling. (c–f) MR images. We decided in
ity, and complete removal of a medial petrous epidermoid favor of the subtemporal epidural approach and succeeded
had been achieved. Five years later, a recurrence was to remove the tumor completely utilizing endoscope-
detected and was initially observed. It showed, however, assisted microsurgical technique. The patient did not have
growth tendency. At presentation, the patient had trigemi- new neurological deficits following surgery
nal hypesthesia and dysesthesia, as well as anacusis on the
Management 519

e f

Fig. 8.5 (continued)

a b

Fig. 8.6 Initial MR images, showing a giant epidermoid approaches—was taken. Due to the severe visual deficit,
with massive supra- and infratentorial extension. (a, b) the supratentorial part was approached initially. The
T1-weighted sequences after gadolinium administration. tumor was removed microsurgically with endoscopic
(c, d) FLAIR sequences. (e–g) T2-sequences. (h–j) CISS assistance, and decompression of the olfactory, optic,
sequences. This 32-year-old female patient presented oculomotor, and abducens nerves, as well as of the pitu-
with decreased bilateral vision, mainly on the left side itary stalk and the internal carotid and basilar artery and
(she could not count fingers >20 cm distance) and right- their branches, was achieved. The patient recovered well
sided homonymous hemianopia. She has been diagnosed after surgery but on the seventh post-op day, developed
of having an epidermoid incidentally 6 years earlier at severe hyponatremia due to a SIADH. The endocrino-
another institution and has been followed conservatively logical examination revealed a slight hypocortisolism
with irregular MRI scans. Considering the size and exten- and hypothyroidism, necessitating substitution. After
sion pattern of the lesion, a decision in favor of two-step regulation of the electrolyte homeostasis, she was
tumor removal—via a frontotemporal and retrosigmoid discharged
520 8 Cerebellopontine Angle Epidermoids

c d

e f

Fig. 8.6 (continued)


Management 521

g h

i j

Fig. 8.6 (continued)


522 8 Cerebellopontine Angle Epidermoids

a b

c d

f
Management 523

a b

Fig. 8.8 CT scan before (a) and after (b) the second patient recovered rapidly and was discharged without new
surgery: removal of the remaining infratentorial tumor via neurological deficits
a retrosigmoid approach with endoscopic assistance. The

Fig. 8.7 MR images prior to the second surgery. The homonymous hemianopia persisted. No double vision or
patient was admitted again in 6 months for the planned other symptoms were noted. The hormonal levels were
second stage. Her vision was significantly better, but the normal besides the hypothyroidism
524 8 Cerebellopontine Angle Epidermoids

Case Illustration 8.1 A microhook was used to remove the tumor


CP Angle Epidermoid 1 from jugular foramen (j); the tumor mass
This 43-year-old male patient presented with could be then mobilized from the lower cra-
long-standing history of hearing loss on the nial nerves (k). Further removal allowed visu-
right side. MRI was performed 2 months prior alization of the choroid plexus of the fourth
to admission and showed an epidermoid tumor ventricle, which was densely adherent to the
in the right CP angle. At admission, the patient epidermoid capsule (l). The PICA was gradu-
had right-sided hypacusis—approximately ally dissected from the capsule. Extreme care
40 dB hearing loss in the main speech area. He was paid to preserve all tiny arterial branches
had no further neurological deficits or com- (m). The tumor capsule was dissected from
plaints. The MRI study demonstrated the the brain stem with the two-hand technique
medium-sized epidermoid that was embedded (BS—brain stem). In the depth of the opera-
in the medial part of the lower CP angle in the tive field, the vertebral artery (VA) was then
space between the brain stem and cerebellum seen. The epidermoid (n) was actually embed-
(the cerebellopontine fissure). It looked inti- ded in the brain stem between the exit/entry
mately related to the most medial parts of the zone of the facial and cochlear nerves. These
seventh and eighth cranial nerves, in particular nerves were preserved anatomically; however,
to their exit/entry zones. On T1- and the cochlear nerve was severely changed by the
T2-weighted images, the lesions were isoin- long-standing compression, and during its dis-
tense to the CSF (a, c, d). No contrast uptake section, the ABR wave V was lost. Although
was demonstrated. The epidermoid, as well as it was extremely adherent to the brain stem,
its relations to the cranial nerves, was clearly the last tumor part was then also removed (o).
seen on the CISS study (e). All neural and vascular structures of the CP
The patient was operated via the standard angle were preserved, including the petrous
RS approach. The CP angle was exposed, and vein (p, q—arrow). In this case, the endo-
the epidermoid located at the level between scopic inspection confirmed the complete-
the seventh and eighth and the lower cranial ness of tumor removal—no residual tumor
nerves (*) was seen (f). It was medial to the was found. Additional advantage of the endo-
lower cranial nerves and had a small exten- scope was that it allowed directly excluding
sion into the jugular foramen. The epidermoid bleeding from infratentorial bridging veins
was debulked using microdissector (g) and (r–t: arrowhead, trochlear nerve; 5, trigemi-
tumor forceps (h). The arrow points at the nal nerve; 7, facial nerve; BS, brain stem; P,
tumor part entering the jugular foramen (i). posterior pyramid).
Management 525

a b

c d

MRI of the patient demonstrating the epidermoid cyst trating the consecutive steps of the epidermoid removal
in the right CP angle (a–e). Intraoperative images illus- (f–t). See the text for explanations
526 8 Cerebellopontine Angle Epidermoids

e f

g h
Management 527

i j

k l
528 8 Cerebellopontine Angle Epidermoids

m n

o p
Management 529

q r

s t
530 8 Cerebellopontine Angle Epidermoids

Case Illustration 8.2 caudally. Tumor removal was performed ini-


CP Angle Epidermoid 2 tially purely endoscopically. Later on, in
This 31-year-old female patient presented order to dissect the tightly attached tumor
with left-sided facial hypesthesia (V1,2), pro- capsule from the brain stem, we switched to
gressive facial weakness (HB grade IV), and microscopic visualization. The angled endo-
hearing loss (PTA 20 dB). The diagnosis of a scope was used to inspect those parts of the
CP angle cystic tumor was made 1 year earlier operative field that remained hidden when the
at another facility, but the patient was preg- microscope was used, and a remnant at
nant, and follow-up was recommended. the brain stem, medial to the exit zone of the
During the last several months, however, the facial nerve, was identified and removed.
hearing deteriorated, and a new MRI, pre- Complete removal of the epidermoid was thus
sented on figures a–g, was made. achieved.
The patient was operated via the RS The patient had an uneventful postoperative
approach (h–s). The dura was incised along period. Her hearing level remained unchanged.
the transverse and sigmoid sinuses, and the Intensive facial training was started, and the
posterior aspect of the tumor was seen. The facial nerve function slightly improved. The
seventh–eighth nerve complex was dislocated postoperative MRI is presented on figures T–U.
Management 531

a b

c d

(a–g) MRI showed a lesion in the left CP angle with view. Note the preserved vessels within the tumor and
characteristics, compatible with an epidermoid. (a, b) the petrous vein. (l) Further bimanual removal and dis-
T1-weighted sequence after gadolinium enhancement. section of the epidermoid capsule. (m) The major part of
(c, d) T2-weighted sequence. (e) Proton density sequence. the tumor is removed; the trigeminal (5) and the seventh–
(f) T2-CISS sequence. (g) FLAIR sequence. (h–s) eighth cranial nerves are free from tumor. Piece of the
Intraoperative images: (h) initial microscopic view of the tumor capsule was densely adherent to the brain stem
tumor, located superior to the seventh–eighth cranial (m, n—asterisk; BS brain stem). (o) This part was
nerves (7–8), reaching superior the tentorium (T). removed under microscopic visualization. (p–r)
P petrous bone, R retractor. (i) Corresponding endo- Endoscopic control at the end of surgery and the corre-
scopic view. (j) Tumor removal under pure endoscopic sponding microscopic view (s). (t, u) Postoperative MR
observation. (k) Gradually the brain stem came into images, demonstrating the completeness of removal
532 8 Cerebellopontine Angle Epidermoids

e f

h
g

i
j
Management 533

k l

m n

o p
534 8 Cerebellopontine Angle Epidermoids

q r

s t

u
Management 535

Case Illustration 8.3 Tumor removal continued medially along


CP Angle Epidermoid 3 these nerves up to the brain stem. This part
One year prior to presentation, this 55-year-old of the epidermoids’ capsule was very adher-
male patient had a transient ischemic attack. ent to the pia of brain stem. Still, using the
MR examination of the brain was performed, two-hand dissection technique, the capsule
and a cystic lesion in the right CP angle was could be completely dissected, and all small
visualized. Radiological follow-up was rec- vessels were preserved. As noted earlier in
ommended. Since several months, however, epidermoids—in contrast to VS—major ves-
he noticed some hearing deterioration and sels can be embedded in the tumor mass. In
numbness of the right half of the face. At the current case, a complex arterial network
admission, he had a trigeminal hypesthesia was found deep in the lower tumor part. The
and paresthesia (V1–3) and hearing loss on tumor had to be removed between the ves-
the right side (audiogram: hearing loss of sels, and its capsule had to be dissected from
35 dB in the main speech area). MRI revealed the vessel wall.
a cystic lesion in the upper right CP angle The inspection with angled endoscope dem-
(a–e). The characteristics of the lesion were onstrated complete removal of the epidermoid
compatible to those of an epidermoid. in the CP angle. A remnant was seen, however,
The patient was operated via the RS in the IAC and removed with a microhook
approach. Initially, only the epidermoid, under microscopic control. The endoscope was
extending lateral to the IAC, could be seen. used again at the end to confirm the complete-
The cranial nerves were hidden—they were ness of tumor removal.
either embedded in the tumor or compressed The patient recovered rapidly from surgery
medially. An extremely careful tumor debulk- and had no new neurological deficits. Hearing
ing was started. The fifth cranial nerve—and was preserved at the preoperative level. At the
later the seventh–eighth nerve complex— sixth postoperative day, a subcutaneous CSF
could be identified using the tentorium and collection appeared and was treated with a
the porus acusticus internus as landmarks. lumbar drainage for 6 days.
536 8 Cerebellopontine Angle Epidermoids

a b

c d

(a–d) Preoperative MRI—(a, b): T1-weighetd tumor (i–k) and from the brain stem (l). (m) Endoscopic
sequence after contrast administration. (c, d) Native examination demonstrating small remaining tumor in
T1-weighted sequence. (e) T2-weighted sequence. the IAC (arrow). (n, o) Removal of the remnant under
(f–q) Intraoperative endoscopic and microscopic microscopic control. (p, q) Complete removal of the
images. (f) Initial view of the CP angle and of the epi- epidermoid: endoscopic panoramic view at the end of
dermoid. (g) Tumor removal and visualization of the surgery. The arrow points at the IAC, free form tumor;
trigeminal nerve and upper pons (h). Dissection of arrowhead—Dorello’s canal and abducens nerve
the tumor from the multiple arteries embedded in the
Management 537

e f

g h
538 8 Cerebellopontine Angle Epidermoids

i j

k l
Management 539

m n

q
540 8 Cerebellopontine Angle Epidermoids

Case Illustration 8.4 tumor was removed via the level defined by the
CP Angle Epidermoid 4 fifth and seventh–eighth cranial nerves.
Fifty-one-year-old female with a history of Although a complete removal was presumed,
headache and facial pain on the right side the inspection with an angled endoscope (30°)
since 2 years. MR examination was performed revealed tumor remnant at the brain stem, ante-
due to exacerbation of her complaints, and a rior to the REZ of trigeminal nerve. This part
right-sided CP angle mass was visualized. was removed under microscopic control and
Neurological examination at presentation verified with the endoscope. By pointing the
revealed no abnormalities, besides the strong endoscope more anteriorly, a small tumor rem-
trigeminal pain (V1,2) on the right side. Her nant, localized just lateral to the lower basilar
MRI is presented on figures a–c. artery, was seen and removed under direct
The tumor was exposed via the RS avenue endoscopic control. The last three figures dem-
(d–q): it was found to be located medial to the onstrate the complete tumor removal and the
fifth and seventh–eighth cranial nerves. preservation of all neural and vascular struc-
Therefore, it was approached initially via the tures of the CP angle. A rare anatomical variant
upper CP angle level (between the tentorium was seen: a small vessel passed through the
and the trigeminal nerve), taking care not to split abducens nerve.
injure the petrosal vein. Later on tumor, removal The postoperative period was uneventful,
was continued via the level between the trigem- and the patient had no neurological deficits.
inal and the vestibulocochlear nerves. Gradually, The pain syndrome was initially controlled
the basilar artery could be visualized in the with medications. At 6 months follow-up, the
depth of the operative field. Then, the remaining pain had completely disappeared.
Management 541

a b

d
c

Epidermoid tumor, located in the medial CP angle on nerves. (h, i) Gradually, the basilar artery was visual-
the right side. Preoperative T1-weighted MR images: ized (arrowheads). (j) The endoscope allowed to
native (a) and after gadolinium administration (b). (c) detect a small remnant at the brain stem, which was
FLAIR sequence. Intraoperative images, illustrating hidden by the trigeminal nerve. (k) Removal of the
the consecutive steps of tumor removal. (d) Initial remnant under microscopic control. (l, m) Small piece
microscopic view of the CP angle and the epidermoid of the tumor (arrow) was seen at the clivus, close to
(arrows), medial to the trigeminal (5) and vestibuloco- the basilar artery and removed under endoscopic con-
chlear (7–8) nerves. (e, f) The tumor was approached trol (n). (o) The abducens nerve was split, and a vessel
initially via the superior CP angle level, above the was passing through it. Endoscopic (p) and micro-
trigeminal nerve, taking care not to injure the petrous scopic (q) view of the CP angle at the end of surgery
vein (asterisk). (g) Further tumor removal via the level (CP—choroid plexus). (r, s) Postoperative T1- and
between the trigeminal and the vestibulocochlear T2-weighted MRI—complete tumor removal
542 8 Cerebellopontine Angle Epidermoids

e f

g h
Management 543

i j

m n
544 8 Cerebellopontine Angle Epidermoids

o p

q r

s
References 545

References 15. Lalwani AK (1992) Meningiomas, epidermoids, and


other nonacoustic tumors of the cerebellopontine
angle. Otolaryngol Clin North Am 25(3):707–728
1. Abramson RC, Morawetz RB, Schlitt M (1989)
16. Liu P, Saida Y, Yoshioka H, Itai Y (2003) MR imaging
Multiple complications from an intracranial epider-
of epidermoids at the cerebellopontine angle. Magn
moid cyst: case report and literature review.
Reson Med Sci 2(3):109–115. doi:JST.JSTAGE/
Neurosurgery 24(4):574–578
mrms/2.109 [pii]
2. Ahmed I, Auguste KI, Vachhrajani S, Dirks PB, Drake
17. Maccarty CS, Leavens ME, Love JG, Kernohan JW
JM, Rutka JT (2009) Neurosurgical management of
(1959) Dermoid and epidermoid tumors in the central
intracranial epidermoid tumors in children. Clinical
nervous system of adults. Surg Gynecol Obstet 108(2):
article. J Neurosurg Pediatr 4(2):91–96.
191–198
doi:10.3171/2009.4.PEDS08489 [pii]
18. Mallucci CL, Ward V, Carney AS, O’Donoghue GM,
3. Altschuler EM, Jungreis CA, Sekhar LN, Jannetta PJ,
Robertson I (1999) Clinical features and outcomes in
Sheptak PE (1990) Operative treatment of intracranial
patients with non-acoustic cerebellopontine angle
epidermoid cysts and cholesterol granulomas: report of
tumours. J Neurol Neurosurg Psychiatry 66(6):768–771
21 cases. Neurosurgery 26(4):606–613; discussion 614
19. Mohanty A, Kolluri VR, Santosh V (1996) Squamous
4. Annet L, Duprez T, Grandin C, Dooms G, Collard A,
cell carcinomatous change in a posterior fossa epider-
Cosnard G (2002) Apparent diffusion coefficient
moid: case report with a review of the literature. Br J
measurements within intracranial epidermoid cysts in
Neurosurg 10(5):493–495
six patients. Neuroradiology 44(4):326–328.
20. Mohanty A, Venkatrama SK, Rao BR, Chandramouli
doi:10.1007/s00234-001-0726-2
BA, Jayakumar PN, Das BS (1997) Experience with
5. Berger MS, Wilson CB (1985) Epidermoid cysts of
cerebellopontine angle epidermoids. Neurosurgery
the posterior fossa. J Neurosurg 62(2):214–219.
40(1):24–29; discussion 29–30
doi:10.3171/jns.1985.62.2.0214
21. Murakami N, Matsushima T, Kuba H, Ikezaki K,
6. Bonneville F, Sarrazin JL, Marsot-Dupuch K,
Morioka T, Mihara F, Inamura T, Fukui M (1999)
Iffenecker C, Cordoliani YS, Doyon D, Bonneville JF
Combining steady-state constructive interference and
(2001) Unusual lesions of the cerebellopontine angle:
diffusion-weighted magnetic resonance imaging in
a segmental approach. Radiographics 21(2):419–438
the surgical treatment of epidermoid tumors.
7. Cantu RC, Wright RL (1968) Aseptic meningitic syn-
Neurosurg Rev 22(2–3):159–162
drome with cauda equina epidermoid tumor. J Pediatr
22. Nagashima C, Takahama M, Sakaguchi A (1982)
73(1):114–116
Dense cerebellopontine epidermoid cyst. Surg Neurol
8. Dechambre S, Duprez T, Lecouvet F, Raftopoulos C,
17(3):172–177
Gosnard G (1999) Diffusion-weighted MRI postop-
23. Osborn AG, Preece MT (2006) Intracranial cysts: radio-
erative assessment of an epidermoid tumour in the
logic-pathologic correlation and imaging approach.
cerebellopontine angle. Neuroradiology 41(11):
Radiology 239(3):650–664. doi:239/3/650 [pii]
829–831. doi:90410829.234 [pii]
24. Safavi-Abbasi S, Di Rocco F, Bambakidis N, Talley
9. Dee RH, Kishore PR, Young HF (1980) Radiological
MC, Gharabaghi A, Luedemann W, Samii M, Samii A
evaluation of cerebello-pontine angle epidermoid
(2008) Has management of epidermoid tumors of the
tumor. Surg Neurol 13(4):293–296
cerebellopontine angle improved? A surgical synopsis
10. deSouza CE, deSouza R, da Costa S, Sperling N,
of the past and present. Skull Base 18(2):85–98.
Yoon TH, Abdelhamid MM, Sharma RR, Goel A
doi:10.1055/s-2007-991108
(1989) Cerebellopontine angle epidermoid cysts:
25. Samii M, Tatagiba M, Piquer J, Carvalho GA (1996)
a report on 30 cases. J Neurol Neurosurg Psychiatry
Surgical treatment of epidermoid cysts of the cerebel-
52(8):986–990
lopontine angle. J Neurosurg 84(1):14–19.
11. Dutt SN, Mirza S, Chavda SV, Irving RM (2002)
doi:10.3171/jns.1996.84.1.0014
Radiologic differentiation of intracranial epidermoids
26. Schroeder HW, Oertel J, Gaab MR (2004) Endoscope-
from arachnoid cysts. Otol Neurotol 23(1):84–92
assisted microsurgical resection of epidermoid tumors
12. Goel A, Muzumdar D, Desai K (2006) Anterior tento-
of the cerebellopontine angle. J Neurosurg 101(2):
rium-based epidermoid tumours: results of radical
227–232. doi:10.3171/jns.2004.101.2.0227
surgical treatment in 96 cases. Br J Neurosurg
27. Sirin S, Gonul E, Kahraman S, Timurkaynak E (2005)
20(3):139–145. doi:XQ74738410212029 [pii]
Imaging of posterior fossa epidermoid tumors. Clin
13. Kallmes DF, Provenzale JM, Cloft HJ, McClendon RE
Neurol Neurosurg 107(6):461–467. doi:S0303-
(1997) Typical and atypical MR imaging features of
8467(04)00184-2 [pii]
intracranial epidermoid tumors. AJR Am J Roentgenol
28. Talacchi A, Sala F, Alessandrini F, Turazzi S, Bricolo
169(3):883–887
A (1998) Assessment and surgical management of
14. Kobata H, Kondo A, Iwasaki K (2002) Cerebellopontine
posterior fossa epidermoid tumors: report of 28 cases.
angle epidermoids presenting with cranial nerve
Neurosurgery 42(2):242–251; discussion 251–242
hyperactive dysfunction: pathogenesis and long-term
29. Yamakawa K, Shitara N, Genka S, Manaka S,
surgical results in 30 patients. Neurosurgery
Takakura K (1989) Clinical course and surgical
50(2):276–285; discussion 285–276
546 8 Cerebellopontine Angle Epidermoids

prognosis of 33 cases of intracranial epidermoid 31. Zamani AA (2000) Cerebellopontine angle tumors:
tumors. Neurosurgery 24(4):568–573 role of magnetic resonance imaging. Top Magn Reson
30. Yasargil MG, Abernathey CD, Sarioglu AC (1989) Imaging 11(2):98–107
Microneurosurgical treatment of intracranial dermoid
and epidermoid tumors. Neurosurgery 24(4):561–567
Nonvestibular Schwannomas
9

Schwannomas originate most frequently from the the middle fossa; type B tumors arise from the
sensory cranial nerves. In the absence of root of the trigeminal nerve and are located pre-
neurofibromatosis, the motor nerves are involved dominately in the posterior fossa; and type C
extremely rare [8, 73]. The most frequent nonves- tumors have significant components in both mid-
tibular schwannomas are those originating from dle and posterior fossae (hourglass or dumbbell
the trigeminal nerve, facial nerve, and lower cra- type). A fourth category—type D—was added
nial nerves. Nonvestibular schwannomas consti- later on (Fig. 9.1d); it includes tumors originat-
tute 2–3 % of all CP angle tumors and 8–10 % of ing more peripherally from the distal branches of
all intracranial schwannomas [26, 74]. the trigeminal nerve. They have a smaller intrac-
ranial part and a predominant extracranial exten-
sion to the orbit, infratemporal fossa, or
Trigeminal Schwannomas pterygopalatine fossa [6, 43, 50].
Type A tumors occur in 24–50 % of the
General Features patients, type B occur in 8–30 %, and type C in
20–76 % [31, 43, 65]. Type D tumors are
Trigeminal schwannomas have been described extremely rare—less than 50 cases have been
for the first time by Dixon in 1846 [3]. In 1918, reported in the literature [2, 61]. This classification
Frazier reported the first successful removal is very useful for the selection of surgical
of a primary trigeminal (gasserian) ganglion approach and for predicting the outcome.
neurinoma. Trigeminal schwannomas are the
second most frequent intracranial schwannoma,
comprising 0.8–8 % of all intracranial schwan- Neuroimaging
nomas and 0.07–0.5 % of all intracranial tumors
[9, 10, 31, 43, 50, 65, 84]. The diagnosis of trigeminal schwannomas is
These slow-growing tumors may arise from based on CT scan setting and on MRI, both native
the trigeminal nerve root, the trigeminal gan- and after gadolinium application. On CT, the
glion, or its peripheral branches. In 1955, tumor generally appears isodense or slightly
Jefferson [31] proposed a classification scheme hyperdense and enhances uniformly after admin-
for trigeminal schwannomas that—with some istration of contrast material [47, 50, 77].
modifications—is currently generally applied [3, In type A tumors, sharp-edged bone defects in
13, 23, 43]. According to tumor location, three the middle fossa floor, involving the foramen
distinct types were identified (Fig. 9.1a–c). Type ovale and the foramen spinosum, might be seen.
A tumors arise from the trigeminal ganglion In case of significant medial and anterior exten-
(gasserian ganglion) and are located mainly in sion, the lateral aspect of sella turcica, anterior

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 547


DOI 10.1007/978-3-642-35422-9_9, © Springer-Verlag Berlin Heidelberg 2013
548 9 Nonvestibular Schwannomas

a b

c d

Fig. 9.1 (a) Type A schwannomas, arising from the fossa. (c) Type C schwannomas—hourglass or dumbbell
trigeminal ganglion and are located mainly in the middle type with middle and posterior fossae parts. (d) Type D
fossa. (b) Type B schwannomas, arising from the trigeminal schwannomas, arising from the distal branches of the
nerve root and are located predominately in the posterior trigeminal nerve with a predominant extracranial extension

clinoid process, dorsum sellae, and superior hypointense on T1-weighted images and hyper-
orbital fissure may be eroded [15, 31, 52]. In type intense on T2-weighted sequences. They
B and C schwannomas, erosion of the petrous enhance homogeneously after the administra-
apex and of the inferomedial surface of the tion of gadolinium [61, 65, 86]. Cystic compo-
petrous bone might be seen. nents have been identified in up to 26 % of the
On MRI, trigeminal schwannomas appear as cases in some series [3]. Axial, sagittal,
well as circumscribed lesions, isointense or and coronal views are essential for precise
Trigeminal Schwannomas 549

evaluation of the tumor extension and for plan- of the nerve; trigeminal motor deficit (33–60 %);
ning the operative approach. facial pain (15–34 %); headache (20–44 %);
Conventional cerebral angiography currently diplopia (15–52 %); and—more rarely—hearing
is seldom required. Trigeminal schwannomas are loss, ataxia, and various cranial nerve palsies [13,
generally less vascular tumors, and embolization 23, 50, 76]. Interestingly, up to 10 % of patients
is needed in exceptional cases. Magnetic reso- may not have trigeminal dysfunction as a pre-
nance angiography demonstrates reliably the dis- senting feature [13, 43, 50].
placement of major vessels, for example, Wasting of the temporal and pterygoid mus-
displacement of the intracavernous and petrosal cles is a relatively common finding and is
segments of the internal carotid artery and stretch- observed in 24–60 % of patients. The corneal
ing of the carotid siphon in anteriorly situated reflex is depressed or absent in 36–82 % of
tumors [23, 76]. Rarely, the carotid artery may be patients. Very large tumors cause symptoms of
engulfed by the tumor. Posterior fossa (type B) increased intracranial pressure, papilledema, and/
and dumbbell-shaped (type C) tumors frequently or contralateral pyramidal signs. Cranial nerve
elevate and displace medially the posterior cere- palsies occur usually secondary to tumor com-
bral and the superior cerebellar arteries. These pression. In some series, as many as 50–52 % of
tumors can appear as encircled by the posterior the patients had oculomotor nerve palsies by the
cerebral artery in the superior aspect and by the time of diagnosis [3, 38, 43]. Dysfunction of the
superior cerebellar artery inferior [59]. The basi- abducens nerve is seen in up to 40 %; this nerve
lar artery is displaced usually in medial and pos- is the second most frequently affected cranial
terior direction. nerve after the trigeminal one.
Tumors that grow anterior may penetrate the The facial pain in patients with trigeminal
cavernous sinus wall and expand within the sinus, schwannomas can vary in intensity and type—
leading to characteristic displacement of the from dull constant to severe lancinating pain. It
carotid artery, described above [50, 65]. In rare differs from the pain in patients suffering from
cases, the tumor may penetrate the temporal dura trigeminal neuralgia, and the two conditions should
and expand intradurally. Type B and C trigeminal not be confused. These tumors very rarely cause
schwannomas extend out of the porus trigeminus typical trigeminal neuralgia. Most patients com-
or through the eroded medial petrous bone intra- plain of constant pain or of paroxysmal lancinat-
durally into the posterior fossa. ing facial pain with attacks that last longer and do
The differential diagnosis of trigeminal not always have specific triggering mechanisms.
schwannomas includes meningiomas, epider- In contrast to trigeminal neuralgia, the pain related
moids, vestibular schwannomas, chordomas, to trigeminal schwannomas does not respond well
chondrosarcomas, lymphomas, and skull-base to medications, such as carbamazepine [13, 85].
metastases. Although trigeminal schwannomas are typi-
cally benign lesions, histological malignancy has
been reported with varying frequency—from 0 to
Clinical Presentation 7.9 % [13, 17, 23, 50].

The average age at diagnosis is similar to that in


patients with other intracranial schwannomas— Management
typically the fourth to fifth decades of life.
A characteristic clinical syndrome caused by In the last decades, remarkable improvement in
trigeminal schwannomas does not exist: clinical the surgical management of trigeminal schwan-
presentation is related rather to their location and nomas has been achieved in terms of complete-
size. Most common symptoms and signs are ness of removal, avoidance of major morbidity,
trigeminal sensory deficit—paresthesia or numb- and preservation of neural functions [3, 13, 17,
ness (74–96 %), often in more than one division 23, 65, 76]. The reported rate of total or near-total
550 9 Nonvestibular Schwannomas

removal in largest series is above 70 %. Complete extradural–intradural approach, combined with


tumor removal is possible even in case of large drilling the bone margins of the superior orbital
schwannomas because they most frequently dis- fissure, lateral wall of the cavernous sinus, and
place surrounding neurovascular structures rather Meckels’ cave, are recommended for intracav-
than engulf them. ernous lesions [14, 16, 25]. Dumbbell tumors
The optimal management of patients with are operated via the petrosal, extended petrosal,
trigeminal schwannomas, according to our experi- or combined petrosal approaches [3, 13, 72].
ence, is their complete surgical removal, which The subtemporal (subtemporal–transtentorial)
leads to definitive cure of the patient. The preserva- approach with incision of the tentorium has been
tion of neurological functions, however, has always recommended for tumors extending through the
a priority. Therefore, exceptions to complete porus trigeminus into the posterior fossa, which
removal are those cases in which the tumor infiltrates remain cephalad to the seventh–eighth nerve
the neurovascular structures or a clear dissection complex [28, 76, 82]. The anterior petrosal or
plane to the brain stem is absent. Patients in bad Kawase approach, which includes resecting the
general condition, who cannot tolerate prolonged pyramidal apex, is proposed for better expos-
surgical procedures, as well as those unwilling sur- ing the tumor and the petroclinoid region down
gery, may be followed (small tumors, stable minor to the inferior petrosal sinus [33]. Some authors
neurological deficit) or treated with radiosurgery. even recommend the subtemporal extradural–
intradural approach combined with presigmoid
Surgical Treatment posterior petrosectomy or with an anterior petro-
Inadequate tumor exposure has been long regarded sectomy [4, 51, 68, 69] as the optimal approach,
as a major reason for incomplete tumor removal. which provides excellent exposure of the tumor
Various skull-base techniques and extensive with minimal brain retraction.
approaches, which were supposed to facilitate With increasing experience, however, it became
tumor removal, have been therefore introduced. obvious that the goals of surgery—complete tumor
Some early reports suggested some surgical benefit removal and preservation of neurological func-
when using a “skull-base approach.” Taha et al. tions—could be well achieved using more simple
[76] compared their experience with the conven- and safe approaches. The senior author favored the
tional approaches, such as frontotemporal combined presigmoid–subtemporal approach for
transsylvian, subtemporal–intradural, subtempo- type C tumors previously (Case Illustration 9.2).
ral–transtentorial, and suboccipital, and the In an early series, published in 1995 [65], this
skull-base approaches (frontotemporal extradural– approach was used in 42 % of the cases, while the
intradural, frontoorbitozygomatic, subtemporal retrosigmoid was used in just 8 %. Nowadays,
anterior petrosal, and presigmoid posterior petro- however, this approach is not considered suitable
sal) for the excision of trigeminal schwannomas. for trigeminal schwannomas removal because of
In a series of 15 patients published in 1995, they the high risk of approach-related morbidity. The
found that tumor exposure, ease of dissection, and retrosigmoid suprameatal approach is a safer and
radicality of removal were superior with skull-base simpler alternative, allowing sufficient exposure
approaches. Residual or recurrent tumors were of the tumor. The main restrictions to complete
found in 65 % of patients following conventional removal are the tumors’ biological characteristics
approaches compared with 10 % of patients fol- and surgeons’ experience rather than the extent of
lowing skull-base approaches. the approach or of the tumor exposure.
Trigeminal neurinomas of the middle
fossa have been operated via the frontotem- Type A Trigeminal Schwannomas
poral transsylvian, cranioorbitozygomatic, (Predominant Location in the Middle
subtemporal–intradural, zygomatic middle fossa, Cranial Fossa)
or the combined subtemporal–intradural and extra- (a) Frontotemporal (pterional) transsylvian
dural approaches [3, 20, 76]. The extradural ante- approach. This flexible approach provides
rior temporopolar approach and the subtemporal adequate exposure to most type A tumors.
Trigeminal Schwannomas 551

A low temporal craniotomy and early opening (b) Infratemporal extradural technique. Trigeminal
of the basal cisterns and of the Sylvian fissure schwannomas located within the Meckels’
to allow sufficient egress of CSF are manda- cave or in the posterior cavernous sinus can
tory to avoid excessive brain retraction. Unless be approached via the infratemporal extradural
the tumor extends into the infratemporal fossa, technique (Fig. 9.2). In case the tumor has lim-
an additional zygomatic osteotomy is not ited extension into the posterior fossa, it can be
required. removed by incising the tentorium.

a b

Fig. 9.2 Type A trigeminal schwannoma, located mainly carotid artery could be appreciated (c). The subtemporal
in the Meckels’ cave (a). The bone-window CT demon- extradural approach was chosen and performed under
strated clearly the extent of erosion of the petrous apex navigational guidance (d). The postoperative MRI (e, f)
(b). Using the BrainLAB software, the relation of the confirmed the completeness of tumor removal
tumor to the petrous and the cavernous segments of the
552 9 Nonvestibular Schwannomas

Fig. 9.2 (continued)


Trigeminal Schwannomas 553

e f

Fig. 9.2 (continued)

Type B Trigeminal Schwannomas (see chapter Retrosigmoid Suprameatal


(Predominant Posterior Fossa Location) Approach and Case Illustration 9.1).
(a) Retrosigmoid approach. The standard RS Trigeminal schwannomas are located
approach, as described in the book, is typically medial to the seventh–eighth cra-
sufficient for tumors, located within the pos- nial nerve complex. The working corridor is
terior fossa or those having minimal exten- between these nerves and the tentorium.
sion into the Meckels’ cave. Extreme care should be therefore applied
(b) Retrosigmoid suprameatal approach. The not to injure the nerves during tumor
additional removal of the suprameatal tuber- removal or when inserting/removing the
cle exposes the Meckels’ cave tumor portion instruments.
554 9 Nonvestibular Schwannomas

Case Illustration 9.1 (j, k), exposing the contents of the Meckels’
Retrosigmoid Suprameatal Approach cave (l—arrows). This allowed mobilization
Twenty-seven-year-old female patient pre- of the remaining tumor part, its dissection
sented with MR images showing a polycyclic from the surrounding structures—including
trigeminal schwannoma on the right side with the oculomotor and abducens nerves—and
a predominant CP angle tumor and smaller complete tumor removal (m–o). The trigemi-
portions in the Meckels’ cave (a–f). The nal nerve was splayed by the tumor, as seen on
examination was performed because of (m–o), but could be preserved. Only the fas-
unspecific complaints of slight vertigo and cicles from which the tumor originated had to
numbness in the left hand. Her neurological be sacrificed. Importantly, after tumor resec-
status was normal at admission. Still, because tion, all blood clots in the CP angle have to be
of the tumor large size and the severe brain thoroughly removed. Thus, a hidden bleeding
stem compression, surgery was the only man- point/vessel can be identified and coagulated;
agement option. The retrosigmoid suprameatal secondly, a potential obstruction to the CSF
approach was selected. flow is avoided. After the removal of a large
The tumor was exposed between the sev- clot through the level between the seventh and
enth and eighth cranial nerves and tentorium eighth and the lower cranial nerves, the clivus
(g), and internal decompression with CUSA and the basilar artery could be visualized
and suction was done (h). Then, the capsule (p, q).
was separated carefully from the seventh– The routinely performed CT on the day
eighth cranial nerve complex and from the after surgery (r) did not reveal any pathologi-
brain stem. This dissection was performed cal findings. In the first several days after sur-
using the 2-hand technique, as shown on (i): gery, the patient had severe nausea and
the surgeon holds the tumor with one hand and vomiting that had to be managed with antie-
exerts slight traction (in the present case with metics. Within 1 week, these complaints sub-
a microdissector), while the other hand is used sided. She had trigeminal hypesthesia, mainly
to peel the arachnoidea of the capsule. Despite in the V2 area; slight hearing loss; and HB
the very intense adhesions of the capsule to grade II facial palsy that improved during the
the brain stem, the CP angle tumor part could duration of the hospital stay. At follow-up, the
be completely removed. Then, the suprameatal patient complained of slight trigeminal hypes-
tubercle was drilled off with a diamond drill thesia and decreased but functional hearing.
Trigeminal Schwannomas 555

a b

d
c

Preoperative MR study of the patient. (a–c) T1-weighted The suprameatal tubercle (dotted line) was removed to
sequence after contrast administration. (d, e) T2-weighted gain access to the middle fossa tumor (l, arrows). (m)
sequences. (f) CISS sequence. (g–r) Intraoperative Thus, the trigeminal nerve fibers could be better mobi-
images, illustrating the steps of surgery. (g) Initial view lized and dissected from the tumor (asterisk (clivus)).
of the tumor in the CP angle (Tu tumor, T tentorium, R (n, o) Most of the fibers of the splayed trigeminal
retractor, P pyramid, 7 seventh–eighth cranial nerve nerve were not involved by the tumor. (p, q) Complete
complex). (h) Initial internal debulking with the ultra- tumor removal; preservation of all neurovascular struc-
sonic aspirator (CUSA) and the suction (S). (i) Once tures (asterisk (basilar artery)). The basilar artery could
sufficient internal decompression was achieved, the cap- be visualized only after removal of the blood coagulum
sule was dissected from the surrounding structures. (j, k) (C). (r) Postoperative control CT scan
556 9 Nonvestibular Schwannomas

e f

g h

i
j
Trigeminal Schwannomas 557

k l

m
n

o p
558 9 Nonvestibular Schwannomas

q r
Trigeminal Schwannomas 559

Type C Trigeminal Schwannoma brain stem and cranial nerves under direct
(Dumbbell Tumors) visual control. Thereafter, the petrous apex
(a) Retrosigmoid suprameatal approach. The is resected intradurally by drilling off the
approach is usually sufficient for tumors with suprameatal tubercle. The Meckels’ cave
smaller middle fossa part. can be thus opened as widely as necessary,
(b) Retrosigmoid suprameatal approach with/ according to the tumor expansion and the
without additional resection of the tento- individual anatomical characteristics. An
rium and use of angled endoscope (Figs. 9.3, important advantage of the approach is the
9.4, and 9.5). The approach allows for safe enhanced possibility to preserve the
removal of the most challenging posterior abducens nerve, which is frequently com-
fossa tumor part and its dissection form the pressed/dislocated by the tumor. In contrast

a b

c d

Fig. 9.3 Type C trigeminal schwannoma with a very large middle cranial fossa part. (a, b) T1-images after contrast
administration and (c, d) CISS images
560 9 Nonvestibular Schwannomas

a b

c d

Fig. 9.4 Removal of the tumor presented on the previous off (c). Thus, sufficient exposure of the tumor, including
figure via the retrosigmoid suprameatal endoscope- its Meckels’ cave part, was gained, and it could be deb-
assisted approach. The tumor is seen in the depth of the ulked (d, e) and removed (f). The endoscopic inspection
surgical field, hidden by the suprameatal tubercle (a). The did not reveal any tumor remnants (g, h)
dura of the tubercle was excised (b), and it was drilled
Trigeminal Schwannomas 561

ee f

g h

Fig. 9.4 (continued)

to the approaches from superior and lateral Very important advancement in the last two
(e.g., the anterior transpetrosal approach or decades was the introduction of the
the subtemporal transtentorial approach), endoscope-assisted technique. The use of angled
the nerve can be identified early at its brain endoscopes allows for reliable visualization of the
stem exit zone and can be followed up to whole tumor, including its middle fossa part, and
Dorello’s canal during tumor removal. The its removal under microscopic or endoscopic guid-
exposure of the supratentorial tumor part ance (see Case Illustration 9.3). Even tumors with
can be further enlarged by resection of the considerable extension in the middle cranial fossa
tentorium. Care should be taken not to can be thus removed (Figs. 9.3, 9.4, and 9.5). The
injure the superior cerebellar artery, which transpetrosal approaches, although used previously
is in close contact with the tumor, and of by the senior author (see Case Illustrations 9.2 and
the trochlear nerve. 9.4), are currently rarely indicated.
562 9 Nonvestibular Schwannomas

a b

c d

Fig. 9.5 Postoperative control images of the patient presented on Figs. 9.3 and 9.4. (a) CT scan and (b–d) MR
images
Trigeminal Schwannomas 563

Case Illustration 9.2 surrounding neural structures (d). The tumor


Presigmoid–Subtemporal Approach could be removed completely (e). The patient
This is a case operated by the senior author in did not have new neurological deficits and
the early 1990s. The patient had a dumbbell made an uneventful recovery. Nevertheless,
type C tumor with large tumor portions in both nowadays this tumor would be operated via
the middle and posterior cranial fossae. The the much safer retrosigmoid suprameatal
combined presigmoid–subtemporal approach approach with endoscopic assistance to ensure
(b, c) was selected that allowed for perfect the completeness of tumor removal, in partic-
exposure of the whole tumor and its ular in the area of the Meckels’ cave.

a b

(a) Preoperative MRI, demonstrating the extension of lum afforded a perfect visualization of the tumor and
the tumor. (b) Skin incision. (c) One-piece bone flap of all surrounding structures. (f) Tumor removal. (g)
was made. (d) Extent of bone removal. (e) Elevation of The tumor has been completely removed, and most of
the temporal lobe and lateral retraction of the cerebel- the trigeminal nerve fibers have been preserved
564 9 Nonvestibular Schwannomas

c d

f g
Trigeminal Schwannomas 565

Case Illustration 9.3 intense adhesions after the first surgery (c).
Retrosigmoid Suprameatal Approach with The anatomical structures could not be recog-
Endoscope Assistance nized initially, and the tumor was approached
This 36-year-old female patient presented to along the posterior pyramid with very careful
our department with a history of headache blunt and sharp dissection and frequent use of
since many years and facial pain in the area of electrical stimulation (d). Then, the cystic
V2,3. A dumbbell trigeminal schwannoma tumor contents were evacuated, internal
was diagnosed, and she had been operated in decompression was performed and the CP
another institution—the tumor part in the CP angle tumor part was removed (e–g). Using
angle was removed partially. After surgery, angled endoscope (30°), the remaining tumor
the patient had trigeminal nerve hypesthesia, portion in the Meckels’ cave could be seen (h)
vertigo, hearing loss, as well as gait instabil- but could not be safely removed. Therefore,
ity. Due to the high-dose steroid treatment, she the suprameatal tubercle (dotted line on figure
developed iatrogenic diabetes mellitus. At i) was drilled off (j, k). Using angled microin-
presentation, she had V1 and V2 anesthesia struments, it could be dissected from the sur-
and V3 hypesthesia, atrophy of the mastica- rounding structures and from the trigeminal
tory muscles, and hearing loss (20 dB PTA). nerve fascicles and removed as a large piece
The MRI (a, b) showed a type C partially cys- under pure endoscopic guidance (l–o). The
tic trigeminal schwannoma, located in the remaining smaller tumor pieces in the anterior
Meckels’ cave and in the posterior cranial part of the Meckels’ cave (p—asterisk) were
fossa. A decision was taken to approach the then removed with an endoscopic guidance (q)
tumor via the retrosigmoid suprameatal route. and complete tumor resection achieved (r).
A retrosigmoid recraniotomy was per- After surgery, the only new neurological
formed (c–r). Tumor exposure in the CP angle deficit was a transient diplopia due to a partial
proved to be extremely difficult due to the abducens nerve dysfunction.

a b

(a, b) Preoperative MRI showing a type C trigeminal See the text for explanations. (5 trigeminal nerve P
schwannoma, located in the Meckels’ cave and in the pyramid, T tentorium, R retractor, Tu tumor, MC
posterior cranial fossa. (c–r) Intraoperative images. Meckels’ cave, * small tumor remnants)
566 9 Nonvestibular Schwannomas

c d

e f

g h
Trigeminal Schwannomas 567

i j

k l

m n
568 9 Nonvestibular Schwannomas

o p

q r
Trigeminal Schwannomas 569

Type D Trigeminal Schwannomas General Operative Technique


(Predominant Infratemporal Trigeminal schwannomas are typically well encap-
Extracranial Location) sulated; have a smooth, lobulated appearance; and
These schwannomas usually originate from the are typically yellowish. Although relatively avas-
mandibular or maxillary divisions of the trigem- cular schwannomas with soft consistency predom-
inal nerve and extend into the infratemporal inate, some tumors are firm and/or well
fossa through the foramen ovale or foramen vascularized. Once the tumor is exposed, its
rotundum. We favor the extradural subtempo- removal follows the standard scheme: initial intra-
ral–infratemporal approach with additional capsular debulking and then dissection of the cap-
removal of the zygomatic arch (Fig. 9.6) or the sule from adjacent neural structures in the
frontotemporal intradural approach with endo- arachnoid plane. Most schwannomas have a good
scopic assistance (Fig. 9.7). In case the tumor arachnoid plane and can be dissected even from
grows intradurally and compresses the tempo- the healthy fascicles of the trigeminal nerve. In
ral lobe, an additional temporal or frontotempo- contrast to neurofibromas, which cause fusiform
ral craniotomy is necessary. On Figs. 9.8 and enlargement of the whole nerve, schwannomas do
9.9, a rare trigeminal schwannoma, arising from not involve all fascicles of the nerve and tend to
the intraorbital fascicles of the nerve, is grow eccentrically. Hence, only the fascicles from
presented. which the tumor originates should be sacrificed
avoiding severe trigeminal deficits—anesthesia,
masticatory problems, or disabling keratitis.

a b

c d

Fig. 9.6 Trigeminal schwannoma, extending into fossa schwannoma. The tumor was exposed and removed
infratemporalis (surgery in the early 1990s). (a, b) via the transzygomatic subtemporal extradural approach
CT and T2-weighted MR images of a type D trigeminal (c, d). 5 trigeminal nerve; arrowheads tumor
570 9 Nonvestibular Schwannomas

a b

c d

Fig. 9.7 Type D trigeminal schwannoma with extracra- window CT, demonstrating the enlarged oval foramen.
nial extension into the infratemporal fossa via the enlarged The tumor could be removed completely without causing
oval foramen. (a–c) T1-weighted sequences MR images additional neurological deficits via the frontotemporal
following contrast administration. (d, e) T2-weighted intradural approach
sequence. (f, g) CISS sequence. (h) Preoperative bone-
Trigeminal Schwannomas 571

e f

g h

Fig. 9.7 (continued)


572 9 Nonvestibular Schwannomas

a b

Fig. 9.8 Trigeminal schwannoma type D, arising from tion of the tumor lateral and inferior the optic nerve and
the intraorbital fascicles of the nerve (the patient was bulb. The steps of its removal are presented in the next
operated in the 1980s). (a, b) CT demonstrating the loca- figure

a b

Fig. 9.9 Trigeminal schwannoma type D (continuation initially, the tumor is debulked, and only afterward, the
of the previous figure). (a) Preoperative photo of the capsule is dissected and removed. Image (d) shows the
patient. (b) Skin incision, marked with arrowheads and debulked tumor (arrows—tumor capsule). (e, f) Complete
exposure of the tumor (Tu) (c). Of note, identical tech- tumor removal (R—retractor)
nique is applied, regardless of the tumor origin/location:
Trigeminal Schwannomas 573

c d

e f

Fig. 9.9 (continued)


574 9 Nonvestibular Schwannomas

Case Illustration 9.4 was highly pigmented and originated from the
Melanotic Trigeminal Schwannoma fascicles of the nerve within the Meckels’
The CT and MRI images (a, b) of this patient cave. It could be removed completely with
show a type C trigeminal schwannoma with preservation of the adjacent cranial nerves,
characteristics that do not differ from those of including most fascicles of the trigeminal
classical tumors. The patient was operated via nerve (d). Histopathological diagnosis was
the presigmoid subtemporal approach. On WHO grade I melanotic schwannoma. The
figure (c), a microscopic photograph of the follow-up of the patient is more than 10 years,
exposed tumor, medial to the main trunk of and up to now, no recurrences have been
the trigeminal nerve, is presented. The tumor detected.

a b

Preoperative CT and MRI, demonstrating the tumor and its extension (a, b). Intraoperative microscopic view of
the tumor (c). (d) Complete tumor removal
Trigeminal Schwannomas 575

c d
576 9 Nonvestibular Schwannomas

Tumors involving the cavernous sinus can be example, diplopia, facial pain, hypacusis, or
dissected from its wall and removed safely using ataxia. In respect to trigeminal nerve sensory
the current operative techniques [9, 17, 84]. The function, a transient postoperative deterioration
experience of the senior author has shown, how- may be observed that tends to improve in approx-
ever, that very tinny intracavernous residual parts imately half of the cases.
might be indiscernible and could lead to tumor Surgery-related cranial nerve morbidity—
recurrence. As reported previously in all eight most frequently of the ocular motor, cochlear
patients with tumor invading the cavernous sinus, nerves, and facial nerves—is usually caused by
complete tumor removal was achieved [65]. At operative manipulations and/or tumor dissection.
follow-up MRI, however, in two cases, tumor It is related mostly to the operative approach and
recurrence was detected. The risk of recurrence is to the surgeons’ experience.
highest in trigeminal schwannomas located in the
cavernous sinus, followed by those located in Radiotherapy
Meckels’ cave and the porus trigeminus, and is Stereotactic radiosurgery has been promoted as
least in posterior fossa tumors [76]. an alternative primary or adjuvant management
It has been pointed out that in case of incom- option that leads to a high rate of tumor growth
plete resection, the recurrence rate is higher in control. It is reported to be an effective treatment
trigeminal schwannomas than in vestibular modality for small- and medium-size trigeminal
schwannomas [22]. Still, the natural history of schwannomas [29, 60]. In one large prospective
tumor remnants is not known: some authors study, presented by Pan et al. [60], good response
found a high incidence of symptomatic tumor to the treatment—tumor shrinkage more than
growth, while other report on long periods of one-third in its greater diameter—was noted in
remission [50, 63]. 88.5 % of tumors smaller than 30 mm. The review
Pigmented or melanotic trigeminal schwanno- of the relevant literature, however, shows that the
mas are exceedingly rare (see Case Illustration rate of tumor progression, despite the radiosur-
9.4). Melanotic schwannomas are very rare gery, is >9 %. The probability for improvement
peripheral nerve tumors with a predilection for of existing neurological symptoms is less in case
spinal nerve involvement. Trigeminal nerve ori- of radiosurgery when compared to surgery.
gin of the tumor has been described in a few Furthermore, the rate of cranial nerve morbidity
cases. Characteristic is their heavy pigmentation, is higher when radiosurgery is applied.
as evident in the presented case, which is due to Kano et al. [32] evaluated 33 consecutive
deposit of melanin pigment in the cytoplasm. patients with trigeminal schwannomas treated via
Melanotic schwannomas might be a component gamma knife surgery and found that at an average
of the autosomal dominant Carney syndrome that of 6 years, the rate of progression-free survival at
includes spotted mucocutaneous pigmentation, 1, 5, and 10 years after SRS was 97.0, 82.0, and
myxomas (cardial, cutaneous, and mammary), 82.0 %, respectively. Three patients (9.1 %) had
and endocrine hyperactivity [11]. A prevalence symptomatic disease progression. Phi et al. [62]
of growth hormone producing pituitary ade- achieved tumor growth control in 95 % of their
nomas, causing acromegaly, has been described patients. Twenty-seven percent experienced new
in such patients. Importantly, a higher rate of or worsening cranial neuropathies, and in 46 %,
malignant features has been reported in melan- tumor expansion after radiosurgery was observed.
otic schwannomas. The authors conclude that cranial neuropathies
are bothersome complications of radiosurgery.
Outcome Worsening of the disease was observed in 12 % of
Neurological symptoms, caused by trigeminal the patients treated by Sheehan et al. [70].
schwannomas, are related generally to their com- Due to these considerations, we regard complete
pression rather than infiltration. Surgery, there- tumor resection with preservation of brain and cra-
fore, may lead to relieve of these symptoms, for nial nerve function the ideal treatment. Radiosurgery
Oculomotor Nerve Schwannoma 577

may be an alternative in case of small tumors in Trochlear Schwannomas


elderly, somatically unstable patients.
King JS published the first case of surgical treat-
ment of trigeminal schwannoma in 1976 [35].
Oculomotor Nerve Schwannoma Less than 30 cases of pathologically verified tro-
chlear nerve schwannomas have been reported up
In 1927, Kovacs described an isolated oculomotor to now in the literature [21, 36, 57]. They origi-
nerve schwannoma observed at autopsy [37]. nate usually from the intradural cisternal part of
Since then, 35 cases have been reported in the lit- the nerve and more rarely from its extradural cav-
erature [27, 78]. Schwannomas can arise from any ernous part and are subdivided into cisternal, cav-
segment of the oculomotor nerve. Celli et al. [12] ernous, and cisternocavernous groups [18]. The
categorized them accordingly into cisternal, cav- tumor expands in the ambient and interpeduncu-
ernous, and cisternocavernous types. These tumors lar cisterns or grows toward the CP angle.
are located most frequently within the interpedun- Anterior extension into the parasellar space has
cular cistern and cavernous sinus. Large tumors been also observed [81].
may have parasellar and suprasellar extension. Patients with trochlear schwannomas usually
Cisternal tumors expand in the prepontine cistern present with trochlear nerve palsy, trigeminal
and may compress the brain stem [44, 66]. nerve signs/symptoms, headache, long-tract
Patients with oculomotor schwannomas have signs, ataxia, or symptoms from other cranial
almost always a certain degree of diplopia due to nerves [1, 18, 21, 35]. Interestingly, only 50 %
oculomotor palsy. Other symptoms are related to of the patients have trochlear nerve palsy at pre-
compression of neighboring cranial nerves or—in sentation, which can be explained by the devel-
case of large lesions—to intracranial hyperten- opment of compensatory mechanisms for the
sion [56, 78]. lost function during the slow tumor growth
The still-limited experience with surgery of [18, 49]. The nerve of origin is determined reli-
these tumors has shown that cisternal lesions can ably only intraoperatively, but the possibility of
be removed while preserving the anatomical con- trochlear schwannomas should be at least
tinuity of the nerve with good chances for func- considered.
tional recovery. Complete resection of the Trochlear schwannomas can be accessed via
cavernous schwannomas, however, might be different approaches, according to their exten-
impossible without nerve sacrifice. Therefore, sion, clinical symptoms, and surgeons’ prefer-
the management plan should be discussed ence [21]. (See Case Illustration 9.5). The
together with the patient, considering his neuro- trochlear nerve palsy persists after surgery in
logical and general condition, as well as tumor more than 40 % after surgery, but a gradual reso-
size, location, and growth tendency. lution/compensation usually occurs.
578 9 Nonvestibular Schwannomas

Case Illustration 9.5 bradycardia. Therefore, the cyst was drained


Trochlear Nerve Schwannoma initially, which lead to relaxation of the cere-
This 52-year-old female patient presented bellum (b). The solid tumor portion with the
with trigeminal hypesthesia, double vision typical appearance of schwannoma was
due to trochlear nerve palsy, facial nerve palsy located in the ambient, crural, and upper CP
(House–Brackmann grade III), decreased cisterns. The tumor originated from the cister-
hearing on the left side (40 dB at 1.5 Hz), and nal part of the trochlear nerve just before its
gait instability for 10 months. She had long- entrance in the free edge of the tentorium.
tract signs and a grade 4/5 central right-sided Initial debulking using an ultrasound aspirator
hemiparesis. The patient underwent an open and platelet knife was performed, and—fol-
biopsy at another institution with the presum- lowing relieve of the tension—the capsule was
able diagnosis of pilocytic astrocytoma. The dissected in the arachnoid plane from the sur-
MRI showed a lesion with a cystic and com- rounding structures. The trochlear nerve was
ponents (a). The 2.5-cm large solid tumor part completely destroyed by the tumor, and its
was isointense on T1-weighted images and distal segment could not be identified. Total
hypointense on T2-weighted images with tumor removal was achieved, and the histo-
intense contrast enhancement and was well logical examination revealed a WHO grade I
circumscribed. The cystic part was located schwannoma.
dorsally. After surgery, the patient did not have new
The tumor was approached via the RS ave- neurological deficits and had an uneventful
nue. Intense adhesions following the previous recovery (c). At follow-up, the trigeminal
surgery were encountered that precluded nerve dysfunction, the facial nerve palsy,
opening of the cerebellomedullary cistern. ataxia, and long-tract signs recovered com-
Furthermore, the attempt to retract the cere- pletely. However, the right-sided trochlear
bellar hemisphere led to significant palsy and the hypacusis persisted.

(a) Preoperative imaging. (b) Intraoperative view of the tumor (T tentorium; C cerebellum; P pyramid).
(c) Postoperative follow-up MRI (From Gerganov et al. [21]. Copyright © 2007, with permission from Elsevier)
Trochlear Schwannomas 579

c
580 9 Nonvestibular Schwannomas

Abducens Nerve Schwannoma functional disturbance of the abducens nerve was


seen, which has been explained by the develop-
Since the first description of an abducens nerve ment of compensatory mechanisms.
schwannoma by Bing-huan in 1981, 18 such The neuroradiological features of these
cases have been presented in the literature. tumors are similar to those of other schwanno-
According to the classification of Tung et al. [79], mas. Therefore, preoperative differentiation
based on the tumor origin, the abducens schwan- between oculomotor, trochlear, trigeminal, and
nomas are two types: arising within the cavern- abducens nerve schwannomas is generally not
ous sinus or arising along the course of the nerve possible. A variety of operative approaches have
in the prepontine area. Most tumors belong to the been applied. In some of the reported cases, the
first group and involve the cavernous sinus. tumor could be removed from the nerve fasci-
Abducens schwannomas, in contrast to all other cles of origin, preserving the main trunk. Such
cranial nerve schwannomas, originate away from favorable outcome could be explained by the
the glial–Schwann sheath junction [80]. Patients presence of anatomical variations, such as split-
present typically with double vision due to ting of the nerve or the existence of two separate
abducens nerve palsy. In case of large tumors, nerve trunks [30, 55]. In 3 reported cases, the
symptoms from compression of adjacent neural abducens nerve could be preserved, and at fol-
structures and/or intracranial hypertension low-up, complete recovery of its function was
develop [58]. In some rare cases [54], no obvious achieved [53, 54, 80].
Abducens Nerve Schwannoma 581

Case Illustration 9.6 compress the lower cranial nerves inferolater-


Abducens Nerve Schwannoma ally and the seventh–eighth cranial nerve com-
This 42-year-old man presented with a history plex laterally (d, e). It was debulked, and
of headache and episodes of dizziness and despite the strong adhesion of its capsule to
vertigo since several months. His neurological the abducens nerve, it could be completely
examination revealed only a slight left-sided removed, preserving the anatomical integrity
hypacusis. The MRI (a–c) showed a solid of the nerve (f, g). The only neurological
lesion with intense contrast enhancement that deficit that appeared after surgery was a par-
caused significant compression of the brain tial abducens nerve paresis on the left side. At
stem at the level of the lower pons and upper 1-year follow-up, however, the function of the
medulla oblongata. nerve recovered completely.
The patient was operated via the retrosig-
moid approach. The tumor was found to

a b

(a–c) Preoperative MRI study, demonstrating the con- (IX–XI) and (e) complete tumor removal. All anatomi-
trast-enhancing tumor in the prepontine space. (d, e) cal structures have been preserved. (f, g) Postoperative
Intraoperative images. (d) Initial exposure of the tumor MRI, proving the completeness of tumor removal
and its relations to the facial and vestibulocochlear (Reprinted from Nakamura et al. [54]. Copyright ©
nerve complex (VII–VIII) and the lower cranial nerves 2002, with permission, Elsevier)
582 9 Nonvestibular Schwannomas

d e

f g
Facial Schwannomas 583

Facial Schwannomas ganglion), and meatal segment lesions. A more


practically oriented classification is that of Sarma
In 1930, Schmidt described for the first time a et al. [66], which is based on their surgical ana-
schwannoma originating from the facial nerve tomic features: (1) cerebellopontine angle, (2)
[24]. Facial schwannomas are slow-growing geniculate, and (3) tympanomastoidal lesions
tumors that may originate from any part of the (Figs. 9.10, 9.11, and 9.12). Frequently, however,
nerve—from the CP angle to its extracranial the tumor involves more than one segment of the
peripheral portion (see figures and the case nerve. Facial nerve schwannomas account for
illustrations)—but have a predilection for the less than 2 % of all schwannomas [75]. Twenty
geniculate segment [41, 75, 83]. Lipkin et al. [45] percent of them are located within the CP angle
subdivided facial schwannomas into tympanic, and comprise approximately 3 % of all CP angle
vertical, labyrinthine (including the geniculate tumors [34].

a b

Fig. 9.10 Facial schwannoma, located predominantly in the middle cranial fossa with a smaller component in the IAC,
operated by the senior author in the early 1990s
584 9 Nonvestibular Schwannomas

a b

Fig. 9.11 Facial schwannoma in the mastoid segment of tumor-involved segment of the facial nerve and its com-
the nerve. (a) CT scan demonstrating the smooth bone plete removal. (g, h) Sural nerve graft was adjusted at the
widening (arrow). (b, c) MRI images demonstrating the healthy proximal and distal edges of the nerve (g, arrows)
contrast-enhancing lesion in the area (arrow). (d) The and fixed with fibrin glue (h, asterisks). (i, j) Photos of the
approach was performed under neuronavigational guid- patient 1 year later
ance. (e, f) Intraoperative images demonstrating the
Facial Schwannomas 585

e f

g h

Fig. 9.11 (continued)


586 9 Nonvestibular Schwannomas

i j

Fig. 9.11 (continued)


Facial Schwannomas 587

a b

c d

Fig. 9.12 Facial schwannoma arising in the area of the the CT images. The tumor was removed completely via
ganglion geniculi and extending to the middle cranial the middle fossa approach. (a, b) Bone-window CT scans.
fossa and into the IAC. Note the destruction of the middle (c, b) T1-weighted contrast-enhanced MR images. (e, h)
cranial fossa floor and the communication to the IAC on T2-weighted images
588 9 Nonvestibular Schwannomas

e f

g h

Fig. 9.12 (continued)


Facial Schwannomas 589

Case Illustration 9.7 that it could be a case of facial nerve schwan-


Facial Schwannoma in the IAC noma was discussed with the patient before
This 56-year-old male patient presented with surgery. The tumor was approached via the
a history of hearing loss on the right side and retrosigmoid route (b–o). It originated from
vertigo. His neurological examination the CP angle segment of the facial nerve. In
revealed slight HB grade 2 facial nerve palsy. sake of radicality, the nerve was transected in
He had functional hearing with a decrease of its healthy parts. Nerve reconstruction was
25 dB in the main speech area. MRI showed a then performed with interpositional sural
contrast-enhancing lesion in the right IAC nerve graft. The vestibular and the cochlear
(a), which was slightly dilated. Presentation nerves were preserved, and after surgery,
with facial nerve palsy is rather untypical for hearing level was unchanged. The postopera-
VS, especially of this size. The possibility tive period was uneventful.

Facial schwannoma located in the right IAC. (Aa, Ab) The origin of the tumor from the facial nerve (7) is
T1-weighted contrast-enhanced MRI of the tumor, well seen on image. (h, i) Complete tumor removal
appearing as a vestibular schwannoma. No radiologi- with preservation of the vestibular and the cochlear
cal clues on its eventual pathological type. (b) Initial nerves. The distal facial nerve stump is seen in the lat-
exposure of the CP angle (arrowhead (seventh–eighth eral part of the IAC. (j) In order to identify a healthy
nerves); r (retractor); arrows (incision of the dura proximal stump, the facial nerve exit zone at the brain
around the posterior lip of the IAC)). (c) The dura stem had to be dissected (LCN—lower cranial nerves).
around the posterior lip has been stripped off and The intact vestibulocochlear nerve complex was used
incised. (d) The IAC has been widely opened. The ves- as a guide. (k) Proximal facial nerve stump. (l) Sural
tibular nerves (8) and the tumor (tu) are seen. A small nerve graft of appropriate length has been approxi-
part of the dura, covering the medial part of IAC, has mated between the proximal and distal facial stumps
not been still incised (*). Its excision revealed the (small arrows) and fixed with fibrin glue (m, n). (o)
whole intracanalicular and part of the CP angle tumor The IAC is filled with small fat pieces, sealed with
part (e). (f–h) Dissection of the tumor from the ves- fibrin glue. During this procedure, great care had to be
tibular and cochlear nerves and its gradual removal. exerted not to dislocate the graft
590 9 Nonvestibular Schwannomas

b c

d e

Tu Tu

f g

Tu

8
Facial Schwannomas 591

h i

j k

LCN

l
592 9 Nonvestibular Schwannomas

m n

o
Facial Schwannomas 593

The preoperative neuroradiological work-up is be tumor location (Figs. 9.10, 9.11, and 9.12). An
similar to that for other CP angle tumors: bone- example for this is the widening of the fallopian
window thin-slice CT, native and contrast- canal seen on bone-window CT.
enhanced MRI, as well as flexion/extension X-ray The clinical presentation of patients with such
of the spine (semi-sitting position!). Several radio- tumors is variable and is related to tumor location
logical patterns of facial schwannomas have been and size. Patients with facial schwannomas, in
reported, but none of them are pathognomonic contrast to those with VSs, typically have some
(Fig. 9.13). The only radiological clue to differen- degree of facial palsy at presentation. Therefore, in
tiate a facial from a vestibular schwannoma may every patient with a CP angle tumor and facial

a b

c d

Fig. 9.13 Patient with a facial schwannoma on the left T1-weighted sequences after gadolinium application. (e)
side, located in the CP angle and IAC. The findings are not T2-weighted sequence. (f, g) T2-CISS sequences. At sur-
specific, and a differential diagnosis to vestibular schwan- gery, the tumor turned out to originate from the facial nerve.
noma, based on imaging, cannot be made. (a) Bone-window It was removed completely, and the nerve was reconstructed
CT scan; note the widened IAC and the high location of the with an interposition graft. The postoperative CT—the day
jugular bulb. (b) T1-weighted native MRI. (c, d) after surgery—is shown on (h) and (i)
594 9 Nonvestibular Schwannomas

e f

g h

Fig. 9.13 (continued)

palsy, an origin of the schwannoma from the facial facial nerve sacrifice and reconstruction with a
nerve should be suspected, especially if the palsy sural nerve graft should be considered. Normal
is progressive in nature. The patient should be facial function, however, does not exclude a diag-
counseled accordingly, and the option of eventual nosis of facial schwannoma—27–54 % of the
Facial Schwannomas 595

patients do not have facial palsy at presentation pronounced, some surgeons recommend conser-
[64, 67]. Hearing loss, vertigo, or tinnitus are the vative surgery with the hope of preserving the
other common initial symptoms [24, 40]. Rarely remaining facial function as long as possible. The
patients present with hemifacial spasm [66]. The proposed surgical options are tumor stripping with
interval between onset of symptoms and diagnosis at least partial anatomical preservation of the facial
is usually long. Sherman et al. [71] pointed that the nerve or—if this is impossible—only decompres-
reason for delayed diagnosis might be initial con- sion/tumor debulking [5]. Other surgeons recom-
fusion with Bell’s palsy. These authors recom- mend immediate surgery and tumor removal after
mend MRI examination in all patients with the diagnosis because of the increased risk to hear-
progressive weakness of the facial nerve, as well ing loss related to further tumor growth [66].
as in patients with suspected Bell’s palsy in case of Our opinion is that the best treatment of facial
progressive worsening of the facial function or in nerve schwannomas is complete tumor removal
case no improvement after 6 months is observed. with resection of the involved facial nerve seg-
In case of pronounced facial palsy (HB grade ment and restoration of the facial function (see
III or worse) or enlarging tumors, tumor removal Figs. 9.14 and 9.15 and Case Illustration 9.7).
with nerve reconstruction is the only reasonable Earlier reconstruction of the facial nerve might
option [48]. The issue if and when to operate a lead to better recovery (see Case Illustration 9.8).
patient with small tumor and normal facial nerve Preservation of hearing is mandatory. In some
function or with mild symptoms is discussed con- patients without facial nerve deficits before sur-
troversially in the literature. Some authors recom- gery, it may be possible to resect only the
mend an initial wait-and-see strategy until the loss involved portion of the nerve and to preserve
of facial function develops [19, 46]. The rationale some of the fibers. Thereby, the facial nerve
for this strategy is the fact that these tumors may function can be preserved. In case a facial palsy
grow quite slowly. If the facial palsy is less exists preoperatively, however, more extensive

a b

Fig. 9.14 Giant facial nerve schwannoma in a 53-year- petrous bone and its massive destruction. The patient has
old female patient, presenting with facial hypesthesia, been operated via the RS approach, and the tumor could
anacusis, HB grade II facial nerve weakness, swallowing be removed completely; the origin of the schwannoma
difficulty, severe gait ataxia, and headache. Preoperative was the facial nerve, which had to be therefore sacrificed
images (a–f): note the extension of the tumor in the and then reconstructed
596 9 Nonvestibular Schwannomas

c d

e f

Fig. 9.14 (continued)

involvement of the nerve fibers is likely. Radical Rare finding is the more diffuse infiltration of
surgery in such case necessitates nerve transec- the nerve, usually appearing as pearl-like clusters
tion and reconstruction. The healthy proximal along its course. In such case, healthy nerve
and distal segments of the nerve have to be stumps do not exist, and primary nerve recon-
exposed, even by performing partial mastoidec- struction is impossible. In these patients, decom-
tomy. Tensionless end-to-end anastomosis usu- pressive surgery to retain residual function is an
ally requires the use of a nerve graft from the option. Once the functional loss becomes more
sural nerve (see chapter on Facial Nerve severe, complete tumor removal and hypoglos-
Reconstruction). sal–facial anastomosis are performed.
Facial Schwannomas 597

a b

c d

Fig. 9.15 Postoperative images of the patient from tracheostoma and PEG were placed. Within 3 weeks, she
Fig. 9.14. (a) CT scan 1 day after surgery. (b) CT scan 4 could resume oral feeding and at discharge could walk
days later. (c, d) MRI 4 months after surgery. Immediately without help. Four months later, a hypoglossal–facial
after tumor removal, she had worsening of swallowing; a nerve anastomosis was performed
598 9 Nonvestibular Schwannomas

Case Illustration 9.8 continued with internal debulking of the CP


Giant Facial Nerve Schwannoma angle tumor part. The tumor had a hard consis-
Nine-year-old girl complaining of progres- tency, and only in the depth, close to the brain
sively deteriorating hearing since 2 years. stem, was somewhat softer. The tumor capsule
Neurological examination revealed anacusis was gradually dissected from the compressed
and gaze nystagmus on the right side and severe lower cranial nerves, and the facial nerve was
gait ataxia. The initial CT and MR findings visualized at the brain stem. It could not be,
are presented on (a–f): giant homogeneously however, recognized anatomically in its more
enhancing CP angle tumor with massive lateral segment: it practically disappeared in
destruction of the petrous bone and extension the tumor. Electrical stimulation of the whole
in an anterosuperior direction up to the dura of medial tumor part provoked vivid facial EMG
the middle fossa, in anteromedial direction to response, although nerve fibers could not be
the petrous segment of the carotid artery, and identified, which confirmed the diagnosis of
caudally to the jugular foramen and significant facial nerve schwannoma. Taking into account
brain stem compression but no hydrocephalus. the patients’ age, it was decided to proceed
No clinical or radiological signs of NF-2 have with tumor removal and to sacrifice the nerve.
been detected, and the presumptive diagnosis The tumor was removed completely; then
was VS, despite the very young patients’ age. healthy medial and lateral stumps of the facial
Surgical removal of the tumor was planned via nerve were prepared, and a 2-cm long sural
the RS approach in the semi-sitting position. nerve graft was used to bridge them.
Transesophageal echocardiography revealed a The postoperative period was uneventful,
hemodynamically insignificant opened fora- and the patient was discharged 13 days after
men ovale, which did not hinder the planned surgery. Her facial nerve showed significant
surgery. recovery at the follow-up examination 12
The tumor was exposed via the typical RS months later (t–v).
approach (g–q). The petrous tumor part was Summary: The clinical presentation of this
covered by a very thin layer of bone and dura. young patient and the neuroradiological exam-
They were removed widely exposing this inations did not give any clues to the patho-
tumor portion. Caudally, the tumor capsule logical tumor type. Complete tumor removal,
was attached to the jugular bulb and had to be which was the goal of surgery, could be
dissected from it. It was debulked, and a small achieved only if the facial nerve was sacrificed.
segment of the facial nerve could be identified In young patients, the degree of functional
in the fundus area of the IAC. Medially, it dis- facial recovery after direct end-to-end nerve
appeared in the tumor mass. Therefore, we reconstruction in the CP angle is very high.
Facial Schwannomas 599

a b

c d

(a–f) Preoperative images, demonstrating a giant initially. (i) Internal decompression of the CP angle
intensely enhancing tumor in the right CP angle with tumor portion followed. (j) The tumor has been
large intrapetrous part. The extensive destruction of removed completely and the brain stem—decom-
the petrous bone is well visible on the bone-window pressed. (k, l) The facial nerve was infiltrated by the
high-resolution CT scan (a, b). (c–e) T1-weighted MR tumor. The proximal stump is seen with some attached
sequence after contrast administration and (f) CISS tumor (arrows) on the images; preparation of the prox-
sequence. (g–q) Intraoperative images, illustrating the imal (m) and of the distal (n) nerve stumps (PS proxi-
consecutive steps of tumor removal. (g) RS approach mal stump, DS distal stump); a graft of appropriate
with exposure of the edges of the sigmoid (SS) and length is prepared (o) and fixed to the stumps with
transverse sinuses (TS). (h) The tumor portion in the fibrin glue (p). (q) Fat is used to prevent CSF leak.
pyramid was covered by a very thin bone lamina, Photo of the patient immediately after surgery (r, s)
which was drilled off. The dotted line shows the and 12 months later (t–v)
amount of bone removal. This part was removed
600 9 Nonvestibular Schwannomas

e f

g h
TS P

SS

i j
Facial Schwannomas 601

k l

m n
PS

DS
602 9 Nonvestibular Schwannomas

o p

q
Facial Schwannomas 603

r s

t u v
604 9 Nonvestibular Schwannomas

Cochlear Schwannomas 5. Angeli SI, Brackmann DE (1997) Is surgical excision


of facial nerve schwannomas always indicated?
Otolaryngol Head Neck Surg 117(6):S144–S147.
Schwannomas arising from the cochlear nerve doi:S0194-5998(97)70084-6 [pii]
are extremely rare. They are discovered usually 6. Arena S, Hilal EY (1976) Neurilemomas of the
at surgery, but high-resolution MRI, in particular infratemporal space: report of a case and review of the
the 3D-CISS mode, showing enhancement of the literature. Arch Otolaryngol 102(3):180–184
7. Barbieri M, Bruzzo M, Mora R, Meller R, Chays A,
cochlear division of the eighth cranial nerve or Magnan J (2001) Cochlear schwannomas. Skull Base
cochlear tumors in the inferior compartment of 11(4):241–244
the anterior wall of the IAC with or without 8. Berger MS, Edwards MS, Bingham WG (1982)
extension into the cochlea, may suggest such ori- Hypoglossal neurilemmoma: case report and review
of the literature. Neurosurgery 10(5):617–620
gin [39, 42]. 9. Bordi L, Compton J, Symon L (1989) Trigeminal neu-
Patients with cochlear schwannomas usually roma. A report of eleven cases. Surg Neurol
present with profound hearing loss and tinnitus. 31(4):272–276
Complaints of vertigo are less typical. Some 10. Cantini R, Giorgetti W, Valleriani AM, Burchianti M
(1987) Trigeminal schwannomas in adolescence.
authors even point that in case of rapidly progres- Pediatr Neurosci 13(4):198–201
sive hearing loss and absence of vertigo, the diag- 11. Carrasco CA, Rojas-Salazar D, Chiorino R, Venega
nosis cochlear nerve tumor has to be considered JC, Wohllk N (2006) Melanotic nonpsammomatous
[7]. Factors guiding the management of such trigeminal schwannoma as the first manifestation of
Carney complex: case report. Neurosurgery
patients are their general condition, hearing level 59(6):E1334–E1335. doi:10.1227/01.NEU.0000
on the side of the tumor, and on the healthy side, 245608.07570.D2; discussion E1335
as well as tumor size. While in case of profound 12. Celli P, Ferrante L, Acqui M, Mastronardi L,
hearing loss surgery is the best option, in case of Fortuna A, Palma L (1992) Neurinoma of the third,
fourth, and sixth cranial nerves: a survey and report
less severe loss, especially if the hearing on the of a new fourth nerve case. Surg Neurol 38(3):
other side is also compromised, a period of obser- 216–224
vation might be warranted. 13. Day JD, Fukushima T (1998) The surgical manage-
ment of trigeminal neuromas. Neurosurgery 42(2):
233–240; discussion 240–231
14. Day JD, Fukushima T, Giannotta SL (1994)
Lower Cranial Nerve Schwannomas Microanatomical study of the extradural middle
fossa approach to the petroclival and posterior cavern-
This topic is discussed in the chapter tumors of ous sinus region: description of the rhomboid con-
struct. Neurosurgery 34(6):1009–1016; discussion
the jugular foramen. 1016
15. de Benedittis G, Bernasconi V, Ettorre G (1977)
Tumours of the fifth cranial nerve. Acta Neurochir
(Wien) 38(1–2):37–64
16. Dolenc VV (1985) A combined epi- and subdural
References direct approach to carotid-ophthalmic artery aneu-
rysms. J Neurosurg 62(5):667–672. doi:10.3171/
1. Abe T, Iwata T, Shimazu M, Matsumoto K (1994) jns.1985.62.5.0667
Trochlear nerve neurinoma associated with a giant 17. Dolenc VV (1994) Frontotemporal epidural approach
thrombosed dissecting aneurysm of the contralateral to trigeminal neurinomas. Acta Neurochir (Wien)
vertebral artery. Surg Neurol 42(5):438–441 130(1–4):55–65
2. Akhaddar A, El-Mostarchid B, Zrara I, Boucetta M 18. Du R, Dhoot J, McDermott MW, Gupta N (2003)
(2002) Intracranial trigeminal neuroma involving the Cystic schwannoma of the anterior tentorial hiatus.
infratemporal fossa: case report and review of the litera- Case report and review of the literature. Pediatr
ture. Neurosurgery 50(3):633–637; discussion 637–638 Neurosurg 38(4):167–173. doi:10.1159/000069094
3. Al-Mefty O, Ayoubi S, Gaber E (2002) Trigeminal 19. Fenton JE, Chin RY, Fagan PA, Sterkers O, Sterkers
schwannomas: removal of dumbbell-shaped tumors JM (2004) Facial nerve outcome in non-vestibular
through the expanded Meckel cave and outcomes of schwannoma tumour surgery. Acta Otorhinolaryngol
cranial nerve function. J Neurosurg 96(3):453–463. Belg 58(2):103–107
doi:10.3171/jns.2002.96.3.0453 20. Fukaya R, Yoshida K, Ohira T, Kawase T (2010)
4. Al-Mefty O, Fox JL, Smith RR (1988) Petrosal Trigeminal schwannomas: experience with 57 cases
approach for petroclival meningiomas. Neurosurgery and a review of the literature. Neurosurg Rev
22(3):510–517 34(2):159–171. doi:10.1007/s10143-010-0289-y
References 605

21. Gerganov V, Amir S, Koerbel A, Brandes A, Stan 36. Kohama M, Murakami K, Endo T, Watanabe M,
A, Madjid S (2007) Cystic trochlear nerve schwan- Tominaga T (2009) Surgical and histological observa-
noma. Case report. Surg Neurol 68(2):221–225. tions of trochlear neurinoma: case report. Neurol Med
doi:S0090-3019(06)00963-3 [pii] Chir (Tokyo) 49(5):217–220. doi:JST.JSTAGE/
22. Goel A (1995) Infratemporal fossa interdural approach nmc/49.217 [pii]
for trigeminal neurinomas. Acta Neurochir (Wien) 37. Kovacs W (1927) Ueber ein solitares Neurinoma des
136(1–2):99–102 Nervus oculomotorius. Zentralbl Allgemeine Pathol
23. Goel A, Muzumdar D, Raman C (2003) Trigeminal Patholog Anat 40:518–522
neuroma: analysis of surgical experience with 73 38. Krayenbuhl H (1959) Neurinoma of trigeminal nerve.
cases. Neurosurgery 52(4):783–790; discussion 790 Bull Schweiz Akad Med Wiss 15(1):89–100
24. Hajjaj M, Linthicum FH Jr (1996) Facial nerve 39. Krueger WW, Kemper JV (2000) Cochlear schwan-
schwannoma: nerve fibre dissemination. J Laryngol nomas. Skull Base Surg 10(2):87–88
Otol 110(7):632–633 40. Lalwani AK (1992) Meningiomas, epidermoids,
25. Hakuba A, Tanaka K, Suzuki T, Nishimura S (1989) and other nonacoustic tumors of the cerebellopontine
A combined orbitozygomatic infratemporal epidural angle. Otolaryngol Clin North Am 25(3):707–728
and subdural approach for lesions involving the entire 41. Latack JT, Gabrielsen TO, Knake JE, Kemink JL,
cavernous sinus. J Neurosurg 71(5 Pt 1):699–704. Graham MD, Gebarski SS, Yang PJ (1983) Facial
doi:10.3171/jns.1989.71.5.0699 nerve neuromas: radiologic evaluation. Radiology
26. Hamm KD, Gross MW, Fahrig A, Surber G, Henzel 149(3):731–739
M, Kleinert G, Grabenbauer GG, Engenhart-Cabillic 42. Leonetti JP (1998) Cochlear neuromas. Am J Otol
R (2008) Stereotactic radiotherapy for the treatment 19(4):499–502
of nonacoustic schwannomas. Neurosurgery 62(5 43. Lesoin F, Rousseaux M, Villette L, Autricque A,
Suppl):A29–A36. doi:10.1227/01.neu.0000325934. Dhellemmes P, Pellerin P, Vaneecloo JM, Leys D,
16229.03; discussion A36 Jomin M (1986) Neurinomas of the trigeminal nerve.
27. Hironaka Y, Nakase H, Motoyama Y, Mishima H, Acta Neurochir (Wien) 82(3–4):118–122
Park YS, Hirabayashi H, Sakaki T (2010) Orbital 44. Leunda G, Vaquero J, Cabezudo J, Garcia-Uria J,
schwannoma extending to the lateral wall of the cav- Bravo G (1982) Schwannoma of the oculomotor
ernous sinus through the superior orbital fissure – case nerves. Report of four cases. J Neurosurg 57(4):563–
report. Neurol Med Chir (Tokyo) 50(2):154–157. 565. doi:10.3171/jns.1982.57.4.0563
doi:JST.JSTAGE/nmc/50.154 [pii] 45. Lipkin AF, Coker NJ, Jenkins HA, Alford BR (1987)
28. House WF, Hitselberger WE, Horn KL (1986) The Intracranial and intratemporal facial neuroma.
middle fossa transpetrous approach to the anterior- Otolaryngol Head Neck Surg 96(1):71–79
superior cerebellopontine angle. Am J Otol 7(1):1–4 46. Liu R, Fagan P (2001) Facial nerve schwannoma: sur-
29. Huang CF, Kondziolka D, Flickinger JC, Lunsford LD gical excision versus conservative management. Ann
(1999) Stereotactic radiosurgery for trigeminal schwan- Otol Rhinol Laryngol 110(11):1025–1029
nomas. Neurosurgery 45(1):11–16; discussion 16 47. Lunardi P, Missori P, Gagliardi FM, Fraioli B
30. Iaconetta G, Fusco M, Cavallo LM, Cappabianca (1989) Trigeminal schwannoma with infratemporal
P, Samii M, Tschabitscher M (2007) The abducens extension. Case report. J Neurosurg Sci 33(3):
nerve: microanatomic and endoscopic study. 293–295
Neurosurgery 61(3 Suppl):7–14. doi:10.1227/01.neu. 48. Marzo SJ, Zender CA, Leonetti JP (2009) Facial nerve
0000289706.42061.19; discussion 14 schwannoma. Curr Opin Otolaryngol Head Neck Surg
31. Jefferson G (1953) The trigeminal neurinomas with 17(5):346–350. doi:10.1097/MOO.0b013e32832ea999
some remarks on malignant invasion of the gasserian 49. Matsui T, Morikawa E, Morimoto T, Asano T (2002)
ganglion. Clin Neurosurg 1:11–54 Presigmoid transpetrosal approach for the treatment
32. Kano H, Niranjan A, Kondziolka D, Flickinger JC, of a large trochlear nerve schwannoma–case report.
Dade Lunsford L (2009) Stereotactic radiosurgery for Neurol Med Chir (Tokyo) 42(1):31–35. doi:JST.
trigeminal schwannoma: tumor control and functional JSTAGE/nmc/42.31 [pii]
preservation Clinical article. J Neurosurg 110(3): 50. McCormick PC, Bello JA, Post KD (1988) Trigeminal
553–558 schwannoma. Surgical series of 14 cases with review
33. Kawase T, Shiobara R, Toya S (1991) Anterior transpet- of the literature. J Neurosurg 69(6):850–860.
rosal-transtentorial approach for sphenopetroclival doi:10.3171/jns.1988.69.6.0850
meningiomas: surgical method and results in 10 patients. 51. McElveen JT Jr, Dorfman BE, Fukushima T (2001)
Neurosurgery 28(6):869–875; discussion 875–866 Petroclival tumors: a synthesis. Otolaryngol Clin
34. Kertesz TR, Shelton C, Wiggins RH, Salzman KL, North Am 34(6):1219–1230, x
Glastonbury CM, Harnsberger R (2001) Intratemporal 52. Mello LR, Tanzer A (1972) Some aspects of trigemi-
facial nerve neuroma: anatomical location and radio- nal neurinomas. Neuroradiology 4(4):215–221
logical features. Laryngoscope 111(7):1250–1256. 53. Nakagawa T, Uchida K, Ozveren MF, Kawase T
doi:10.1097/00005537-200107000-00020 (2004) Abducens schwannoma inside the cavernous
35. King JS (1976) Trochlear nerve sheath tumor; case sinus proper: case report. Surg Neurol 61(6):559–
report. J Neurosurg 44(2):245–247. doi:10.3171/jns. 563. doi:10.1016/S0090-3019(03)00580-9; discus-
1976.44.2.0245 sion 563
606 9 Nonvestibular Schwannomas

54. Nakamura M, Carvalho GA, Samii M (2002) 70. Sheehan J, Yen CP, Arkha Y, Schlesinger D, Steiner L
Abducens nerve schwannoma: a case report and (2007) Gamma knife surgery for trigeminal schwan-
review of the literature. Surg Neurol 57(3):183–188. noma. J Neurosurg 106(5):839–845. doi:10.3171/
doi:S009030190100670X [pii]; discussion 188–189 jns.2007.106.5.839
55. Nathan H, Ouaknine G, Kosary IZ (1974) The abducens 71. Sherman JD, Dagnew E, Pensak ML, van Loveren
nerve. Anatomical variations in its course. J Neurosurg HR, Tew JM Jr (2002) Facial nerve neuromas: report
41(5):561–566. doi:10.3171/jns.1974.41.5.0561 of 10 cases and review of the literature. Neurosurgery
56. Netuka D, Benes V (2003) Oculomotor nerve schwan- 50(3):450–456
noma. Br J Neurosurg 17(2):168–173 72. Spetzler RF, Daspit CP, Pappas CT (1992) The com-
57. Ohba S, Miwa T, Kawase T (2006) Trochlear nerve bined supra- and infratentorial approach for lesions of
schwannoma with intratumoral hemorrhage: case the petrous and clival regions: experience with 46
report. Neurosurgery 58(4):E791. doi:10.1227/01. cases. J Neurosurg 76(4):588–599. doi:10.3171/
NEU.0000204307.99246.17; discussion E791 jns.1992.76.4.0588
58. Okada Y, Shima T, Nishida M, Okita S (1997) Large 73. Spinnato S, Talacchi A, Musumeci A, Turazzi S,
sixth nerve neuroma involving the prepontine region: Bricolo A (1998) Dumbbell-shaped hypoglossal
case report. Neurosurgery 40(3):608–610 neurinoma: surgical removal via a dorsolateral
59. Paillas JE, Grisoli F, Farnarier P (1974) Trigeminal transcondylar approach. A case report and review of
nerve neurinomas. Apropos of 8 cases. Neurochirurgie the literature. Acta Neurochir (Wien) 140(8):827–832
20(1):41–54 74. Springborg JB, Poulsgaard L, Thomsen J (2008)
60. Pan L, Wang EM, Zhang N, Zhou LF, Wang BJ, Dong Nonvestibular schwannoma tumors in the cerebello-
YF, Dai JZ, Cai PW (2005) Long-term results of pontine angle: a structured approach and management
Leksell gamma knife surgery for trigeminal schwan- guidelines. Skull Base 18(4):217–227. doi:10.1055/s-
nomas. J Neurosurg 102(Suppl):220–224 2007-1016959
61. Paquis P, Castillo L, Lonjon M, Santini J, Grellier P 75. Symon L, Cheesman AD, Kawauchi M, Bordi L
(1998) Extracranial trigeminal schwannomas with (1993) Neuromas of the facial nerve: a report of 12
middle temporal fossa development. Neurochirurgie cases. Br J Neurosurg 7(1):13–22
44(1):38–45. doi:MDOI-JNG-03-1998-44-1-0028- 76. Taha JM, Tew JM Jr, van Loveren HR, Keller JT, el-
3770-101019-ART70 [pii] Kalliny M (1995) Comparison of conventional and
62. Phi JH, Kim DG, Chung HT, Lee J, Paek SH, Jung skull base surgical approaches for the excision of
HW (2009) Radiosurgical treatment of vestibular trigeminal neurinomas. J Neurosurg 82(5):719–725.
schwannomas in patients with neurofibromatosis type doi:10.3171/jns.1995.82.5.0719
2: tumor control and hearing preservation. Cancer 77. Tancioni F, Gaetani P, Villani L, Zappoli F, Rodriguez
115(2):390–398. doi:10.1002/cncr.24036 Y, Baena R (1995) Neurinoma of the trigeminal root
63. Pollock BE, Kondziolka D, Flickinger JC, Maitz A, and atypical trigeminal neuralgia: case report and
Lunsford LD (1993) Preservation of cranial nerve review of the literature. Surg Neurol 44(1):36–42.
function after radiosurgery for nonacoustic schwan- doi:0090301995001735 [pii]
nomas. Neurosurgery 33(4):597–601 78. Tanriover N, Kemerdere R, Kafadar AM, Muham-
64. Saleh E, Achilli V, Naguib M, Taibah AK, Russo A, medrezai S, Akar Z (2007) Oculomotor nerve schwan-
Sanna M, Mazzoni A (1995) Facial nerve neuromas: noma located in the oculomotor cistern. Surg Neurol
diagnosis and management. Am J Otol 16(4): 67(1):83–88. doi:S0090-3019(06)00432-0 [pii]; dis-
521–526 cussion 88
65. Samii M, Migliori MM, Tatagiba M, Babu R (1995) 79. Tung H, Chen T, Weiss MH (1991) Sixth nerve
Surgical treatment of trigeminal schwannomas. schwannomas. Report of two cases. J Neurosurg
J Neurosurg 82(5):711–718. doi:10.3171/jns.1995.82. 75(4):638–641. doi:10.3171/jns.1991.75.4.0638
5.0711 80. Vachata P, Sames M (2009) Abducens nerve schwan-
66. Sarma S, Sekhar LN, Schessel DA (2002) noma mimicking intrinsic brainstem tumor. Acta
Nonvestibular schwannomas of the brain: a 7-year Neurochir (Wien) 151(10):1281–1287. doi:10.1007/
experience. Neurosurgery 50(3):437–448; discussion s00701-009-0302-9
438–439 81. Veshchev I, Spektor S (2002) Trochlear nerve neu-
67. Sataloff RT, Frattali MA, Myers DL (1995) Intracranial roma manifested with intractable atypical facial pain:
facial neuromas: total tumor removal with facial nerve case report. Neurosurgery 50(4):889–891; discussion
preservation: a new surgical technique. Ear Nose 891–882
Throat J 74(4):244–246, 248–256 82. Visot A, Derome P, De Leon JL (1992) Spheno-
68. Sekhar LN, Pomeranz S, Sen CN (1991) Extradural cavernous and infratemporal trigeminal neurinomas:
petrous bone and petroclival neoplasms. Acta surgical series of 15 cases. Skull Base Surg 2(3):
Neurochir Suppl (Wien) 53:183–192 142–149
69. Sekhar LN, Schessel DA, Bucur SD, Raso JL, Wright 83. Yamaki T, Morimoto S, Ohtaki M, Sakatani K, Sakai
DC (1999) Partial labyrinthectomy petrous apicec- J, Himi T, Harabuchi Y, Tanabe S, Hashi K (1998)
tomy approach to neoplastic and vascular lesions of Intracranial facial nerve neurinoma: surgical strategy
the petroclival area. Neurosurgery 44(3):537–550; of tumor removal and functional reconstruction. Surg
discussion 550–532 Neurol 49(5):538–546. doi:S0090301997000256 [pii]
References 607

84. Yasui T, Hakuba A, Kim SH, Nishimura S (1989) 86. Yoshida K, Kawase T (1999) Trigeminal neurinomas
Trigeminal neurinomas: operative approach in eight extending into multiple fossae: surgical methods and
cases. J Neurosurg 71(4):506–511. doi:10.3171/ review of the literature. J Neurosurg 91(2):202–211.
jns.1989.71.4.0506 doi:10.3171/jns.1999.91.2.0202
85. Yonas H, Jannetta PJ (1980) Neurinoma of the trigem-
inal root and atypical trigeminal neuralgia: their com-
monality. Neurosurgery 6(3):273–277
Jugular Foramen Tumors
10

General Features JF tumors tend to extend through the path of


least resistance. They may extend within the CP
The management of jugular foramen (JF) tumors angle and involve the petrous bone, may grow
remains a challenge despite the considerable anterior to the pons with or without involvement
progress over the last decades. Their location of the clivus, or expand inferior even below the
deep in a confined space of the skull base and the foramen magnum. Some dumbbell-shaped
close proximity or even involvement of vital neu- tumors, usually meningiomas and schwannomas,
ral structures and/or major vessels complicates have both intra- and extracranial parts [76, 77].
surgical removal. The access to the JF area is
restricted by the surrounding structures: the
carotid artery anterior, the vertebral artery infe- Neuroimaging
rior, the hypoglossal nerve medially and the mas-
toid, styloid process, transverse process of C1, The radiological assessment confirms the pres-
and facial nerve laterally [116]. Interdisciplinary ence of a jugular foramen mass and—in most
cooperation offers the possibility to select the cases—allows for precise preoperative assess-
best treatment option for each individual patient. ment of the tumor type. Accurate preoperative
Jugular foramen tumors arise either primary diagnosis is of great value for the decision-making
within the foramen—glomus jugulare tumors and process and for patient counseling (Table 10.1).
schwannomas of the 9th–11th cranial nerves—or High-resolution CT and MRI scans should
from neighboring structures and involve the fora- be obtained in each patient. Thin-slice (1 mm)
men secondarily: meningiomas, chordomas, chon- bone-window CT allows to evaluate the tumor
drosarcomas, cholesteatomas, endolymphatic sac for calcifications and the bone structures for
tumor, chondroblastoma, lymphangioma, temporal hyperostosis or bone erosion. Tumors in the area
bone carcinoma, sarcoma, and metastases. Primary causing significant bone changes are schwanno-
bone tumors may also present as JF lesions: giant- mas, glomus tumors, meningiomas, aneurysmal
cell tumor, aneurismal bone cysts, osteoblas- bone cysts, chondrosarcomas, endolymphatic
toma, plasmacytomas, ossifying fibroma, osteoid sac tumors, giant-cell tumors, and metastases
osteoma, and fibrous dysplasia [63, 64]. The most [63]. Native and contrast-enhanced MRI provides
common primary tumors are glomus jugulare, fol- information regarding not only tumor features but
lowed by schwannomas and meningiomas [28, 38, also on its extension and precise relationship with
84]—these three types compose more than 80 % of neighboring structures. MR angiography permits
all JF tumors [95]. The most common primary ori- to evaluate tumor vascularization; MR venogra-
gin of JF metastases is the head and neck, followed phy depicts the venous circulation and patency/
by breast, lung, and prostate tumors [41]. occlusion of the sigmoid sinus and jugular vein.

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 609


DOI 10.1007/978-3-642-35422-9_10, © Springer-Verlag Berlin Heidelberg 2013
610 10 Jugular Foramen Tumors

Table. 10.1 Typical radiological characteristics of the most frequent JF tumors


CT Bone-window CT MRI Angiography
Schwannoma Isodense or slightly Enlargement of JF, smooth T1: hypo–isointense Mild/moderate
hyperdense well-defined bony margins vascularity
No evidence of bone T2: hyperintense
infiltration Strong contrast
enhancement
Glomus tumor Isodense or slightly Permeative “moth-eaten” T1: salt-and-pepper Extreme vascularity
hyperdense erosion of JF appearance
Irregular bony margins T2: hyperintense Prolonged vascular
blush
Loss of density in the Intense contrast AV shunting
affected bone enhancement
Intratumoral flow voids
Meningioma Slightly Maintained architecture T1: iso–hypointense Mild to moderate
hyperintense vascularity
Hyperostosis/sclerotic T2: iso–hyperintense No arteriovenous
changes of the JF Uniform contrast shunting
enhancement
Absence of vascular
flow voids
Dural tail sign
Modified from Löwenheim et al. [63]

Turbulent flow in a hypertrophic jugular bulb or is involved or encased by the tumor, balloon
slow/absent flow in case of jugular bulb thrombo- occlusion test might be required.
sis must be distinguished from JF tumors. The dif-
ferential diagnosis is based on evaluation of MR
signal characteristics and application of vascular Clinical Presentation and Diagnosis
flow-sensitive techniques, such as phase-contrast
MR venography [20, 84]. Fat saturation technique The diagnosis of JF tumors is made usually by
allows improved detection of marrow involve- ENT surgeons: the most frequent initial com-
ment and has been found useful for evaluation of plaints of these patients are related to cochlear
skull-base abnormalities [38]. nerve dysfunction (hearing loss or tinnitus) and
The degree of tumor vascularity and the nature lower cranial nerves dysfunction (swallowing
of the tumor should guide the need for preopera- difficulty, hoarseness). Frequently, the clinical
tive selective angiography and embolization. picture is variable due to extension into or com-
Intratumoral flow voids on MRI indicate high vas- pression of surrounding structures: CP angle,
cularity. In such cases, digital subtraction angiog- brain stem, middle ear, or para-/retropharyngeal
raphy may be done—for diagnostic purposes and space. The lesion can differentially affect any of
as a guide for preoperative embolization [85]. the low cranial nerves and cause a number of JF
Preoperative embolization might be extremely use- syndromes, which are rarely seen isolated in
ful in cases of highly vascularized tumors [74, 85]. patients with JF tumors. The three typical syn-
The precise knowledge of the tumor-feeding ves- dromes are JF or Vernet’ syndrome (unilateral
sels and their relationship to the carotid and verte- 9th + 10th + 11th cranial nerve involvement), pos-
bral arteries, as well as to venous structures, are terior laterocondylar or Collet–Sicard syndrome
important for the approach planning. Further fac- (unilateral 9th + 10th + 11th + 12th cranial nerve),
tors to consider are the tumor relation to brain and posterior retropharyngeal or Villaret’s syn-
stem, lower cranial nerves, middle ear, petrous drome (involvement of 9th + 10th + 11th + 12th
apex, and clivus. In case the internal carotid artery cranial nerves and Horner’s syndrome).
Management 611

The most frequent initial symptoms are pulsa- exclude the possibility that they are actually
tile tinnitus (25–84 %), conductive hearing loss involved by the tumor. Further important issues
(66–77 %), headache (20–66 %), hoarseness are tumor type and size, patients’ general and
(35–38 %), facial paralysis (15–25 %), vertigo neurological status, as well as his expectations.
(37 %), hoarseness (25–44 %), swallowing Thus, in case of normally functioning lower cra-
difficulty (37–50 %), shoulder weakness (22 %), nial nerves, a tightly adherent tumor part, for
paralysis of the tongue (19–31 %), trigeminal example, in the jugular foramen, might be left.
hypesthesia (13 %), and—in more advanced Contrary, if a severe lower cranial nerve dysfunc-
cases—cerebellar (up to 44 %) and long-tract tion is already present, complete tumor removal
symptoms [28, 84, 94, 98]. should be favored.
Although most authors report that the clinical External beam radiotherapy and radiosurgery
presentation of various JF lesions is similar [28, 38], are primary management options in the elderly
other found some differences. Ramina et al.[85] medically unstable patients and in those unwill-
reported that in glomus jugulare tumors the ini- ing to have surgery. They are used as an adjunc-
tial symptoms are hearing loss and tinnitus, while tive treatment in case of unresectable tumor
schwannomas and meningiomas may present remnants or recurrences [37].
with lower cranial nerves deficit. The function of
the lower cranial nerves is affected in 20–27 % of
glomus jugulare tumors, in 35–50 % of menin- Operative Approaches
giomas, and in 44–50 % of schwannomas [28,
64]. One finding that might be important in the A good knowledge of the normal and pathologi-
differential diagnosis is the retrotympanic mass: cal microanatomy of the JF area is a major pre-
it is found in 91 % of glomus jugulare tumors, in requisite for successful surgical management.
43 % of meningiomas, and rarely (11 %) in According to the classical concept, the jugular
schwannomas [28]. foramen consists of two parts: pars nervosa or
nervous part and pars venosa or venous part. Pars
nervosa is located anteromedially and contains
Management the inferior petrosal sinus, meningeal branches of
the ascending pharyngeal artery, vena canaliculi
Management options of JF tumors include obser- cochlea, and glossopharyngeal nerve, while the
vation, radiation therapy and surgery, or combi- pars venosa, located posterolaterally, contains the
nation thereof. The ideal one is complete surgical jugular bulb and vagus and accessory nerves [5,
excision with preservation of all neurological 7, 26]. The vertical portion of the facial nerve
functions, which leads to definitive healing of the straddles the jugular bulb. Recent microanatomi-
patients, provided the tumor is benign [1, 2, 30, cal studies have shown, however, that this
40, 53, 84]. In malignant tumors and in those simplified concept lacks practical significance.
infiltrating essential structures, subtotal removal The JF consists actually of three compartments,
with adjuvant radiotherapy may be the most rea- defined by dural septations. The third one is
sonable option. Subtotal tumor removal is pre- located between the two venous compartments at
ferred also in cases in which radical surgery the level of the interjugular process [57, 88]. This
carries high risk of new neurologic deficits: interjugular compartment contains the 9th, 10th,
extensive infiltration of the skull base or of the and 11th cranial nerves, running through two
cavernous sinus, absence of a cleavage plane dural perforations: the glossopharyngeal meatus
between the tumor and the brain stem or its (9th nerve) and the vagal meatus (10th and 11th
infiltration, extreme adherence to cranial nerves nerves). The great interindividual variability, in
in patients with normal preoperative function, or regard to the course of the cranial nerves within
encasement of neurovascular structures. Normal the JF and venous anatomy, should be considered
cranial nerve function before surgery does not by the surgeon [103].
612 10 Jugular Foramen Tumors

The operative approach to the jugular fora- The main principle that we follow when select-
men should provide adequate access to the area ing the operative approach is that it should be tai-
and enable the surgeon to perform safe tumor lored to the specific characteristics of each patient in
removal. Generous exposure of all surrounding terms of tumor location, type, size and extension
structures, however, is not a prerequisite for suc- pattern, involvement and degree of erosion/destruc-
cessful surgery. The approach should be tailored to tion of the bone structures, and patients’ neurologi-
the individual patients and tumor characteristics. cal status and expectations. Moreover, surgery
Importantly, it should not cause additional mor- should be regarded as part of the whole manage-
bidity or major discomfort to the patient. Multiple ment—in some tumors it can heal the patient, while
approaches to the area of the jugular foramen, in others, it is a component of the multimodal treat-
depending on the tumor extension pattern, have ment plan. The approach should allow achieving
been developed and are still used: infratemporal the goals of surgery but—on the other hand—should
fossa, modified infratemporal fossa, transmas- not be related to high procedure-related morbidity.
toid, widened transcochlear, translabyrinthine, far Electrophysiological monitoring of the 9th,
lateral, trans-sigmoid, transjugular, suboccipital– 11th, and 12th cranial nerves is performed in addi-
retromastoid, petro-occipital–trans-sigmoid, jux- tion to the routine monitoring of SSEP, facial nerve
tacondylar, mastoid, and lateral neck approaches EMG, and BAEP. The electrodes are placed in the soft
[1, 9, 24, 30, 31, 36, 77, 81, 94, 98]. palate, trapezius muscle, and tongue, respectively.
The infratemporal approach, which is a Endotracheal tube electrodes are utilized in rare cases
refinement of the cervicomastoid approach of to map and monitor the function of vagus nerve.
Gallard and Rebattu [94], was described by Fisch
in 1978 [29]. It allows direct view of the jugular Lateral Suboccipital (Retrosigmoid)
foramen and wide exposure of the tumor. The Approach
approach was modified later to enable resection of Tumors confined to the posterior cranial fossa, as
both tumor components in a single stage [56, 81]. well as most of those located primarily in the
This approach is regarded by some surgeons as jugular foramen with a smaller intracranial part,
the optimal one for resection of more vascular are approached intradurally via the lateral suboc-
tumors, such as glomus jugulare tumors [99]. cipital route. The technique of the approach is
Drawbacks of this technique is that it necessitates described in the chapter on RS approach. In case
sacrifice of the middle ear structures, closure of of jugular foramen tumors, in contrast to VS, the
the ear canal, which can cause conductive hear- dural incision should not be continued necessar-
ing loss and anterior rerouting of the descending ily to the level of the transverse sinus. The main
facial nerve, related to a certain risk of functional anatomical landmarks and neural structures are
deficits [13, 77]. With some later technical identified, and the intracranial tumor portion is
improvements, the risks for permanent facial approached initially. In a manner similar to the
palsy have been considerably reduced. Leonetti one applied in VS, the tumor is debulked with
and Brackmann [62] developed a technique for ultrasonic aspiration or with suction and platelet
mobilizing the facial nerve during infratemporal knife. The dissection of the tumor capsule from
fossa surgery and preserving its vasculature at the the cranial nerves is performed only when
stylomastoid foramen. Pensak and Jackler [82] sufficient internal decompression has been
described the fallopian bridge technique, which achieved and is performed always in the arach-
allows tumor removal without mobilization or noid plane. The two-hand technique is used—the
transposition of the facial nerve. In their series, tumor capsule is held with one hand and the dis-
normal facial function at 6 months follow-up had section is performed with the other hand (usually
92 % of the cases. In case of smaller jugular fora- the dominant one). The intraforaminal part of the
men schwannomas and meningiomas with lim- tumor is approached afterward. If it is small, it
ited extension, more conservative approaches can be gently pulled out of the foramen into the
have been proposed. CP angle and removed. Opening its dorsal wall,
Management 613

in a technique analogous to the one used to open smaller burrs in the depth of the foramen. The
the IAC in case of VS removal, can extend the last bone layer or lamina is removed with a small
approach to the jugular foramen (see Case platelet knife in order to avoid damage to under-
Illustration 10.1). The senior author has applied lying structures. The intraforaminal tumor part is
this technique since the 1980s. The amount of debulked initially, and then its capsule is dis-
safe bone removal is evaluated on the bone- sected from the cranial nerves and/or vessels. The
window thin-slice CT and tailored to the particu- utilization of angled rigid endoscopes (since 2005
lar tumor extension. The dura overlying the at the INI) allowed for removal of tumors with
posterosuperior part of the foramen is stripped even larger intraforaminal extension (see below).
off and cut away. High-speed drill is used to Following tumor removal, in case the jugular
remove the bone in 0.1-mm increments under foramen has been opened, it should also be sealed
direct visual control. Drilling starts usually at the with multiple small fat pieces fixed with fibrin
superior margin of the foramen and proceeds glue. The dura is closed in a watertight fashion,
posteriorly. Large-sized diamond burr (4–5 mm and the opened mastoid air cells are also sealed
in diameter) are used initially and changed to with fat pieces and fibrin glue.
614 10 Jugular Foramen Tumors

Case Illustration 10.1 foramen jugulare, are presented on figures a–c.


Cystic Jugular Foramen Schwannoma The patient was operated in the semi-sitting
Removal (Drilling of the dorsal Wall of the position: a retrosigmoid craniotomy was
jugular foramen) performed, and the CP angle tumor portion was
This patient—operated in 1988—was one of removed. Then, the dura dorsal to the jugular
the first cases in which the technique of open- foramen was excised, and the underlying bone
ing the dorsal jugular foramen wall was applied. drilled off. Thus, the intraforaminal tumor por-
He presented with swallowing deficits and tion was well exposed and completely removed.
hoarseness. The preoperative MR images, The patient recovered rapidly from surgery and
showing the cystic tumor with extension in the did not have new neurological deficits.

a b

(a–c) Preoperative MR images. (d–j) Intraoperative (7–8). (g) The arrowheads delineate the jugular fora-
images. (d) Initial view of the tumor (Tu) in the CP men. (h) Drilling of the dorsal foramen wall and expo-
angle. (e–f) Removal of the CP angle tumor part, visu- sure of the intraforaminal tumor portion (small arrows
alization of the seventh–eighth cranial nerve complex on i). (j) Complete tumor removal
Management 615

d e

f g

h i

j
616 10 Jugular Foramen Tumors

Combined Cervical and Lateral Craniectomy, Mastoidectomy/Petrosectomy


Suboccipital Intra–extradural A retromastoid craniectomy is made, as described
Transmastoid Infralabyrinthine Approach earlier. Additional mastoidectomy is performed
The approach is used in tumors with cervical with a diamond drill to expose the sigmoid sinus
extension or very large inaccessible intraforam- from the superior petrosal sinus to the jugular
inal part. It consists of several steps: bulb. The jugular foramen can be further exposed
1. Skin incision and identification of the anterior dorsolaterally by drilling the posterior part of the
border of the sternocleidomastoid muscle occipital condyle.
2. Exposure of the mastoid The tumor extension and the degree of involve-
3. Neck dissection and exposure of the cervical ment of the pyramid, middle, and/or inner ear
region, vessels, and nerves define the extent of bone removal. Whenever
4. Exposure of the facial nerve needed, a radical mastoidectomy is performed,
5. Retrosigmoid craniectomy and mastoidectomy and the sigmoid sinus is exposed widely, down to
6. Exposure of the sigmoid sinus the jugular foramen. The pyramid is drilled fur-
7. Intradural exposure ther anteromedial to the fallopian canal, superior
8. Extracranial tumor removal to the styloid process and inferior to the laby-
9. Intracranial tumor removal rinth. The antrum, the lateral semicircular canal,
10. Reconstruction of the dura and skull base and the vertical portion of the facial nerve are
exposed. Mobilization and rerouting of the facial
Neck Dissection and Exposure of the Cervical nerve are necessary only if the tumor extends to
Region, Vessels, and Nerves the middle ear cavity or into the carotid canal. In
The patient is placed supine on the operating order to avoid severe facial nerve palsy, care is
table. The head is fixed with a Mayfield head taken to preserve the vascularized fascia and soft
holder and turned 60° to the contralateral side. A tissue attached to the nerve at the stylomastoid
slightly curved retroauricular skin incision is per- foramen.
formed, starting in the temporal region and reach- In deaf patients with extensive tumors, the
ing the anterior border of sternocleidomastoid posterior wall of the external auditory meatus is
muscle. A dissection plane is created between the drilled, and the external auditory canal is cut at
parotid gland and the anterior border of the ster- the osteocartilaginous junction, thereby gaining
nocleidomastoid muscle, preserving the greater access to the tumor portion in the ear.
auricular nerve. The facial nerve is identified
between the mastoid process and angle of man- Intradural Exposure
dible, ventral to the stylomastoid foramen. The dura might be incised posterior or anterior to
Helpful landmark for its identification is the the sigmoid sinus; the incision is then extended to
“pointer” of the cartilaginous auditory canal. The the jugular foramen. The dura, transverse and
sternocleidomastoid muscle is detached from the sigmoid sinuses, and the jugular bulb are retracted
mastoid and reflected inferiorly. The posterior anteriorly. The cerebellopontine cistern is then
belly of the digastrics muscle is also cut at the opened to release the CSF, and the cerebellum is
skull base and reflected inferiorly. retracted gently, exposing the intracranial tumor
The hypoglossal nerve is located inside the portion.
carotid triangle, medially to the posterior belly of
the digastric muscle. The caudal cranial nerves Tumor Removal
are exposed in the neck, along with the internal The extracranial portion of the tumor is usually
jugular vein, the common carotid artery with the resected first, following the identification of the
external and internal carotid arteries, and fol- crucial neck structures. The varying direction of
lowed to the skull base. displacement and/or involvement of the major
Management 617

vessels in different tumors should be consid- methyl methacrylate (Palacos). The sterno-
ered. Glomus jugulare tumors may surround or cleidomastoid muscle is sutured back at its
even infiltrate the walls of the internal carotid insertion on the occipital bone, the wound cav-
arteries, while schwannomas frequently displace ity is filled with hemostatic sponges and/or fat
it anteriorly. pieces, and the wound is closed in layers. In
The intracranial portion of the tumor is deb- case the external auditory canal had been cut, it
ulked with the ultrasonic aspiration and dissected is closed in two layers.
from the cranial nerves and vessels. With this
technique, a wide exposure of the jugular fora-
men is achieved, allowing removal of the whole Endoscope-Assisted or Pure Endoscopic
intraforaminal tumor portion. The most lateral Tumor Removal
parts might be visualized and removed with endo-
scopic control, as described earlier. Schwannomas An important advancement in the recent years
and meningiomas of the jugular foramen usually was the introduction of the rod lens endoscopes
displace the nerves ventrally and the jugular bulb and the development of appropriate instrumen-
dorsally and inferiorly, while in case of glomus tation. Endoscopy is currently an integral part
tumors, the relations between the tumor and sur- of the neurosurgical armamentarium in case of
rounding structures cannot be predicted. Tumors jugular foramen tumors. Angled endoscopes
arising from the dome of the jugular bulb can allow to visualize and remove, whether under
extend intraluminally—in the sigmoid sinus and microscopic or under pure endoscopic control,
internal jugular vein. In such cases, the sigmoid tumor parts, extending or located “behind the
sinus and the internal jugular vein have to be corner.” Importantly, the microscope and the
ligated and opened in order to achieve complete endoscope should not be considered as mutu-
tumor removal. The anterior jugular bulb wall ally excluding tools but rather should be used
helps to preserve the lower cranial nerves. judiciously to achieve the goals of surgery.
In case of benign noninvasive lesions, Complete resection of such tumors required
complete removal of the tumor and infiltrated previously complex combined approaches or
bone should be attempted. Contrary, in case two separate operative steps, which were related
of malignant tumors or if a cleavage plane is to certain risk of approach-related morbidity.
absent, complete “oncological” removal is fre- Currently, the extracranial and the jugular fora-
quently not possible without the risk of severe men parts of the tumors can be removed via the
neurological deficits and the amount of resec- standard cervical transmastoid approach, while
tion should be carefully planned. If the facial the intracranial part can be removed with an
nerve has been destroyed by the tumor, healthy endoscope-assisted/pure endoscopic technique:
proximal and distal ends are identified and an the “outside–inside technique.” Thus, the neces-
interposition sural nerve graft is used for its sity of a retrosigmoid approach to access this
reconstruction. part is obviated (see Case Illustration 10.2).
Tumors with a major intracranial intradural
Reconstruction of the Dura part, especially if they cause brain stem com-
and Skull Base pression, should be approached and removed
After the tumor has been resected, the jugular via the retrosigmoid route. The intraforaminal
foramen is sealed with several fat pieces and tumor extension in such cases can be resected
fibrin glue. It can be further reinforced from out- with an endoscope-assisted/pure endoscopic
side with the posterior belly of the digastric technique: the “inside–outside technique” (see
muscle. The dura is closed in a watertight fash- Case Illustration: Endoscope-Assisted transcer-
ion, and the posterior fossa is reconstructed with vical Tumor Removal)
618 10 Jugular Foramen Tumors

Case Illustration 10.2 The tumor was seen encroaching the digastric
Endoscope-Assisted Transcervical Tumor muscle from above (m, n). Its capsule was
Removal then incised, and intracapsular removal under
The patient presented with a large recurrent microscopic control was performed (o, p). In
chondrosarcoma in the left CP angle and jug- order to visualize and reach the more rostral
ular foramen areas with a major extracranial parts, an angled endoscope was utilized. Using
extension along the internal jugular vein (a–l). angled instruments and endoscopic visualiza-
She has been operated three times previously: tion, the whole tumor, including its intradural
24 years, 10 years, and 4 years ago via differ- part in the CP angle, could be completely
ent approaches, including a retrosigmoid one. removed (q, r). Enlargement of the bone open-
At presentation, she had an HB grade IV facial ing was not necessary. The integrity of the
nerve palsy (since the last surgery), trigemi- lower cranial nerves and of seventh–eighth
nal nerve hypesthesia (V1,V2), swallowing nerves was preserved. Fat pieces and fibrin
difficulty, and dysphonia. glue were used to seal the dural opening.
Considering the size of the tumor and the Thus, the tumor could be removed completely
growth tendency of its intracranial part, a deci- in one stage, and a transcranial approach was
sion in favor of surgery was taken. The surgical not necessary.
strategy was to approach initially the extracra- After the surgery, temporary worsening of
nial tumor part transcervically and follow it the swallowing deficit was noted. Still, the
proximally through the bone corridor created function recovered rapidly, and in 3 weeks,
by the growing tumor (see the bone-window the oral feeding could be started. At 3 months
CT scans on figure a). In case the intradural follow-up examination, the patient reported
part could not be safely removed, a lateral that swallowing is even better than prior to
suboccipital craniotomy could be additionally surgery. The facial nerve palsy remained
made. The internal carotid artery and the lower unchanged. The postoperative MRI is pre-
cranial nerves were exposed at the beginning. sented on figures s–x.
Management 619

a b

c d

Preoperative CT (a) and MR images (b–l) of the tumor part, visible with the microscope. (q)
patient, demonstrating the extension of the tumor. See Visualization of tumor remnants with the angled endo-
the text for details. Intraoperative images: (m, n) expo- scope and their removal with angled instruments (r).
sure of the tumor and incision of its capsule. (o) Gradual (s–x) MRI performed 3 weeks after surgery. The tumor
internal decompression. (p) Complete removal of the cavity is filled with fat tissue
620 10 Jugular Foramen Tumors

e f

g h
Management 621

i j

k l
622 10 Jugular Foramen Tumors

m n

p
o

q r
Management 623

s t

u v
624 10 Jugular Foramen Tumors

w x
Postoperative Care and Outcome 625

Extended Endoscopic Endonasal of the esophageal muscles. On the other hand,


Approach laryngeal closure during swallowing, which in
healthy persons prevents aspiration, is inhibited.
The extended endoscopic endonasal approach to Especially endangered for aspiration are patients
the JF has been developed in the recent years as with palsy of both ninth and tenth nerves.
an alternative to the microsurgical technique. Its Bilateral vagus nerve injuries lead additionally
theoretical advantage is reduction of the risks to stridor and respiratory insufficiency due to the
related to manipulation of neurovascular struc- inability to abduct the vocal cords and, hence,
tures [108, 125]. The approach is regarded as to open the larynx. The hypoglossal nerve has
especially useful in case of caudal and medial the lowest incidence of iatrogenic injury. Its uni-
tumor extension. Early experience has shown that lateral dysfunction causes dysarthria and swal-
the technique is feasible and does not restrict the lowing difficulties. Bilateral dysfunction causes
access and—respectively—the possibility to significant swallowing dysfunction due to dis-
remove the tumor. The direct access to the carotid turbed swallowing initiation, which is mediated
artery in its subpetrous and retropharyngeal parts by the hypoglossal nerve.
allows for early devascularization of the tumor. Lower cranial nerve dysfunction severely
The experience with this approach is still limited impacts the patients’ quality of life and might
and insufficient to prove its supremacy to micro- increase the risk of other, potentially life-threatening,
surgical or endoscope-assisted tumor removal in complications. In order to avoid such outcome,
terms of neurological outcome and radicality of every effort should be made to preserve the
tumor removal. A major problem that remains is nerves at surgery and to manage the patients
the difficult reconstruction of the dura and the with neurological deficits properly. The main
skull base in tumors with intracranial extension prerequisites for preservation of lower cranial
and the respectively high CSF leak rate [125]. nerves are the following: careful selection of
surgical indications and goals; choice of most
appropriate operative approach; detailed knowl-
edge of lower cranial nerve microanatomy;
Postoperative Care and Outcome early identification of all crucial structures,
using the anatomical landmarks; and constant
Lower Cranial Nerves Dysfunction intraoperative nerve monitoring. Dissection of
the tumor capsule should be performed only
The most frequent and dangerous operative com- after sufficient internal tumor decompression
plication is the occurrence of new lower cranial has been achieved and should always proceed in
nerve deficit. Injuries to these nerves lead to the correct arachnoid plane.
dysphagia and/or dysphonia (9th, 10th cranial
nerves), scapular winging and shoulder droop
(11th nerve), and weakness and atrophy of the Postoperative Care
tongue (12th nerve). These nerves are vulnerable
to injury along their intracranial, extracranial, Slowly developing swallowing dysfunction dur-
and foraminal segments. The glossopharyngeal ing tumor growth can be generally well compen-
nerve is regarded as the most vulnerable to sur- sated for by the healthy contralateral side. New
gical injury due to its medial position within postoperative lower cranial nerve dysfunction,
the jugular foramen. Its most important func- however, is related to a very high risk of compli-
tion is sensory innervation of the upper phar- cations, such as aspiration pneumonia. The swal-
ynx: it triggers the swallow reflex. In case the lowing difficulty and the impaired ability to
vagus nerve is intact, however, it acts as a sec- cough and expectorate promote frequent aspira-
ond trigger of the swallow reflex. Injury to the tions with the related risk of aspiration pneumo-
vagus nerve disrupts the peristaltic contractions nia and other pulmonary complications.
626 10 Jugular Foramen Tumors

Patients operated for jugular foramen tumors Fayad et al. [28] presented their experience
are usually intubated nasally. The risk of aspira- with 83 patients with jugular foramen tumors.
tion is related to the patients’ level of conscious- Total tumor removal was achieved in 81 % of the
ness, and too early extubation after surgery cases. The occurrence of new lower cranial nerve
should be avoided. Nasal tubes are well tolerated deficits was highest in meningiomas (50 %),
and should be removed only when the patient is followed by schwannoma (22 %) and glomus
really awake and cooperative. In case of swal- jugulare tumors (19 %). Normal or near-normal
lowing dysfunction, oral feeding is prohibited. facial function at last follow-up had 88 % of the
The reason for the swallowing dysfunction patients. Although the conclusion of the authors
should be identified and specific examina- is that total resection of jugular foramen tumors
tions carried out: endoscopic and/or radiologi- can be a curative treatment, subtotal removal
cal evaluation of the swallowing act. The later may be required to preserve neural functions.
study provides information on the mechanics Ramina et al. [85] reviewed retrospectively 102
of swallowing, the laryngeal function, and the patients with jugular foramen tumors. Complete
risk of aspiration. Nasogastric tube feeding is tumor removal was achieved in 77.5 % of the
continued—even for up to several weeks— patients with paragangliomas and in all patients
until swallowing recovers, and resumption of with schwannomas. Lower cranial nerve deficit
adequate oral intake is safe. Tracheostomy occurred in 10 % but was transient in 3.8 %.
should be considered in patients with swallow- The mortality rate in the series was 4.2 %. In the
ing difficulty and recurrent aspiration. It should series of Lustig and Jackler [64], new postopera-
be performed as early as possible if clinical tive CN dysfunction occurred most frequently
signs of persistent aspiration are present. Early in patients with meningiomas (55 %), followed
vocal cord medialization may be an alternative by those with glomus tumors (35 %) and with
to tracheostomy. If prolonged recovery is antici- schwannomas (15 %).
pated, a gastrostomy is necessary as a long-term Another common complication, related mostly
means for providing nutrition. Currently, it is to the technique used to reconstruct the dura and
performed as a percutaneous endoscopic proce- the skull base, is the CSF leak and the related
dure. Speech–language (logopedic) therapy is infectious complications. The incidence of CSF
helpful to promote velopharyngeal function and leak in the literature varies from 3.9 to 13.3 %
teach compensatory articulation techniques and [52, 85, 98].
should be initiated early after surgery.
Tumor type has been found to correlate with
the risk of developing lower cranial nerve dys- Radiotherapy/Radiosurgery
function. Glomus jugulare tumors often compress
the lower cranial nerves, without necessarily Radiosurgery and stereotactic radiotherapy are
infiltrating them. Schwannomas originate from alternative treatment options for patients with
one of the lower cranial nerves, and preop- smaller JF tumors. The experience is expanding,
erative palsy is often present. The risk for new and many groups report good results. Compared
neurological deficits is highest in case of menin- to surgery in the JF area, good standards of radio-
giomas. In the series of Lustig and Jackler [64], surgical treatment are much earlier achieved and
new deficit was registered in approximately 60 % are not so directly related to the individual expe-
of the meningiomas, 30 % of glomus jugulare rience. The outcome after radiosurgery, espe-
tumors, and in 15 % of schwannomas. Oghalai cially at long-term follow-up, however, is not
et al. [77] presented a series of 28 patients with superior to that of surgical resection, performed
jugular foramen tumors with intracranial exten- by experienced surgeon. Furthermore, the goal of
sion: new postoperative lower cranial palsy was radiation therapy is tumor control, not cure.
found only in patients with meningiomas (40 %) Additional drawbacks of radiation therapy are the
and schwannomas (36 %). following: lack of tissue diagnosis, delay from
Jugular Foramen Schwannomas 627

treatment to its effect, and the risks of radiation,


especially in case of younger patients. Therefore,
we consider primary radiosurgery/radiotherapy
as an option only for those unwilling or unable to
undergo surgery.

Jugular Foramen Schwannomas

JF schwannomas originate either from the


glossopharyngeal, vagus or accessory nerves, or
from the cervical sympathetic chain within the
jugular foramen [50, 58, 109]. They are relatively
uncommon and in the absence of neurofibromatosis
comprise 2.9–4 % of all intracranial schwannomas
[94, 96, 98, 113]. In the senior authors’ series, Fig. 10.1 Small schwannoma in the area of the right JF
the ratio of JF to vestibular schwannomas was (T2-weighted MR image). The diagnosis was made
1:24 [94]. relatively early due to the patients’ complaints of swal-
lowing difficulty
JF schwannomas originate most frequently
from the 9th cranial nerve and most rarely from
the 11th nerve. As noted by some authors, how- proximal or distal within the jugular foramen.
ever, the exact nerve of origin is often difficult to More proximal origin along the nerve usually
determine and is not always reported [23, 44, leads to predominant intracranial tumor exten-
113]. Song et al. [109] estimated that up to 90 % sion, while tumors with more distal sites of origin
of the cases reported in the literature are identified tend to grow intraforaminally and extracranially
to originate from the glossopharyngeal or vagus [23]. Song et al. [109] pointed that the tumor
nerves (Figs. 10.1 and 10.2). The reason for this grows in the direction of least pressure or
predilection is still unknown. According to one resistance, but the expansion can be modified by
hypothesis, Schwann cells in the vicinity of the specific anatomical structures acting as a barrier
respective ganglia are more prone to tumor devel- to tumor growth, for example, the inferior
opment. The superior and inferior ganglia of the petrosal sinus.
glossopharyngeal nerve are both located within
the JF; the superior ganglion of the vagus nerve is
located within the JF, while its inferior ganglion Classification
is located below the extracranial orifice of the
foramen [64, 109]. On the other hand, the acces- Several classifications of JF schwannomas related
sory nerve has no ganglia within the JF. to tumor location and extension pattern have been
JF schwannomas are more common in females introduced and are currently used by different
and are diagnosed usually in the fourth to sixth surgeons [19, 31, 58, 69]. We present the one that
decade of life [23, 31, 58]. They are slow-growing we use in our practice [94], which is useful for
noninfiltrative lesions. Malignant schwannomas approach selection and outcome prediction:
in this region are rare but have been reported [8]. • Type A—the tumor is located primarily intracra-
JF schwannomas comprise a highly variable nially with minimal enlargement of the JF.
group not only in terms of nerve of origin. The • Type B—the tumor is primarily in the JF with
tumor itself may have intracranial, intraforaminal, a smaller intracranial portion.
and/or extracranial components or combination • Type C—the tumor is primarily extracranial.
thereof. The variable site of origin along the nerve • Type D—“dumbbell”-shaped tumor with
could explain these differences, for example, extra- and intracranial components.
628 10 Jugular Foramen Tumors

a b c

d e f

g h

Fig. 10.2 MRI of a patient with a vagus nerve schwan- it from other JF schwannomas. The tumor was removed
noma. The preoperative images (a–e) provide no clue on completely via the retrosigmoid approach. At surgery, it
the possible nerve of origin. (a–c) T1-weighted contrast- was found that it originated from the fibers of the vagus
enhanced sequences. (d) T2-weighted sequence. (e) T2 nerve. The control MR images 12 months later are shown
dark fluid sequence. Imaging does not allow to distinguish on figure (f–h)

Clinical Presentation by the tumors’ size, location, and effect (e.g.,


The early clinical symptoms in patients with compression) on the surrounding structures
JF schwannomas are related to the tumors’ ori- [63]. In the senior authors’ series, the most fre-
gin, while in later stages they are determined quent initial symptom was headache (66 %),
Jugular Foramen Schwannomas 629

followed by hearing loss (50 %), hoarseness Neuroimaging


(38 %), and dysphagia (38 %). At presentation,
81 % of the patients had lower cranial nerve On native CT scans, JF schwannomas appear
deficit, 75 % had hearing loss, 44 % had cer- isodense or slightly hyperdense and show moder-
ebellar symptoms, 31 % had tongue hemiat- ate enhancement after administration of contrast
rophy, 25 % had facial palsy, and 13 % had medium. Bone-window thin-slice CT demonstrates
altered facial sensation [94]. The lower cranial the enlargement of JF with smooth well-defined
nerve dysfunction does not necessarily cor- bony margins and no evidence of bone infiltration
relate with the nerve of origin because the JF (see the case illustrations to the chapter) [7].
compartment is so small that all cranial nerves JF schwannomas are iso- to slightly hypoin-
may be affected at the same time. tense on T1-weighted MRI and have a mild or
JF schwannomas should be differentiated moderate hyperintensity on T2-weighted MRI.
preoperatively from VS and glomus jugulare They show dense enhancement after contrast
tumors. Type A tumors tend to present as a administration. The tumor is well circumscribed
CP angle mass and cause hearing loss, tinni- and has a round or elongated configuration that
tus, vertigo, and ataxia, thus mimicking a ves- follows the course of the respective cranial nerves
tibular schwannoma [94]. Preoperative facial [27, 65, 96]. The lack of IAC involvement differ-
nerve weakness, however, has been reported in entiates them from VS. The “dumbbell” shape—
20–25 % of patients with JF schwannomas [69, 94], if present—is almost pathognomonic [63]. Based
while in VS, such presentation is not typical. on the preoperative neuroimaging data, it is, how-
Patients with type B tumors present with tinni- ever, impossible to determine the nerve of origin
tus, deafness, middle ear mass, and lower cra- of the schwannoma, even if the tumor is of small
nial nerve and/or 12th nerve dysfunction, while size (Figs. 10.1, 10.2, 10.3, 10.4, 10.5, and 10.6).
patients with type C tumors present with a pal- MR angiography is generally sufficient to present
pable neck mass, mass of the lateral pharyngeal the vasculature of and around the tumor. Highly
wall, or with a variety of JF syndromes [67, 105]. vascularized schwannomas are exceptional, and
Dumbbell-shaped tumors frequently present as preoperative conventional angiography with or
a neck mass or dysphagia/dysphonia. without embolization is generally not required.

a b

Fig. 10.3 Type A JF schwannoma with a large intracranial part and a smaller JF part. (T1-weighted MRI after gado-
linium application)
630 10 Jugular Foramen Tumors

a b

Fig. 10.4 Axial and coronal T1-weighted MRI after gadolinium application, demonstrating a type B JF schwannoma,
located predominantly within the jugular foramen (arrow)

a b

Fig. 10.5 T1-weighted contrast-enhanced and T2-weighted MRI contrast application of a large type C schwannoma
Jugular Foramen Schwannomas 631

a b

c d

Fig. 10.6 Type D schwannoma—“dumbbell”-shaped tumor (T1-weighted MRI after gadolinium application)
632 10 Jugular Foramen Tumors

Case Illustration 10.3 Considering the fact that the major tumor
JF Schwannoma with a Predominant part was located intradurally in the CP angle
Intracranial Growth and caused brain stem and cerebellar com-
This 53-year-old male patient presented with pression with respective neurological signs, a
complaints of headache, hearing loss on the decision in favor of the RS approach was
right side, and dizziness since 12 months. taken. Initially, the CP angle tumor part was
Neurological examination revealed a slight debulked, and its capsule was dissected from
abducens nerve palsy and complete hearing all surrounding structures in the arachnoid
loss on the right side, as well as gait ataxia. plane. Then, the jugular foramen was opened,
Bone-window CT showed an area of erosion and the intraforaminal tumor part was removed
of the petrous bone with smooth borders, as completely. The anatomical integrity of the
usually seen in case of slow-growing schwan- lower cranial nerves was preserved.
nomas (a, b). MRI demonstrated a large tumor After the surgery, the patient did not have
in the right jugular foramen area with a pre- new neurological deficits (o, p: postoperative
dominant intradural intracranial extension, CT scan early after surgery). He recovered
irregular shape, and inhomogeneous contrast rapidly and at discharge had just slight gait
uptake (c–e). instability and hearing loss.

Preoperative CT (a, b) and T1-weighted MRI after capsule were performed intermittently, and gradually
contrast application (c–e), demonstrating a large tumor the facial nerve (asterisks) was seen and dissected from
in the right jugular foramen area. The tumor has an the tumor (h). Gradually, the whole CP angle tumor
inhomogeneous signal on the MR images; it is predom- portion was removed. (i) The brain stem, free of tumor,
inantly located intracranially intradurally and has a and almost the entire abducens nerve up to the Dorello’s
smaller extension into the jugular foramen and the canal (6) are visible. (j) Tumor removal around the
petrous bone. Severe compression of the brain stem basilar artery. (k) The dorsal wall of the foramen has
and of the cerebellum is also visible. The bone erosion, been removed. (Tu tumor, protruding from the jugular
well seen on the bone-window CT, has the typical for foramen); the small arrowheads indicate the amount of
lower cranial nerve schwannoma’s smooth margins. bone drilling. (l) The intraforaminal tumor part was
Intraoperative images. (f) The dura has been incised then removed. (m, n) The jugular foramen, free from
along the sigmoid sinus. (g) The cerebellum is retracted tumor, prior to the placement of fat pieces to prevent
very slightly to expose only a small part of the tumor CSF leak. (o, p) Postoperative CT. The schwannoma
(Tu). (C cerebellum, D dura). Internal decompression has been removed completely via the retrosigmoid
was performed to decrease the pressure, and only after- approach. The hyperdense signal on the postoperative
ward, a larger part of the tumor was exposed. Further CT is caused by the fat pieces, placed to prevent CSF
internal decompression and dissection of the tumor leak through the petrous bone
Jugular Foramen Schwannomas 633

a b

c d

e
634 10 Jugular Foramen Tumors

f g

h i
Jugular Foramen Schwannomas 635

j k

l m
636 10 Jugular Foramen Tumors

n o

p
Jugular Foramen Schwannomas 637

Histologically, JF schwannomas are similar to hypervascular, a staged removal via two different
other intracranial schwannomas; variable nerves complimentary approaches is justified (see Case
of origin or tumor location do not modify their Illustration 10.10). Preoperative embolization of
biological characteristics. Most JF schwannomas the arterial feeders in such extensive tumors is of
are solid. Approximately 15–24 % may have cys- great help and should be an integral part of the
tic degeneration or be primarily cystic [23, 58]. management.
Patients with such tumors are usually younger In our series, all tumors were removed com-
and present with hearing loss, ataxia, headache, pletely [94] without operative mortality. Transient
and minor dysphagia. worsening of the facial nerve function was seen
Radiologically, cystic schwannomas are sub- in 19 % of patients, and transient vagus and
divided into type 1 lesions (single large cysts with hypoglossal nerve dysfunction were each seen
thin ringlike enhancement of the tumor wall) and in 6 % of patients. All cranial nerve dysfunction
type 2 lesions (multiple cysts with very irregular, had improved considerably at the last follow-up
thick enhancement of the tumor walls). Such examination.
tumors generally extend into the CP angle and The intraoperative management of cystic JF
rarely cause bony enlargement of the JF [23]. schwannomas is somewhat different. If the cyst
is opened and decompressed, it may be difficult
to recognize the plane between the tumor capsule
Management and the arachnoid membrane. The initial step of
surgery is therefore identification of the interface
The goal of surgery of JF schwannomas is their between the tumor and the surrounding struc-
complete excision—this can be achieved in tures (see Case Illustration 10.8). The arachnoid
most cases without producing new neurological plane in cystic schwannomas, which have thick
deficits. Reoperations are related to higher risks tumor capsules, can be recognized by pulling
of injury to the lower cranial nerves second- slightly the tumor capsule with a microdissector.
ary to scarring. Therefore, exceptions to com- Of note, the brain stem surface may appear very
plete removal should be made only in case of fragile after stripping off the tumor, increasing
infiltration of a given cranial nerve or absence of the risk of postoperative bleeding or neurologi-
arachnoid dissection plane. cal deficit. Patients with cystic JF schwannomas,
Type A schwannomas are approached via in contrast to those with cystic VS, have a bet-
a RS craniectomy (see Case Illustrations 10.3, ter outcome, in terms of neurological function-
10.4, and 10.6), while in types B to D tumors, ing. We have noted even hearing improvement
a single-stage combined cervical and lateral sub- in more than half of the operated patients with
occipital intra–extradural transmastoid infral- preoperative hearing loss.
abyrinthine approach is used (see Fig. 10.7 and Patients with JF schwannomas can be treated
Case Illustrations 10.5 and 10.7). In recent years, with radiosurgery or with stereotactic radiother-
such tumors are removed via an endoscope- apy—either as a primary treatment modality.
assisted intracranial or transcervical approaches Although VS and JF are histologically identical,
(see Case Illustration 10.9). In the JF, the lower they have different location and originate from dif-
cranial nerves and jugular bulb are typically dis- ferent nerves. Therefore, the results of radiosurgery
placed caudally. Infiltration of the cranial nerves of VS cannot be generalized to JF schwannomas.
is exceptional, and with meticulous microsur- The experience with JF schwannomas is relatively
gical technique, radical tumor removal can be limited, and the follow-up is not sufficient to make
achieved with minimal or no morbidity. Despite definite recommendations. Zhang et al. [123,
their benign histological structure, some schwan- 124] presented their experience with 27 patients
nomas tend to cause extensive bone destruc- with JF schwannomas treated with gamma knife
tion and to adhere intensely to surrounding radiosurgery. Their mean tumor volume was
structures. In such cases, especially if the tumor is 13.5 cm3; the mean marginal dose was 14.6 Gy
638 10 Jugular Foramen Tumors

a b

c d

e f

Fig. 10.7 This patient with a JF schwannoma type D transmastoid infralabyrinthine approach. (c) Skin incision,
(a, b: preoperative CT and MRI) was operated by the exposure (d) Complete tumor removal. (e) Reconstruction of
senior author in the late 1980s. The tumor was removed the dura (f). Postoperative CT (g)
completely using the combined cervical and extradural
Jugular Foramen Schwannomas 639

patients with JF schwannomas treated with gamma


g
knife radiosurgery over a period of 10 years.
During a mean follow-up of 3.5 years, the tumors
decreased in 8/17 patients, remained unchanged in
8/17 patients, and increased in one patient. Martin
et al. [68] presented a large series of 34 patients
with 35 tumors that underwent radiosurgery. The
mean duration of follow-up was 83 months, dur-
ing which the tumors regressed in 17 patients,
remained stable in 16, and progressed in two. The
5- and 10-year actuarial control rates were 97 and
94 %, respectively. Although the long-term tumor
control rates were favorable, the study has a more
limited value in regard to neurological outcome, in
particular the risk of new cranial nerve deficits: 22
patients (65 %) had previous microsurgical resec-
Fig. 10.7 (continued) tion, and all patients experienced various cranial
neuropathies.
and was prescribed to the 35–55 % isodose line. We reserve primary radiosurgery for elderly
The patients were followed for a mean of 38.7 medically unstable patients that are unable to
months: 44 % decreased in size, 52 % remained undergo a complex surgery due to their somatic
unchanged, and in 4 % tumor growth was observed. condition and for patients unwilling to surgical
Muthukumar et al. [75] reported on a series of 17 tumor removal.
640 10 Jugular Foramen Tumors

Case Illustration 10.4 nerve complex. Afterward, the dorsal wall


JF Schwannoma Type A (RS Approach and of the JF was opened, and the intraforaminal
Opening of the JF) tumor part was removed under microscopic
This 64-year-old patient presented with and endoscopic visualization. Complete
severe right-sided hearing loss, facial nerve tumor removal could be achieved, and the
palsy (HB grade III), and slight swallow- lower cranial nerves were preserved. After
ing difficulty. He was operated via the RS surgery a temporary deterioration of swal-
approach. The CP angle tumor part was lowing function was noted. At discharge,
first debulked and then dissected from the however, the swallowing recovered to the
brain stem and from the seventh–eighth preoperative level.

(a) CT of the PCF, demonstrating the enlarged JF on the complex was found to be compressed and dislocated supe-
right side. The foramen is with smooth well-defined riorly. (i) The tumor portion in the JF area (arrow) was
bony margins; the petrous bone is not infiltrated. (b–f) then approached. (j, k) Gradually the lower cranial
MRI of the same patient, showing a partially cystic nerves were freed (small arrow). (l) In order to gain
lesion in the JF with the imaging characteristics of a sufficient access to the intraforaminal part (arrow), the
schwannoma. (b) Native T1-weighted sequence. (c, d) dorsal wall of the JF was drilled off with diamond drill
T1-weighted sequences after gadolinium administra- (the drilled area is marked with the two arrows). (m)
tion. (e, f) T2-weighted sequences. The tumor is located Microscopic view of the JF with some remaining tumor
predominantly in the JF and extends into the posterior distally. (n) The use of an angled endoscope allowed to
cranial fossa, compressing the cerebellum. (g–r) extend the view into the lateral part of the foramen. (o)
Intraoperative images illustrating the consecutive steps The JF was further drilled off under direct endoscopic
of surgery. (g) Initial view of the tumor (Tu), following control. (p) Endoscope-controlled tumor removal. (q)
dural incision and slight superolateral retraction of the Complete tumor removal, verified with the endoscope.
cerebellum. (h) The tumor was debulked and gradually (r) Plugging of the opened JF with multiple fat pieces
dissected from the brain stem. The seventh–eighth nerve and fibrin glue—microscopic view
Jugular Foramen Schwannomas 641

a b

c d

e f
642 10 Jugular Foramen Tumors

g h

i j
Jugular Foramen Schwannomas 643

k l

m n
644 10 Jugular Foramen Tumors

o p

q r
Jugular Foramen Schwannomas 645

Case Illustration 10.5 the JF was exposed sufficiently and the lower
JF Schwannoma Type II Combined cranial nerves were seen, the tumor capsule
Cervical and Extradural Transmastoid was coagulated and internal decompression
Infralabyrinthine Approach was performed. The tumor capsule could be
This 35-year-old male patient presented with then dissected from the surrounding structures
complete hearing loss and abducens nerve and including the carotid artery, and complete
recurrent nerve palsies on the right side. He tumor removal was accomplished. A broad
had been operated initially at another institu- scar tissue layer, a sequel of the initial intra-
tion: the intracranial part of the tumor (a dural surgery, was seen to separate the JF from
schwannoma) has been removed via the RS the CP angle. This layer was free from tumor
approach. The MRI demonstrated a primarily and was not opened in order to prevent CSF
extracranial tumor remnant with imaging char- leak or injury to the intracranial structures.
acteristics compatible with a JF schwannoma Furthermore, the need of complex reconstruc-
(a–c). The lesion was approached via the com- tion could be thus avoided. The patient made
bined cervical and extradural transmastoid an uneventful recovery and was discharged 2
infralabyrinthine approach. The facial nerve weeks after surgery. The only new deficit was
was exposed and dissected in its mastoid seg- slight facial nerve palsy—HB grade 3/6 at dis-
ment and dislocated slightly anteriorly. Once charge—recovered completely at follow-up.

a b

Remnant of a schwannoma located extradurally in the area of the JF. The relationship of the tumor to the petrous
carotid artery is well seen on the axial slice (a)
646 10 Jugular Foramen Tumors

Case Illustration 10.6 and originated from the cisternal segment of


JF Schwannoma Type A (Pure Intracranial the glossopharyngeal nerve. The seventh–
Location) eighth nerve complex was slightly dislocated
This 55-year-old male patient presented with superiorly. Complete tumor removal with
an 18-month history of low back pain with preservation of the integrity of all structures
irradiation into the right leg. MRI of the lum- was achieved. A rare anatomical finding—
bar spine showed a spinal tumor, most proba- duplication of the abducens nerve—was seen.
bly a schwannoma at L2 level. MRI of the The incidence of this variant is reported to be
whole neuraxis was also performed to exclude 5–28.6 % in cadaveric studies. In this case, the
further lesions, and the CP angle lesion, pre- nerve originated at the pontomedullary sulcus
sented on the figure, was detected. The lesion as two independent trunks. A small arterial
had characteristics of a schwannoma located branch of the AICA was found to pass between
in the caudal part of the CP angle without the two nerve trunks in the vicinity of the canal
obvious penetration into the IAC (a–d). of Dorello. The possibility of this variant has
Therefore, the presumptive diagnosis was to be kept in mind when working in the area in
schwannoma of the lower cranial nerves, order to avoid nerve interruption/injury.
which was supported by the fact that the After surgery the patient had only slight
cochlear function was normal. The neurologi- hearing deficit (PTA of 30 dB). The lumbar
cal examination was entirely negative. tumor was removed uneventfully 10 days later.
The tumor was approached via the RS route The histopathological finding was schwan-
(e–l). It was located entirely in the CP angle noma (WHO grade I).

(a–d) JF schwannoma type A with pure intracranial loca- glossopharyngeal nerve (black arrow). A rare anatomical
tion. (e–g) Initial view of the tumor in the CP angle. Its variant was seen (j, k)—a duplicated abducens nerve,
relation to the seventh to eighth cranial nerves and to originating at the pontomedullary sulcus as two inde-
the lower cranial nerves (arrowhead) is well seen. (Tu pendent trunks. A small arterial branch of the AICA
tumor, P pyramid). The tumor was debulked with the was found to pass between the two nerve trunks in the
CUSA (h) and dissected from the seventh to eighth vicinity of the canal of Dorello (j, white arrow).
nerve complex. The facial nerve (FN) and the vestibu- *Vertebral artery. (l) Complete tumor removal with
locochlear nerves (VC) can be well seen (i). The tumor preserved neurovascular structures (arrowheads:
originated from the proximal cisternal segment of the glossopharyngeal nerve)
Jugular Foramen Schwannomas 647

a b

c d

e f
648 10 Jugular Foramen Tumors

g h

i j

k
l
Jugular Foramen Schwannomas 649

Case Illustration 10.7 The tumor could be removed completely


Accessory Nerve Schwannoma via the combined cervical and lateral suboc-
This 7-year-old boy presented with a diagnosis cipital intra–extradural transmastoid infral-
of an accessory nerve schwannoma. He had abyrinthine approach. The facial nerve was
been operated twice 2 years ago elsewhere, and so strongly adherent to the tumor that it had
later on a VP shunt has been implanted. The to be resected and reconstructed with sural
tumor had been removed partially, but the rem- nerve interposition graft. Besides that, the
nant showed a steady growth tendency (a–d). patient did not have any new neurological
At presentation the patient had slightly ataxic deficits. The postoperative MRI is demon-
gait, incomplete binasal hemianopsia, facial strated on figures e and f: note the fat placed
nerve paresis (grade 4/6 HB), complete hearing in the resection cavity in order to prevent
loss on the left side, and incomplete left-sided CSF leak.
accessory and hypoglossal nerve paresis.
650 10 Jugular Foramen Tumors

a b

c d

(a–d) Preoperative contrast-enhanced T1-weighted the area of the pyramid corresponds to the fat tissue
MRI of the patient. The MR venography (d) showed an used for the reconstruction and for preventing CSF
occlusion of the sigmoid sinus distal to its knee. (e, f) leak. The widened ventricular system had no clinical
Postoperative T1-weighted MRI demonstrating the significance and at follow-up proved to be stable
complete tumor removal. The high intensity signal in
Jugular Foramen Schwannomas 651

e f
652 10 Jugular Foramen Tumors

Case Illustration 10.8 The tumor was partially cystic and located
Cystic JF Schwannoma medial to the lower cranial nerves. It was
This 49-year-old male patient has been oper- accessed between the fibers of the individual
ated elsewhere 2 years ago for a tumor in the nerves and through the level below the
jugular foramen area (histopathology: WHO seventh–eighth nerve complex and the lower
grade II ependymoma). The tumor was cranial nerves. First, it was decompressed
removed partially, and radiotherapy was per- internally, and then its capsule, which was
formed. Due to radiological evidence of tumor very tightly adherent to adjacent neural struc-
growth, he underwent subsequently radiosur- tures, was carefully dissected. As pointed ear-
gery. Nevertheless, the last follow-up MRI lier, radiotherapy renders the cranial vessels
showed further tumor growth, which corre- extremely vulnerable. Utmost care was taken
lated to a progressive neurological deteriora- to preserve both the major arteries and the
tion. At presentation to our institute, he had small but vital perforators. The tumor part
hearing loss (30 dB in the main speech area), attached to the brain stem could be also
swallowing difficulty, severe gait ataxia, and a removed without violating the pial layer.
slight right-sided central hemiparesis. MR Complete tumor removal was achieved, and
examination showed a cystic tumor exhibiting all neurovascular structures were preserved.
a ring-enhancement pattern after contrast After surgery the dysphagia worsened, and
administration (a–e). The tumor was located the patient had a protracted postoperative period
medially at the brain stem in close proximity in the intensive care unit. A tracheotomy and
to the lower cranial nerves entry/exit zones. later on a percutaneous endoscopic gastros-
All potential risks, related to the previous tomy were placed. The patient suffered severe
surgery and radiation therapy, were thoroughly pulmonary embolism, as well as infectious
discussed with the patient and his family. complications, and received appropriate ther-
Considering the biological activity of the apy. Gradually his condition began to improve,
tumor, its steady growth tendency, and the and he could be transferred to a normal ward.
corresponding neurological deterioration, a Swallowing improved, and the tracheostoma
decision for an operative tumor removal was could be closed. At discharge he could walk
taken. The standard RS approach with addi- and care of himself independently. Oral feed-
tional opening of the foramen magnum was ing, however, was still impossible. The follow-
selected (f–m). up period of this patient is less than 1 month.
Jugular Foramen Schwannomas 653

a b

c
d

(a–e) Preoperative MRI of the patient, demonstrat- (g) The caudally located cystic portion was opened,
ing an intracranially located cystic lower cranial and the C1 root could be seen. (h, i) Internal decom-
nerve schwannoma. (a–c) T1-weighted sequences pression of the tumor between the fibers of the lower
after gadolinium administration. (d, e) T2-weigted cranial nerves and above the C1 root. (j, k) The
sequences. (f–m) Intraoperative images illustrating tumor capsule was adherent to the vertebral artery
the steps of tumor removal. (f) Initial view of the but could be dissected, and all perforators were pre-
tumor, located medially to the lower cranial nerves served (l). Complete tumor removal with anatomical
Tu tumor; LCN lower cranial nerves; 11 abducens preservation of all neurovascular structures in the
nerve. The C1 nerve root is hidden by the tumor. area (m)
654 10 Jugular Foramen Tumors

f g

h i
Jugular Foramen Schwannomas 655

j k

l
m
656 10 Jugular Foramen Tumors

Case Illustration 10.9 cranial nerves could be dissected from the


JF Schwannoma Removed via a Retrosigmoid tumor and preserved anatomically. Then, the
Endoscope-Assisted Approach tumor capsule was dissected from the brain
Thirty-two-year-old female patient with right- stem, from the basilar, and the right vertebral
sided hearing loss and facial nerve palsy (HB arteries. The intraforaminal tumor part could
grade IV), swallowing difficulty, and gait insta- be well demonstrated with the angled endo-
bility presented with radiological evidence of a scope and removed under pure endoscopic
large jugular foramen tumor. MRI demonstrated control with appropriate angled instruments.
a very large tumor in the area of the jugular Due to the wide dilatation of the jugular
foramen with a major intradural part and with foramen, the endoscope allowed not only to
significant extracranial extension (a–g). The visualize but also to remove the extracranial
CT and bone-window CT (a–c) demonstrated tumor part completely. The tumor cavity was
dilated jugular foramen and extensive erosion of initially coated with Gelfoams and then filled
the adjacent petrous bone. The signal character- with multiple fat pieces, which were sealed
istics of the tumor, the intense contrast uptake, with fibrin glue. The postoperative period
and the smooth bony margins were compatible was unremarkable. The swallowing difficulty
with those of schwannoma. persisted, and hoarseness appeared. A slight
Considering the degree of intracranial tumor tendency toward improvement of facial nerve
growth and the compression of brain stem, the function was noted before discharge. At 1-year
RS approach was selected. Initially, the dura follow-up examination, she had an HB grade
dorsal to the jugular foramen was excised, III facial nerve function; the gait stability
and the roof of the foramen was drilled off, recovered completely. She could eat normally
thereby exposing the intraforaminal tumor but reported to have occasional swallowing
part. Internal decompression and dissection of problems, especially with hard food. MRI
the tumor capsule were performed intermit- study was unremarkable and did not demon-
tently in all directions. The seventh–eighth strate tumor residual/recurrence.
Jugular Foramen Schwannomas 657

a b

c d

(a–g) Preoperative CT and MRI of the patient: a large T2-weighted MR images. (h–n) Early postoperative MRI
jugular foramen tumor with intra- and extracranial exten- performed 5 days after surgery. The heterogeneous signal
sion is visible. Substantial erosion of the petrous bone on the various sequences is caused by the Gelfoam, fat
with smooth well-defined bony margins, and no evidence tissue, and the typical radiological artifacts, appearing
of bone infiltration can be seen on the bone-window CT early after surgery. (h, i) CT scan. (j–l) T1-weighted MR
images. The signal characteristics of the tumor and the sequence after gadolinium application. (m, n)
pattern of bone erosion are highly suggestive of schwan- T2-weighted sequences. The ring enhancement does not
noma. (a–c) CT, including bone-window mode. (d, e) reflect tumor remnant, as evident when the corresponding
T1-weighted MR image after contrast application. (f, g) T2-weighted signal characteristics are evaluated
658 10 Jugular Foramen Tumors

e f

g h

i j
Jugular Foramen Schwannomas 659

k l

m n
660 10 Jugular Foramen Tumors

Case Illustration 10.10 vascularized. Still, it could be decompressed


Giant JF Schwannoma Removed in Two internally, and its lower part could be removed.
Steps The strong bleeding from the tumor prohibited
This 15-year-old male patient presented with a more radical surgery.
history of hearing loss on the right side for 3 Prior to the second surgery, which was
years and occasional swallowing difficulty. done 2 weeks after the first one via the ret-
Neurological examination revealed right-sided rosigmoid approach, superselective catheter-
hypacusis (20 dB in the main speech area), pare- ization and embolization of the tumor feeders
sis of the right hypoglossal nerve, and a slight were performed (k–n). Despite the emboliza-
deviation of the soft palate to the left side with a tion, the lesion was still strongly vascularized.
weak gag reflex. CT and MRI studies demon- Following thorough internal debulking, the
strated a huge skull-base lesion, extending from intradural part could be dissected from the
the jugular foramen to the sphenoid sinus (5.3 × brain stem and cranial nerves and removed
4.6 × 5.2 cm in diameter) with signal character- completely. Then, the exposure of the petrous
istics of schwannoma (a–h). The tumor caused tumor part was enhanced by additional bone
extensive destruction of the skull base and had removal around the IAC and by resection of
large intradural and extracranial portions. the petrous apex. Complete tumor removal
Considering the size and the extension of was achieved using microscopic and endo-
the tumor, as well as the relatively good neuro- scopic technique, and all cranial nerves could
logical condition of the patient, a decision was be preserved anatomically. The nerve of ori-
taken for a staged tumor removal via two dif- gin of this tumor could not be identified with
ferent approaches. At the first stage, the tumor certainty. The exposed bone of the pyramid
was approached via the transmastoid presig- was covered with fat pieces, fixed with fibrin
moid approach (i, j). A retroauricular retro- glue, in order to avoid CSF leak, while the
mastoid skin incision was done and extended huge cavity was filled with multiple sponges
caudally; the stylomastoid foramen and the (Gelita-s) and Tabotamp (o, p).
facial nerve were identified; a mastoidectomy Histopathological examination proved that
was performed, the proximal facial nerve seg- the tumor was a WHO grade I schwannoma.
ment was identified, and the sigmoid sinus was After surgery the lower cranial nerve dysfunc-
exposed up to the jugular bulb in the jugular tion was more pronounced but showed gradual
foramen. Thus, sufficient portion of the tumor improvement, and within 1 week peroral feed-
was exposed to allow its safe removal. The ing was initiated. The hearing on the right side
tumor turned out to be extremely and strongly was completely lost.
Jugular Foramen Schwannomas 661

a b

c d

(a–h) Giant skull-base tumor, extending in the posterior angiography demonstrating the hypervascular tumor
and middle cranial fossae, with extensive bone destruc- and the feeders, coming from the ascending pharyngeal,
tion, as visible on the CT images (a, b). (c) T1-weighted medial meningeal, and accessory meningeal arteries
native image. (d, e) Axial T1-weighted images after (k, l). The petrous segment of the ICA was narrowed and
contrast application. (f) Axial T2-weighted image. (g) dislocated. The occlusion test showed sufficient blood
Coronal and sagittal. (h) T1-weighted images after supply from the contralateral anterior and posterior com-
contrast application. Note the severe brain stem and municating arteries. (m, n) Only partial embolization of
temporal lobe compression. (i, j) Bone-window CT the feeding arteries was possible. (o, p) CT scans, per-
demonstrating the amount of petrous bone resection formed 1 day after the last surgery. The tumor cavity is
and the approach to the tumor. (k–n) Digital subtraction filled with multiple sponges (Gelita-s) and Tabotamp
662 10 Jugular Foramen Tumors

e f

g h

i j
Jugular Foramen Schwannomas 663

k l

m n

o p
664 10 Jugular Foramen Tumors

Hypoglossal Nerve Schwannomas correlation between these tumors and the para-
ganglion tissue described by Guild [90]. In 1962,
In 1933, De Martel et al. [111] first reported a Alford and Guilford classified these lesions as glo-
hypoglossal nerve schwannoma. These tumors mus tympanicum and glomus jugulare tumors [3].
can be completely intracranial, intracranial– Glomus tumors, which are also called paragan-
extracranial, or completely extracranial [112]. gliomas, chemodectomas, and nonchromaffin
Symptoms differ depending on the location tumors, are relatively rare: they represent 0.012 %
of the tumor. Tongue wasting is present in the of all tumors diagnosed in humans [60]. Four typ-
majority of patients at the time of presentation. ical primary locations are known in the head and
Suboccipital headaches, exacerbated by neck neck region: middle ear cavity (glomus tympani-
movements, are early symptoms of intracranial cum), dome of the jugular bulb (glomus jugulare),
tumors [11]. Patients with intracranial tumors ganglion nodosum of the vagus nerve (glomus
may also exhibit jugular foramen syndrome or vagale), and the carotid body near the carotid
symptoms of brain stem compression. Patients bifurcation (carotid body tumor). Glomus tumors
with extracranial tumors may present with a are the second most common temporal bone tumor
mass in the upper neck or parapharyngeal area, in after VS. Glomus jugulare tumors comprise
addition to wasting of the tongue [112]. Surgical 60–80 % of all primary JF tumors [10, 71].
strategies depend on tumor location. Preoperative Females are affected more frequently than male
angiography is important to establish the position persons [3, 110] with peak age of presentation
and dominance of the vertebral arteries and the being the fourth–fifth decade. It has been esti-
anatomic features of the venous sinuses. In the mated that up to 20 % of glomus tumors are famil-
immediate postoperative period, the possibility ial. Familial tumors are multicentric or bilateral in
of aspiration attributable to paresis of CNs IX approximately 25–78 % of the cases [119].
and X must be considered [117]. The glomus jugulare tumors are histologically
benign lesions, which rarely grow rapidly and are
usually encapsulated. Still, if untreated, they are
Glomus Jugulare Tumors related to high morbidity and mortality rates.

Glomus jugulare tumors have long fascinated


surgeons and are still regarded as one of the most Pathogenesis and Classification
challenging skull-base lesions. The advancement
in microsurgical techniques and intensive care Glomus jugulare tumors arise from paraganglia
management; the improvements in neuroradiol- cells situated near the jugular foramen. The para-
ogy, in particular the use of selective preoperative ganglion tissue is regarded as part of the extra-
embolization; and the team approach allowed to adrenal neuroendocrine system. In the temporal
reduce significantly the operative morbidity and bone, the vast majority of paraganglia are found
to achieve good long-term outcome. within the adventitia of the dome of jugular bulb
(glomus jugulare) and over the promontory in the
middle ear (glomus tympanicum). They can be also
General Features found along the course of the glossopharyngeal
nerve, the Jacobson nerve (the tympanic branch of
The first descriptions of glomus tissue are credited the glossopharyngeal nerve), or the Arnold nerve
to Valentin in 1840 and Krause in 1878 [12]. The (the auricular branch of the vagus nerve) [18, 43].
carotid body-like structures in the temporal bone Histologically, paragangliomas in all locations are
were described in 1941 by Guild who named them similar.
“glomus jugulare bodies” [42]. Three years later— Familial glomus tumors show an autosomal
in 1945—Rosenwasser published the first descrip- dominant inheritance pattern with maternal
tion of a middle ear paraganglioma and found a imprinting effect: the trait is transmitted from the
Glomus Jugulare Tumors 665

father to his offspring. Two putative regions on Table. 10.2 Fisch classification of glomus tumors of the
chromosome 11—PGL1 at 11q23.1 and PGL2 at temporal region
11q13.1—contain the candidate genes [47]. It Class Description
has been shown that both of these loci are associ- A Tumors limited to middle ear space
ated with maternal imprinting [46]. Genetic B Tumors limited to middle ear space or mastoid
w/o involvement of the infralabyrinthine space
screening of high-risk individuals in affected
of the temporal bone
families allows early detection and management C Tumors involving the infralabyrinthine and
of multifocal or bilateral disease [101]. apical spaces, w/ extension into the apex
Glomus jugulare tumors arise in the lateral D1 Tumors w/ intracranial extension < 2 cm
skull base and expand along the paths of least D2 Tumors w/ intracranial extension > 2 cm
resistance. Although histologically benign, they
are locally invasive and may invade adjacent bone,
blood vessels, dura mater, and cranial nerves. Table 10.3 Glasscock–Jackson classification of glomus
jugulare tumors [54]
They may cause destruction of the pyramid,
including the labyrinth into the hypotympanum, Grade Description
mesotympanum and sinus tympani, the juguloca- I Small tumor involving jugular bulb,
middle ear, and mastoid
rotid spine, and the hypoglossal canal [87]. These
II Tumor extending under internal
tumors may grow intracranially and penetrate the auditory canal; may have intracranial
dura of the posterior cranial fossa or extend along extension
the cranial nerves around the jugular bulb, infe- III Tumor extending into petrous apex;
rior petrosal sinus, and carotid artery. More rarely, may have intracranial extension
they penetrate the dura of the middle cranial fossa IV Tumor extending beyond petrous
bone into clivus or infratemporal
[55, 99]. Extracranial extension into and/or along
fossa; may have intracranial extension
the jugular vein may cause obstruction of the
venous drainage or produce a pharyngeal or neck
mass. the tumor (Fig. 10.8e). The tumor enhances
This intimate association of glomus jugulare intensely after gadolinium administration and
tumors to the lower cranial nerves and inner ear lacks dural tail sign.
structures, the involvement of major vessels, the Glomus jugulare tumors are predominantly
hypervascularity of the tumor, as well as its pro- supplied by the external carotid artery system,
pensity for intracranial extension, are the main mainly by the ascending pharyngeal artery, fol-
factors complicating operative removal [29, 47]. lowed by the posterior auricular, stylomastoid,
The currently most widely applied classifi- and occipital branches. Less frequently, it has
cation schemes reflect this complex extension feeders from the internal carotid artery (caroti-
patterns (Tables 10.2 and 10.3). cotympanic branches of intrapetrous carotid) or
the vertebrobasilary system (posteroinferior cere-
Neuroimaging bellar artery) [85]. The conventional angiographic
The bone-window CT (Fig. 10.8a) shows skull- appearance is of a hypervascular mass with an
base infiltration and destruction that cause an intense characteristic tumor “blush,” which is
irregular “moth-eaten”-like erosion pattern and coarser and less pronounced than in meningiomas.
enlargement of the JF [63, 79, 100]. On Early draining veins, representing early arterio-
T1-weighted MR images, a typical salt-and-pep- venous shunting, are seen. Angiography demon-
per appearance is seen with hyperintense foci, strates the relationship of the tumor to the carotid
which represents serpentine flow void pattern or artery, its vascularity, and feeding vessels. The
hemorrhagic foci (Fig. 10.8b–d) [63, 78, 85]. On carotid artery may be encased, narrowed, or even
T2-weighted sequences, the tumor is hyperin- occluded. The venous phase study provides infor-
tense and shows flow voids within the mass, cor- mation regarding the patency of the sigmoid sinus,
responding to regions of high blood flow within jugular bulb, and jugular vein, as well as on the
666 10 Jugular Foramen Tumors

size and dominance of the contralateral trans- Clinical Presentation


verse–sigmoid sinuses.
MR angiography and MR venography that The clinical presentation of glomus jugulare
have been implemented are less invasive tools to tumors is related to their compressive effect, local
study the vascular supply of the tumor and the invasiveness, and high vascularity. Typical signs
jugular vein intraluminal patency [79]. In one and symptoms are hearing loss, pulsatile tinnitus,
study, 3D time-of-flight MR angiography showed and lower cranial nerve palsies. The most com-
a sensitivity and specificity of 90 and 92 %, mon presenting sign is unilateral conductive hear-
respectively, when compared with the digital ing loss, typically caused by invasion of the middle
substraction angiography [118]. ear, followed by pulsatile tinnitus secondary to the

a b

c d

Fig. 10.8 Glomus jugulare tumor. CT images. Native T1-weighted contrast-enhanced images. (i) T2-weighted
(a). After contrast administration (b). Bone-window scan image. (j, k) MR angiography
(c). MR imaging. (d, e) T1-weighted images. (f–h)
Glomus Jugulare Tumors 667

e f

g h

i j k

Fig. 10.8 (continued)


668 10 Jugular Foramen Tumors

tumor’s rich vascularity. Lower cranial nerve dys- these modalities. Initial observation or “watchful
function is seen in patients with large tumors. waiting” may be another option. The known pro-
Notably, slowly developing palsies can be well pensity of glomus tumors to grow and cause symp-
compensated by the patients. Other symptoms are toms, however, restricts its application to some
determined by the tumor growth pattern and the selected cases, for example, moribund asymptom-
direction of extension. The facial nerve is affected atic patients. A more proactive approach is gener-
most commonly (in up to 33 %), followed by the ally favored, and surgery remains the mainstay of
vagus, glossopharyngeal, hypoglossal, and spinal treatment [2, 28, 85, 100].
accessory nerves [66]. Compression of surround- The current multimodal management of glo-
ing neural structures may cause nystagmus, ataxia, mus jugulare tumors is aimed at improving local
gait instability, and—in more advanced cases— tumor control and minimizing the resultant mor-
hemiparesis or symptoms of increased intracra- bidity. It includes preoperative embolization of
nial pressure. Large tumors with anterior extension feeding arteries and judicious microsurgical tumor
may encase or even invade the walls of the carotid resection with attempted preservation of neuro-
artery, causing Horner’s syndrome. logical functions. In most patients with confined
Glomus tumors are known to produce vasoac- glomus jugulare tumors, the goal of complete
tive substances. Clinically significant hormonal tumor resection with preservation of function is
secretion occurs in 1–4 % of the patients [18, 52]. feasible. In case of infiltration of the cranial nerves
These could lead to potentially serious complica- or brain stem, subtotal tumor resection is per-
tions, such as severe hypertension crisis. If the formed. Similarly, in case the carotid artery is
patient presents with signs suggestive of a hor- infiltrated, subtotal resection is preferred to carotid
monally active tumor, such as labile hyperten- resection and reconstruction. Tumor remnants
sion, tachycardia, and facial flushing, a thorough that have a propensity to grow at follow-up are
evaluation is mandatory. In case the level of subjected to radiosurgery/radiotherapy.
secretion of vasoactive substances is too low, it Patients with more extensive glomus jugulare
may not be recognized clinically. Therefore, tumors pose specific challenges. In such cases,
some authors propose screening for excess cate- the neurological condition of the patient at pre-
cholamine secretion in all patients with glomus sentation is of paramount importance for the
jugulare tumors [2]. Twenty-four-hour urine decision-making process. The attempt of radical
sample and the plasma should be analyzed for surgery is inevitably related to a very high risk of
catecholamines, metanephrines, and normeta- injury to the lower cranial nerves. Surgery is
nephrines. Patients in whom the catecholamine therefore recommended only in patients present-
level is four times higher than normal require pre- ing with compromised lower cranial nerve func-
operative prophylaxis, usually with an alpha and tion. Due to the phenomenon of brain plasticity,
beta catecholamine blocker. Adrenal imaging during the gradual loss of ipsilateral function, the
should be performed in patients with hyperse- contralateral nerves take over the control of swal-
creting tumors in order to exclude coexisting lowing. Such patients are not so endangered from
adrenal norepinephrine-secreting tumors. aspiration and the related complications. In con-
trast, an acute lower cranial nerve dysfunction,
for example, occurring at surgery, can be gener-
Management ally poorly compensated for. Surgery in patients
with normal swallowing function should be per-
General Management Principles formed only in case of significant brain stem
The optimal treatment of glomus jugulare tumors compression, and its goal should be merely
is still a matter of debate. The options are microsur- decompression of neural structures. We believe
gical resection, tumor embolization, radiosurgery/ that the treatment of choice of such patients is
stereotactic radiotherapy, conventional fractionated embolization, which generally ensures tumor
external beam radiotherapy, or a combination of growth control or even leads to tumor shrinkage.
Glomus Jugulare Tumors 669

If the tumor demonstrates further growth later on, presented a series of four patients operated with a
the procedure may be repeated. Glomus jugulare similar technique [35]. In 1966, Denecke reported
tumors that cannot be controlled with this method on the further improvement of the surgical tech-
should be treated with radiosurgery or radiother- nique, allowing its application in more extensive
apy, depending on tumor size and extension. lesions [25]. In 1968, House introduced the com-
Radiation therapy causes slow worsening of bined neck and simple mastoidectomy technique
lower cranial nerve function over many month or [51], while Portmann advocated the team approach
years that enables the development of compensa- (ENT and neurosurgeon) and surgery in two
tory mechanisms. stages [61]. Kempe et al. described the successful
use of suboccipital craniectomy and standard
Surgery of Glomus Jugulare Tumors mastoidectomy in tumors involving both the tem-
The first exploration of the jugular bulb was poral bone and the posterior fossa [59]. Similar
reported by Seiffert in 1934. Early attempts for a technique with additional exposure of the internal
systematic surgical approach to glomus jugulare carotid artery via the glenoid fossa was reported
tumors were made in 1941–1945 by Guild and by Hilding and Greenberg [49]. Glasscock et al.
Rosenwasser [91]. In 1951, Semmes performed [39], in 1974, described an approach which com-
the first successful neurosurgical removal of a bined the wide exposure of the skull base of
glomus jugulare tumor via a suboccipital approach Shapiro and Neues and the use of the extended
and reported it in 1953 [102]. Capps reported on facial recess, described by House. Gardner et al.
five cases of successfully operated tumors and [34] presented their concept of multidisciplinary
described some important technical advances in cooperation in 1977. Their technique consisted of
1952 [21]. Due to the high morbidity/mortality three steps: exposure of the skull base through the
rate of these early surgical attempts, alternative neck, bone removal within the temporal bone and
management options have been developed in the jugular fossa, and tumor removal/reconstruction
1950s. Williams et al. [122] reported on the Mayo of the wound.
Clinic experience with radiotherapy of glomus In the late 1970s and early 1980s, new
tumors in 1955, and this method became tempo- approaches have been described by Graham,
rarily the mainstay of treatment. In the same Gardner, Fish, Menzel, Draf, and the senior author
period, neuroradiology made considerable prog- [93, 95]. In 1978, U. Fisch described an approach
ress, mainly due to the work of Valvassori. In that allowed accessing and controlling the inter-
1960, Meacham and Capps [70] and Thoms et al. nal carotid artery within the temporal bone—the
[115] described their experience with removing infratemporal approach [29]. Thus, even larger
glomus jugulare tumors via a suboccipital route, glomus jugulare tumors could be safely resected.
which actually revived the surgical management The classic infratemporal fossa type A approach
of these tumors. They used the suboccipital [30] affords an excellent exposure of the jugular
approach and found that tumor extension deter- bulb area. It consists of a cervical approach with
mines the radicality of surgery. Since then, multi- control of the carotid artery and jugular vein, a
ple surgical approaches to the JF have been mastoidectomy with skeletonization of the jugular
developed and refined with the goal of more com- bulb, closure of the external auditory canal, and
plete and safe tumor resection [2, 14, 36, 62, 100]. anterior rerouting of the facial nerve. Later on—in
In 1964, Shapiro and Neues [104] reported on the 1987—Al Mefty et al. [1] described a combined
surgical resection of a recurrent glomus jugulare infratemporal and posterior fossa approach for
tumor with minimal blood loss and good neuro- the removal of giant glomus jugulare tumors that
logical outcome. Complete tumor resection was have a large intracranial component.
possible by removing the jugular bulb and trans- The anterior transposition of the facial nerve,
location of the facial nerve. One year later, Gejrot as described by Capps [22] and refined later by
670 10 Jugular Foramen Tumors

several authors [16, 30, 54], provides wide expo- suboccipital approach may be sufficient: the
sure and early control of the internal carotid intraforaminal tumor part can be removed by
artery; however, the cost is temporary or even per- drilling the wall of the jugular foramen or by
manent facial weakness. A major issue related to applying the angled endoscope. In case of tumors
these extended lateral approaches is the safe man- with predominant extracranial extension, the cer-
agement of the facial nerve and of the lower cra- vical approach is usually sufficient. The naviga-
nial nerves during the surgical procedure without tional guidance may be of great help, especially
compromising the exposure of the surgical mass. if the tumor has destroyed the normal anatomical
Glomus jugulare tumors with intracranial exten- landmarks (Fig. 10.11).
sion have been resected via the MFA, subtempo-
ral–infratemporal transmastoid, retrolabyrinthine, Embolization
translabyrinthine, transcochlear, transcondylar, Parallel to the advancement of surgery, major
juxtacondylar, or combined approaches [14, 28, progress has occurred in the endovascular embo-
55, 80, 85, 100]. The reported rate of gross total lization techniques of glomus jugulare tumors,
tumor removal in large clinical series is 77–92 %, promoted mainly by Hilal, Michelsen, and
depending on the tumor’ stage. The main source Lajaunas. Hilal and Michelsen [48] and Brismar
of morbidity after glomus jugulare surgery is and Cronqvist [17] first described the preopera-
lower cranial nerve dysfunction [2]. tive superselective embolization of major feeding
The ideal operative technique should allow for vessels of glomus jugulare tumors in separate
control of the carotid artery, coagulation or liga- publications. In 1979, Simpson et al. [107]
tion of the afferent vessels, proximal control of reported the use of preoperative embolization in
the sigmoid sinus and internal jugular vein, glomus jugulare tumors in an effort to reduce
identification and preservation of the lower cra- intraoperative blood loss. Since then, its efficiency
nial nerves, and possibility for reliable dural clo- in reducing intraoperative bleeding, facilitating
sure and reconstruction of the operative area complete surgical resection, and shortening oper-
[48]—Fig. 10.9. On the other hand, of paramount ative time has been well established (see Case
importance is the avoidance of significant Illustration 10.11) [84]. Embolization is an essen-
approach-related morbidity. With increasing tial component of the current management of
experience, it became obvious that the achieve- large and complex glomus jugulare tumors (see
ment of best outcome requires individually tar- Case Illustration 10.11). The recommended delay
geted approach selection. between embolization and surgery should be at
In complex glomus jugulare tumors with cra- least 2–3 days to allow embolization-related
nial and cervical extension, we favor the combined edema to subside but no longer than 2–4 weeks
cervical and lateral suboccipital intra–extradural because of the gradual recanalization of feeding
transmastoid infralabyrinthine approach, as descri- vessels. Of note, some adverse effects of embo-
bed earlier (Figs. 10.10 and 10.12). lization on the complexity of surgery have been
A major recent advancement was the intro- reported: the vessels might be more susceptible
duction of the endoscope as an adjuvant visual- to injury, and the tumor might be hardened and
ization tool. With the endoscope-assisted therefore more difficult to remove.
technique or with pure endoscopy, the tumor por-
tion in the jugular foramen can be removed under Radiotherapy/Radiosurgery
direct visual control. Thus, in tumors with lim- Glomus jugulare tumors have been increasingly
ited intracranial extension and no adhesion to frequently treated in the last decades with frac-
neural/vascular structures, the need of additional tionated radiotherapy, radiosurgery, or stereotac-
craniotomy can be avoided. In tumors with pre- tic radiotherapy. These treatment modes have
dominant intracranial extension, the retrosigmoid been applied as primary management, as an
Glomus Jugulare Tumors 671

a b

c
d

Fig. 10.9 Giant glomus jugulare tumor, operated by the tumor feeders and to have proximal control of the carotid
senior author in 1980s. The CT scan (a) shows the extent artery (g, h). (i) Complete tumor removal with preserva-
of petrous bone erosion. (b, c) Preoperative angiography. tion of the carotid artery. (j) Skin closure. (k) Postoperative
The tumor was visible at pharyngoscopy (d) and otoscopy angiography—intact carotid artery. (l) The patient has
(e). At that time, embolization of the tumor was not fea- been followed up for almost three decades and had no
sible, what necessitated wide neck dissection to access the signs of recurrence
672 10 Jugular Foramen Tumors

e h

Fig. 10.9 (continued)


Glomus Jugulare Tumors 673

adjunct to surgery, or as a salvage therapy in case


k
of recurrences [121]. The goal of radiotherapy/
radiosurgery is obtaining tumor growth control
and providing symptom relief. Glomus tumors
are regarded as radioresistant, and the radiation
is supposed to control tumor growth by
influencing the highly developed vasculature of
the tumors.
Multiple clinical studies on glomus tumors
treated primarily with radiosurgery described
high rates of tumor control with only rare cases
of tumor progression [15, 33, 72]. Stereotactic
radiation for glomus jugulare tumors is generally
well tolerated. In contrast to VS and menin-
giomas, in case of glomus tumors, a symptomatic
improvement, especially of tinnitus and otalgia,
l can be achieved [28]. The reported tumor control
rates vary between 71 and 100 %, and the symp-
tom control rate is 80–100 %.
No study reported, however, long-term out-
come results—the mean follow-up period was
up to 60 months [32, 72, 83, 106]. Recurrences
of glomus jugulare tumors can arise up to 25
years after the initial treatment. Radiotherapy,
as stated previously in the text, aims at achiev-
ing tumor growth control. It does not affect the
“chief” cells, which keep the tumor alive [45].
Further drawback is that the catecholamine
secretion is not affected by the application of
radiation. Therefore, we consider radiosurgery
as a primary management option in patients
with advanced age and with contraindication
for other therapy.
Radiation therapy may be an adjunctive
option in the rare aggressive glomus jugulare
tumors. Its role in patients with extensive tumors
and normal lower cranial nerve function has
been described earlier in this chapter. As salvage
therapy, radiosurgery/radiotherapy is an accept-
able option in case surgery of the recurrence is
deemed too risky.
Fig. 10.9 (continued)
674 10 Jugular Foramen Tumors

a b

Fig. 10.10 Glomus jugulare tumor (a, b), removed com- (c) Cervical exposure. (d) Cranial part of the approach. (e)
pletely via the combined cervical and lateral suboccipital The tumor has been removed. (f) Tumor specimen. (g)
intra–extradural transmastoid infralabyrinthine approach. Postoperative audiogram
Glomus Jugulare Tumors 675

c d

e f

Fig. 10.10 (continued)


676 10 Jugular Foramen Tumors

a b

Fig. 10.11 Neuronavigation screenshots made during the resection of a glomus jugulare tumor. The position of the
pointer tip demonstrates the extent of tumor removal
Glomus Jugulare Tumors 677

a b

c d

Fig. 10.12 Large glomus jugulare tumor, operated by the mastoid muscle and posterior to the ear. (f) Exposure of the
senior author in 1992 (a) CT scan. (b) MR image after con- mastoid, neck dissection, and exposure of the cervical ves-
trast administration. Initially, tumor embolization was per- sels and nerves. (g) Retrosigmoid craniectomy and mas-
formed (angiography before (c) and after the embolization toidectomy have been performed, the sigmoid sinus are
(d)), and then the combined cervical and lateral suboccipi- well visible. (h) The tumor is exposed intradurally. (i)
tal intra–extradural transmastoid infralabyrinthine approach Resection of the jugular vein with the tumor. (j) Complete
was used to expose and remove the tumor completely. (e) tumor removal. (k) Watertight reconstruction of the dura.
Skin incision along the anterior border of the sternocleido- Postoperative imaging: (l–o) CT scans. (p) MR image
678 10 Jugular Foramen Tumors

e f

g h

i j

Fig. 10.12 (continued)


Glomus Jugulare Tumors 679

k n

m
p

Fig. 10.12 (continued)


680 10 Jugular Foramen Tumors

Case Illustration 10.11 A decision was then taken to attempt tumor


Glomus Jugulare Tumor (Embolization resection via the RS route. Once the skin was
and Surgery) incised, the subcutaneous air collection was
This 42-year-old male patient presented with encountered—the air penetrated through
a history of pulsatile tinnitus in the left ear multiple small holes of the underlying bone.
since 20 months and retroauricular pain since Typical retrosigmoid approach was per-
3–4 months. One month earlier, spontaneous formed, and a jugular foramen tumor that
subcutaneous emphysema behind his left ear extended in the CP angle and in the IAC was
appeared. Neurological examination revealed seen. Taking care not to injure the com-
slight ataxia and decreased hearing on the left pressed seventh and eighth cranial nerves,
side—more than 60-dB loss in the main speech internal debulking was performed. Then, the
area. CT and MR examinations demonstrated dura of the pyramid was excised, and the pet-
a tumor with irregular shape in the area of the rosal tumor part, extending from the jugular
left jugular foramen and bone destruction of foramen inferior to the petrous carotid artery
the adjacent petrous bone (a–j). The epidural anterolaterally and to the petrous apex medi-
emphysema was clearly visible. The tumor ally, was exposed and removed via an
showed strong and homogeneous contrast inframeatal approach (described in the chap-
uptake after administration of gadolinium (a, ter). The posterior and inferior walls of the
b, e, f). Interestingly, neither the MRI nor the IAC were then also drilled off, and the intra-
angiographic characteristics (j) of the tumor canalicular tumor was dissected from the
did correspond to that of a typical glomus nerves and removed. Complete tumor
tumor. removal was thus achieved, and the bone
Superselective embolization of tumor feed- opening was filled with multiple fat pieces
ers was performed (k–p), and 1 week later, the sealed with fibrin glue (r–u: postoperative
tumor was approached via a transmastoid MRI control).
approach with the goal of obtaining a tissue After surgery the patient had a swallowing
sample and—respectively—of a definitive difficulty that, however, improved within 3
histopathological diagnosis (q). The lesion days, and normal peroral feeding was started.
proved to be a glomus jugulare tumor. Hearing level remained unchanged.

(a–j) Preoperative CT and MR images showing the occipital artery, from the petrosal branch of the middle
jugular foramen tumor. The main tumor part is extra- meningeal artery, and from the posterior auricular
dural, but an intradural part and an extracranial artery. These vessels were selectively catheterized
parapharyngeal extension are visible. Note the tumor with microcatheters, and the tumor was embolized
relation to the carotid artery (g–j). (a, b) Bone-window with polyvinyl alcohol. (m–p) Control angiography
CT demonstrating the extensive petrous bone destruc- after the embolization showing substantial devascular-
tion and the air collection. (c–i) T1-weighted sequences ization of the tumor. (f) Venous phase. (q) Bone-
after gadolinium administration. (j) MR angiography. window CT, performed after the transmastoid biopsy
(k–p) Preoperative DSA demonstrating the highly approach. (r–u) Postoperative MR images demon-
vascular tumor and its feeders. Main vascular supply strating the complete removal of both the intra- and of
comes from the meningeal branch of the ascending the extracranial tumor parts. (r–t) T1-weighted
pharyngeal artery, from the stylomastoid branch of the sequences. (u) T2-weighted sequence
Glomus Jugulare Tumors 681

a b

c
d

e
f
682 10 Jugular Foramen Tumors

g h

i j
Glomus Jugulare Tumors 683

k l

m n
684 10 Jugular Foramen Tumors

o p

q
Glomus Jugulare Tumors 685

r s

t u
686 10 Jugular Foramen Tumors

Meningiomas of the Jugular Foramen CT, MRI, and MR angiography, including the
MR venography. On CT, meningiomas appear as
General Features slightly hyperintense masses. The surrounding
bone structures have relatively preserved archi-
Meningiomas of JF are either primary or tecture, but occasionally hyperostosis/sclerotic
secondary—extending to the JF from adjacent changes of the JF are seen [38]. Meningiomas, in
areas. Primary jugular foramen meningiomas are contrast to schwannomas and glomus tumors,
very rare and are estimated to comprise 4–9 % of all may have calcifications [63].
posterior fossa meningiomas [4, 89, 97]. They are JF meningiomas are typically iso- to hypoin-
considered to arise from the arachnoid-lining cells tense on T1-weighted MRI and have intermediate
of the jugular bulb within the jugular fossa [73, 97]. to increased signal intensity on T2-weighted images,
JF meningiomas are usually benign and have a depending on histology composition [65, 97]. No
slow insidious growth. These tumors occasionally flow voids are noted. The tumor enhances strongly
are locally aggressive and infiltrate the temporal after contrast application. Meningiomas may have
bone and nerve tissue [6, 73]. From the JF area, the a dura tail, which is key feature for their differen-
tumor tends to extend superior into the CP angle, tial diagnosis [38, 65].
inferior toward the foramen magnum or medially, The venous flow and the patency of the sig-
causing brain stem compression. It may further moid sinus and jugular vein can be reliably evalu-
invade the temporal bone and middle ear or extend ated on MR venography. Meningiomas, as well
extracranially to the neck region. The extracranial as glomus jugulare tumors, tend to infiltrate the
extension is usually limited to the carotid space, jugular vein; in contrast, schwannomas usually
and carotid encasement and/or jugular vein occlu- only compress it [73, 89, 114]. DSA and embo-
sion may occur [38]. The classification, presented lization of the arterial feeders might be consid-
by Bakar [6], which is based on the degree of ered in selected highly vascular tumors.
tumor extension, clearly illustrates the great vari- Primary and secondary JF meningiomas have
ability of JF meningiomas: different growth pattern that is reflected by the neu-
• Type I—primarily within the JF roimaging findings. Those with secondary exten-
• Type II—primarily extension to the CP angle/ sion into the JF are usually solid, well-circumscribed
intracranially sessile lesions with a broad dural base. Their gross
• Type IIa—extension to the CP angle/intracranially appearance is en plaque or globular (Fig. 10.13).
and middle ear Meningiomas arising primarily within the JF have a
• Type III—primarily extension to the neck more infiltrative growth pattern with extension into
• Type IIIa—extension to neck and middle ear the surrounding skull base and/or protrusion into
• Type IV—extension to the CP angle/intracra- the skull-base foramina (see Case Illustration 10.12)
nial region and neck (dumbbell shape) [38, 73]. In large tumors, however, it is impossible
• Type IVa—extension to the CP angle/intracranial to differentiate between primary and secondary JF
region, neck, and middle ear meningiomas (Fig. 10.14). Some meningiomas
tend to infiltrate centrifugally the surrounding skull
base in all directions [65]. MacDonald et al. [65]
Neuroimaging presented five patients with late diagnosis: in all of
them, the posterior fossa, middle ear cavity, inter-
The comprehensive evaluation of patients with JF nal auditory canal, hypoglossal canal, and carotid
meningiomas includes thin-slice bone-window space were involved.
Meningiomas of the Jugular Foramen 687

a b

c d

e f

Fig. 10.13 Preoperative CT (a, b) and MRI (c, d) images selected. Initially, the intradural tumor was exposed (e) and
of a patient with a left-sided jugular foramen meningioma removed (f). Then the pyramid was drilled (g), and the
with a large intra- and extracranial parts. Considering the remaining tumor removed completely. The postoperative
absence of hearing on the tumor side and the growth pattern CT scans (h, i) demonstrate the extent of bone removal
of the tumor, the transmastoid–transpetrous approach was
688 10 Jugular Foramen Tumors

g h

Fig. 10.13 (continued)

Fig. 10.14 Meningioma extending from the JF (arrow)


to the foramen magnum area
Meningiomas of the Jugular Foramen 689

Case Illustration 10.12 was unwilling, however, to accept any risk of


Primary JF Meningioma new neurological dysfunction. The standard
This 44-year-old patient had been operated 8 RS approach was used to expose the area of
years prior to presentation at another facility— the jugular foramen. A relatively small tumor
a partial removal of his jugular foramen men- bulging from the foramen into the CP angle was
ingioma had been performed. Four years later, seen and removed. The 10th and 11th cranial
he had been treated with radiosurgery because nerves were severely adherent to the tumor and
of growth of the tumor remnant. The follow- the scar tissue, formed after the previous sur-
up MRIs 2 and 3 years later showed a slow gery and radiosurgery. All lower cranial nerves
but constant growth tendency of the extrafo- showed good response to electrical stimula-
raminal tumor part. At presentation, he had tion. The jugular foramen could be entered
severe accessory nerve palsy on the left side via a small corridor between the nerve fibers,
with atrophy of the trapezius muscle and slight and a part of the tumor could be removed.
hearing loss. He reported occasional swallow- Considering the very high risk of damaging
ing disturbance, but the examination, includ- the nerves and the related risk of severe dys-
ing fiber-optic endoscopy, did not reveal any function, it was decided not to proceed with
obvious dysfunction of the ninth and tenth tumor removal. After the surgery, a transient
cranial nerves. The possible management swallowing difficulty was observed, but within
options were discussed with the patient and 4 days, normal feeding could be resumed. The
his family thoroughly, and—considering the histological diagnosis was meningothelial
steady growth tendency—a decision was taken meningioma grade 1. At follow-up 12 months
to attempt removal of the tumor. The patient later, the tumor remnant remained unchanged.
690 10 Jugular Foramen Tumors

a b

(a–c) Primary JF meningioma with a limited extension on T2-weighted images (b) and shows a strong homo-
into the CP angle. The tumor is isointense on geneous contrast uptake (c)
T1-weighted images (a) and isointense to hypointense
Meningiomas of the Jugular Foramen 691

Clinical Manifestation radicality of surgery, every effort should be made


to avoid new severe deficits and deterioration of
The clinical manifestation of JF meningiomas is patient’s quality of life. In case of tumor
related to their size and exact location and can infiltration of the cranial nerves or brain stem or
mimic the presentation of glomus jugulare tumors. in case clear dissection plane is absent, a less
Initial symptoms are due to lower cranial nerves or than total resection is the most reasonable option.
cochlear nerve dysfunction. At presentation the The biological activity of small tumor remnants
most frequent symptoms/signs are hearing loss cannot be reliably predicted. Frequently they do
(52–62 %), middle ear mass (23 %), dysphagia not show growth tendency for a long time period.
(23–50 %), deficit of the accessory nerve (6–38 %), Therefore, we do not recommend immediate
tinnitus (17 %), dizziness (17 %), hoarseness postoperative radiotherapy even in case of known
(14 %), tongue paresis (5–12 %), ataxia (12 %), remnants. A follow-up MRI is performed in 6
and headache (6–25 %) [6]. Middle ear mass may months, and radiotherapy—usually stereotactic
be identified on otoscopy (in type IIa, IIIa, and radiosurgery—is applied in case the remnant
IVa). The clinical picture is similar to that of other shows progressive growth.
jugular foramen tumors and is hence not sufficient Meningiomas on the side of a single ipsilat-
for preoperative differential diagnosis [86]. eral sigmoid sinus/bulb or even in case of domi-
nant ipsilateral sinus should be removed with
utmost care. Very important issue to consider is
Operative Treatment the preoperative neurological status of the
patients. Thus, in case the tumor involves the
The selection of an operative approach is based pyramid and the patient is deaf and has other cra-
on the following parameters: tumor size; loca- nial nerve deficits, the transmastoid–transpetrous
tion; extension; patient’s presenting symptoms, approach might be most suitable to achieve the
in particular hearing function; facial nerve and goals of surgery (Fig. 10.13).
lower cranial nerves function; and goal of sur-
gery. A wide variety of operative approaches has Outcome and Follow-Up
been promoted in the literature: infratemporal The most frequent new neurological deficits after
fossa type A approach; translabyrinthine; transco- surgery are lower cranial nerve, hearing loss, and
chlear; transcondylar–suprajugular, transcondy- facial nerve dysfunction. The facial nerve is espe-
lar–retrojugular, or transcondylar–transjugular; cially endangered if a procedure involving its
petro-occipital–transmastoid; and retrosigmoid wide dissection or transposition is performed.
approach—alone or in combination. Meningioma patients tend to have worse postop-
The goal of surgery of JF meningiomas is their erative lower cranial nerve outcomes than patients
complete removal and preservation of neurologi- with glomus tumors or schwannomas [38, 64].
cal functions. We favor the RS as the optimal To avoid neurological disasters, tumor relation to
operative approach that provides sufficient tumor the brain stem should be assessed very carefully.
exposure in most cases (see chapters on RS Meningiomas of the jugular foramen, similar to
approach). The jugular foramen can be opened— all other skull-base meningiomas, may be either
as described earlier—to access the intraforaminal infiltrative or have a well-developed and pre-
tumor part (see Case Illustration 10.13). The served arachnoid plane (see the discussion in the
involved bony structures, as well as the infiltrated chapter on CP angle meningiomas). In case the
dura, should be removed as far as safely possible. pia covering is infiltrated, no attempt at complete
In case of extensive tumors, involving the pyra- resection should be made.
mid and middle ear or extending extracranially, The recurrence rate in case of JF meningiomas
the approach should be modified accordingly. is reported to lie between 12.5 and 25 % [4, 73,
Considering the typical for skull-base meningio- 86, 97] and is higher in case of incomplete tumor
ma’s propensity to persist or recur despite the removal. These tumors, however, tend to recur
692 10 Jugular Foramen Tumors

despite of the extent of resection. Even after com- The outcome in patients with malignant
plete tumor removal, Molony et al. [73] observed meningiomas or with aggressive histological
recurrences in two of eight patients, Vrionis in tumor type (papillary, rhabdoid, microcys-
3/4 patients [120], Ramina in 1/5 patients [86], tic, and clear cells meningiomas) is poor: the
and Arnautovic and Al Mefty [4] in 1/8 patients. tumors tend to recur irrespective of the com-
A judicious tumor removal and vigilant follow- pleteness of their resection. Early radiotherapy
up with regular MR imaging is therefore abso- is recommended because it could provide bet-
lutely mandatory. The preferred treatment of ter tumor control and can postpone the time to
tumor recurrences is their surgical removal, pro- recurrence [86, 92].
vided it can be safely performed. Remnants con- Primary radiosurgery is an option, acceptable
sidered high surgical risks are best managed with only in patients with small meningiomas, inca-
radiotherapy/radiosurgery. pable, or unwilling to undergo surgical removal.
Meningiomas of the Jugular Foramen 693

Case Illustration 10.13 seventh to eighth cranial nerves and supe-


JF Meningioma with a Large CP Angle rior to the lower cranial nerves, which were
Part severely compressed. The tumor capsule
This 60-year-old patient presented with a his- was incised, and the tumor debulked using
tory of left-sided hearing loss since 12 months alternately the CUSA, platelet knife, tumor
and tinnitus since 7 months. His neurological forceps, and suction–irrigation. Then, the
status was completely normal besides a capsule was then dissected from the brain
decreased hearing on the left side (40d PTA). stem and from the CP angle vessels. In order
The MRI showed a contrast-enhancing tumor to gain access to the intraforaminal tumor
in the left CP angle, located mainly inframeatal part, the roof (dorsal wall) of the jugu-
and extending into the jugular foramen (a–f). lar foramen was widely opened. Thus, the
The lesion had imaging characteristics of a whole tumor could be removed completely
meningioma. The widened jugular foramen with preservation of all cranial nerves and
(CT scan) indicated that the tumor could be vessels. The histopathological diagnosis
originating in the area of the jugular foramen. was meningothelial meningioma (WHO
The RS approach was performed as usual, grade 1). The postoperative period was
exposing the edges of transverse and sig- uneventful. No new neurological deficits
moid sinuses (g–n). The highly vascular- appeared after surgery, and the hearing level
ized meningioma was located inferior to the remained unchanged (40 dB PTA).
694 10 Jugular Foramen Tumors

a b

c d

CT (a, b) and MRI (c–f) images demonstrating a primary diamond drill head was used to open the foramen widely
JF meningioma on the right side. (g) RS craniotomy, (k) and thereby to expose the whole tumor—(l) (Tu). The
exposing the edges of the transverse and sigmoid sinuses tumor was completely removed, and all neurovascular
(asterisks). (h, i) Initial view of the highly vascularized structures were preserved. (m) The intact AICA and
meningioma located inferior and lateral to the seventh to abducens nerve, dissected from the tumor. (n) Final view
eighth cranial nerve complex (not visible initially) and of the CP angle, demonstrating the complete tumor
superior to the lower cranial nerves (arrows). Once the removal, the preservation of the seventh to eighth cranial
tumor mass in the CP angle was removed, the jugular nerves and of the lower cranial nerves (LCN). The opened
foramen had to be opened to gain access to the intrafo- jugular foramen was afterward plugged with multiple fat
raminal tumor part. (j) The dura overlying the dorsal wall pieces to prevent CSF leak. (o) CT scan, performed on
of the jugular foramen was incised (arrowheads). The the first postoperative day
Meningiomas of the Jugular Foramen 695

e f
696 10 Jugular Foramen Tumors

g h

i j
Meningiomas of the Jugular Foramen 697

k l

m
n

o
698 10 Jugular Foramen Tumors

References 18. Brown LA (1953) Glomus jugulare tumor of the mid-


dle ear; clinical aspects. Laryngoscope 63(4):281–
292. doi:10.1288/00005537-195304000-00001
1. Al-Mefty O, Fox JL, Rifai A, Smith RR (1987) A
19. Bulsara KR, Sameshima T, Friedman AH, Fukushima
combined infratemporal and posterior fossa approach
T (2008) Microsurgical management of 53 jugular
for the removal of giant glomus tumors and chondro-
foramen schwannomas: lessons learned incorporated
sarcomas. Surg Neurol 28(6):423–431
into a modified grading system. J Neurosurg
2. Al-Mefty O, Teixeira A (2002) Complex tumors of
109(5):794–803. doi:10.3171/JNS/2008/109/11/0794
the glomus jugulare: criteria, treatment, and outcome.
20. Caldemeyer KS, Mathews VP, Azzarelli B, Smith RR
J Neurosurg 97(6):1356–1366
(1997) The jugular foramen: a review of anatomy,
3. Alford BR, Guilford FR (1962) A comprehensive study
masses, and imaging characteristics. Radiographics
of tumors of the glomus jugulare. Laryngoscope
17(5):1123–1139
72:765–805. doi:10.1288/00005537-196206000-00006
21. Capps FC (1952) Glomus jugulare tumours of the
4. Arnautovic KI, Al-Mefty O (2002) Primary menin-
middle ear. J Laryngol Otol 66(7):302–314
giomas of the jugular fossa. J Neurosurg 97(1):12–20.
22. Capps FC (1958) Chemodectoma or tumor of the glo-
doi:10.3171/jns.2002.97.1.0012
mus jugulare and tympanic bodies. AMA Arch
5. Ayeni SA, Ohata K, Tanaka K, Hakuba A (1995) The
Otolaryngol 67(5):556–559
microsurgical anatomy of the jugular foramen. J
23. Carvalho GA, Tatagiba M, Samii M (2000) Cystic
Neurosurg 83(5):903–909. doi:10.3171/jns.1995.83.
schwannomas of the jugular foramen: clinical and
5.0903
surgical remarks. Neurosurgery 46(3):560–566
6. Bakar B (2010) Jugular foramen meningiomas: review
24. Clemis JD (1980) Neurogenic tumors of the skull
of the major surgical series. Neurol Med Chir (Tokyo)
base. Otolaryngol Head Neck Surg 88(5):511–518
50(2):89–96
25. Denecke HJ (1966) On the surgery of diffuse glomus
7. Bakar B (2008) The jugular foramen schwannomas:
tumors in the region of the foramen jugulare. Arch
review of the large surgical series. J Korean Neurosurg
Klin Exp Ohren Nasen Kehlkopfheilkd 187(2):
Soc 44(5):285–294. doi:10.3340/jkns.2008.44.5.285
656–662
8. Balasubramaniam C (1999) A case of malignant
26. Dichiro G, Fisher RL, Nelson KB (1964) The jugular
tumour of the jugular foramen in a young infant.
foramen. J Neurosurg 21:447–460. doi:10.3171/
Childs Nerv Syst 15(6–7):347–350
jns.1964.21.6.0447
9. Bartal AD, Djaldetti MM, Mandel EM, Lerner MA
27. Eldevik OP, Gabrielsen TO, Jacobsen EA (2000)
(1973) Dumb-bell neurinoma of the hypoglossal
Imaging findings in schwannomas of the jugular fora-
nerve. J Neurol Neurosurg Psychiatry 36(4):592–595
men. AJNR Am J Neuroradiol 21(6):1139–1144
10. Batsakis JG, Farber ER (1967) Chemodectomas of the
28. Fayad JN, Keles B, Brackmann DE (2009) Jugular
head and neck. Univ Mich Med Cent J
foramen tumors: clinical characteristics and treatment
33(3):123–130
outcomes. Otol Neurotol 31(2):299–305. doi:10.1097/
11. Berger MS, Edwards MS, Bingham WG (1982)
MAO.0b013e3181be6495
Hypoglossal neurilemmoma: case report and review
29. Fisch U (1978) Infratemporal fossa approach to
of the literature. Neurosurgery 10(5):617–620
tumours of the temporal bone and base of the skull.
12. Bickerstaff ER, Howell JS (1953) The neurological
J Laryngol Otol 92(11):949–967
importance of tumours of the glomus jugulare. Brain
30. Fisch U (1982) Infratemporal fossa approach for glo-
76(4):576–593
mus tumors of the temporal bone. Ann Otol Rhinol
13. Borba LA, Ale-Bark S, London C (2004) Surgical
Laryngol 91(5 Pt 1):474–479
treatment of glomus jugulare tumors without rerout-
31. Franklin DJ, Moore GF, Fisch U (1989) Jugular fora-
ing of the facial nerve: an infralabyrinthine approach.
men peripheral nerve sheath tumors. Laryngoscope
Neurosurg Focus 17(2):E8. doi:170208 [pii]
99(10 Pt 1):1081–1087. doi:10.1288/00005537-
14. Borba LA, Araujo JC, de Oliveira JG, Filho MG,
198210000-00020
Moro MS, Tirapelli LF, Colli BO (2010) Surgical
32. Galbraith JG, Butler JS, Harty JA (2011) Recurrent
management of glomus jugulare tumors: a proposal
spontaneous hip dislocation in a patient with
for approach selection based on tumor relationships
neurofibromatosis type 1: a case report. J Med Case
with the facial nerve. J Neurosurg 112(1):88–98.
Reports 5(1):106. doi:10.1186/1752-1947-5-106.
doi:10.3171/2008.10.JNS08612
1752-1947-5-106 [pii]
15. Boyle JO, Shimm DS, Coulthard SW (1990) Radiation
33. Ganz JC, Abdelkarim K (2009) Glomus jugulare
therapy for paragangliomas of the temporal
tumours: certain clinical and radiological aspects
bone. Laryngoscope 100(8):896–901. doi:10.1288/
observed following Gamma Knife radiosurgery. Acta
00005537-199008000-00018
Neurochir (Wien) 151(5):423–426. doi:10.1007/
16. Brackmann DE (1987) The facial nerve in the
s00701-009-0268-7
infratemporal approach. Otolaryngol Head Neck Surg
34. Gardner G, Cocke EW Jr, Robertson JT, Trumbull
97(1):15–17
ML, Palmer RE (1977) Combined approach surgery
17. Brismar J, Cronqvist S (1978) Therapeutic emboliza-
for removal of glomus jugulare tumors. Laryngoscope
tion in the external carotid artery region. Acta Radiol
87(5 Pt 1):665–688
Diagn (Stockh) 19(5):715–731
References 699

35. Gejrot T (1965) Surgical treatment of glomus jugulare 51. House WF, Glasscock ME 3rd (1968) Glomus tym-
tumours with special reference to the diagnostic value panicum tumors. Arch Otolaryngol 87(5):550–554
of retrograde jugularography. Acta Otolaryngol 52. Jackson C (2001) Glomus tympanicum and glomus
60:150–168 jugulare tumors. Otolaryngol Clin North Am
36. George B, Lot G, Tran Ba Huy P (1995) The jux- 34:941–970
tacondylar approach to the jugular foramen (without 53. Jackson CG (1993) Neurotologic skull base surgery
petrous bone drilling). Surg Neurol 44(3):279–284. for glomus tumors. Diagnosis for treatment planning
doi:0090-3019(95)00174-3 [pii] and treatment options. Laryngoscope 103(11 Pt 2
37. Gerosa M, Visca A, Rizzo P, Foroni R, Nicolato A, Suppl 60):17–22
Bricolo A (2006) Glomus jugulare tumors: the option 54. Jackson CG, Glasscock ME 3rd, Nissen AJ, Schwaber
of gamma knife radiosurgery. Neurosurgery 59(3): MK (1982) Glomus tumor surgery: the approach,
561–569. doi:10.1227/01.NEU.0000228682.92552. results, and problems. Otolaryngol Clin North Am
CA. 00006123-200609000-00008 [pii]; discussion 15(4):897–916
561–569 55. Jackson CG, Kaylie DM, Coppit G, Gardner EK (2004)
38. Gilbert ME, Shelton C, McDonald A, Salzman KL, Glomus jugulare tumors with intracranial extension.
Harnsberger HR, Sharma PK, Jensen RL (2004) Neurosurg Focus 17(2):E7. doi:170207 [pii]
Meningioma of the jugular foramen: glomus jugulare 56. Jenkins HA, Fisch U (1981) Glomus tumors of the
mimic and surgical challenge. Laryngoscope 114(1):25– temporal region. Technique of surgical resection.
32. doi:10.1097/00005537-200401000-00004 Arch Otolaryngol 107(4):209–214
39. Glasscock ME 3rd, Harris PF, Newsome G (1974) 57. Katsuta T, Rhoton AL Jr, Matsushima T (1997) The
Glomus tumors: diagnosis and treatment. Laryngoscope jugular foramen: microsurgical anatomy and opera-
84(11):2006–2032 tive approaches. Neurosurgery 41(1):149–201; dis-
40. Green JD Jr, Brackmann DE, Nguyen CD, Arriaga MA, cussion 201–142
Telischi FF, De la Cruz A (1994) Surgical management 58. Kaye AH, Hahn JF, Kinney SE, Hardy RW Jr, Bay JW
of previously untreated glomus jugulare tumors. (1984) Jugular foramen schwannomas. J Neurosurg
Laryngoscope 104(8 Pt 1):917–921. 60(5):1045–1053. doi:10.3171/jns.1984.60.5.1045
doi:10.1288/00005537-199408000-00001 59. Kempe LG, VanderArk GD, Smith DR (1971) The neuro-
41. Greenberg HS, Deck MD, Vikram B, Chu FC, Posner surgical treatment of glomus jugulare tumors. J Neurosurg
JB (1981) Metastasis to the base of the skull: clinical 35(1):59–64. doi:10.3171/jns.1971.35.1.0059
findings in 43 patients. Neurology 31(5):530–537 60. Lack EE, Cubilla AL, Woodruff JM, Farr HW (1977)
42. Guild S (1941) A hitherto unrecognized structure, the Paragangliomas of the head and neck region: a clini-
glomus jugularis, in man. Anat Rec 79:28 cal study of 69 patients. Cancer 39(2):397–409
43. Gulya AJ (1993) The glomus tumor and its biology. 61. Leman P, Portmann M, Cohadon F, Leifer C, Traissac,
Laryngoscope 103(11 Pt 2 Suppl 60):7–15 Castel (1968) Oto-neurosurgical collaboration in the
44. Hakuba A, Hashi K, Fujitani K, Ikuno H, Nakamura total ablation of a tympano-jugular glomus tumor
T, Inoue Y (1979) Jugular foramen neurinomas. Surg with intracranial extension. Neurochirurgie 14(7):
Neurol 11(2):83–94 828–830
45. Hawthorne MR, Makek MS, Harris JP, Fisch U (1988) 62. Leonetti JP, Brackmann DE, Prass RL (1989)
The histopathological and clinical features of irradi- Improved preservation of facial nerve function in the
ated and nonirradiated temporal paragangliomas. infratemporal approach to the skull base. Otolaryngol
Laryngoscope 98(3):325–331. doi:10.1288/00005537- Head Neck Surg 101(1):74–78
198803000-00018 63. Lowenheim H, Koerbel A, Ebner FH, Kumagami H,
46. Heth J (2004) The basic science of glomus jugulare Ernemann U, Tatagiba M (2006) Differentiating imaging
tumors. Neurosurg Focus 17(2):E2. doi:170202 [pii] findings in primary and secondary tumors of the jugular
47. Heutink P, van der Mey AG, Sandkuijl LA, van Gils foramen. Neurosurg Rev 29(1):1–11. doi:10.1007/
AP, Bardoel A, Breedveld GJ, van Vliet M, van s10143-005-0420-7; discussion 12–13
Ommen GJ, Cornelisse CJ, Oostra BA et al (1992) A 64. Lustig LR, Jackler RK (1996) The variable relation-
gene subject to genomic imprinting and responsible ship between the lower cranial nerves and jugular
for hereditary paragangliomas maps to chromosome foramen tumors: implications for neural preservation.
11q23-qter. Hum Mol Genet 1(1):7–10 Am J Otol 17(4):658–668
48. Hilal SK, Michelsen JW (1975) Therapeutic percuta- 65. Macdonald AJ, Salzman KL, Harnsberger HR, Gilbert
neous embolization for extra-axial vascular lesions of E, Shelton C (2004) Primary jugular foramen menin-
the head, neck, and spine. J Neurosurg 43(3):275–287. gioma: imaging appearance and differentiating fea-
doi:10.3171/jns.1975.43.3.0275 tures. AJR Am J Roentgenol 182(2):373–377
49. Hilding DA, Greenberg A (1971) Surgery for large 66. Makek M, Franklin DJ, Zhao JC, Fisch U (1990)
glomus jugulare tumor. The combined suboccipital, Neural infiltration of glomus temporale tumors. Am J
transtemporal approach. Arch Otolaryngol 93(3): Otol 11(1):1–5
|227–231 67. Maniglia AJ, Chandler JR, Goodwin WJ Jr, Parker JC
50. Horn KL, House WF, Hitselberger WE (1985) Jr (1979) Schwannomas of the parapharyngeal space
Schwannomas of the jugular foramen. Laryngoscope and jugular foramen. Laryngoscope 89(9 Pt
95(7 Pt 1):761–765 1):1405–1414
700 10 Jugular Foramen Tumors

68. Martin JJ, Kondziolka D, Flickinger JC, Mathieu D, 83. Pollock BE (2004) Stereotactic radiosurgery in
Niranjan A, Lunsford LD (2007) Cranial nerve preser- patients with glomus jugulare tumors. Neurosurg
vation and outcomes after stereotactic radiosurgery for Focus 17(2):E10. doi:170210 [pii]
jugular foramen schwannomas. Neurosurgery 61(1): 84. Ramina R, Maniglia JJ, Fernandes YB, Paschoal JR,
76–81. doi:10.1227/01.neu.0000279726.90650.6d. Pfeilsticker LN, CoelhoNeto M (2005) Tumors of the
00006123-200707000-00008 [pii]; discussion 81 jugular foramen: diagnosis and management.
69. Mazzoni A, Sanna M, Saleh E, Achilli V (1997) Neurosurgery 57(1 Suppl):59–68; discussion
Lower cranial nerve schwannomas involving the jug- 59–68
ular foramen. Ann Otol Rhinol Laryngol 106(5): 85. Ramina R, Maniglia JJ, Fernandes YB, Paschoal JR,
370–379 Pfeilsticker LN, Neto MC, Borges G (2004) Jugular
70. Meacham WF, Capps JM (1960) Intracranial glomus- foramen tumors: diagnosis and treatment. Neurosurg
jugulare tumor with successful surgical removal. Focus 17(2):E5. doi:170205 [pii]
J Neurosurg 17:157–160 86. Ramina R, Neto MC, Fernandes YB, Aguiar PH, de
71. Megerian CA, McKenna MJ, Nadol JB Jr (1995) Meneses MS, Torres LF (2006) Meningiomas of the
Non-paraganglioma jugular foramen lesions mas- jugular foramen. Neurosurg Rev 29(1):55–60.
querading as glomus jugulare tumors. Am J Otol doi:10.1007/s10143-005-0415-4
16(1):94–98 87. Rao AB, Koeller KK, Adair CF (1999) From the
72. Miller JP, Semaan MT, Maciunas RJ, Einstein DB, archives of the AFIP. Paragangliomas of the head and
Megerian CA (2009) Radiosurgery for glomus jugu- neck: radiologic-pathologic correlation. Armed Forces
lare tumors. Otolaryngol Clin North Am 42(4):689– Institute of Pathology. Radiographics
706. doi:10.1016/j.otc.2009.04.004. S0030-6665 19(6):1605–1632
(09)00059-0 [pii] 88. Rhoton AL Jr (2000) Jugular foramen. Neurosurgery
73. Molony TB, Brackmann DE, Lo WW (1992) 47(3 Suppl):S267–S285
Meningiomas of the jugular foramen. Otolaryngol 89. Roberti F, Sekhar LN, Kalavakonda C, Wright DC
Head Neck Surg 106(2):128–136 (2001) Posterior fossa meningiomas: surgical experi-
74. Murphy TP, Brackmann DE (1989) Effects of preop- ence in 161 cases. Surg Neurol 56(1):8–20; discussion
erative embolization on glomus jugulare tumors. 20–21. doi:S0090-3019(01)00479-7 [pii]
Laryngoscope 99(12):1244–1247. doi:10.1288/ 90. Rosenwasser H (1945) Carotid body like tumor
00005537-198912000-00007 involving the middle ear and mastoid bone. Arch
75. Muthukumar N, Kondziolka D, Lunsford LD, Otolaryngol 41:64–67
Flickinger JC (1999) Stereotactic radiosurgery for 91. Ruben RJ (2007) The history of the glomus tumors –
jugular foramen schwannomas. Surg Neurol nonchromaffim chemodectoma: a glimpse of biomed-
52(2):172–179. doi:S0090-3019(99)00031-2 [pii] ical Camelot. Acta Otolaryngol 127(4):411–416.
76. Oghalai JS, Buxbaum JL, Jackler RK, McDermott MW doi:10.1080/00016480601002088. 777054949 [pii]
(2005) Skull base chondrosarcoma originating from the 92. Salcman M (1991) Malignant meningiomas. In: A-M O
petroclival junction. Otol Neurotol 26(5):1052–1060. (ed) Meningiomas. Raven Press, New York, pp 75–85
doi:00129492-200509000-00037 [pii] 93. Samii M (1989) Tumors of the internal auditory canal
77. Oghalai JS, Leung MK, Jackler RK, McDermott MW and cerebellopontine angle. In: Samii M, Draf W
(2004) Transjugular craniotomy for the management (eds) Surgery of the skull base: an interdisciplinary
of jugular foramen tumors with intracranial extension. approach. Springer, Berlin, pp 377–395
Otol Neurotol 25(4):570–579; discussion 579. 94. Samii M, Babu RP, Tatagiba M, Sepehrnia A
doi:00129492-200407000-00026 [pii] (1995) Surgical treatment of jugular foramen
78. Olsen WL, Dillon WP, Kelly WM, Norman D, Brant- schwannomas. J Neurosurg 82(6):924–932.
Zawadzki M, Newton TH (1987) MR imaging of para- doi:10.3171/jns.1995.82.6.0924
gangliomas. AJR Am J Roentgenol 148(1):201–204 95. Samii M, Bini W (1993) Surgical strategy for jugular
79. Ong CK, Fook-Hin Chong V (2009) Imaging of jugu- foramen tumors. In: Sekhar LN, Janecka IP (eds)
lar foramen. Neuroimaging Clin N Am 19(3):469– Surgery of cranial base tumors. Raven Press, New
482. doi:10.1016/j.nic.2009.06.007. S1052-5149 York, pp 379–387
(09)00043-4 [pii] 96. Sanna M, Bacciu A, Falcioni M, Taibah A (2006)
80. Patel SJ, Sekhar LN, Cass SP, Hirsch BE (1994) Surgical management of jugular foramen schwanno-
Combined approaches for resection of extensive glo- mas with hearing and facial nerve function preserva-
mus jugulare tumors. A review of 12 cases. J Neurosurg tion: a series of 23 cases and review of the literature.
80(6):1026–1038. doi:10.3171/jns.1994.80.6.1026 Laryngoscope 116(12):2191–2204. doi:10.1097/01.
81. Pellet W, Cannoni M, Pech A (1988) The widened mlg.0000246193.84319.e5. 00005537-200612000-
transcochlear approach to jugular foramen tumors. 00017 [pii]
J Neurosurg 69(6):887–894. doi:10.3171/jns.1988.69. 97. Sanna M, Bacciu A, Falcioni M, Taibah A, Piazza P
6.0887 (2007) Surgical management of jugular foramen
82. Pensak ML, Jackler RK (1997) Removal of jugular meningiomas: a series of 13 cases and review of
foramen tumors: the fallopian bridge technique. the literature. Laryngoscope 117(10):1710–1719.
Otolaryngol Head Neck Surg 117(6):586–591. doi:10.1097/MLG.0b013e3180cc20a3. 00005537-
doi:S0194-5998(97)70037-8 [pii] 200710000-00003 [pii]
References 701

98. Sanna M, De Donato G, Di Lella F, Falcioni M, 112. Sutay S, Tekinsoy B, Ceryan K, Aksu Y (1993)
Aggrawal N, Romano G (2009) Nonvascular lesions Submaxillary hypoglossal neurilemmoma.
of the jugular foramen: the gruppo otologico experi- J Laryngol Otol 107(10):953–954
ence. Skull Base 19(1):57–74. doi:10.1055/s- 113. Tan LC, Bordi L, Symon L, Cheesman AD (1990)
0028-1103124 Jugular foramen neuromas: a review of 14 cases.
99. Sanna M, De Donato G, Taibah A, Russo A, Falcioni Surg Neurol 34(4):205–211. doi:0090-3019(90)
M, Mancini F (1999) Infratemporal fossa approaches 90130-H [pii]
to the lateral skull base. Keio J Med 48(4):189–200 114. Tekkok IH, Ozcan OE, Turan E, Onol B (1997)
100. Sanna M, Jain Y, De Donato G, Rohit, Lauda L, Jugular foramen meningioma. Report of a case and
Taibah A (2004) Management of jugular paragan- review of the literature. J Neurosurg Sci 41(3):
gliomas: the Gruppo Otologico experience. Otol 283–292
Neurotol 25(5):797–804. doi:00129492-200409000- 115. Thoms OJ, Shaw DT, Trowbridge WV (1960)
00025 [pii] Glomus jugulare tumor: report of a case with surgi-
101. Semaan MT, Megerian CA (2008) Current assess- cal removal. J Neurosurg 17:500–504. doi:10.3171/
ment and management of glomus tumors. Curr Opin jns.1960.17.3.0500
Otolaryngol Head Neck Surg 16(5):420–426. 116. Tummala RP, Coscarella E, Morcos JJ (2005)
doi:10.1097/MOO.0b013e32830c4595. 00020840- Transpetrosal approaches to the posterior fossa.
200810000-00006 [pii] Neurosurg Focus 19(2):E6. doi:190206 [pii]
102. Semmes R (1951) Discussion of Alexander E Jr, 117. Ulso C, Sehested P, Overgaard J (1981) Intracranial
Beamer PR, Williams JO: Tumor of the glomus jug- hypoglossal neurinoma: diagnosis and postoperative
ulare with extension into the middle ear. J Neurosurg care. Surg Neurol 16(1):65–68
8:515–523 118. van den Berg R, Schepers A, de Bruine FT, Liauw L,
103. Sen C, Hague K, Kacchara R, Jenkins A, Das S, Mertens BJ, van der Mey AG, van Buchem MA
Catalano P (2001) Jugular foramen: microscopic (2004) The value of MR angiography techniques in
anatomic features and implications for neural preser- the detection of head and neck paragangliomas. Eur
vation with reference to glomus tumors involving the J Radiol 52(3):240–245. doi:10.1016/j.
temporal bone. Neurosurgery 48(4):838–847; dis- ejrad.2003.12.002. S0720048X03003747 [pii]
cussion 847–838 119. van der Mey AG, Maaswinkel-Mooy PD, Cornelisse
104. Shapiro MJ, Neues DK (1964) Technique for CJ, Schmidt PH, van de Kamp JJ (1989) Genomic
removal of glomus jugulare tumors. Arch Otolaryngol imprinting in hereditary glomus tumours: evidence
79:219–224 for new genetic theory. Lancet 2(8675):1291–1294
105. Shearman CP, Pearman K, Hockley AD (1986) 120. Vrionis FD, Robertson JH, Gardner G, Heilman CB
Dumb-bell schwannoma of the jugular foramen. (1999) Temporal bone meningiomas. Skull Base
Childs Nerv Syst 2(1):52–53 Surg 9(2):127–139
106. Sheehan J, Kondziolka D, Flickinger J, Lunsford LD 121. Willen SN, Einstein DB, Maciunas RJ, Megerian
(2005) Gamma knife surgery for glomus jugulare CA (2005) Treatment of glomus jugulare tumors in
tumors: an intermediate report on efficacy and safety. patients with advanced age: planned limited surgical
J Neurosurg 102(Suppl):241–246 resection followed by staged gamma knife radiosur-
107. Simpson GT 2nd, Konrad HR, Takahashi M, House J gery: a preliminary report. Otol Neurotol 26(6):1229–
(1979) Immediate postembolization excision of glo- 1234. doi:00129492-200511000-00026 [pii]
mus jugulare tumors: advantages of new combined 122. Williams HL, Childs DS Jr, Parkhill EM, Pugh DG
techniques. Arch Otolaryngol 105(11):639–643 (1955) Chemodectomas of the glomus jugulare
108. Snyderman CH, Pant H, Carrau RL, Prevedello D, (nonchromaffin paragangliomas) with especial ref-
Gardner P, Kassam AB (2009) What are the limits of erence to their response to roentgen therapy. Ann
endoscopic sinus surgery? The expanded endonasal Otol Rhinol Laryngol 64(2):546–566
approach to the skull base. Keio J Med 58(3):152–160 123. Zhang H, Cai C, Wang S, Liu H, Ye Y, Chen X (2007)
109. Song MH, Lee HY, Jeon JS, Lee JD, Lee HK, Lee Extracranial head and neck schwannomas: a clinical
WS (2008) Jugular foramen schwannoma: analysis analysis of 33 patients. Laryngoscope 117(2):278–
on its origin and location. Otol Neurotol 29(3):387– 281. doi:10.1097/01.mlg.0000249929.60975.a7.
391. doi:10.1097/MAO.0b013e318164cb83 00005537-200702000-00017 [pii]
110. Spector GJ, Ciralsky RH, Ogura JH (1975) Glomus 124. Zhang N, Pan L, Dai JZ, Wang BJ, Wang EM, Cai
tumors in the head and neck: III. Analysis of clinical PW (2002) Gamma knife radiosurgery for jugular
manifestations. Ann Otol Rhinol Laryngol 84(1 Pt foramen schwannomas. J Neurosurg 97(5 Suppl):456–
1):73–79 458. doi:10.3171/jns.2002.97.supplement5.0456
111. Spinnato S, Talacchi A, Musumeci A, Turazzi S, 125. Zimmer LA, Hirsch BE, Kassam A, Horowitz M,
Bricolo A (1998) Dumbbell-shaped hypoglossal Snyderman CH (2006) Resection of a recurrent para-
neurinoma: surgical removal via a dorsolateral ganglioma via an endoscopic transnasal approach to
transcondylar approach. A case report and review of the jugular fossa. Otol Neurotol 27(3):398–402.
the literature. Acta Neurochir (Wien) 140(8):827–832 doi:00129492-200604000-00017 [pii]
Other Cerebellopontine
Angle Tumors 11

Intra-Axial Tumors Extending and goal of surgery. Besides the standard ret-
Secondarily to the Cerebellopontine rosigmoid approach, its modification—the lat-
Angle eral suboccipital approach to the craniocervical
junction, as described in the chapter on foramen
Intra-axial tumors originating in the brain stem, magnum meningiomas, can be utilized. Tumors
cerebellum, or fourth ventricle can have an exo- with both CP angle and fourth ventricular part
phytic part or become large enough to involve can be removed via a midline suboccipital cran-
the CP angle [4]. Clinically they can manifest as iotomy with opening of the foramen magnum.
a CP angle mass. Most common are astrocy- The fourth ventricle is accessed via the telovelar
tomas, ependymomas, and medulloblastomas, route and—once the intraventricular tumor part
followed by choroid plexus papillomas, lympho- is removed—the CP angle is reached via the
mas, hemangioblastomas, gangliogliomas, and dilated foramen of Luschka (see Case Illustration
dysembryoplastic neuroepithelial tumors [8, 11.3). Alternatively, tumors with larger CP angle
16, 30]. Their clinical presentation is nonspecific, part may be approached via the retrosigmoid
and the presenting symptoms are not related to route: once the cisternal part is removed, the
the tumor type: they depend rather upon the fourth ventricle may be reached via the enlarged
affected cranial nerves or neural structures. The foramen of Luschka (see Case Illustration
preoperative diagnosis of a CP angle tumor is 11.1).
based mainly on neuroimaging [4, 42].
Characteristic findings are narrow or undetect-
able CP angle cistern, absent brain–tumor Brain Stem Gliomas
interface, and peritumoral edema.
It has been estimated that 0.3–2 % of all CP Brain stem gliomas are highly heterogeneous
angle lesions originate from primary brain tissue tumors that occur at any age. Still, they affect
[42]. Surgery rarely leads to cure of patients with primarily children, with the mean age of diag-
such tumors and should be regarded as a compo- nosis at 7–9 years. These tumors account for
nent of the whole management concept. Its goal 10–20 % of all pediatric central nervous sys-
is to provide histopathological diagnosis and to tem tumors [18]. Relatively rarely they mani-
remove the lesion as completely as safely possi- fest as asymmetric expansion of the brain stem
ble, thereby achieving volume reduction, brain into the CP angle or have an exophytic part in
stem decompression, and restoration of the CSF the area—Fig. 11.1 [43]. Extension into and
circulation pathways. The operative approach is even enlargement of the IAC has also been
selected considering tumor size and extension, reported [31, 45]. Primary CP angle gliomas
neurological and general condition of the patient, are exceedingly rare; they arise from the

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 703


DOI 10.1007/978-3-642-35422-9_11, © Springer-Verlag Berlin Heidelberg 2013
704 11 Other Cerebellopontine Angle Tumors

vestibulocochlear nerve [3, 11, 20] or from the On MR imaging, brain stem gliomas are
trigeminal nerve [2]. Most cases reported in the hypointense in T1-weighted images and hyper-
literature are either fibrillary or gemistocytic intense on T2-weighted images and may be sur-
astrocytomas [2, 5]. rounded by hyperintense edematous area [4]. The
Three cases of primary extra-axial pilocytic CISS sequence is very useful for delineating CP
astrocytoma of the CP angle have been reported angle anatomy and tumor borders. The signal
[2, 3, 31]. These tumors were completely sepa- characteristics and the extent of contrast enhance-
rated from the brain stem, and a clear tissue plane ment are related to tumors’ grade. Mirone et al.
between the tumor capsule and the brain stem [31] pointed that a high index of suspicion is
could be identified. warranted when evaluating a CP angle mass in a

a b

c d

Fig. 11.1 Pilocytic astrocytoma of the lower brain stem via a median suboccipital craniotomy and telovelar
in a 20-year-old patient. The tumor has a large pontomed- approach to the fourth ventricle (e–h: postoperative MR
ullary cystic part and an exophytic contrast-enhancing images)
part in the left CP angle (a–d). It was removed completely
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 705

e f

g h

Fig. 11.1 (continued)

child. Blurring of the margin between the tumor Surgery is part of the management of brain
and the brain stem or cerebellum, extensive peri- stem gliomas. Its goal is to verify the histo-
tumoral edema, and widening of the lateral recess logical tumor type, to perform tumor debulk-
of the fourth ventricle should raise the suspicion ing, or to remove the safely accessible tumor
for an intrinsic tumor with secondary CP angle part (see Figs. 11.2, 11.3, and 11.4 and Case
extension [31]. Illustration 11.1).
706 11 Other Cerebellopontine Angle Tumors

a b

c d

Fig. 11.2 Heterogeneous lesion with MR characteristics stern sequence. (b, d) T2-weighted image. (c) T1-weighted
of a low-grade glioma. It originated from the pedunculus image after contrast administration. See next figures for
cerebelli above the level of the seventh to eighth cranial details on tumor removal
nerves and extended into the left CP angle. (a) T2-weighted
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 707

a b

Tu

7-8

c d

Fig. 11.3 Intraoperative images illustrating the steps of complex, R retractor, arrowhead petrous vein). (b–d) In
tumor removal (patient from previous figure). (a) Initial order to avoid major neurological deficit, the tumor was
view of the tumor from a retrosigmoid perspective, present- removed only partially. Histological examination showed
ing as expansion of the brain stem above the root exit zone that it was a low-grade glioma
of the facial nerve (Tu tumor, 7–8 vestibulocochlear nerve
708 11 Other Cerebellopontine Angle Tumors

a b

Fig. 11.4 Low-grade exophytic astrocytoma of the cervi- view of the tumor. (e, f) Gradual removal and exposure of
cal medulla with intracranial extension into the lower left the vertebral artery. (g) Complete tumor removal and pres-
CP angle. (a–c) Preoperative images. The patient has been ervation of all structures
operated in the early 1990s by the senior author. (d) Initial
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 709

d e

f g

Fig. 11.4 (continued)


710 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.1 Electrical stimulation was repeatedly applied to


Brain Stem Low-Grade Glioma Removed identify the essential areas of the brain stem.
via the Lateral Suboccipital Approach with Tumor removal was then continued in a medial
Opening of the Foramen Magnum direction through the enlarged foramen of
Twenty-six-year-old male patient presented Luschka. The intra-axial tumor part, which
with complaints of general weakness, vertigo, infiltrated the medulla oblongata, was not removed
deteriorating hearing on the right side, and occa- in order to prevent major deficits; the nerve root
sional swallowing difficulty since more than 12 entry/exit zone of the lower cranial nerves was
months. Neurological examination revealed gait located exactly in this area, which explained the
instability, hearing loss, rotatory nystagmus, and preoperative dysphagia. The tumor portion in the
lack of pharyngeal reflex on the right side. MRI fourth ventricle and in the CP angle was removed,
demonstrated a large brain stem tumor with and decompression of the seventh to eighth cranial
extension in the right CP angle and fourth ven- nerves was achieved. Histopathological examina-
tricle through the foramen of Luschka (a–e). tion revealed a diffuse WHO grade II astrocytoma.
The patient was operated via a lateral suboc- Immediately after surgery, the patient had
cipital approach with opening of the foramen mag- facial nerve weakness (HB grade III); the dys-
num (f–p). Following the dura incision along the phagia was more pronounced and a tracheos-
sigmoid sinus, the tonsil was elevated, and the tomy had to be performed. PEG tube was also
tumor part protruding into the CP angle was seen. inserted. Within the hospital stay, his gait
This part was gradually resected, which allowed improved significantly. In 2 months the tra-
exposing and dissecting the seventh to eighth cheostoma could be closed and the facial
nerve complex and later of the abducens nerve. nerve function improved to HB grade II.

a b

(a–e) Preoperative MR examination of the patient, lum has been slightly elevated, exposing the expanded
demonstrating the tumor of the lower brainstem and its infiltrated by the tumor medulla (asteriks) and the lower
extension into the CP angle and 4th ventricle through cranial nerves (LCN). (i) The exophytic tumor part (Tu)
the foramen of Luschka. (a, b) T1-weighted sequence, was seen more rostrally to the LCN; the LCN entry/exit
following contrast administration. (c, d) T2-weighted zone was in the tumor-involved area of the medulla (j).
and (e) T2-weighted CISS sequences. The tumor (k) Electrical stimulation was frequently applied.
causes dilatation of the CP angle lateral to the tumor. (l–m) Gradual resection of the exophytic tumor, which
(f–p) Intraoperative images. (f) Lateral suboccipi- allowed to visualize the 7–8th nerve complex (n) and
tal approach with opening of the foramen magnum later – of the abducent nerve (o) (6). (p) Tumor removal
(SS – sigmoid sinus). (g) The dura is opened along was then continued in a medial direction through the
the sigmoid sinus (C – cerebellum). (h) The cerebel- enlarged foramen of Luschka (V – 4th ventricle)
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 711

c d

h
712 11 Other Cerebellopontine Angle Tumors

i j

k l
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 713

m n

o p
714 11 Other Cerebellopontine Angle Tumors

Choroid Plexus Papillomas mass with lobulations and frond-like irregular


pattern—a cauliflower-like appearance [4, 24].
Choroid plexus papillomas represent 0.4–1 % of Calcifications are seen in 4–20 % of the tumors
all intracranial neoplasms [44]. They originate [39]. Their radiological differential diagnosis is
from the epithelial cells of the choroid plexus and difficult if a frond-like pattern, calcifications,
are similar histologically to the normal choroid and/or enlargement of foramen of Luschka are
plexus [21]. Malignant forms are very rare. not present. Plexus papillomas are isointense on
Choroid plexus papillomas occur mainly in T1- and T2-weighted MR images, have irregular
children and are located in the lateral ventricles. structure, and enhance intensely after contrast
In adults their most common location is the fourth injection. They are often associated with hydro-
ventricle. Papillomas originating from the chor- cephalus, caused by obstruction of the CSF flow
oid plexus of the fourth ventricle can extend via or by CSF hypersecretion by the tumor [33]. In
the foramina of Luschka into the CP angle. Very the series of Talacchi et al. [46], 8 of 12 CP angle
rarely they develop primarily in the CP angle [10, choroid plexus papillomas had hydrocephalus.
13, 25]. In 1 series of 245 patients of choroid The optimal treatment of plexus papillomas is
plexus papillomas, only 17 were located in the surgical removal: complete tumor removal is
CP angle [39]. related to an excellent prognosis [13, 21] (see
These tumors present clinically with signs and Case Illustration 11.2). The tumor may be very
symptoms of a CP angle lesion that are related to adherent to the brain stem, however, and total
its size and precise location. On CT imaging, they removal with preservation of neurological func-
present as well-defined homogeneously enhancing tions may be challenging [25].
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 715

Case Illustration 11.2 direction of the brain stem, which allowed


CP Angle Plexus Papilloma exposing the tumor. The tumor was adherent
This 65-year-old male patient presented with to the brain stem and to the lower cranial
complaints from headache, right-sided weak- nerves and engulfed the vertebral artery with
ness, and gait instability, as well as occasional its small branches. Still, it could be gradually
swallowing difficulty. MR examination (a–e) dissected and removed completely. Frozen
demonstrated a tumor in the lower CP angle, section analysis showed that it was an adeno-
reaching the level of the foramen magnum. It carcinoma metastasis.
showed intense and homogeneous contrast The postoperative period was uneventful.
enhancement and appeared to be dura based. No new neurological deficits appeared. During
Meningioma was hence the most plausible the hospital stay, the strength recovered com-
diagnosis. pletely and the patient was able to walk inde-
The patient was operated via the lateral pendently. The definitive histopathological
suboccipital approach with opening of the study showed that the tumor was an atypical
foramen magnum (f–j). Initially the tumor (WHO grade II) plexus papilloma. Regular
could not be seen, as it was located medially follow-up with MRI every 6 months was rec-
and was hidden by the flocculus. The flocculus ommended. At the first follow-up examina-
was partially resected and retracted, and the tion, the patient had no neurological deficits
lower cranial nerves were followed in the and MRI signs of recurrence.

a b

Preoperative MR images, clearly demonstrating the flocculus, LCN lower cranial nerves, asteriks PICA). (g, h)
tumor and its location. (a) T1-weighted native sequence. The PICA has been dissected and the flocculus slightly
(b and c) T1-weighted sequences following gadolinium elevated, exposing the underlying tumor medial to the
administration. (d) T2-weighted sequence. (e) T2- CISS lower cranial nerves and jugular foramen (Tu).
sequence. Intraoperative images demonstrating the steps (i) Following removal of the main tumor bulk, its caudal
of tumor removal. (f) Initial view of the CP angle (ret- portion was removed between the fibers of the ninth to
rosigmoid approach). The tumor is still hidden by the tenth and the 11th cranial nerves. The vertebral artery
flocculus (arrow seventh to eighth cranial nerves, flocc (VA) was also dissected free from the tumor (j)
716 11 Other Cerebellopontine Angle Tumors

c d

f
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 717

g h

i j
718 11 Other Cerebellopontine Angle Tumors

Ependymomas enhancing or nonenhancing areas [50]. The


T2-weighted and the T2-CISS sequences may
Approximately 70 % of intracranial ependymo- demonstrate the encasement of the major arteries
mas are infratentorial, arising from differentiated and the relation of the tumor to cranial nerves
ependymal cells lining the fourth ventricle and its (Fig. 11.4). Spinal MRI is useful to rule out meta-
lateral recesses. Up to 15 % of these tumors may static disease. The main differential diagnosis of
extend secondarily into the CP angle through the ependymomas is medulloblastoma and astrocy-
foramina of Luschka—Fig. 11.5 [50]. Primary toma. Medulloblastomas, which rarely extend
CP angle ependymomas originate presumably through the fourth ventricular exit foramina, have
from the ependymal cell rests within the foramen a more bulbous extension compared to the thin
of Luschka and extend both laterally into the CP “tongues” of tissue seen in ependymomas.
angle and medially into the fourth ventricle [7, Ependymomas have a propensity for CSF spread.
9, 17]. They rarely invade the floor of the fourth MR study of the whole neuraxis and CSF sam-
ventricle but have a propensity to encase the pling should be performed to evaluate the extent
lower cranial nerves, the posteroinferior cerebel- of CNS involvement and for tumor staging.
lar artery, and the basilar artery [12]. CP angle ependymomas, especially in child-
Radiologically ependymomas appear as irreg- hood, are often quite large before they become
ular lobulated extra-axial tumors in the posterior clinically apparent. Their typical presentation is
fossa (Fig. 11.4). They are hypointense on rather intracranial hypertension than cranial nerve
T1-weighted MR images and hyperintense on deficits.
T2-weighted MR images and have an intermedi- The current standard of care for patients with
ate-to-high FLAIR signal intensity [4, 50]. ependymoma is maximal surgical resection and
Ependymomas have heterogeneous internal evaluation of the presence of other foci [26, 34].
structure due to the presence of calcifications, CP angle ependymomas pose particular surgical
hemorrhages, necrosis, and/or intratumoral cysts. challenges due to the frequent encasement of cra-
The tumor demonstrates irregular enhancement: nial nerves and vessels [41]. Still the major deter-
the strong enhancement of the soft tissue compo- minant of outcome and long-term survival is the
nents of the tumor is intermixed with poorly extent of tumor resection, and every effort should

a b

Fig. 11.5 Ependymoma of the fourth ventricle, extending into the left CP angle via the foramen of Luschka
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 719

be made to achieve a gross total resection—if role of craniospinal irradiation in patients with
safely possible [28, 41] (see Case Illustrations local disease and no evidence of metastasis is con-
11.3 and 11.4). Early postoperative MRI control troversial. In cases of metastatic disease, however,
should be performed to assess the degree of resec- it is certainly indicated. The efficacy of chemo-
tion. A rim of enhancement along the resection therapy is uncertain; it may be recommended only
cavity margins appears soon following surgery. in case of failed surgery and radiotherapy.
Hence, the imaging should be performed less than Regular follow-up of the patients is important
24 h after surgery. In case a residual tumor is to detect early possible recurrences. Most recur-
detected that is deemed safely removable, reop- rences are local at the site of the primary tumor.
eration should be considered. If the residual can- Recurrent ependymomas are managed best by
not be removed completely, radiotherapy is reoperation with involved-field radiotherapy.
usually administered. The necessity of adjuvant Main prognostic factors of survival are com-
postoperative therapy is related to multiple fac- pleteness of resection, patients’ age, histological
tors, such as extent of resection, preoperative tumor grade, and tumor location. Complete
staging, histologic tumor grade, tumor staging, removal (or “gross total resection”) is correlated
and patients’ age. Radiotherapy should be consid- with improved prognosis. Children older than 3
ered in all aggressive tumors and in case of subto- or 4 years of age and infratentorial ependymomas
tally resected nonanaplastic ependymomas. The have a better general prognosis.
720 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.3 capsule was dissected stepwise from the PICA
Ependymoma with Extension in the CP Angle on both sides, from the vertebral arteries, the
Removed Through the Dilated Foramen C1 nerve roots, and the accessory nerve. The
of Luschka removal with an Endoscope- cerebellar tonsils were elevated, and further
Assisted Microsurgical Technique debulking of the intraventricular tumor part
A 22-year-old female patient presented with followed. The tumor extension in the CP angle
complaints of headache since 1 year and could be also removed through the dilated
increasing neck stiffness, nausea, and vomit- foramen of Luschka, applying a combined
ing since 1 month. Her neurological evalua- microsurgical and endoscopic technique. Due
tion revealed truncal ataxia and hyperreflexia to infiltration of the floor of the fourth ventri-
in all four extremities. MRI examination dem- cle in the left lateral recess—the probable site
onstrated a large tumor, occupying the fourth of tumor origin—complete tumor resection
ventricle, extending caudally to the C2 level, was not safely possible. The postoperative
and expanding into the CP angle through the period was uneventful. No new neurological
enlarged left foramen of Luschka (a–f). The deficits appeared, besides a slight temporary
tumor was hypointense on T1-sequences and swallowing difficulty. Figures k–p show the
hyperintense on T2-sequences and had highly early postoperative MRI control. In 4 days,
inhomogeneous internal structure. however, oral feeding was successfully initi-
The patient was operated via the midline ated. Histopathological diagnosis was ana-
suboccipital approach with C1 laminectomy plastic ependymoma (WHO grade III).
(g–i). The tumor part, protruding through the Adjuvant chemo- and radiotherapy was, there-
foramen of Magendie, was debulked initially fore, recommended according to the HIT 2000
with the ultrasonic aspirator. Then, the tumor study protocol.

a b

(a–f) Extensive ependymoma of the 4th ventricle with stay sutures, exposing the tumor (h, i). Elevation of the
extension into the left CP angle through the enlarged lower pole of the tumor allowed to visualize the C1
foramen of Luschka – preoperative MR images. (a–c) roots and the lower cranial nerves (j). (k–p)
T1-weighted sequences following Gadolinium admin- Postoperative MR images, demonstrating the extent of
istration. (d, e) T2-weighted sequences. (f) CISS tumor removal. (k–n) T1-weighted sequences follow-
sequence. (g–j) Intraoperative images of the tumor: ing Gadolinium administration. (o) T2-weighted
the dura is incised in the midline (g) and fixed with 2 sequences. (p) T2- CISS sequence
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 721

c d

e f

g h
722 11 Other Cerebellopontine Angle Tumors

i j

k l

m n
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 723

o p
724 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.4 deterioration, a decision for surgery was


Complete Removal of an Anaplastic taken. In this particular case, the RS approach
Ependymoma seemed to be less appropriate: (1) the large
This 39-year-old male patient had been operated postoperative parenchymal defect provided
twice for a posterior cranial fossa ependymoma sufficient working space and (2) retraction of
at another neurosurgical facility—10 and 7 years the cerebellum would certainly endanger its
earlier. The last histopathological diagnosis was remaining neurological functions and had to
anaplastic ependymoma; combined chemo- and be avoided. The tumor was approached, there-
radiotherapy followed. Five to 6 weeks prior fore, via a median suboccipital craniotomy. It
to presentation, he experienced rapid hearing was very firm and vascularized. The vertebral
deterioration on the right side, accompanied and basilar arteries were engulfed by the
by vertigo. Neurological examination showed tumor, and the lower cranial nerves were
hypesthesia in V1, V2, and V3 branches and compressed and dislocated caudally. Still,
severe hearing loss on the right side, as well following initial internal decompression, it
as dysmetria (right side) and gait ataxia. MR could be dissected from surrounding struc-
examination demonstrated a large tumor in the tures. The seventh to eighth cranial nerves
right CP angle (a–d) with imaging characteris- were also severely compressed but not
tics, compatible with a recurrent ependymoma. infiltrated, which allowed for their safe dis-
Large postoperative parenchymal defect involv- section and preservation. The tumor could be
ing the vermis and part of the right cerebellar removed completely, as shown on the postop-
hemisphere were further seen. A certain growth erative MR imaging (e–h).
tendency was obvious when the tumor size was After the surgery, the patient had swallow-
compared to the previous images. ing difficulties, which rapidly improved.
Considering the size and location of the Interestingly, the hearing also improved to a
tumor and the progressive neurological functional level.

a b

Preoperative MRI of the patient. (a, b) T1-weighted images following gadolinium administration (e, f)
sequences following contrast administration. (c, d) and T2-weighted images (g, h), performed 6 months
T2-weighted sequences. (e–h) Postoperative T1-weighted postoperatively
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 725

c d

e f

g h
726 11 Other Cerebellopontine Angle Tumors

Subependymomas involved by tumors originating in the fourth


ventricle [22].
Subependymomas are low-grade (WHO grade 1) Neuroimaging features of subependymomas
glial neoplasms typically located in the sub- are nonspecific: they are generally isointense or
ependymal regions of the ventricles [6, 15]. hypointense on T1-weighted MR images and
They may become symptomatic at any age hyperintense on T2-weighted MR images [6, 15,
but have a peak incidence in middle-aged and 40]. Heterogeneity is typical and is seen in up to
elderly patients [37]. The tumor is characterized 67 % of the patients [40]. Calcifications are seen
by low proliferative activity and—according to on CT in up to 50 % of the patients [6]. Following
some authors—should be classified as a hama- gadolinium administration, most subependymo-
rtoma rather than as a true neoplasm [35]. mas show at least some enhancement [40].
Subependymomas cause slowly progressive clin- Notably, 21 % of subependymomas do not
ical symptoms or remain asymptomatic and are enhance. Hoeffel et al. [15] even point that when
common incidental finding at autopsy. an adult patient presents with an intraventricular
These tumors generally arise from the floor tumor that is isodense or hypodense and shows
of the fourth ventricle near the foramen of minimal or no enhancement, a subependymoma
Monro or—very rarely—are in relation to the should suspected. Symptomatic or enlarging sub-
central canal of the spinal cord [6]. In contrast ependymomas should be treated actively—com-
to ependymomas, which tend to be paraven- plete surgical removal is the optimal management
tricular, subependymomas tend to be intraven- form and leads to excellent long-term outcome
tricular. The CP angle might be secondarily (see Case Illustration 11.5).
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 727

Case Illustration 11.5 the right CP angle via the foramen of Luschka.
Fourth Ventricle Subependymoma with The patient was operated via the median sub-
Extension in the CP Angle occipital approach, and the tumor, which
This 47-year-old male patient presented with proved to be a subependymoma, could be
complaints of headache and nausea since sev- removed completely (see figures c–j for details
eral months. His neurological examination on the operative technique). After the surgery,
revealed no major abnormalities. The MRI, the patient did not have any neurological
presented on figures a and b showed a tumor in deficits and recovered rapidly. The postopera-
the lower fourth ventricle with extension into tive MRI is presented on figures k and l.

a b

c d

(a, b) Preoperative MR images (a) T1-weighted images tight primary closure. (e) The tumor, protruding trough
following gadolinium administration and CISS images the foramen of Magendie, was visualized. It was deb-
(b). The arrow points at the small tumor extension into ulked carefully, preserving the PICA and its branches.
the right CP angle. (c–j) Consecutive steps of the Then, the tumor capsule was dissected from the sur-
microsurgical tumor removal. (c) Median suboccipital roundings (f–h). (i) The tumor part, extending into the
craniotomy with resection of foramen magnum was left foramen of Luschka and CP angle, was then dis-
performed. (d) The dura was incised parallel to the sected from the cranial nerves and from the vertebral
midline to evade the occipital sinus. Two stay sutures artery and completely resected. The origin of the tumor
have been placed on both sides to provide the required was the left recess of the fourth ventricle (j). (k, l)
exposure. We avoid the classical “Y”-shaped incision Postoperative MRI: complete tumor removal
of the dura because of the difficulty to achieve water-
728 11 Other Cerebellopontine Angle Tumors

e f

g h
Intra-Axial Tumors Extending Secondarily to the Cerebellopontine Angle 729

i j

k l
730 11 Other Cerebellopontine Angle Tumors

Medulloblastomas maximal but safe cytoreduction and restoration


of the CSF circulation. Unlike ependymomas that
Medulloblastomas are neuroepithelial tumors may be more infiltrative to cranial nerves,
arising from remnants of the primitive neuroec- medulloblastomas extend laterally and tend to
toderm in the roof of fourth ventricle. They grow displace (rather than encase) them. Normal arach-
usually in the cerebellar vermis, fill, and often noid plane is usually present, enabling tumor dis-
invade the floor of the ventricle [38]. Less com- section. The combined radio- and chemotherapy
monly the tumor arises in the cerebellar hemi- has been shown to improve the 5-year survival
sphere. The CP angle may be involved due to rate to 55–76 % for high-risk patients and to
lateral tumor extension via the foramina of 70–80 % for standard-risk patients [14].
Luschka or due to exophytic growth of the
tumor, originating from the cerebellum or from
the pons [1, 49]. Lymphomas
On MRI medulloblastomas are hypointense
on T1- and iso-weighted sequences or hyperin- Less than 20 cases of CP angle lymphomas
tense on T2- weighted sequences with possible have been reported in the literature. They
cystic or necrotic components and surrounding develop in the CP angle as primary extra-axial
area of edema [29]. After administration of gado- lymphoma, as a leptomeningeal lymphoma, or
linium, the tumor enhances homogeneously. In as an extension from an intra-axial lymphoma.
adults, however, a more heterogeneous pattern of Lymphomas are hypointense on T1-weighted
enhancement may be seen. MRI of the spine is MR images and hyperintense on T2-weighted
obligatory in order to rule out drop metastases. MR images and enhance after contrast agent
Cytological examination of CSF, obtained via injection [19, 48]. Perifocal edema is typically
lumbar puncture, from the ventricular drain or present and may suggest the intra-axial origin
from the cisterna magna at the time of surgery, is of the tumor. Aggressive removal is not recom-
mandatory for tumor staging. Symptoms dura- mended and adjuvant radiotherapy should be
tion prior to presentation is usually very short. always considered [32].
Patients present frequently with signs of intracra-
nial hypertension, as well as with symptoms or
signs of cerebellar, brain stem, and/or cranial Rare Extra-axial Cerebellopontine
nerve dysfunction. Angle Lesions
Less than 20 cases of CP angle medulloblasto-
mas have been reported in the world literature Extra-axial tumors and other mass lesions devel-
[23, 27, 47]. Some authors suggest that these oping within the CP angle are separated from the
medulloblastomas arise from remnants of the brain parenchyma (brain stem and cerebellum)—
external granular layer in the cerebellar hemi- in contrast to secondary CP angle tumors—by a
sphere, mainly the flocculus, which faces the CP cleft of cerebrospinal fluid. In larger tumors/
angle or from proliferating residue of the lateral lesions, such a cleft is absent, but a characteristic
medullary velum. Medulloblastomas cannot be radiological feature is the presence of vessels
reliably differentiated from other CP angle lesions between the lesion and the parenchyma [64].
before surgery due to their similar clinical and The most frequent extra-axial tumors are
neuroradiological characteristics. schwannomas, meningiomas, and arachnoid cysts.
Clinical outcome of patients with medullo- The remaining lesions comprise less than 1 %:
blastoma varies according to age, postoperative metastases, melanomas and melanocytic tumors,
tumor residuum, and metastatic stage [14]. They craniopharyngioma, pituitary adenoma, lym-
are best managed with surgery, followed by cran- phoma, chloroma, hemangioblastoma, respiratory
iospinal radiotherapy [14, 36]. The role of che- cyst, solitary fibrous tumor, aneurysm, cavern-
motherapy is controversial. Goal of surgery is oma, sarcoidosis, tuberculosis, Erdheim–Chester
Rare Extra-axial Cerebellopontine Angle Lesions 731

disease, neurocysticercosis, and hydatid cysts [64, Arachnoid cysts have smooth and well-defined
83, 96, 119, 141, 166]. borders [107]. These cysts may be difficult
to differentiate from epidermoids of the CP
angle on conventional T1-weighted images
Arachnoid Cysts and T2-weighted images and from other cys-
tic lesions, such as cystic schwannomas, cystic
Arachnoid cysts are congenital collections of meningiomas, and neurocysticercosis (see the
CSF contained within the splitted or duplicated appropriate chapters). Arachnoid cysts are gen-
arachnoidal membrane. According to the most erally more homogenous, have less mass effect,
widely accepted theories, they originate from and may cause adjacent bony reaction, usually
arachnoid splitting due to alteration of the CSF bone scalloping [65]. Furthermore—in contrast
pressure and flow or from duplication of the to epidermoids—they displace adjacent arter-
arachnoid membrane [137]. Duplicated arach- ies and cranial nerves rather than encase them
noid membrane is found histologically in the [63]. Definitive clues for their correct diagnosis
majority of the cases. are provided by fluid-attenuated inversion recov-
The natural history of arachnoid cysts is not ery imaging, diffusion-weighted imaging, and
well known: some of these cysts are quiescent CISS spin-echo MR images [88]. Occasionally,
throughout life, some remain asymptomatic for elevated signal intensity on T1-weighted images
many years before showing clinical manifesta- can be seen, which is due to a higher protein con-
tions, while other enlarge progressively and cause tent of the cyst [144]. Dynamic MRI technique,
compression of adjacent structures or CSF flow as well as CT cisternogram, could allow differen-
disturbance. Spontaneous reduction is observed tiating between communicating and noncommu-
very rarely [142]. Cyst enlargement may be due nicating with the CSF arachnoid cysts.
to active secretion from the cyst wall and passive The large majority of arachnoid cysts are
fluid diffusion into the cyst due to the existing asymptomatic and found incidentally. They may
osmotic gradient or intracystic hemorrhage. become symptomatic by causing compression or
Communicating cysts may grow due to the pres- stretching of surrounding structures or—more
ence of a functional one-way valve between the rarely—by causing hydrocephalus [100, 144].
subarachnoid space and the cyst. Spontaneous or traumatic intracystic hemorrhage
Ten to 11 % of all of intracranial arachnoid has rarely been described in the posterior fossa
cysts are located in the CP angle, which is the [103]. The most frequent presenting symptom in
second most common location of such cysts after the series of Jallo et al. [107] was headache, while
the Sylvian fissure area [57, 60, 142, 144, 147]. in the series of the senior author it was vertigo/
Arachnoid cysts may be located only in the CP dizziness [144], followed by hearing loss, gait dis-
angle (58 % of the cases according to our previ- turbance, tinnitus, and trigeminal dysfunction.
ous study [144]) or may extend dorsally to the The management of patients with arachnoid
brain stem (25 %) or even into the IAC (17 %). cysts is determined by their clinical and biologi-
Rarely, arachnoid cysts in adults may remain cal behavior. Asymptomatic small cysts may be
confined within the IAC [68, 147]. The senior followed with regular MRI, clinical, and audio-
author has operated three patients with pure intra- logical control. In case enlargement of the cyst is
canalicular arachnoid cysts. noted, surgery may be indicated. Symptomatic
The typical CT and MR appearance of arach- cysts, especially those causing clinical worsen-
noid cysts is that of a noncalcified homogeneous ing, should be treated actively, since their removal
extra-axial lesion, with imaging characteris- leads to clinical improvement and the related
tics similar to those of CSF. It is hypointense morbidity rate is very low [107, 144].
on T1-weighted sequences and hyperintense Surgical options include cyst aspiration, endo-
on T2-weighted images and does not enhance scopic or microsurgical cyst fenestration, cysto-
after administration of gadolinium [65, 116]. peritoneal shunt, cyst marsupialization into the
732 11 Other Cerebellopontine Angle Tumors

subarachnoid space, and complete or partial cyst cyst wall and the surrounding neurovascular
resection [51, 107, 121, 144]. structures is found, small parts of the capsule
We favor open surgery via the RS approach, may be left unremoved (see Case Illustrative
which the safest technique. It allows to confirm 11.6). In cases of posterior fossa communicating
the diagnosis, to coagulate any arachnoidal blood arachnoid cysts with associated hydrocephalus, a
vessels, and to fenestrate multiloculated cysts VP shunting may be sufficient. However, if pre-
[107]. Some authors recommend resecting only operative symptoms do not recover and the cyst
the medial, lateral, and posterior membranes of does not shrink, open surgery should be consid-
the cyst and fenestrating the anterior membrane ered [144].
into the cisterns of the CP angle. Based on the Generally, most symptoms regress after sur-
experience with 28 surgically treated cases (MS), gery. In our experience, the headache, vertigo,
we believe that radical resection of the cyst and dizziness, and ataxia improved in the majority of
careful removal of the cyst parts that are adherent patients. Hearing dysfunction, however, improved
to vessels and cranial nerves reduce the risk of only rarely: hypacusis improved in two patients,
recurrences (see Case Illustrative 11.7). Of remained unchanged in three patients, and
course, if extreme dense adherence between the became worse in two patients.
Rare Extra-axial Cerebellopontine Angle Lesions 733

Case Illustration 11.6 nerve complex (7–8) and the fifth nerve (f; T
Arachnoid Cyst (Incomplete Resection tentorium). The outer membrane of the cyst
of the Medial Cyst Wall) was relatively firm and non-translucent. It was
This is a 22-year-old female patient, com- opened and resected with microscissors (g).
plaining of hearing loss on the right side, The inner or medial wall was densely attached
headache, and vertigo for 15 months. The neu- to the seventh to eighth cranial nerves, as well
rological examination revealed no pathologi- as to the AICA branch, passing superior to the
cal signs besides a profound right-sided nerves (h). An attempt to resect the medial
hearing loss. MR examination demonstrated a cyst wall was therefore not made. Therefore, a
cystic lesion in the right CP angle, compatible wide fenestration to the medial subarachnoid
with an arachnoid cyst (a–e). cisterns was made (figures i, j). The patient
The CP angle was exposed via the RS recovered very rapidly from surgery and had
approach, and the arachnoid cyst was seen in no new neurological deficits. Postoperative
its upper part between the seventh and eighth MR images: k–n.

a b

(a–e) MR images of the patient, demonstrating the cyst nerves and brainstem. (f–j) Intraoperative images, pre-
in the CP angle. The contents of the cyst have signal senting the removal of the cyst. See the text for expla-
characteristics, identical with those of the CSF. Note nations. (k–n) Postoperative MRI: T1-weighted (k, l),
the mass effect on the adjacent neural structures: cranial T2-weighted (m), and T2- CISS sequences (n)
734 11 Other Cerebellopontine Angle Tumors

c d

h
Rare Extra-axial Cerebellopontine Angle Lesions 735

i j

k l

n
736 11 Other Cerebellopontine Angle Tumors

Case Illustrative 11.7 cranial nerves (g). Initially, a small fenestra-


Arachnoid Cyst (Complete Removal) tion of its outer membrane away from the cra-
Nine-year-old girl complaining of headache nial nerves at a nonvascular site was made
since 4–5 months. Her neurological status was with microscissors (h). The remaining mem-
normal. MRI showed a cystic lesion in the right brane was resected under direct control using
CP angle with diameter of 2.6 × 1.8 × 1.7 cm. both hands: the left hand was holding the cyst
Notably, a small contrast-enhancing part was and the right hand was holding the microscis-
seen at the upper pole of the cyst, and the pre- sors. No mural nodule was found, and the
sumptive histological diagnosis was pilocytic results from the histological examination
astrocytoma. Therefore, the decision for surgi- revealed an arachnoid cyst (i, j). Then the
cal removal of the lesion via the RS approach remaining part of the membrane was carefully
was taken. dissected from the lower cranial nerves and
The cyst was located in the lower CP angle, from the brain stem and removed in order to
between the seventh and eighth and the lower prevent future recurrences (k, l).

a b

Preoperative MRI: (a, b) T1-weghted sequence after See the text for explanations. (m–o) Postoperative
gadolinium administration. (c) T2-weighted sequences. T1-weighted images after gadolinium administration
(d, e) CISS sequences. (f) Dark fluid sequence. (g–l) (m, n) and T2-weighted MR images (o)
Consecutive steps of surgery (arrow - arachnoid cyst).
Rare Extra-axial Cerebellopontine Angle Lesions 737

c d

e f
738 11 Other Cerebellopontine Angle Tumors

g h

i j

k l
Rare Extra-axial Cerebellopontine Angle Lesions 739

m n

o
740 11 Other Cerebellopontine Angle Tumors

Dermoids angle is, therefore, not a favorable location of


dermoids.
Dermoids are rare congenital nonneoplastic cys- Dermoids grow slowly and tend to produce
tic lesions with a capsule composed of epidermal symptoms due to local compressive effect in the
elements and dermal derivatives, such as hair fol- CP angle or cause obstruction of CSF pathways
licles, sebaceous, and even sweat glands. They (Fig. 11.6). Hydrocephalus is frequently the
contain thick, buttery material that results from cause of initial signs and symptoms. Infectious
epithelial desquamation and sebaceous gland complications—bacterial meningitis or cerebel-
secretion, as well as variable amount of hair or lar abscess—occurring due to bacterial contami-
even teeth [69]. A dermal sinus tract may connect nation via the dermal tract, may be the initial
the cyst to the skin surface. Dermoid cysts are presentation of these tumors [164]. Compression
less common than epidermoid cysts. They tend to of the trigeminal nerve can cause trigeminal neu-
be located in the midline, while epidermoid cysts ralgia [108]. Rare complication is rupture of the
tend to occur in a more lateral position. The CP dermoid and distribution of its content along the

a b

c d

Fig. 11.6 CT scan (a, b) and T1-weighted MR contrast- Note the extension in both CP angles and the severe com-
enhanced images (c, d) of a patient with very large der- pressive effect on the brain stem
moid cyst of the posterior and left middle cranial fossae.
Rare Extra-axial Cerebellopontine Angle Lesions 741

CSF. This can remain asymptomatic or cause a radical removal and preservation of all neurologi-
wide range of symptoms including aseptic men- cal functions.
ingitis, hydrocephalus, cranial nerve deficit, or As in all CP angle tumors, the cyst is opened
epilepsy [70, 133]. and internal decompression is performed initially.
Dermoid cysts have various CT and MRI Then the cyst wall is then dissected. En bloc
appearance depending on the fat content of the removal, which is recommended for dermoids in
tumor (Fig. 11.6). They are well-circumscribed other locations, is not safely feasible in the CP
lesions with a thick peripheral occasionally angle. Similarly to CP angle epidermoids, if a
enhancing capsule [63]. Most frequently they small part of the capsule is very densely attached
have very high signal intensity on T1-weighted to a cranial nerve or to the brain stem, it should be
and low signal intensity on T2-weighted MR left. If the dermoid is associated with dermal
images. As pointed by Yasargil et al. [165], sinus tracts, both should be removed during the
dermoid tumors have a similar appearance to surgery. The sinus tract should be dissected very
epidermoids but with a wider range of attenua- carefully from the surrounding healthy tissue and
tion values. Calcifications are seen in approxi- bone and excised.
mately 20 % of the dermoids but are very rare Dermoids have a very low growth potential
in epidermoid tumors [98]. Diffusion-weighted and the risk for recurrence is low. Spillage of cyst
MR imaging allows for their differentiation: contents is to be limited to prevent recurrences or
the typical for dermoids hyperintensity aseptic meningitis. The risk is, however, rela-
is related to the decreased water proton diffu- tively low. In one case operated by the senior
sion [133]. author, extensively disseminated fat particles in
CP angle dermoids may be a greater operative the subarachnoid space were found after surgery
challenge than epidermoids due to the younger (see Case Illustration 11.8). During the 8 years of
age of most patients and the more rapid tumor follow-up, neither neurological deterioration nor
growth [133, 165]. Their capsule is thicker than growth of the multiple lesions was seen [70]. The
that of epidermoids and may be tightly attached secondary trigeminal neuralgia has a good out-
to adjacent structures. Goal of surgery is their come after the removal of the dermoid [108].
742 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.8 moid tumor. After surgery the patient had
CP Angle Dermoid Tumor incomplete oculomotor paresis and slight hypa-
A 36-year-old woman presented with right- cusis on the right side. On the fifth postopera-
sided trigeminal neuralgia (V3) for 4 years. tive day, aseptic meningitis was diagnosed.
Besides a V3 hypesthesia on the right side, her Treatment with dexamethasone (8 mg, 3 times/
neurological examination was unremarkable. day for 1 week) led to rapid resolution of his
CT and MRI were performed and demon- symptoms. The accurate surgical manipula-
strated a relatively large tumor in the medial tions and the thorough irrigation of the opera-
CP angle area on the right side that compressed tive field after tumor removal did not prevent
severely the brain stem (a). The lesion was spillage of the cyst contents in this particular
hyperintense on T1-weighted and heteroge- case, which led to subarachnoid dissemination
neous—iso- and hypointense—on T2-weighted of fatty material (a–d) and postoperative chem-
MR sequences (b–d). ical meningitis. Nevertheless, the patient recov-
The tumor was exposed via the RS approach: ered quickly and was discharged in a good
it was partially cystic and contained yellow condition on the 15th postoperative day. The
fluid. Its capsule could be dissected completely neurological deficits resolved completely
from the involved third, fourth, fifth, seventh, within several months. Annual MRI follow-up
and eighth cranial nerves and from the brain studies showed no changes in the size and loca-
stem. The histopathological diagnosis was der- tion of the subarachnoid fat particles.

a b

(a) CT scan showing the strongly hypodense tumor. noid dissemination of fatty material, most probably
(b, c) The lesion was hyperintense on T1-weighted from the cyst contents. The disseminated lipid droplets
MR images. (d) On T2-weighted sequences, the tumor have hyperintense signal on T1-weighted images and
was more heterogeneous with isointense and hypoin- hypointense signal on T2-weighted images (Reprinted
tense zones. (e–h) T1-weighted and T2-weighted MR from: Carvalho et al. [70]. Copyright © 2000, with
sequences, demonstrating the widespread subarach- permission from Wolters Kluwer Health)
Rare Extra-axial Cerebellopontine Angle Lesions 743

c d

e f

g h
744 11 Other Cerebellopontine Angle Tumors

Hamartomas histopathological examination, usually intraop-


eratively when a frozen section examination is
Intracranial hamartomas are considered to be performed. These tumors are typically firmer
nonneoplastic lesions formed as a result of abnor- than VSs and are more tightly attached to the
mal embryologic development. Glioneural hama- seventh and eighth nerve complex. Attempt at
rtomas involving the IAC are very infrequent: complete tumor resection may be related to high
only seven cases have been reported in the litera- risk of injury to neural structures. Therefore, in
ture [97]. case the tumor cannot be safely dissected or the
Radiologically hamartomas cannot be reli- dissection causes lasting changes of the electro-
ably distinguished from VSs. Their clinical pre- physiological monitoring parameters (EMG,
sentation is also nonspecific and is related to BAEP, or facial nerve MEP), this most adherent
facial or vestibulocochlear nerves dysfunction part of the lesion should be left unremoved (see
[71]. The diagnosis of hamartomas is made at Case Illustration 11.9).
Rare Extra-axial Cerebellopontine Angle Lesions 745

Case Illustration 11.9 and adjacent structures was missing, and both
IAC Hamartoma the facial and cochlear nerves were infiltrated.
A 40-year-old man presented with progressive Even the slightest manipulations of the lesion
hearing loss and tinnitus on the right side for a caused major changes of the BAER (c) and of
period of 16 months. The neurological exami- the facial nerve EMG. Complete resection
nation revealed only hypacusis on the right with functional nerve preservation of these
side: the hearing loss according to the audio- nerves was hence deemed impossible and the
gram was 30 dB in the main speech area. tumor was removed partially (d, e). The
Enhanced MR T1-weighted images demon- patient recovered very rapidly from surgery.
strated a small lesion in the lateral one-third of He had normal facial nerve function and a
the IAC, which was supposed to be VS (a, b). slightly deteriorated hearing function—hear-
The patient was operated via the RS ing loss of 40 dB in the main speech area.
approach, and the posterior wall of the IAC Histopathological result was glioneural hama-
was removed widely up to the fundus. The rtoma. The residual lesion remained
tumor was found to have very hard consis- unchanged for the period of follow-up (>18
tency. Arachnoid plane between the tumor months).

a b

(a, b) Native and contrast-enhanced T1-weighted MR view of the lesion, located in the lateral third of the
study of the patient. The lesion is seen only after gad- IAC (the posterior wall of the IAC has been removed
olinium administration (b, arrow). (c) BAEP moni- widely up to the fundus). (e) Partial removal of the
toring, demonstrating the reversible changes that lesion and preservation of the facial and cochlear
occur due to various surgical manipulations. Note the nerves (Reprinted from: Carvalho et al. [71].
repeated decrease of the amplitude and loss of com- Copyright © 2003, with permission, Wolters Kluwer
ponents, in particular of waves I, II, and III. (d) Initial Health)
746 11 Other Cerebellopontine Angle Tumors

II IV
C I III V
Healthy side

Tumor side, start

After drilling: hard tumor in IAC

Mobilization in IAC: wave I splitting

Dissection in IAC:
wave I amplitude decrease
Puting medial, away from function:
no waves

Release: V returns

Tumor biopsy: I, III and V present

Observation

Further resection: I and III lost

Release

Slow recovery, histology unclear

Decision: priority to function


partial resection

Complete wave recovery

150 w

0 10 m
Rare Extra-axial Cerebellopontine Angle Lesions 747

d e
748 11 Other Cerebellopontine Angle Tumors

Lipomas T1 sequences, the tumor is hyperintense and does


not enhance after gadolinium application [67, 82].
Lipomas are rare extra-axial CP angle tumors: No signal on fat-suppression sequences is charac-
approximately 100 cases have been reported in teristically seen.
the literature [105]. They are estimated to repre- Patients with CP angle lipomas are followed
sent 0.14 % of all CP angle and IAC tumors with regular neuroimaging and clinical control.
[158]. Patients with CP lipomas typically present Surgical treatment is rarely indicated—in case of
with cranial nerve symptoms/signs: hearing loss, intractable cranial neuropathies or brain stem com-
tinnitus, dizziness, trigeminal neuralgia, and/or pression [67, 105, 122, 158]. Another indication
facial neuropathies [67]. for surgery may be to achieve definitive histologi-
Lipomas are reliably diagnosed with T1-weighted cal diagnosis, for example, in case liposarcoma
and fat-suppression MRI (Fig. 11.7a, b). On native is suspected [82]. Lipomas are typically well

a b

Fig. 11.7 This 50-year-old female presented with a his- on T2, no mass effect or contrast enhancement). At sur-
tory of hearing loss, vertigo, tinnitus, and gait instability gery, the diagnosis was confirmed and a partial removal
for 2 years. CT (a) and MRI (b) demonstrated characteris- (internal debulking) was performed (c) (Reprinted from:
tic abnormal signal within the CP angle (arrow) that was Schuhmann et al. [146])
consistent with lipoma (hyperintense on T1, hypointense
Rare Extra-axial Cerebellopontine Angle Lesions 749

vascularized, do not respect natural borders, and are or the respiratory buds. Their frequent associa-
densely adherent or infiltrate surrounding tissues, tion with other congenital malformations is
in particular the brain stem and cranial nerves. The regarded as an indirect confirmation of this the-
attempt to remove them completely may be very ory [91]. The walls of the cyst is lined with sim-
hazardous. The goal of surgery should be rather ple or pseudostratified cuboidal-to-columnar
decompression of neural structures than complete epithelium with a basement membrane resem-
tumor removal. Tankere et al. [158] reviewed the bling those of respiratory and intestinal tracts
treatment outcome in 98 patients with CP lipomas mucosa, while the cyst is filled with mucin-like
(literature and own material). In most cases (67 %), secretions [101]. This fluid secretion leads to pro-
partial tumor removal was performed. After sur- gressive cyst enlargement. The intracystic fluid is
gery, the existing neurological symptoms remained usually milky, xanthochromic, or transparent
unchanged in 9.2 % and improved in 50 % of the [91]. The endodermal origin of neurenteric cysts
patients. New postoperative deficits developed in determines their immunohistochemical charac-
two-thirds of the patients. Preservation of hearing teristics. They usually stain positively for keratin,
was possible in only 26 % of the patients. carcinoembryonic antigen, and epithelial mem-
brane antigen and negatively for glial fibrillary
acidic protein, neuron-specific enolase, S100,
Neurenteric Cysts and vimentin.

Neurenteric cysts, also known as enterogeneous, Neuroimaging


endodermal, respiratory, or bronchogenic cysts, The imaging characteristics of neurenteric cysts
are rare benign congenital lesions of the central can be quite variable due to the presence of pro-
nervous system [59, 89, 143]. They occur at any teinaceous or other hydrophilic contents [75]. CT
level of the neuraxis but most commonly are usually reveals a well-defined, noncalcified
found in the lower cervical and upper thoracic hypodense lesion. On T1-weighted MR images,
regions [53]. They are generally located intradur- they may appear hypointense, isointense, or
ally ventral to the spinal cord [85]. Intracranial slightly hyperintense, depending on the content
neurenteric cysts are even more rare with the of the cyst. They are typically hyperintense on
majority—more than 90 %—located in the poste- T2-weighted and FLAIR images. Peripheral rim
rior fossa [52, 59, 89]. The typical location of enhancement of the cyst wall is very rarely seen
such cysts is along the midline: anterior to the [63, 91, 143].
brain stem (50 %), within the fourth ventricle The signal intensity of the cyst depends on its
(21 %), or extending into the cervical canal protein content. It can rarely mimic CSF when
(18 %) [59]. The CP angle is involved in 17 %. this content is low [74], but it is often isointense to
Spinal neurenteric cysts are associated fre- hyperintense relative to brain parenchyma on
quently (in 40–69 % of the cases) with other con- T1-weighted images and hyperintense on
genital malformations: vertebral body dysgenesis, T2-weighted images when the protein concentra-
meningomyeloceles, segmentation abnormalities, tion is high. Neurenteric cysts have low signal
spina bifida, and respiratory or enteric tract malfor- intensity on DWI sequences (Fig. 11.8g). These
mations [131]. They may be connected by a fibrous imaging features may enable their differentiation
tract, fistula, or cleft to structures derived from the from epidermoid cysts, which may exhibit similar
primitive gut in the thoracic or abdominal cavities intensities on conventional sequences but have
[62]. Contrary to that, intracranial neurenteric cysts characteristic high signal intensity on DWI [149].
are typically isolated congenital malformations.
Neurenteric cysts are thought to arise during Clinical Presentation
the embryologic development of the notochord Neuroepithelial cysts are frequently detected
due to a failed dissolution of the transient neuren- incidentally. Occasionally they may produce pro-
teric canal between the notochord and the foregut gressive neurological symptoms and/or chronic
750 11 Other Cerebellopontine Angle Tumors

a b

c d

Fig. 11.8 Typical MR characteristics of neurenteric cysts. (a) T1-weighted native sequence. (b–d) T1-weighted
sequences after gadolinium administration. (e) T2-CISS sequence. (f) FLAIR sequence. (g) DWI sequence

increased intracranial pressure [59]. Clinical Management


symptoms are due to the enlargement of the cyst, The treatment of choice of neurenteric cysts is
caused by active secretion by the goblets cells complete excision of both the cyst contents and its
and compression of neighboring neurovascular capsule (see Case Illustrations 11.10 and 11.11).
structures. Rarely, small microcysts can rupture Importantly, the dissection should not cause injury
into the subarachnoid space, leading to recurrent to the neighboring neurovascular structures
aseptic meningitis [143]. [59, 91]. In case of strong adhesion to surrounding
CP angle cysts lead to dysfunction of the fifth, structures, small parts of the cyst wall may be left
seventh, eighth, or of the lower cranial nerves and in place to avoid postoperative morbidity (see Case
corresponding symptoms. Further common com- Illustration 11.10). Close clinical and neuroradio-
plaints are headaches, gait disturbance, and motor logical follow-up of these patients should be per-
and sensory disturbances. formed to detect early possible cyst recurrence.
Rare Extra-axial Cerebellopontine Angle Lesions 751

e f

g
752 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.10 cerebellum (e). The cysts’ capsule was opened,
Neurenteric Cyst 1 and the fluid contents, which were yellowish
The CP angle tumor in this patient was discov- and milky, were aspirated (f, g). The capsule
ered incidentally after a road traffic accident. was then dissected stepwise; the more solid
At presentation he did not have any symptoms tumor parts were exposed and also removed
and the neurological examination was normal. (h, i). Although the capsule was found to be
MRI showed a lesion with a predominant loca- tightly adherent to the brain stem and basilar
tion in the left CP angle with an extension to artery at certain points, it could be dissected
the contralateral side and encasement of the from them and excised. The very narrow CP
basilar artery (a–d). The lesion was molded angle provided insufficient access to the con-
around the brain stem with a relatively less tralateral tumor part without endangering the
compressive effect, indicating its possible soft function of the cochlear and facial nerves,
consistence. The presumptive diagnosis was even if the angled endoscope was used (j).
epidermoid tumor. After thorough discussion Therefore, no attempt was made to remove the
with the patient and his family, it was decided contralateral extension of the cyst.
to proceed with surgery in order to avoid neu- Histopathologically, the lesion was found to
rological deterioration in the future. The man- be a neurenteric (respiratory) cyst with focal
agement concept was to remove the major squamous epithelium metaplasia without ana-
tumor bulk via a left-sided retrosigmoid plastic features and hemosiderin in the cyst
approach and—if necessary—to remove the wall, indicating old hemorrhages.
remaining part via a second right-sided The postoperative period was uneventful;
retrosigmoid craniotomy later on. the patient did not have any neurological
The RS approach was performed, the lat- deficit and recovered rapidly. The CT per-
eral cerebellomedullary cistern was opened, formed 1 day after surgery is shown on figure
and sufficient CSF was drained, thus exposing k. During a follow-up of more than 60 months,
the cystic tumor without any retraction of the the cyst remnant remained unchanged.

a b

Preoperative CT (a) and MR images (b–d) of the aspiration of its yellowish contents via the level
lesion, which was hyperintense on both T1- and between the seventh and eighth and the lower cranial
T2-weighted sequences. Consecutive steps of the cyst nerves (f, g) and caudal to the lower cranial nerves
removal. (e) Initial view of the cyst (arrows) located (h). Resection of the safely accessible cyst capsule (i).
medial to the seventh–eighth (7–8) and the lower (j) View of the CP angle at the end of surgery. (k)
cranial nerves (asterik). (f) Opening of the cyst and Postoperative CT scan (first postoperative day)
Rare Extra-axial Cerebellopontine Angle Lesions 753

c
d

e f

g
h
754 11 Other Cerebellopontine Angle Tumors

i j

k
Rare Extra-axial Cerebellopontine Angle Lesions 755

Case Illustration 11.11 Following the opening of the dura, a multicys-


Neurenteric Cyst 2 tic tumor was seen, located around the verte-
This 46-year-old female patient presented bral artery and C1 rootlets. The cysts’ fluid
with a history of fluctuating weakness of the was thick and milky; its wall was adherent to
right leg for 6 months. Her neurological exam- the brain stem and vertebral artery. Still, using
ination revealed no abnormalities. MR imag- the bimanual preparation technique, the lesion
ing demonstrated an extra-axial lesion in the could be removed completely. An angled
foramen magnum area that extended caudally endoscope was used at the end to examine the
to the C2 level. The lesion was located ante- whole surgical field, including the premedul-
rior and lateral to the medulla and spinal cord lary space and to verify the completeness of
and had a slight compressive effect. It was resection.
isointense on T1-weighted images and slightly The patient recovered quickly from the sur-
hypointense on T2-weighted images (a–d). gery and did not have any neurological deficits.
The patient was operated via a lateral suboc- Her early postoperative MRI is presented on
cipital approach with a limited opening of the figures e–g. Histopathological examination
foramen magnum, C1 hemilaminectomy, and proved that the lesion was a neurenteric (respi-
partial C2 hemilaminectomy on the right side. ratory) cyst without signs of malignancy.

a b

c d

(a–d) Preoperative T1- and T2- weighted MR images, demonstrating the extension of the lesion and its relation
to the surrounding structures. (e–g) Postoperative MRI control: no remnants can be visualized
756 11 Other Cerebellopontine Angle Tumors

e f

g
Rare Extra-axial Cerebellopontine Angle Lesions 757

Hemangioblastomas radical surgery can cause severe morbidity.


Other tumors arise from the choroid plexus,
Cushing and Bailey introduced the term heman- develop in the ventricle, and do not invade the
gioblastoma in 1928: it refers to a benign vas- brain stem. The rare cervicomedullary heman-
cular neoplasm that occurs most frequently in gioblastomas may extend secondary into the
the posterior cranial fossa. Hemangioblastomas CP angle.
account for 8–12 % of all tumors in this area With respect to their internal structure, heman-
[78, 79, 139]. They are slow-growing tumors gioblastomas can be cystic, solid, or mixed [152].
of uncertain histological origin and occur spo- Most frequently they are composed of a cherry
radically or as a part of Von Hippel–Lindau red solid nodule, associated with a cyst that con-
disease (VHL). Sporadic tumors present at tains a clear yellowish fluid.
40–50 years of age and involve predominantly Hemangioblastomas are most reliably diag-
the cerebellum. Hemangioblastomas in patients nosed on MRI (Fig. 11.9). On native T1-weighted
with VHL present earlier—at 20–30 years of sequences, the solid tumor part is hypointense or
age—and involve the cerebellum, brain stem, isointense; on T2-weighted images, it is hyperin-
or spinal cord [128]. Such hemangioblastomas tense [63, 117]. Strong enhancement after admin-
may be multiple. According to some studies, istration of contrast media with clear delineation
up to 20 % of patients with VHL have lower from surrounding neural tissue is typical for
brain stem or craniocervical hemangioblasto- hemangioblastomas. The cyst—if present—is
mas [78, 92, 135]. Some of them arise in the sharply demarcated and has homogeneous CSF-
area of the obex or in the posterolateral aspect like signal intensity: hyperintense on T1-weighted
of medulla oblongata and may infiltrate the images and hyperintense on T2-weighted images
floor of fourth ventricle. Distinct margin to [63]. Cerebral angiography, which is currently
neural tissue is usually absent, and attempt at largely replaced by MR angiography, demonstrates

a b

Fig. 11.9 MR images of a patient with multiple heman- type, and the rather mild symptoms, a decision was made
gioblastomas, including a large right-sided cystic CP to perform tumor embolization. In case of further growth
angle hemangioblastoma. An opened biopsy has been or neurological deterioration, surgical excision would be
performed at another institution and an Ommaya reser- recommended. The follow-up of the patient is 12 months
voir was inserted. The patient presented because of con- and the tumor is stable. (a–c) T1-weighted sequences
tinuous tumor enlargement. Considering the radiological after gadolinium administration. (d, e) T2-weighted
tumor characteristics, in particular the absence of clear sequences. (f) Multiple scattered supra- and intracranial
plain in relation to the brain stem, the histological tumor hemangioblastomas
758 11 Other Cerebellopontine Angle Tumors

c d

e f

Fig. 11.9 (continued)

the highly vascular tumor blush. High-flow vessels benefits, such as in patients with poor general
may be observed on MRI as flow voids at the condition. In case of critical location of the tumor
periphery of the mass [63]. A thorough medical with infiltration of important neurovascular struc-
evaluation for other stigmata of VLH, including tures, the attempt at complete removal may have
MR study of the neuraxis, abdominal CT scan or devastating consequences (see Case Illustration
ultrasound, and ophthalmological examination, as 11.12). Staged multimodal management includ-
well as detailed family history, to exclude multiple ing embolization, surgery, and radiosurgery can
hemangioblastomas should be performed prior to be beneficial in such patients [73, 154].
surgery. Asymptomatic lesions in patients with multiple
Surgical removal is considered the standard hemangioblastomas may be initially observed
treatment of hemangioblastomas [78, 154]. They with regular MRI examinations to rule out tumor
are benign tumors and may be cured by complete enlargement.
surgical excision. Exceptions are those cases, in Preoperative endovascular embolization of the
which the risk of operation outweighs its potential solid tumor component before its resection is very
Rare Extra-axial Cerebellopontine Angle Lesions 759

useful in large hypervascular hemangioblastomas • The tumor is usually well demarcated from
of the posterior fossa—see Case Illustration 11.12 the surrounding brain or spinal cord and has a
[155]. It reduces the tumor vascular supply and pre- clear gliotic margin.
vents major intraoperative bleeding, minimizing the • Its removal is performed by circumferential
risk of neurological morbidity [156, 160]. However, dissection along the gliotic margin and divid-
as noted by some authors [115], even superselective ing its vascular supply.
embolization of large hemangioblastomas, espe- • The surface of the tumor may be coagulated,
cially those located in the medulla oblongata, is not thereby shrinking the tumor. Penetration of
without infarction hazards and is not always safely the tumor itself should be avoided because of
feasible. Occasionally the embolization inhibits its extreme vascularity.
tumor growth control and even causes tumor shrink- • Attempt to remove a hemangioma piecemeal
age. Surgery may be postponed and is required only can lead to uncontrollable hemorrhage.
if the tumor continues to enlarge. • The cyst if present is not composed of tumor
The operative strategy should be selected indi- cells; hence, its wall should not be removed.
vidually, according to the location and size of the • In case a distinct dissection plane is absent or
tumor and of its feeding vessels. Important points the tumor infiltrates neural or major vascular
to consider are: structures, no attempt should be made to
• Early identification and coagulation of feed- remove it completely.
ing vessels. Radiotherapy (fractionated radiotherapy or
• Arterial feeders should be coagulated prior to radiosurgery) is an adjunct following incomplete
draining veins. tumor removal or alternative treatment in patients
• In case a large cyst exists, its decompression in poor general condition that obviates major sur-
allows for improved access and easier delinea- gery [109, 111]. Of note, hemangioblastomas
tion of the interface between tumor and neural with large cystic components are not good candi-
parenchyma. dates for radiosurgery.
760 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.12 feeders form the vertebral artery was made.
CP Angle Hemangioblastoma However, selective occlusion of the tumor
This 25-year-old male patient had been oper- feeders only turned out to be impossible.
ated in another neurosurgical facility for a The patient was operated via the RS
large tumor in the lower CP angle 5 months approach. Exposure of the hemangioblastoma
prior to presentation. A biopsy was taken required meticulous dissection of the severe
and—due to extremely strong bleeding from adhesions caused by the previous surgery. The
the tumor—several clips were placed; the sur- tumor was extremely vascularized and even
gery was interrupted. The patient remained slightest manipulation caused strong bleeding
ventilated for several weeks and then slowly (e). Due to its large size, critical location, and
recovered. Histopathological diagnosis was infiltration of adjacent structures, circumferen-
hemangioblastoma. At presentation he com- tial dissection was not a reasonable option.
plained of deteriorating gait and frequent Alternating bipolar coagulation and removal of
vomiting. He had horizontal nystagmus and the tumor with the ultrasonic aspirator were
incomplete hypoglossal palsy on the right therefore initiated (f). Gradually the caudal and
side. Gag reflexes were absent. Babinski sign dorsal parts were dissected and resected from
was positive on the left side. The patient had the brain stem. The lower cranial nerves were
severe gait and truncal ataxia and was unable engulfed by the tumor. Nevertheless, we pro-
to walk independently. His previous history ceeded with tumor removal until the seventh to
was remarkable for “treatment of angiomas” eighth nerves were seen. In the root exit zone
(lack of precise records) on both eyes. of the facial nerve, the adhesions were so
MRI study showed a large strongly enhanc- strong that further resection would inevitably
ing tumor in the lower brain stem area and CP damage the nerve. The caudal limit of the
angle (a–d). The tumor extended via the lat- resection was the tumor part, engulfing the ver-
eral recess into the fourth ventricle and caused tebral artery and its small branches. In general,
significant brain stem compression. DSA significant volume reduction of the tumor and
demonstrated a highly vascular lesion with brain stem decompression was achieved (g).
supply from the vertebral and the external Following surgery, the patient had incom-
carotid arteries. plete abducens nerve palsy on the right side
Considering the tumor size, the severe and was unable to swallow, so that a tracheos-
brain stem compression, and the progressive tomy was placed. After a prolonged intensive
clinical deterioration of the patient, a decision care management (4 weeks), he could be
was made in favor of new attempt to remove transferred to a normal ward. His condition
the lesion surgically. Initially superselective gradually improved. The swallowing recov-
embolization of the external vascular feeders ered and the tracheostoma could be closed. At
was performed. At a second stage immediately discharge the patient was able to take care of
before surgery, an attempt to embolize the himself and walk without support.
Rare Extra-axial Cerebellopontine Angle Lesions 761

a b

c d

Preoperative T2-weighted (a) and T1-weighted MRI of initial view of the tumor (e), partial removal (f), and
the tumor following gadolinium administration (b–d). view at the end of surgery (g). Postoperative T2- (h)
Note the flow voids, corresponding to large vessels in and T1-weighted MR sequences after contrast applica-
the periphery of the tumor (a). Intraoperative images: tion (i–k) demonstrating the extent of tumor resection
762 11 Other Cerebellopontine Angle Tumors

e f

g
Rare Extra-axial Cerebellopontine Angle Lesions 763

h i

j k
764 11 Other Cerebellopontine Angle Tumors

CP Angle Metastases

Most secondary CP angle tumors are benign and are


usually of neuroectodermal origin. As recalled by
Tu et al. [159], Cornil reported the first case of CP
angle metastasis in the 1930s. With the widespread
of modern neuroimaging, secondary metastatic
tumors to the CP angle and IAC are increasingly
frequently being diagnosed [56, 77, 99].
Metastases comprise 0.2–0.7 % of all CP
angle tumors [66, 125]. They spread to the area
either by hematogenous or by leptomeningeal
dissemination [168]. The three most common
primary malignancies that metastasize to the CP
angle are breast and lung carcinoma and mela-
noma [66, 150, 167]. Other tumors reported to
metastasize to the area are colon carcinoma, renal
cell carcinoma, adenocarcinoma, nasopharyngeal Fig. 11.10 Contrast-enhancing lesion in the left CP
carcinoma, thyroid carcinoma, ovarian carci- angle and IAC. The presence of a second round cerebellar
noma, gallbladder carcinoma and lymphoma lesion with similar imaging characteristics raised the sus-
picion of metastases. Clinical work-up revealed multiple
[54, 94, 102, 112, 136, 167]. lung metastatic lesions and the primary focus—a breast
The radiologic appearance of metastatic carcinoma
tumors can mimic that of common benign lesions
of the CP angle such as schwannoma and menin- cancer [90, 150, 159, 162]. Metastases cause
gioma [168]. They are usually smaller lesions, neurological dysfunction by compression or
isointense or hypointense on T1-weighted MR direct infiltration of neural structures. The mass
sequences and hypointense on T2-weighted MR effect of the tumor may be further enhanced by
sequences, and show strong enhancing after gad- the surrounding edema. Hence, even small metas-
olinium administration [63, 162]. Perifocal edema tases—in contrast to benign CP tumors—may
in the adjacent cerebellum/brain stem, which is cause major neurological dysfunction. Facial
seen as high signal on FLAIR and T2-weighted nerve weakness at presentation is relatively fre-
images, can be observed. Warren et al. [162] quently seen in metastatic tumors but is excep-
pointed that metastatic tumors are eccentric from tional in VSs and rare in CP angle meningiomas.
the IAC because they do not arise from the sev- On the other hand, if patients with known pri-
enth to eighth nerves but from the flocculus and mary cancer have symptoms such as vertigo, tin-
choroid plexus. Further clues are the evidence of nitus, hearing loss, or facial palsy, metastasis in
bilateral CP angle lesions and the presence of the CP angle—despite its rarity—should be sus-
multifocal cerebral or neck lesions pected [66, 125]. Huang and Young [102] exam-
[162]—Fig. 11.10. ined radiologically 174 cancer patients with such
Clinically metastatic tumors in the CP angle symptoms and found CP angle tumors in just 6 of
cannot be distinguished from benign tumors. them. Interestingly only 3/6 tumors turned out to
Still, some clinical factors should raise the suspi- be metastatic: a history of previous malignancy
cion of malignancy: rapid clinical deterioration, and a secondarily discovered CP angle lesion
advanced age at presentation, progressive sen- does not necessarily imply a metastasis.
sorineural hearing loss, facial nerve weakness, Generally, metastases to the CP angle are
bilateral cranial nerve symptoms, and history of believed to have rapid clinical course and carry
Rare Extra-axial Cerebellopontine Angle Lesions 765

poor prognosis. In patients with CP angle tumors condition of the patient, primary tumor control,
that turn out to be metastatic, the primary focus presence of multiple metastases, as well as size,
should be identified and managed appropriately. location, and symptoms caused by the CP angle
The management of CP angle tumors in patients tumor. Surgical removal of isolated CP angle
with known cancer should be part of their general metastases along with appropriate chemotherapy
treatment concept. Factors to consider are general and radiation can provide local tumor control.
766 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.13 followed with MRI studies. Several weeks


Metastatic Tumor to the CP Angle prior to presentation, increasing weakness of
This 54-year-old female patient suffered from the extremities and headache appeared. MRI
an invasive ductal carcinoma of the right breast. demonstrated a certain growth tendency of the
Partial breast resection had been performed tumor and a compressive effect on the brain
1 year ago, followed by therapy with Herceptin stem and cerebellum (a, b).
(the HER-2-neu was positive). She has been Considering the size of the tumor, its pro-
diagnosed to have 2 lesions in the posterior gressive enlargement with resultant clinical
cranial fossa, which were highly suspicious for deterioration and the good general condition
metastases. The large tumor in the right cere- of the patient, as well as her personal wish, a
bellar hemisphere—metastasis from the ductal decision was taken to remove the tumor surgi-
carcinoma—has been removed surgically 2 cally. The RS route was selected, and the
months prior to presentation at our department tumor could be removed completely, preserv-
and whole brain radiation was planned. The ing the function of the seventh to eighth cra-
second one, located in the left CP angle, was nial nerves.

a b

(a, b) MRI study of a patient with breast carcinoma metastases


Rare Extra-axial Cerebellopontine Angle Lesions 767

Collision Tumors petrous apex that were also clearly separate.


Similar to these cases, one of our patients had a
Collision tumors are defined as neoplasms that coincidental occurrence of two different tumors
grow in one area, collide, and become intermin- in the CP angle that were not in contact and can-
gled [118, 157]. In the CP angle area, such not be strictly defined as “collision tumors” (see
findings have been occasionally reported. Case Illustration 11.14). According to some
Muzumdar et al. [127] described a case of a col- authors, “combined” or “mixed” tumors have
lision tumor comprising a vestibular schwannoma been found in up to 25 % of all schwannomas in
and a petroclival meningioma in a 30-year-old patients with NF-2 [93]. In sporadic cases, how-
male patient. Both tumors were resected via a ret- ever, the association is rare. In the case described
rosigmoid route: the caudal portion of the mass here, however, no history of previous irradiation
proved to be VS, while the rostral portion in the or association with NF-2 was present. The possi-
petroclival was a meningioma. The authors’ lit- bility of such concomitantly growing tumors in
erature review showed that occurrence of VS and the CP angle should be considered when plan-
a petroclival meningioma in “collision” has never ning the surgical strategy.
been reported. Another mechanism is the devel- The condition should be differentiated from
opment of carcinoma metastases in a primary CP the development of combined schwannoma and
angle tumor, usually in VS. Leeman et al. [118] meningioma in a single tumor, which we have
reported on a case of breast carcinoma metasta- reported earlier [120] in a 59-year-old female
sizing to a CP angle meningioma and forming a patient with VS that was found to infiltrate the
collision tumor. facial nerve—Fig. 11.11. In order to achieve
Izci et al. [106] described a schwannoma and complete tumor removal, the facial nerve was
meningioma in the CP angle in a 57-year-old cut and then reconstructed with an interposi-
patient that were not in continuity. The tumors tion sural nerve graft. Interestingly, the histo-
were removed successfully via the retrosigmoid logical and immunohistochemical examination
approach. Wilms et al. [163] presented a vestibu- showed that the tumor was a typical schwan-
lar schwannoma extending into the IAC and a noma with intermingled islets of meningothelial
meningioma with broad implantation on the meningioma.

Fig. 11.11 MR images of a combined schwannoma and meningioma in a single tumor (Reproduced from: Lüdemann
et al. [120]. Copyright © 2000, with permission from Wolters Kluwer Health)
768 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.14 possibly to remove the posterior cranial fossa


Sporadic Unilateral VS and Petroclival part of the meningioma.
Meningioma The patient was operated via the RS
This patient (55 years old) presented with a approach. Complete removal of the VS and
3-year history of occipital pain and vertigo. preservation of the facial nerve was achieved
Since 1 year she noticed that hearing on the (j–l). The petroclival tumor, which proved to
left side deteriorated progressively. Later on be a meningioma, was engulfing the abducens
bilateral papilledema was diagnosed and MRI nerve. It was removed carefully along this
study was performed, which demonstrated a nerve until the Dorello’s canal was reached.
large CP angle tumor and obstructive hydro- Complete removal of the tumor was not
cephalus. Ventriculoperitoneal shunt was attempted due to the infiltration of the cavern-
therefore placed. At presentation the patient ous sinus. The patient recovered well and at
had trigeminal hypesthesia (V1–3) and com- discharge did not have any new neurological
plete hearing loss on the left side, as well as deficits besides a slight (HB grade II) facial
severe gait instability. MRI examination weakness on the left side. The postoperative
showed 2 separate CP angle tumors: a large MR study is presented on figures f–i.
VS (extension grade T4b) and a petroclival As discussed in detail in the chapter on
tumor, most probably a meningioma that Neurofibromatosis, the occurrence of unilat-
invaded the posterior cavernous sinus (neu- eral VS and additional ipsilateral meningioma
roimaging: a–f, before surgery, and e–i, after may be observed in the mosaic type of NF-2,
surgery). Considering the size of the tumors, caused by a mutation at a later stage of embryo-
the extent of brain stem compression, and the genesis. In this case, however, no clinical clues
clinical condition of the patient, the primary for NF-2 existed. Genetic testing was recom-
goal of surgery was to remove the VS und mended but not performed by the patient.

(a–i) MR images of the patient with two different spo- removed completely and the IAC is plugged with fat,
radic tumors in the left CP angle: VS and petroclival generating the characteristic signal on T1 and T2
meningioma that extends into the posterior cavernous sequences. (j–l) Intraoperative images. (j) The VS has
sinus. Although both tumors show intense contrast been removed completely and the facial nerve (arrows)
enhancement, the signal characteristics are obviously has been preserved. Note the widely opened IAC. (k)
different. (a–c) T1-weighted MR sequences following The IAC has been sealed with fibrin glue and fat
contrast administration. (d) T2- CISS sequence. pieces. (l) In the depth of the operative field—in the
Postoperative images: (e) CT scan 1 day after surgery. petrous apex area—a meningioma was seen (C) and
(f, g) T1-weighted MR sequences following contrast was also removed completely. (5 trigeminal nerve, BS
administration. (h, i) T2-CISS sequences. The VS is brain stem, arrowheads lower cranial nerves)
Rare Extra-axial Cerebellopontine Angle Lesions 769

c
770 11 Other Cerebellopontine Angle Tumors

d e

f g
Rare Extra-axial Cerebellopontine Angle Lesions 771

h i
772 11 Other Cerebellopontine Angle Tumors

j k

l
5

BS
Rare Extra-axial Cerebellopontine Angle Lesions 773

Vascular Lesions (no signal) on all spin-echo sequences. Extreme


hypointensity on T2-weighted MR images is
Aneurysms, arteriovenous malformations, highly suggestive for such lesions. Thrombosed
venectasias, or tortuous segments of the vertebral aneurysms with methemoglobin content have
and basilar arteries or some of their branches can hyperintense signal on T1-weighted images
present as CP angle mass and have to be differen- (Fig. 11.12). Importantly, enhancement of the
tiated from neoplastic lesions [64, 80, 104, 124]. mass can be observed due to organization of the
They become clinically apparent either with acute thrombus and has to be differentiated from neo-
symptoms due to SAH or with more insidious plasms [126].
symptoms of a CP angle mass lesion [80]. Patients with distal anteroinferior cerebellar
or superior cerebellar artery aneurysms are usu-
CP Angle Aneurysms ally managed surgically because the endovascu-
Giant or fusiform aneurysms extending to the CP lar treatment seems to be less suitable.
angle usually arise from the vertebral or basilar
arteries [76, 110]. Peripheral aneurysms of the Developmental Venous Anomalies
anteroinferior cerebellar artery are rare, with a of CP Angle
reported incidence between 0.0003 and 0.5 % Developmental venous anomalies (DVAs) are
[55, 123]. Typically they develop at the meatal loop regarded as extreme variations of normal venous
or from the dorsolateral arterial branch. Peripheral drainage, despite the fact that structural abnor-
aneurysms of the superior cerebellar artery in the malities of their walls have been reported [140].
CP angle also have been reported [114]. Extension They are thought to represent anatomically disor-
into the IAC is exceedingly rare: according to Sun dered but physiologically essential venous drain-
et al. [153], only 13 cases of aneurysms located in age of surrounding normal cerebral tissue
the IAC have been so far reported in the literature. and—due to the high risk of venous stroke—
In case of high-flow aneurysms, MRI demon- should not be directly treated (see Case
strates oval or round masses that have flow void Illustration 11.15).
774 11 Other Cerebellopontine Angle Tumors

a b

Fig. 11.12 T1- and T2-weighted MR images of a giant partially thrombosed aneurysm of the basilar artery, presenting
as a CP angle mass
Rare Extra-axial Cerebellopontine Angle Lesions 775

Case Illustration 11.15 The patient could be extubated soon after


Developmental Venous Anomaly the surgery and did not have any neurological
A 51-year-old male patient presented to the deficits. Few hours later, however, he lost con-
senior author in the early 1990s, complaining sciousness. CT was performed immediately
from vertigo, aggravated by vertical eye move- and revealed massive edema of the brain stem
ments. His neurological examination revealed and the right cerebellar hemisphere (h). The
no deficits. Native and contrast-enhanced CT, patient was intubated and appropriate antie-
MRI, and DSA were performed (a–d) and dematous therapy was started. He regained
demonstrated a lesion in the right cerebellum consciousness quickly but suffered from cere-
with characteristics of a developmental venous bellar ataxia and glossopharyngeus palsy with
anomaly. In the right CP angle, a contrast- dysphagia and oculomotor palsy on the right
enhancing mass that could not be unequivo- side.
cally related to the DVA was seen. On the fifth day after surgery, he was suc-
The patient was operated via the RS approach, cessfully extubated and begun slowly to
and a complex venous malformation tightly improve (i). CT control 30 days later (j)
adherent to and compressing the seventh to eighth showed significant reduction of the edema. All
nerve complex was seen (e, f). The adhesions neurological deficits resolved completely
were carefully dissected; the lesion was mobi- within the following months, and at 6 months
lized away from the nerves and coagulated (g). follow-up, he had no neurological symptoms.

a b

The CT (a, b), MRI (c), and venous phase angiogra- dilated veins and their relation to the seventh to
phy (d) images show the typical appearance of a eighth cranial nerve complex (e, f). (g) The veins
developmental venous anomaly in the right cerebel- have been dissected form the nerves and coagulated.
lum. The tributary and the dilated collector veins are (h–j) CT scans of the patient demonstrating the
well seen. An enhancing mass with unknown rela- extensive venous infarction and its subsequent reso-
tion to the DVI is visible in the right CP angle (b, c). lution (h: second day, i: fifth day, j: 30 days)
(e–g) Intraoperative images demonstrating the
776 11 Other Cerebellopontine Angle Tumors

c d

e f

g
Rare Extra-axial Cerebellopontine Angle Lesions 777

h i

j
778 11 Other Cerebellopontine Angle Tumors

CP Angle Arteriovenous Malformations without intervening neural elements and sur-


Arteriovenous malformations (AVM) develop rounded by a gliotic peripheral capsule. They
primarily within the CP angle or in the IAC very occur with an incidence of 0.3–0.5 % and consti-
rarely. Nozaki et al. [130] reported on eight tute 10–20 % of all vascular malformations [61].
patients with such AVMs: all with hemorrhage as Extra-axial cavernomas of the cranial nerves are
initial presentation. On admission all patients had uncommon.
some cranial nerve palsy and three patients had Cavernomas develop in the IAC or CP angle
cerebellar ataxia. The goal of treatment of CP exceedingly rarely [66, 95, 113, 145]. They may
angle/IAC AVMs is complete excision and avoid- present with mass effect and have to be differen-
ance of new neurological deficits [86, 151]. The tiated from tumors developing in the area.
senior author’s experience encompasses four Cavernomas of the IAC are thought to arise from
intracanalicular AVMs treated surgically the capillary plexus of the epineurium of Scarpa’s
(Fig. 11.13). In all cases, the malformation could ganglion in the IAC—see Case Illustration 11.16
be removed completely and the functional out- [58]. Some lesions may extend from the IAC into
come was excellent. the CP angle. On the other hand, brain stem cav-
ernomas may have an exophytic part, extending
Cavernomas into the CPA (Fig. 11.14).
Cavernous malformations or cavernomas are vas- The clinical presentation of such cavernomas
cular malformations, composed of sinusoidal is related to compression and/or dislocation of
vascular spaces separated by connective tissue the adjacent cranial nerves and is similar to that

a b

c d

Fig. 11.13 (a, b) Intraoperative images of the subsequent opening of the IAC (c, d). The patient presented with
steps of resection of an AVM, extending deep into the increasing hearing deficit and vertigo, which after the sur-
IAC. Its complete and safe removal necessitated wide gery showed a gradual recovery
Rare Extra-axial Cerebellopontine Angle Lesions 779

a b

Fig. 11.14 Pontine cavernoma with a large exophytic removal, the PICA and the DVA (arrowhead) became
parapontine part in a 33-year-old male patient. (a–e) visible. Then, the ventral exophytic part, adherent to the
Preoperative images. Note the large DVA, extending into seventh to eighth cranial nerve complex (*), was
the CP angle. The lesion was removed via a midline pos- removed. Complete removal was possible with preserva-
terior fossa craniotomy. (f–j) Intraoperative images, tion of the DVA (arrowhead) and of the cranial nerves
demonstrating the gradual removal of the cavernoma (BS (*). (k) Postoperative MRI, demonstrating the
brain stem, C cerebellum, R retractor). Following partial intact DVA
780 11 Other Cerebellopontine Angle Tumors

e f
C

BS

Fig. 11.14 (continued)


Rare Extra-axial Cerebellopontine Angle Lesions 781

i j

Fig. 11.14 (continued)

of VSs. In case of bleeding, abrupt onset of symp- sequences and hyperintense signal on
toms or worsening of existing symptoms occurs. T2-weighted sequences with variable contrast
Clinical signs/symptoms include sensorineural enhancement [58, 132, 145]. Typically, caverno-
hearing loss, tinnitus, dizziness, vertigo, and pro- mas are surrounded by a peripheral rim of hemo-
gressive facial nerve palsy [58, 95, 148]. Important siderin that shows hypointensity on all sequences
clues to the diagnosis are the presence of facial [61, 64, 84]. Tumors with hyperintensity on T2
weakness and rapidly progressive or abrupt hear- sequence are rare in the IAC/CP angle. In gen-
ing deficit [84, 148]. A possible vascular genesis eral, the differential diagnosis is between lesions
could be suspected in case of episodic exacerba- containing fat, melanin, or intratumoral hemor-
tion or disproportionate severity of the symptoms rhage: hemorrhagic tumor (particularly VS),
regarding the small size of the lesion. cavernoma, dermoid, lipoma, melanoma, and
Radiologically it may be difficult to distinguish cholesterol granuloma [161]. CT scans may
a cavernoma from a VS or other IAC/CP angle demonstrate calcium deposits and/or IAC
lesions—Fig. 11.15. They are well-circumscribed enlargement in lesion with intracanalicular
lesions of mixed signal intensity on T1-weighted extension.
782 11 Other Cerebellopontine Angle Tumors

a b

Fig. 11.15 Pure intracanalicular cavernoma. (a) CT scan administration. (c) View of the cavernoma after opening
with bone-window setting: the IAC is significantly wid- of the IAC. (d) Complete removal of the cavernoma and
ened. (b) T1-weighted MR image following contrast preservation of all cranial nerves
Rare Extra-axial Cerebellopontine Angle Lesions 783

Case Illustration 11.16 more than 3 years later when he noticed pro-
Cavernoma in the IAC gressive hearing loss (a). Despite the fact that
This 43-year-old patient presented to the the tumor showed a growth tendency, the
senior author in 1992 with a history of gait patient decided to postpone surgery. One year
imbalance. CT and MRI were performed, later, he presented with severe vestibular
which demonstrated widened IAC and an problems, further hearing loss, and tinnitus.
intracanalicular tumor on the left side. Taking The patient was operated via the RS approach
into consideration the possible management (b). The lesion, which turned out to be a cav-
options, the patient decided for follow-up ernoma, was removed completely, and both
with regular MRI and hearing level examina- the facial and the cochlear nerves could be
tions. However, the next MRI was performed preserved.

a b

(a) Preoperative MRI, demonstrating the intracanalicular cavernoma. (b) Intraoperative photomicrograph of the
lesion, located in its lateral part of the IAC
784 11 Other Cerebellopontine Angle Tumors

The treatment of choice of IAC/CP angle cav- should be carefully assessed. Once the IAC is
ernomas is total microsurgical resection in order opened, an attempt is made to identify the facial
to prevent recurrences. At surgery intense adhe- and cochlear nerves medial and lateral to the cav-
sions between the cavernoma and the seventh or ernoma. In large lesions, internal decompression
eighth cranial nerves are often found. Pappas is performed initially, followed by dissection of
et al. [134] found dense attachment of the caver- the capsule from the neural structures.
noma to the facial nerve in five of their six cases. In all 7 patients, operated by the senior author,
Considering the high risk of recurrence and the the lesion was located in the IAC and extended
inevitably related to it loss of function, complete into the CP angle in only 2 cases. Complete cav-
cavernoma removal even with the option of ernoma removal was achieved in a single-stage
reconstruction of the injured/interrupted facial surgical procedure in all cases. The facial nerve
nerve should be aimed even in such cases. could be preserved in 5 cases and the cochlear
Although the issue of the most appropriate tim- nerve in 4 cases. The intimate adherence between
ing of cavernoma surgery is controversially the facial nerve and the lesion rendered complete
addressed in the literature, we believe that early removal without its sacrifice impossible in 2
surgery after the diagnosis is related to best func- cases; one of these patients had pronounced facial
tional outcome: multiple bleedings may irrevers- nerve paresis before surgery. Facial nerve recon-
ibly injure the cranial nerves. Moreover, the struction with a sural nerve graft was performed
chances for preservation of the facial nerve are in both cases.
lower in larger lesions. Early after surgery, some degree of facial palsy
The optimal approach is the RS one—it offers was noted in all patients. At follow-up, however,
an excellent overview of the cavernoma and of it improved to House–Brackmann grade I in 1
the whole CP angle and IAC (see Case Illustration patient, grade II in 1 patient, and grade III in the
11.17). The lesion is identified on the basis of its remaining 5 patients. Hearing preservation was
typical reddish-blue color. Its relation to the cra- not an issue because all patients had severe hear-
nial nerves and the degree of IAC extension ing loss at presentation.
Rare Extra-axial Cerebellopontine Angle Lesions 785

Case Illustration 11.17 A reddish-blue lesion was seen, which was inti-
IAC Cavernoma Extending in the CP Angle mately adherent to the seventh to eighth nerves
Twenty-three-year-old female patient presented and highly suspicious for a cavernoma. The
with a history of progressive hearing loss. Two lesion was found to originate within the IAC
weeks prior to admission, she experienced sud- and to protrude along the nerves in the CP angle.
den facial weakness on the left side (HB grade It could be completely removed, and—despite
III) that in 5 days worsened to HB grade IV. the dense adhesions to the nerves—both the
Audiogram revealed left-sided hearing loss of facial and the cochlear nerves were preserved
60–70 dB. CT showed widening of the left IAC, anatomically. Histopathological examination
while MRI demonstrated an intra- and extra- proved that the lesion was a cavernoma. The
canalicular lesion with signal characteristics, postoperative MR study is presented on figures
suspicious for recent hemorrhage (a–e). The j and k. The facial palsy improved steadily and
patient was operated via the RS approach (f–i). at 12 months follow-up was HB grade II.

a b

Preoperative images of the cavernoma. (a) Bone- nerve complex, P pyramid. The posterior wall of the
window CT demonstrates an enlargement of the IAC. IAC has been removed, and the intracanalicular part is
(b) CISS sequence. (c) TOF sequence. (d) T2-weighted seen and gradually dissected from the nerves (g).
sequence. (e) T1-weighted sequence. Intraoperative (h) Complete removal of the cavernoma and preserva-
images presenting the stages of cavernoma removal. tion of both cochlear and facial nerves (small arrows).
(f) Initial view of the cavernoma (arrow), protruding Postoperative T1-weighted MR images, following
into the CP angle. Arrowhead—seventh and eighth contrast application (i, j)
786 11 Other Cerebellopontine Angle Tumors

c d

f
e

g
Rare Extra-axial Cerebellopontine Angle Lesions 787

h i

j
788 11 Other Cerebellopontine Angle Tumors

Neurocysticercosis on T1-weighted MR images and hyperintense on


T2-weighted MR images. Symptomatic mucoce-
Neurocysticercosis is caused by infestation of the les may require drainage. Apex petrositis is an
central nervous system by the larval form of inflammatory lesion of the petrous apex second-
Taenia solium—the cysticerci [81, 138]. Their ary to otitis media [189]. Patients present with ear
radiological and clinical manifestations are and/or retroorbital pain and trigeminal and
highly variable and are related to the location of abducens nerve symptoms, which constitute the
the lesion and the host response. Neurocysticercosis Gradenigo’s syndrome. Other symptoms are pho-
in the CP angle have been reported and usually tophobia, excessive lacrimation, and fever. CT
occur in the racemose form [72, 129]. Their scans demonstrate a destructive expansive lesion
pathognomonic radiological sign, which however of the petrous apex with irregular margins. MR
is not frequently seen, is a 1–3-mm mural nodule imaging can show an enhancing mass at the
in the cyst (the invaginated scolex). They may petrous tip [195]. Osteomyelitis of the skull base,
appear as lobulated cysts with no mural nodule or usually observed in diabetics or immunocompro-
enhancement, as focal cisternal widening or as a mised patients, may involve the petrous bone. It
high signal rim due to surrounding adjacent tis- appears as an area of demineralization or as an
sue reaction and have signal intensity similar to irregular lytic lesion on bone-window CT and has
that of CSF with all sequences [87]. The differen- soft tissue density. MR imaging demonstrates an
tial diagnosis includes other cystic lesions: arach- enhancing area with hyperintense signal on
noid cyst, epidermoid cyst, cystic schwannoma, T2-weighted sequences. Aneurysms of the
and cystic meningioma. petrous portion of the carotid artery cause
smoothly marginated bone erosion and present as
complex structures with flow voids and even con-
Temporal Bone and Petrous trast enhancement on MRI.
Apex Lesions

Neoplastic or inflammatory lesions that originate Cholesterol Granulomas


from the posterior surface of the petrous bone or of the Petrous Apex
the petrous apex may present as CP angle masses.
They have to be differentiated from incidental Manasse first reported cholesterol granuloma of
nonoperative findings, such as asymmetric fatty the temporal bone in the mastoid and the middle
marrow, trapped fluid, or variations of the struc- ear in 1894 [231]. Two distinct entities have been
ture of normal bone [239]. The petrous apex is later recognized: the more rare petrous apex and
marrow filled in 84 %, pneumatized in 9 %, and the unaggressive tympanomastoid cholesterol
sclerotic in 7 % of temporal bones [188]. granuloma, which is typically observed in asso-
Asymmetric pneumatization of the petrous apex ciation with chronic otitis media [270].
may be confused with a neoplasm. The differen- Cholesterol granulomas are relatively rare benign
tial diagnosis is based on the absence of bone lesions of the petrous apex. They may enlarge
destruction or expansion on CT, the absence of enough to expand in the CP angle and compro-
contrast enhancement, and the hypointensity of mise cranial nerves function.
the signal on T2-weighted MR sequences [175]. The incidence of cholesterol granuloma of
Cholesterol granulomas are the most common the petrous apex is less than 0.6 cases per mil-
lesions intrinsic to the petrous apex [270]. They lion population per year. Their prevalence has
are estimated to be ten times more common than been assessed to be 30–40 times less than that
cholesteatomas and 40 times more common than of VSs [175, 215, 228]. Cholesterol granu-
petrous apex mucoceles [175]. Mucoceles are loma is a histologic term used to describe a
nonenhancing lesions, developing due to occlu- chronic granulomatous foreign body giant-
sion of the petrous air cells. They are hypointense cell reaction to the by-products of hemoglobin
Temporal Bone and Petrous Apex Lesions 789

degradation with associated fibrosis and vascular bulb, bony labyrinth, posterior wall of the sphe-
proliferation [180]. According to the classic noid, IAC, and facial nerve. On CT cholesterol
obstruction-vacuum theory of the physiopathol- granulomas appear as nonenhancing isodense
ogy of these lesions, the cascade of events is trig- with the brain parenchyma lesions. Bone-
gered by obstruction of the air cell outflow tracts window CT demonstrates well-defined bony
of the petrous apex [205]. This causes transuda- expansile lytic lesion, with remodeling and ero-
tion of blood or rupture of vessels into the unven- sion of the bone (Fig. 11.16a). They have sharp
tilated mucosal space and anaerobic breakdown and smooth margins—Fig. 11.16a. The MR
of red blood cells, which liberates cholesterol. image of cholesterol granulomas is more
The cycle repeats and leads to accumulation of specific—Fig. 11.16b. The lesion is nonenhanc-
cholesterol crystals, which are strong irritant ing and has high signal intensity on both T1-
to surrounding tissue. Ultimately a cholesterol and T2-weighted sequences [241, 242]. Rarely
granuloma is formed [180]. An alternative theory foci of low signal intensity, related to hemosid-
asserts that the causative factor is hemorrhage erin, may be seen. Areas of contrast enhance-
from exposed bone marrow [217]. ment correspond to granulation tissue [249].
Cholesterol granulomas are round or ovoid When desiccated, cholesterol granulomas can
cysts filled with brown-yellow fluid that contains have areas of low signal intensity that give this
globular material, lipids, and cholesterol crystals, homogeneous T1-hyperintense lesion an het-
surrounded by a thick fibrous capsule [270]. erogeneous T2 pattern, which in this location is
Actually, they are not true cysts because they are very suggestive of the diagnosis [279].
not lined with epithelium. The differential diagnosis of cholesterol gran-
uloma includes epidermoid, cholesteatoma, chor-
Neuroimaging doma, chondrosaroma, metastasis, mucocele, and
High-resolution thin-slice CT and MRI exami- petrous apicitis [214]. Cholesteatomas and epi-
nations are mandatory for the preoperative dermoids, in contrast to cholesterol granulomas,
planning [241, 249, 268]. The essential infor- have a low T1 MR signal intensity, which is
mation is the location and size of the cyst, as slightly greater than CSF signal intensity. On
well as its relation to surrounding structures: T2-weighted sequences, however, all three lesions
brain stem, carotid artery, sigmoid sinus, jugular are hyperintense [183].

Fig. 11.16 Bone-window CT demonstrating an area of petrous apex cholesterol granuloma, which has a small
bone erosion with smooth margins in the left petrous apex. extension to the middle cranial fossa. (The patient was
The corresponding MRI slice (T1-weighted image) shows operated in the late 1980s by the senior author)
790 11 Other Cerebellopontine Angle Tumors

Clinical Presentation causing new neurological deficits. Considering


Cholesterol granulomas are slow-growing lesions, the lack of a true epithelial lining, complete surgi-
causing initially vague or indistinct symptoms, cal excision is not essential, especially if this
such as temporal or retroorbital headaches, atypi- would endanger some important structure
cal facial pain, Eustachian tube dysfunction, and [180, 182]. The choice of operative approach
middle ear effusion. Recurrent serous otitis depends on the exact location and extension of
media, caused by the obstruction of the Eustachian the lesion, its relation to adjacent structures, level
tube, may be the primary clinical manifestation. of preoperative hearing, and goal of surgery [228].
The involvement of the adjacent structures Older patients with progressing lesions may be
leads to more specific symptoms: diplopia due to considered for more conservative surgery—cyst
abducens nerve compression, hearing loss and drainage and partial resection—via a less invasive
vestibular complaints, caused by compression of approach, such as the endonasal one.
the eighth cranial nerve in the IAC or CP angle or Lesions located purely in the petrous apex,
destruction of the bony labyrinth, and facial nerve especially those abutting the posterior wall of
dysfunction, which can appear in more advanced the sphenoid, can be accessed via the endonasal
lesions [180, 241]. In a review of 92 patients, (transsphenoidal) endoscope-assisted approach.
Terao et al. [267] found that the most common Alternatively, the pure endoscopic technique may
clinical presentations were hearing loss (37 %), be applied. The cyst can be safely drained and even
vertigo (28.3 %), and headache (27.2 %). resected [209, 236, 252]. With this less invasive
approach, a more extensive surgery is avoided, the
Management labyrinth and, respectively, the hearing are pre-
Cholesterol granuloma can have variable bio- served and the hospitalization costs are reduced.
logical behavior. Accordingly two clinical sub- Some granulomas, however, due to their exten-
types have been described: aggressive and sion and/or location of the carotid artery, are not
nonaggressive lesions [241]. The nonaggressive adequately accessible via this route. The subtem-
lesions are uncommon. They are usually inci- poral approach with extradural resection of the
dentally discovered and do not progress for long petrous apex is one of the alternatives that we
periods. Such lesions do not require interven- favor. Large lesions with extensive bone destruc-
tion: long-term follow-up is recommended with tion may be accessed via the transmastoid infral-
regular MRI and CT control. It has been reported, abyrinthine approach. Cholesterol granulomas,
however, that rapid growth can occur even after extending more caudally, are best approached via
years of stability [269]. a retrosigmoid craniotomy with drilling of the
Symptomatic or growing lesions should be overlying bone with diamond drill. The exact
treated surgically. The management options amount of bone removal is determined by the
range from complete resection to simple drain- extension of the cyst and the degree of bone ero-
age and fenestration. The necessity to reestab- sion. To access cholesterol granulomas, located
lish an adequate aeration of the evacuated cavity below the level of the IAC, the inframeatal bone
is frequently stressed, while the use of stents is should be drilled off intradurally (retrosigmoid
debated [172, 256, 258]. Some authors recom- inframeatal approach, which was elaborated by
mend extensive skull-base approaches aiming the senior author and used initially in 2004 in a
at more radical direct cyst excision: subtempo- patient with a petrous bone chordoma)—see Case
ral transzygomatic, preauricular subtemporal– Illustration 11.18. Lesions with more cranial
infratemporal, or a variant of the transpetrous extension, medial to the IAC, might require some
approaches, such as the translabyrinthine, tran- degree of suprameatal bone removal (retrosigmoid
sotic, transcochlear, infralabyrinthine, or petrosal suprameatal approach).
ones [172, 173, 182, 187, 202, 268]. Following the evacuation of the cyst contents
In our opinion, cholesterol granulomas should and the resection of its wall, the cyst may be
be resected as extensively as possible without inspected endoscopically. The possibility to look
Temporal Bone and Petrous Apex Lesions 791

around the corner with the angled optic and the more than 85 % of the patients. Cranial nerve
superior illumination allows to detect and to dysfunction generally tends to recover after the
remove possible remnants invisible with the micro- evacuation of the cyst.
scope. We recommend obliteration of the cyst cav- Cholesterol granulomas may recur even late
ity with multiple fat pieces, sealed with fibrin glue after surgery. The recurrence rate can reach 15 %
in order to prevent CSF leaks and reoccurrence of according to some publications [182]. Strict fol-
the cyst. As noted by some authors, the most com- low-up of the patients is therefore recommended,
mon cause for recurrence is obstruction of the including clinical, audiometric and vestibular
drainage site by fibrous tissue [268]. Fat oblitera- examinations, and neuroimaging. Decrease of the
tion of the cavity obviates the need for cyst drain- signal intensity on T1-weighed images and reduc-
age. Although some authors recommend using tion of the size of the lesion are the accepted cri-
muscle [202], we have found that it tends to shrink teria for successful evacuation [218]. At
and—in a long run—is less reliable material. follow-up, the fat-suppression techniques allow
New neurologic deficit is exceptional after to differentiate recurrence from cavities filled
surgery, and symptomatic relief is achieved in with fat.
792 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.18 neurological symptoms. Using high-speed


Cholesterol Granuloma Removal via the diamond drill, the petrous bone dorsal to the
Retrosigmoid Intradural Inframeatal jugular foramen and inferior to the IAC was
Approach removed until the underlying lesion was ade-
This 45-year-old patient has been operated quately exposed—the so-called intradural
15 years prior to presentation to our depart- inframeatal approach was performed. The
ment for a cystic tumor of the right pyramid lesion had a bluish appearance, moderately
via a subtemporal–intradural approach. The thick capsule and mixed content, consisting
tumor was removed partially; both epidermoid of a dark brown fluid part and yellowish cho-
and dermoid were suspected, but the histo- lesterol crystals. These finding corresponded
pathological diagnosis remained unclear. mostly to that of a cholesterol granuloma,
During the follow-up, a steady growth ten- which was proven later by the histopatholog-
dency was obvious, but the patient had no ical examination. The entire content of the
major new symptoms. At presentation the cyst could be removed with suction, tumor
patient had just a hypesthesia in the area of the forceps, platelet knifes of variable size, and
first and second branches of the trigeminal angled curettes under alternating microscopic
nerve—probably resulting from the initial and endoscopic control. The capsule was
surgery—and complained of annoying tinni- then dissected from the surrounding bony
tus. The HR bone-window CT showed an structures and removed. Those portions that
extensive zone of erosion of the pyramid with were invisible with the microscope could be
smooth margins (a, b). MRI showed a cystic visualized with the rigid endoscope control
lesion, located medial and inferior to the IAC, and subsequently removed. Once the com-
with characteristics compatible with a choles- pleteness of removal was verified (micro-
terol granuloma (c–h). scopic and endoscopic), the large bone cavity
Considering the extent of caudal exten- was filled with multiple fat pieces sealed with
sion of the lesion, it would not be adequately fibrin glue. Interestingly, the BAEP monitor-
accessed via the subtemporal approach. ing showed twice temporary decreased
A decision was therefore taken to approach it amplitude and increased latency of the wave
via the retrosigmoid intradural inframeatal V during endoscopic observation. These
approach (i–w). Following a standard RS changes recovered almost immediately fol-
craniotomy, dural opening, and release of lowing the removal of the endoscope.
CSF, the CP angle was well exposed. A yel- Following surgery the patient did not have
lowish lesion was seen to protrude intradur- any new neurological symptoms and was dis-
ally from the pyramid in the area immediately charged 12 days after surgery. The audiogram
dorsal to the jugular foramen. The lesion did showed that the preoperative hearing level
not have any relation to the cranial nerves, was preserved. The CT examination early
which could explain the lack of corresponding after surgery is presented on figures x and y.
Temporal Bone and Petrous Apex Lesions 793

a b

d
c

(a, b) Preoperative CT, demonstrating a lytic lesion LCN – lower cranial nerves; C – cerebellum). (l) Removal
of the medial petrous bone with sharp and smooth of the dura (arrows) and drilling (m) of the bone, dorsal
margins. (c–h) MRI of the patient, demonstrat- to the jugular foramen and inferior to the IAC, which
ing precisely the extension of the lesion and its rela- allowed for exposure of the whole lesion (n), Single
tion to surrounding structures (c) FLAIR sequence. arrowheads and double arrowheads. (o, p) Microscopic
(d) T1-weighted sequence. (e, f) CISS sequences. view, indicating complete removal of the lesion; the
(g, h) T1-weighted sequences after contrast adminis- inspection with angled endoscope, however, demon-
tration. (i–w) Intraoperative images, demonstrating the strated some remaining parts, which were removed
consecutive steps of surgery. (i) Standard RS approach under direct endoscopic guidance (q–v). The bone
and dural incision. Initial CSF drainage from the lateral opening was filled at the end with multiple fat pieces
cerebellomedullary cistern (R – retractor; arrowhead – and sealed with fibrin glue (w). (x, y) Postoperative CT,
transverse/sigmoid sinus knee; asteriks – sigmoid sinus). demonstrating the extent of bone removal. The fat tis-
(j, k) Initial view of the CP angle and the tumor (Tu). sue appears as a strongly hypodense signal
(SA – subarcuate artery; 7–8 – 7–8th cranial nerves;
794 11 Other Cerebellopontine Angle Tumors

e f

i
Temporal Bone and Petrous Apex Lesions 795

j k

l m

n
0
796 11 Other Cerebellopontine Angle Tumors

p q

r
s

t
u
Temporal Bone and Petrous Apex Lesions 797

v w

x y
798 11 Other Cerebellopontine Angle Tumors

Chordomas [260]. The petrous apex area was eroded in 54 %,


while in 21 % the erosion extended more later-
Virchow first described chordomas in 1857. ally. Intratumoral calcifications appear irregular
These tumors are derived from rests of the embry- at CT and are thought to represent sequestrae
onal notochord—a primitive cell line, around from bone destruction.
which the skull base and the vertebral column Chordomas are isointense or hypointense
form [203]. Chordomas account for 0.1–0.2 % of lesions on T1-weighted MR images and are typi-
all primary intracranial neoplasms [170, 263]. cally hyperintense on T2 sequences [203,
They are locally aggressive tumors, causing 225, 263]. Contrast enhancement is typically
expansive destruction of the bone at the site of moderate to high [281]. Multiplanar reconstruc-
origin, followed by subsequent infiltration and tion of the images is essential to evaluate pre-
penetration of the dura and eventually intracra- cisely the tumor extension and to plan surgery.
nial extension [225]. Chordomas originate usu- MR angiography demonstrates the main vessels
ally in the vicinity of the clivus (sphenooccipital and their relationship to tumor. Arterial narrow-
bones) and exhibit various extension patterns in ing is rarely observed, due to the fact that these
all directions. Chordomas arising from the body tumors are generally soft and easily to dissect
and the dorsal aspect of the clivus manifest usu- from adjacent vessels [203].
ally as sphenooccipital or petrosal lesions. In a The differential diagnosis of skull-base chor-
recently published analysis [260], we found that domas includes chondrosarcomas, meningiomas,
the most frequent direction of tumor growth was nasopharyngeal malignancies, trigeminal neuro-
into the posterior cranial fossa (81 %). In 35 % of mas, hemangioblastomas, craniopharyngiomas,
these patients, the brain stem was compressed. pituitary adenomas, lymphoma, fibrous dyspla-
Extension in the CP angle (defined as at least sia, and metastases [199, 203]. In contrast to
2-cm posterolateral growth) was seen in 14 %. chordomas, which are midline tumors, the major-
ity of chondrosarcomas arise along the petro-
Clinical Presentation occipital fissure. The rare midline chondrosarcomas
Symptoms are typically related to compression can be hardly differentiated from chordomas pre-
of adjacent structures and vary with lesion loca- operatively. These two pathological entities,
tion and proximity to critical structures [203]. however, should not be mixed: they have specific
Diplopia caused by abducens nerve palsy is the biological features, clinical presentation, natural
most frequent initial symptom—in 46–90 % of evolution, and outcome. Histologically they may
all cases [211, 260, 274]. Other symptoms/signs share some features, but immunohistochemistry
include headache caused by dural stretching allows for their precise differentiation.
and cranial nerve irritation, oculomotor or tro-
chlear palsy, visual changes, facial numbness or Management
weakness, hearing loss, dysphagia, hoarseness, Skull-base chordomas have relatively poor prog-
and hypoglossal nerve palsy. Further tumor nosis; if untreated, the life expectancy from diag-
growth causes symptoms and signs related to nosis is 6 months to 2 years [220]. Despite surgery
brain stem compression and intracranial hyper- and radiotherapy, recurrences occur early—
tension [235, 281]. usually within 2–3 years.
The most effective management of skull-base
Neuroimaging chordomas is maximal surgical excision fol-
High-resolution bone-window CT scanning lowed by heavy particle radiotherapy [192, 194,
reveals lytic bone destruction along with a soft 225, 246]. Controversial is the issue of the extent
tissue mass that is isodense or hypodense com- of surgery. Some surgeons favor an aggressive
pared to brain parenchyma and enhances moder- surgical approach with maximal tumor resection
ately after contrast application. In our series CT based on the assumption that longer survival
showed evidence of clivus destruction in all cases rates have been associated with more extensive
Temporal Bone and Petrous Apex Lesions 799

tumor removal [192, 206, 272]. Growing experi- of surgery is to remove as much of the tumor as
ence has shown, however, that true oncological possible and to decompress the brain stem, so
resection of chordomas can hardly, if ever, be that safe and more efficient radiotherapy can be
achieved [194, 225, 250]. Complete removal of performed [194, 265].
petroclival chordomas with a clear margin of tis- Previously for midline clival chordomas, we
sue is virtually impossible. Furthermore, long- utilized the transethmoid transsphenoidal
term survival rates and recurrence-free intervals approach (Fig. 11.17). Currently, however, we
have not been substantially prolonged after favor the endonasal-combined microsurgical and
extensive surgeries. The alternative strategy is to endoscope-controlled technique (Fig. 11.18).
perform safe cytoreductive resection, thereby With this technique, tumors extending from the
avoiding the significant procedure-related mor- level of dorsum sellae to C2 level can be accessed.
bidity risk related to aggressive surgery. The goal The paramedian tumor extension can be

a b

Fig. 11.17 Giant skull-base chordoma (a–c), removed via the transethmoid transsphenoidal approach (d, e)
800 11 Other Cerebellopontine Angle Tumors

d e

Fig. 11.17 (continued)

visualized and removed under endoscopic guid- The senior author followed this strategy in 4
ance with appropriate angled instruments. patients: in 3 of them, bilateral retrosigmoid sur-
Chordomas with a more lateral extension, geries were necessary and in 1 subtemporal and
however, cannot be safely removed via an endo- RS approaches were made [260].
nasal route. The approach to such tumors is Initial surgery affords the best opportunity for
selected individually, considering the tumor maximal tumor excision, and radicality should be
extension pattern, the degree of bone destruction, aimed, unless it endangers some essential struc-
the presence of intradural extension, the degree ture. The involved bone should be extensively
of brain stem compression, and the neurological removed, whenever feasible. Importantly, tumor
status of the patient. Extradural chordomas that invasion or surgery itself may impact the stability
already caused significant petrous bone destruc- of the craniovertebral junction. The operative
tion, even if they have a limited intradural exten- morbidity is typically related to cranial nerve pal-
sion, are operated via a presigmoid transpetrous sies, CSF leak, hemorrhages, and hydrocephalus.
approach. In case of relatively intact pyramid or The cranial nerves in the CP angle are usually
large intradural tumor part causing brain stem compressed and dislocated by the tumor; they are
compression, the standard RS approach is pre- infiltrated very rarely, which allows for safe dis-
ferred. This approach allows for removal of both section. In large chordomas, however, cranial
the intra- and extradural tumor parts. Furthermore, nerves may be embedded within the tumor tissue.
the tumor can be safely dissected from the brain The main reason for iatrogenic injury remains
stem and from the cranial nerves under direct their difficult or late visualization. Early
visual control, and the risks related to the extended identification of the essential neural and vascular
petrous bone drilling are avoided (see Case structure is, therefore, the prerequisite for their
Illustration 11.19). preservation. In chordomas with intradural exten-
Extensive skull-base chordomas may require a sion, due to the petrous bone destruction or
2-stage removal via different operating corridors. approach-related bone drilling, the risk of CSF
Temporal Bone and Petrous Apex Lesions 801

a b

Fig. 11.18 Endonasal-combined microsurgical and feedback control on the extent of resection and location of
endoscope-controlled removal of a clival chordoma (a–d) possible remnants. The images (f–i) are acquired intraop-
under navigational guidance (e). The surgery was per- eratively after the tumor removal
formed in the INI BrainSuite, which allows for immediate
802 11 Other Cerebellopontine Angle Tumors

Fig. 11.18 (continued)


Temporal Bone and Petrous Apex Lesions 803

f g

i
h

Fig. 11.18 (continued)

leak is very high. We recommend the utilization or carbon–photon therapy) has been shown to
of multiple small fat pieces sealed with fibrin improve local control and survival rates and is
glue to plug the areas of bone destruction and to recommended in case of incomplete tumor
augment the watertight dural closure. The fat can removal, in case of recurrence, or is delivered to
be harvested from the lower part of the incision the surrounding bone following each surgery,
or from the abdomen, in case a larger amount is regardless of the extent of resection [174, 197,
necessary. 262, 266].
Irradiation with heavy particles (protons, car- Chondroid chordoma is a pathological variant
bon ions, helium, and combined proton–photon of chordomas. At surgery these 2 variants cannot
804 11 Other Cerebellopontine Angle Tumors

be differentiated; they both present similar opera- (hyperintensive area). In case of growth ten-
tive challenges. Immunohistochemical staining dency or symptomatic recurrences, repeated sur-
allows to distinguish them. Although it has been gery is indicated with the goal of decompression
speculated that they may have better prognosis of neural structures and obtaining maximal safe
[211], recent studies found that the biological tumor removal. Of note, the operation-related
behavior and outcome of chondroid chordoma is morbidity rate is higher in repeated surgeries,
similar to that of conventional chordoma [171]. especially after radiotherapy. The orientation
Local recurrence of intracranial chordomas is and identification of normal landmarks and
still common regardless of the therapy mode, structures are more difficult, more intense adhe-
and strict follow-up of the patients is therefore sions of the tumor to cranial nerves and/or brain
recommended. A recurrence is best visualized stem are frequently found, and the large vessels
on contrast-enhanced MRI images (area of con- may be very fragile due to radiation-induced
trast enhancement) or on T2-weighted sequences vasculopathy.
Temporal Bone and Petrous Apex Lesions 805

Case Illustration 11.19 Considering the relations to the brain stem,


Chordoma Removal via the Retrosigmoid the two previous surgeries, and the proton
Approach beam therapy, radical tumor removal was not
Forty-two-year-old female patient operated deemed possible. A new surgery via a left-
twice elsewhere before presentation. The first sided RS approach was planned with the goal
surgery was in 2001: partial removal of a of decompressing the brain stem and volume
chordoma via a frontotemporal craniotomy. reduction of the tumor, as far as safely pos-
One year later, the patient was reoperated via sible. The steps of tumor removal are illus-
a left-sided retrosigmoid craniotomy, and trated on figures d–i.
postoperative proton beam treatment has Six days after the surgery, a CSF leak
been performed. Her recent MRI showed an appeared. The patient was reoperated and a
extensive tumor of the clivus with major small opening between the dural stitches was
intradural growth, engulfment of the verte- identified as source of the leak. Fat, harvested
bral and basilar arteries, and compression from the abdomen, and fibrin glue were used
with suspected brain stem infiltration (a–c). to watertight seal this area.

a b

Preoperative MRI, demonstrating the location of the nerve (5). (e) Tumor removal was performed via sev-
tumor, its relation to the basilar artery, and the deep eral CP angle levels: initially between the LCN and the
penetration in the brain stem. (a) Native T1-weighted seventh–eighth nerves, followed by the level below the
sequence. (b) T1-weighted sequence after gadolinium LCN (f) and the level between the seventh–eighth and
administration. (c) T2-weighted sequence. (d) Initial the fifth nerves (g). The ipsilateral vertebral artery
view of the CP angle and of the tumor (Tu), located (VA), initially hidden by the tumor, is visible on (h). (i)
medially to the lower cranial nerves (LCN), the All neural and vascular structures of the CP angle,
seventh–eighth nerve complex (7–8), and the trigeminal preserved after the tumor removal (PV petrosal vein)
806 11 Other Cerebellopontine Angle Tumors

c d

i
Temporal Bone and Petrous Apex Lesions 807

Chondrosarcomas 12 % of chordoma patients, in 32 % of chondroid


chordoma patients, and in 80 % of chondrosar-
Chondrosarcomas (CSA) originate from chon- coma patients.
drocytes in the areas of residual enchondral carti- CSA and chordomas are completely different
lage or from multipotent mesenchimal cells pathological entities with specific biological
[206, 211]. Cranial CSA originate at or near the behavior and prognosis. Histologically these
petroclival area and comprise approximately 6 % tumors are, however, difficult to differentiate.
of all skull-base tumors [171, 193, 222]. In the The diagnosis is based on immunohistochemis-
senior author’s material, CSA were typically try: both CSA and chordoma contain S-100 and
located paramedian and had most frequently pos- vimentin but that only chordoma contains keratin
terior or posterolateral extension [259]. Ninety- and epithelial membrane antigen.
two percent of them expanded into the posterior
cranial fossa and 64 % involved the CP angle. Clinical Presentation
CSA are subdivided into 3 grades based on The initial clinical presentation is related to tumor
their cellularity and nuclear atypia [255]: from location and size (Figs. 11.19, 11.20, and 11.21).
well differentiated (grade 1) to least differentiated Most frequent initial sign is abducens nerve
(grade 3). These categories correlate to the risk of palsy: it was found in more than half of the
recurrence and the survival rate, although in our patients in our series. Less frequent symptoms/
experience, significant difference between grades signs are headache, ataxia, oculomotor nerve
1 and 2 was not noted [259]. Four histological palsy, hypacusis, tinnitus, vertigo, facial palsy,
CSA types have been reported: conventional, ded- swallowing difficulties, and pyramidal signs.
ifferentiated, clear cell, and mesenchymal [255,
278]. Mesenchymal CSA are associated with Management
worse prognosis and have an average life expec- CSA, extending to the CP angle, similar to all
tancy of less than 28 months [193]. other CSA, can be managed with surgery, radio-
therapy, or combination thereof. Extensive and
Neuroimaging aggressive surgical resection, even total en bloc
CSA have a tendency to invade and destruct sur- removal with carotid artery and cranial nerve
rounding bones. The typical CT appearance of sacrifice, has been proposed by some authors as
CSA is that of a destructive lesion with patchy the “ideal” surgical goal [206, 244, 273].
infiltration, occasionally with a rim of calcification. Considering the high risk of functional deficits
Erosion of the clivus is always found, while the related to aggressive surgery and the long-term
petrous apex is destructed in 83 % of the cases outcome, the dominating alternative treatment
[259]. These tumors are hypo- to isointense on concept is to achieve maximal safe cytoreduction
T1-weighted sequences and hyperintense on and brain decompression while preserving func-
T2-weighted sequences with frequent signal het- tion [193, 248, 278]. The rationale for this approach
erogeneity. They usually demonstrate an intense is that small tumor remnants do not influence
but heterogeneous contrast uptake [244, 278]. significantly the risk of tumor recurrence and
CSA should be differentiated from chordoma, patients’ survival. Skull-base CSA are benign
chondroid chordoma, chondroma, meningioma, slow-growing tumors that can be controlled effec-
osteogenic sarcoma, glomus jugulare, and metas- tively with surgical resection. The 5- and 10-year
tases. Neuroimaging data may be insufficient to survival rates in largest series are 85–95 and
differentiate reliably chordoma, chondroid chor- 71–95 %, respectively [171, 193, 248, 261, 278].
doma, and CSA because their characteristics are We believe that the goal of surgery should be
similar [171, 244]. Tumor location, however, may to achieve radical tumor removal and neurovas-
provide a clue: CSA tend to be lateral to chordo- cular decompression, but not at the expense of
mas. In the series of Al-Mefty et al. [171], the compromising neurological functions and quality
petrous bone with the clivus was involved in of life of the patient. The management of each
808 11 Other Cerebellopontine Angle Tumors

a b

Fig. 11.19 Foramen jugular chondrosarcoma. Preoperative with atrophy of trapezius muscle. MRI (a, b) demonstrated
MRI and postoperative bone-window CT of a 69-year-old a cystic lesion in the right jugular foramen without intrac-
patient with a WHO grade I chondrosarcoma of the right ranial extension. The tumor was removed completely
jugular foramen (arrow). The patient presented with pro- via the transmastoid infralabyrinthine transcervical
gressive dysphagia, dysphonia, and accessory nerve palsy approach (c)

patient should be individualized according to his the anterior transpetrosal ones, may be preferred
personal expectation, neurological status, and (see Case Illustration 11.20). Following this con-
tumor characteristics. In CSA of the CP angle, cept, the 5- and 10-year survival rates in the
the major goal is to achieve brain stem decom- senior author’s series were 95 % [259].
pression and—on the second place—to remove Proton beam radiation is an adjunctive man-
the tumor as radically and as safely as possible. agement method [176, 247]. The survival rates
The retrosigmoid or/and the retrosigmoid and the risks of recurrence and metastases, how-
suprameatal approaches have been found to be ever, are similar in patients undergoing only sur-
most suitable for achieving these goals (see Case gery and in those managed with adjunctive proton
Illustrations 11.21 and 11.22). In case of exten- beam radiotherapy. Therefore, such management
sive involvement and destruction of the pyramid, is recommended in grade III CSA or after surgery
alternative approaches, such as the presigmoid or of early recurrences.
Temporal Bone and Petrous Apex Lesions 809

a b

c d

Fig. 11.20 Preoperative T1-weighted axial and coronal petrous apex and has a smaller part in the middle cranial
MR images (a–c) and T2-weighted images (d) of a patient fossa. The tumor was removed via the standard RS
with a CSA with a very large CP angle part, which causes approach (Reproduced from: Samii et al. [259]. Copyright
severe brain stem compression. The tumor involves the © 2008, with permission of Springer-Verlag)

a b

Fig. 11.21 Postoperative T1-weighted MR images (patient from Fig. 11.20) demonstrating the complete removal of
the CP angle tumor part and the brain stem decompression. The remnant in the middle fossa is followed radiologically
810 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.20 temporal craniotomy was performed after


Chondrosarcoma Removal via the Anterior resection of the zygoma and the petrous apex
Petrosal Route was approached extradurally. The eroded bone
This 52-year-old female patient presented covering the area was drilled off with diamond
with a 6-month history of double vision to the drill, and the petrous segment of the carotid
left side. The neurological examination artery was exposed to provide sufficient access
revealed incomplete left-sided abducens nerve to the lesion. Following tumor removal, the
palsy. MRI demonstrated a tumor in the patient had an uneventful recovery. The histo-
petrous apex area on the left side with a lim- pathological diagnosis was grade 1 chondro-
ited extension into the CP angle (a–d). The sarcoma. Radiosurgery was performed in
bone erosion was well visible on the HR bone- order to lower the risk of recurrence, as
window CT scans. Considering the predomi- assumed at that time. Gradually, the abducens
nant tumor location within the petrous bone, nerve function recovered almost completely.
the small CP angle tumor component and the The patient is followed for more than 72
neurological condition of the patient, she was months and has a very good quality of life
operated via an anterior petrosal route. A low without any evidence of tumor activity.

a
b

c
d

(a–d) Preoperative CT (a) and MRI (b– T2-weighted; the left side. (e–f) Postoperative T1-weighted
c– CISS; d– T1-weighted contrast enhanced sequence after gadolinium administration (e) and
sequences), demonstrating a petrous apex lesion on T2-weighted sequence (f)
Temporal Bone and Petrous Apex Lesions 811

e f
812 11 Other Cerebellopontine Angle Tumors

Case Illustration 11.21 T2-weighted sequences and had an intracra-


Chondrosarcoma Removal (Endoscope- nial extension that reached and slightly com-
Assisted Retrosigmoid Suprameatal pressed the lateral midpons. The patient was
Approach) operated via the RS suprameatal approach.
This 32-year-old male patient complained This approach was selected, considering the
from intermittent double vision for 2 months limited bone erosion of the petrous apex and
prior to presentation. Neurological examina- the presence of intradural tumor portion com-
tion revealed abducens nerve palsy on the pressing the brain stem. The lesion, which
right side. High-resolution bone-window CT proved to be a grade I chondrosarcoma, was
showed an area of bone erosion in the medial removed completely using combined micro-
part of the right pyramid (a), while on MRI a scopic and endoscopic control (g–l).
homogeneously enhancing lesion involving Postoperative CT scans are presented on
the petrous apex was seen (b–f). The lesion figures m and n. Early after surgery, the
was isointense to brain parenchyma on abducens nerve function started to improve
T1-weighted sequences and hyperintense on and at follow-up resolved almost completely.

a b

(a–f) Preoperative CT (a), T1-weighted MR image up to the Meckels’ cave and enhancing the approach
following contrast administration (b–d), CISS (e) and to the tumor (i). Following initial internal debulking
T2-weighted MR sequence (f), demonstrating an (j, arrowhead), the tumor was removed completely
enhancing lesion in the area of the petrous apex with (k). Note the preserved abducent nerve (arrow), medial
extension into the medial CP angle. (g–l) Tumor to the tumor. (l) Endoscopic inspection to verify the
removal: (g) initial view of the tumor (Tu) and the sur- completeness of resection. (m, n) Postoperative CT
rounding structures in the CP angle (5 – trigeminal images: 1 day after surgery (m) and 10 days later (n).
nerve; 7–8 – 7–8th nerve complex; S – suprametala Note the moderate pneumocephalus after surgery
tubercle). (h) The suprametal tubercle was drilled off, (m) and its complete resorption (n)
thus exposing the whole length of the trigeminal nerve
Temporal Bone and Petrous Apex Lesions 813

c d

e f

g h
814 11 Other Cerebellopontine Angle Tumors

i j

k l

m n
Temporal Bone and Petrous Apex Lesions 815

Case Illustration 11.22 anteriorly the external auditory canal, antero-


Chondrosarcoma in the Jugular Foramen medially the petrous carotid artery, and supe-
Area (Retrosigmoid Endoscope-Assisted riorly the seventh to eighth nerve complex,
Approach) became evident. Its capsule could be dissected
This 64-year-old male patient has been oper- from all surrounding structures, and the tumor
ated at another hospital 12 years prior to pre- could be removed completely with endoscopic
sentation: a tumor in the jugular foramen area assistance. The space was filled with multiple
on the right side, presumably a schwannoma, gelatin sponges (later on, we begun using fat
has been removed partially. The residual has tissue for such purposes). After surgery the
been followed neuroradiologically at irregular patient recovered quickly and had no new
intervals. The last imaging showed substantial deficits. Considering the invasion of the exter-
increase of its size. At presentation the patient nal ear, a new surgery was performed 10 days
had severe gait ataxia and complete hearing later—the external ear canal was closed and
loss on the right side. MR examination showed an incidentally found cholesteatoma was
an extensive tumor of the right petrous bone removed. The postoperative MRI is shown on
with large extracranial and intracranial parts figures i–l.
(a–h). The tumor was approached via the ret- The tumor turned out to be a grade II chon-
rosigmoid route; it had a thick capsule and drosarcoma and regular MR follow-up was
was filled with gelatinous content. Following recommended. For the period of follow-
dissection of the surrounding scar tissue, the up—48 months—no recurrences have been
actual extension of the tumor, which reached diagnosed.

a b

(a–h) Preoperative MR examination: native T1-weighted CT (i) and MRI (j–l) images demonstrating the com-
(a, b), T2-weighted (c–e) and T1-weighted sequences plete tumor removal
following contrast application (f–h). (i–l) Postoperative
816 11 Other Cerebellopontine Angle Tumors

c d

f
e

h
g
Temporal Bone and Petrous Apex Lesions 817

i j

k l
818 11 Other Cerebellopontine Angle Tumors

Chondroblastomas The histopathological result was chodroblastoma.


The patient did not have new neurological deficits
Chondroblastoma was first reported by Codman after surgery and recovered rapidly. A follow-up
in 1931 and occurs typically in the epiphyses of MRI is planned in 3 months: the decision if to
the long bones. Chondroblastomas involve the undertake radiotherapy will depend on the
skull base extremely rare. According to Hatano findings of this study.
et al. [210], approximately 70 cases of such
tumors involving the temporal bone have been
reported. Chondroblastoma arises from imma- Endolymphatic Sac Tumors
ture cartilage and is typical for the young age
group. Histologically they are benign tumors but Endolymphatic sac tumors (ELST) originate
clinically show malignant behavior with inva- from the endolymphatic sac or duct [212]. Their
sion and/or metastasis to adjacent structures exact anatomical origin is supposed to be the
[238]. Surgery is the mainstay of treatment of osseous portion (vestibular aqueduct) of the
such tumors, and the goal is to achieve total endolymphatic duct/sac system [229]. These
tumor removal, if safely possible [224]. Partial tumors were first established as a distinct patho-
removal or curettage is related to high risk of logical entity by Heffner in 1989 [212] and were
recurrence [223]. Radiation may be effective in recognized as part of the Von Hippel–Lindau
suppressing tumor growth and is regarded as an syndrome by Manski et al. in 1997 [232]. They
adjunctive therapy mode in case of tumor rem- are rare tumors of the posterior temporal bone,
nants. Primary radiotherapy is not a plausible whose incidence is actually underestimated
option: some authors have shown that it may because they have been misdiagnosed as a variety
induce malignant transformation to chondrosar- of other tumors [213].
comas [280]. Previously known as aggressive papillary
In our series, we had one patient with a petrous middle ear tumors, ELST are histologically
bone chodroblastoma, presented on Fig. 11.22. benign slow-growing low-grade adenocarcino-
This 16-year-old patient presented recently with mas. They are locally invasive and frequently
trigeminal hypesthesia (V2), facial palsy (HB expand intracranially in the CP angle area [212,
grade III), and anacusis on the left side. He had 226]. ELST tend to recur locally but do not
been operated 5 months earlier in another depart- metastasize.
ment: tumor removal had been performed via a Though most cases are sporadic, ELST have
retrosigmoid approach; the histopathological been described in association with Von Hippel–
diagnosis was aneurysmal bone cyst. The CT and Lindau disease, an autosomal dominant cancer
MR images at admission are presented on syndrome [275]. The incidence of ELST in
Fig. 11.22a–i. Considering the extension pattern patients with this syndrome is 15 % [191]. In
of the tumor and the wide destruction of the 30 % of the patients with VHL syndrome, ELST
petrous bone, a decision in favor of subtemporal occur bilaterally and cause significant neurologi-
epidural transpetrous tumor removal was taken. cal disability: deafness and severe disequilibrium
The tumor was widely enucleated and dissected [234]. VHL syndrome is linked to a germline
from the surrounding structures, including the mutation of the VHL gene [275, 276], mapped to
internal carotid artery. The most medial and pos- chromosome 3p25. The VHL protein is involved
terior parts, however, were strongly adherent; any in the regulation of the activity of a hypoxia-
attempt to mobilize or dissect them would endan- inducible factor, which enhances cellular metab-
ger the integrity of the facial nerve, of the lower olism and increases the expression of angiogenic
cranial nerves, or of the carotid artery. Therefore, and mitogenic factors. With absent or abnormal
this portion was not removed. The resection VHL protein function, angiogenesis is stimu-
cavity was filled with fat tissue, well visible on lated, which explains the typical hypervascular
the postoperative CT scans (Fig. 11.22j, k). pattern of the VHL-related tumors [219].
Temporal Bone and Petrous Apex Lesions 819

a b

c d

Fig. 11.22 Petrous bone chondroblastoma with a large enhanced images. (h, i) T2-CISS images. The early post-
cp angle and supratentorial extension. Preoperative CT operative CT images are presented on (j, k) (See the text
(a–c) and MR (d–i) images. (d–g) T1-weighted contrast- for details)
820 11 Other Cerebellopontine Angle Tumors

e f

g h

Fig. 11.22 (continued)


Temporal Bone and Petrous Apex Lesions 821

i j

Fig. 11.22 (continued)

Neuroimaging ELST should be differentiated from other


ELST are centered at or near the posteromedial destructive tumors involving the temporal bone:
surface of the petrous bone. Bone-window CT glomus tumor, chordoma, chondrosarcoma, men-
reveals bone erosion with “moth-eaten” bone ingiomas, CP angle choroid plexus papilloma,
margins [191, 227]. Frequently, bone spicules or ceruminous gland tumors, metastatic thyroid or
fragments are seen within the tumor. The radio- renal cell carcinoma, benign adenomatous
graphic finding in case of small ELST is typically tumors, and primary bone tumors [243].
limited bony erosion adjacent to the vestibular
aqueduct. The cystic tumor areas are hyperin- Clinical Presentation
tense on T2- and T1- weighted MR sequences, ELST present most frequently (in up to 95 % of
while the solid portions have heterogeneous the patients) with unilateral hearing loss: of grad-
intensity on both sequences. ELST are generally ual or of sudden onset [191, 232, 234]. The puta-
highly vascular tumors. tive mechanism of hearing loss in case of large
822 11 Other Cerebellopontine Angle Tumors

ELST is tumor invasion of the otic capsule (the Complete tumor excision has been shown to lead
bony covering of the inner ear). Sudden hearing to better results than subtotal or partial excision
loss is supposed to be caused by intralabyrinthine followed by radiotherapy: the cure rate after total
hemorrhage, while the slowly progressing hear- tumor removal is up to 90 %, while in case of
ing loss mimicking that of Menière’s disease is subtotal resection with adjuvant radiation of the
due to endolymphatic hydrops [185]. Other com- residual tumor, it is just 50 % [212, 229, 234].
mon symptoms are tinnitus (90 % of patients), Early surgery has been recommended to
vertigo or disequilibrium (66 %), and aural full- reduce the morbidity related to ELST because of
ness (30 %) [229, 232, 253]. The vestibular the unpredictable onset of audiovestibular mor-
symptoms related to ELST follow usually a pat- bidity, which is not related to tumor size
tern consistent with unilateral vestibulopathy: [229, 234]. In advanced tumors, however, com-
severe symptomatology with subsequent milder plete excision is rarely possible due to involve-
or asymptomatic periods. Possible pathophysio- ment of lower cranial nerves, sigmoid sinus, and/
logical mechanisms are either the “burn out” phe- or petrous portion of the carotid artery [254].
nomenon or the development of central Therefore, high-resolution MR and CT imaging
compensation [191]. of the vestibular aqueduct and endolymphatic sac
Further symptoms in patients with ELSC are area are recommended in all patients with VHL
due to involvement of adjacent cranial nerves: the syndrome.
facial nerve is involved most frequently, followed The RS approach is best suited for the removal
by ninth, tenth, and fifth cranial nerves. Tumor of both small and large ELST (see Case Illustration
expansion causes cerebellar and long-tract signs, 11.25). It allows for radical surgery and—in case
intracranial hypertension, and/or hydrocephalus. of small tumors—offers the possibility for hear-
In some cases, a vascular mass in the external ing preservation. The petrous bone can be drilled
auditory canal or a bluish-red discoloration—in off additionally, as described earlier in the book,
case the tympanic membrane is intact—can be providing adequate access to the whole tumor. In
visualized [212]. The natural evolution of ELST case of large ELST, the dissection of the CP angle
tumors is unpredictable, and remarkable tumor part from the cranial nerves or the brain
variability, ranging from more indolent to very stem is safer when performed under the better
aggressive progress, has been observed (see Case visual control, provided by the RS approach. In
Illustrations 11.23 and 11.24). case the tumor is confined to the petrous bone
and hearing is completely lost due to destruction
Management of the inner ear, the transmastoid transperosal
The ideal treatment of ELST is their complete approach may be an alternative (see Case
resection, which is curative. In small tumors, this Illustrations 11.23 and 11.24). Preoperative
can even alleviate vestibular symptoms and allow embolization of major feeders can be performed
for preservation of residual hearing [191]. to reduce intraoperative bleeding [243].
Temporal Bone and Petrous Apex Lesions 823

Case Illustration 11.23 Considering the hypervascularity of the


Sporadic ELST with Relatively Slow tumor, its extension pattern, and the presence
Development, Removed in Two Steps via of compete facial palsy, a decision was taken
Transmastoid and Retrosigmoid Approaches to perform first embolization of its feeding
Twenty-nine-year-old female patient pre- arteries and then remove it in two separate
sented with a history of progressive hearing operative steps with reconstruction of the
loss on the right side since her 14th year. facial nerve, if necessary.
One year ago, dysesthesia in the area of the The tumor feeders were successfully embo-
right ear and slight facial palsy appeared. She lized, and 4 days later its petrous and extracra-
has been diagnosed of having a large tumor nial portions were removed via a right-sided
involving the petrous bone and the CP angle transmastoidal approach. The proximal seg-
that was removed partially elsewhere via the ment of the facial nerve was infiltrated by the
transmastoid approach. Following surgery tumor. Therefore, it was reconstructed by per-
she had anacusis and complete facial palsy. forming an end-to-end anastomosis between
On the last bone-window CT, an area of bone its distal healthy part and the hypoglossal nerve
erosion in the pyramid with irregular con- (i, j: postoperative CT scans). After surgery,
tours and small bony fragments in the center the patient suffered from minor dysphagia,
of the mass were seen (a). MRI revealed a which improved rapidly. Two weeks later, the
heterogeneous partially cystic tumor, which intracranial tumor part was removed com-
destroyed the lateral petrous bone (b–h). Its pletely via the retrosigmoid route (k–m: post-
solid portions enhanced intensely after gad- operative CT scans).
olinium application. The tumor had major Summarizing, this is a case of a sporadic
intracranial and extracranial extensions in the ELST with relatively slow development for
infrapetrous area, and the comparison with more than 14 years, good outcome of surgery,
previous images showed a certain growth and no recurrence up to present (30 months of
tendency. follow-up).
824 11 Other Cerebellopontine Angle Tumors

a b

d
c

Preoperative CT (a) and MR images of the patient, and bone-window (j) CT scans after the first surgery.
demonstrating the location of the tumor and the extent Native (k) and bone-window CT scans (l, m) follow-
of bone erosion. (b, c) Native T1-weighted sequence. ing the last surgery. The hyperdense signal corresponds
(d–g) T1-weighted sequence following contrast to fat tissue, used to seal the petrous bone
administration. (h) T2-weighted sequence. Native (i)
Temporal Bone and Petrous Apex Lesions 825

f g

h i
826 11 Other Cerebellopontine Angle Tumors

j k

l
Temporal Bone and Petrous Apex Lesions 827

Case Illustration 11.24 parenchymal tumors or cysts. Ophthalmological


VHL Syndrome-Associated ELST with examination, however, revealed a retinal
Aggressive Biological Behavior angioma on the left side so that a suspicion of
This 13-year-old boy presented with nonfunc- VHL syndrome aroused; genetic testing was
tional hearing on the right side that occurred recommended. The patient made an unevent-
suddenly 4 months earlier. The MRI demon- ful recovery from the surgery without new
strated a large destructive tumor involving the neurological deficits and follow-up MR imag-
right petrous bone with extension from the ing showed no signs of recurrence.
external auditory canal to the jugular foramen, Two years later, however, a slight facial
as well as into the CP angle (a). The presump- palsy (HB grade II) appeared and the MRI
tive radiological diagnosis was glomus jugu- showed a large tumor recurrence (b–e).
lare tumor. Digital subtraction angiography Considering the relatively early recurrence, a
was done and showed a highly vascularized decision for radical surgery—removal of the
tumor, supplied predominantly by a strongly tumor and of the affected bone structures via
dilated occipital meningeal artery—a branch the transpetrous approach—was taken. The
of the external occipital artery. Further feeders external auditory canal was closed plastically,
were originating from the ascending pharyn- the facial nerve was identified distal to the sty-
geal artery, the posterior temporal artery, the lomastoid foramen, and a lateral subtotal neuro-
internal carotid artery, and the temporo-occip- navigation-guided petrosectomy was performed.
ital branch of the middle meningeal artery, The facial nerve was exposed along its whole
while a small medial tumor part was supplied petrous segment—it was partially infiltrated by
by the slightly dilated anteroinferior cerebel- the tumor. The affected segment of the nerve
lar artery. The carotid artery was not infiltrated (approximately 1.5 cm) was resected, and its
by the tumor. The venous outflow system was continuity was reconstructed with a sural nerve
not significantly affected, although the right interposition graft. The tumor was gradually
jugular bulb was narrowed due to tumor com- removed, reaching the petrous segment of the
pression. Superselective catheterization with carotid artery and the middle fossa dura. Then,
embolization of the major feeding arteries was the presigmoid dura was opened, and the CP
then performed. angle tumor part was removed, dissecting its
The retrosigmoid approach was selected for capsule from the severely compressed lower
tumor removal, taking into account the absence cranial nerves. The surgical cavity was filled
of tumor extension into the lateral petrous bone with fat tissue, harvested from the abdomen
and the mastoid, as well as the normal facial (postoperative MR images: f–i).
nerve function. This route would allow further Postoperatively the patient, besides the
to perform the dissection of the tumor capsule complete facial palsy, had lower cranial
from the cranial nerves and brain stem under nerves dysfunction, and a PEG tube had to be
direct visual control. In case the petrous tumor placed before discharge. Four months later he
part would remain inaccessible, the approach presented to our outpatient department with
could be extended presigmoidally. complete recovery of the swallowing and
At surgery the tumor was found to be the speech. One year later, his facial nerve
purely extradural and could be completely function improved to HB grade III. On MRI,
removed with preservation of all cranial however, a small round contrast-enhancing
nerves. The histopathological diagnosis was nodule, highly suspicious for a recurrence,
ELST. MRI of the whole neuraxis was there- was seen in the petrous apex area (j, k).
fore performed but showed no further lesions. Due to the good clinical improvement and
Ultrasound of the abdomen ruled out further the absence of any new neurological symp-
828 11 Other Cerebellopontine Angle Tumors

toms, further close follow-up control was more aggressive biological behavior. It
recommended. Six months later, the finding occurred early in life, the clinical history was
remained unchanged. short, and the tumor tended to infiltrate the
Summarizing, this ELST was most proba- neural structures and to recur relatively early,
bly associated to VHL syndrome and had even after complete removal.

(a) Initial MR images of the tumor. (b–e) MRI show- administration). Note the fat tissue used to fill the sur-
ing the large tumor recurrence (T1-weighted images gical cavity. (j, k) Follow-up MRI, showing a small
after gadolinium administration). (f–i) Postoperative round contrast-enhancing nodule in the petrous apex
MR images, demonstrating the completeness of area (arrow), which is highly suspicious for a
tumor removal (T1-weighted images after gadolinium recurrence
Temporal Bone and Petrous Apex Lesions 829

b c

d e

f g
830 11 Other Cerebellopontine Angle Tumors

h i

j k
Temporal Bone and Petrous Apex Lesions 831

Case Illustration 11.25 cranial nerves and brain stem. Initially, the
Sporadic Cystic ELST with a Predominant fluid cyst contents were aspirated; then the
Part in the CP Angle capsule was dissected from the lower cranial
This 43-year-old patient complained since 5 nerves in the area of the jugular foramen. The
years of progressive hearing loss, tinnitus, and dissection proceeded to the brain stem, which
vertigo on the left side. Later on progressive allowed identifying the severely atrophic
gait instability appeared. He has been diag- facial nerve. The trigeminal and the abducens
nosed to have a large left-sided CP angle tumor, nerves could be freed from the tumor and pre-
involving the petrous bone, and was operated served. Gradually the whole tumor capsule
in another neurosurgical facility. The tumor was separated and resected. The petrous
was removed partially, and the histopathologi- tumor portion was removed thereafter, and
cal examination revealed a cholesterol granu- the bone defect was filled with multiple small
loma. He presented to our institute 2 years later fat pieces, sealed with fibrin glue (h, i).
and reported to have a progressive facial weak- The patient did not have new neurological
ness since 12 months and swallowing difficulty deficits after the surgery. He recovered quickly,
since 6 months. Neurological examination and 2 weeks later the planned facial nerve
revealed hypesthesia in the V2 and V3 areas, reconstruction was performed. The histo-
HB grade VI facial nerve palsy, complete hear- pathological and immunohistochemical study
ing loss, and lack of gag reflex. He had further of the specimen showed that it was an endo-
severe gait ataxia and slight right-sided hemi- lymphatic sac tumor. Considering this finding,
paresis (grade 4/5). a thorough screening was performed but no
CT and MRI examination demonstrated a further stigmata of Von Hippel–Lindau dis-
very large tumor in the left CP angle with severe ease were discovered. Active speech therapy
compression of the brain stem (a–g). Although and medialization of the vocal cord in case no
the fourth ventricle was occluded, the ventricu- improvement appeared in 6 months were
lar system was only slightly dilated. Extensive recommended.
destruction of the pyramid was evident. Summarizing, this patient had a very rare
Tumor removal via the retrosigmoid cystic sporadic ELST with a predominant part
approach was planned with subsequent, at a in the CP angle. Despite the previous surgery
later stage, hypoglossal–facial anastomosis and the tight adhesions of the cyst capsule to
for reconstruction of facial nerve function. the brain stem and cranial nerves, the tumor
The tumor was found to be predominantly could be removed completely via the retrosig-
cystic and very tightly adherent to the lower moid approach.
832 11 Other Cerebellopontine Angle Tumors

a b

c
d

(a–g) Preoperative CT of the patient (a, b) and MRI Gadolinium), demonstrating the extension of the
((c) Axial T1-weighted sequence. (d) Coronal tumor, the destruction of the pyramid and the degree
T1-weighted sequence. (e) Axial T2-weighted of brainstem compression. (h, i) Postoperative CT.
sequence. (f) Axial T1-weighted sequence with The fat plug is seen as a more hypodense signal
Gadolinium. (g) Coronal T1-weighted sequence with
Temporal Bone and Petrous Apex Lesions 833

f g

h
i
834 11 Other Cerebellopontine Angle Tumors

Fibrous Dysplasia FD should be differentiated form inflammatory


or neoplastic lesions, especially if T2-weighted
Fibrous dysplasia (FD) is an idiopathic benign images show high signal intensities. In case the
slowly progressive disorder, in which mature bone expansion conforms to the general shape of
bone is replaced by fibro-osseous tissue [204]. the bone of origin, the diagnosis may be more
This developmental anomaly can affect any bone straightforward [190]. The more rare localized
in the body: it represents 2.5 % of all bone tumors bone involvement, however, might represent a
and 7 % of the benign bone tumors [201]. FD diagnostic challenge. The radiological differential
typically presents during the first three decades diagnosis of FD includes chordoma, chondrosar-
of life. coma, chondroblastoma, carcinomas nasopharyn-
Histologically, FD consists of varying amounts geal, metastasis, plasmacytoma, giant-cell tumors,
of spindle cell bundles and irregular bony trabe- hemangiomas, and Paget’s disease [169].
culae of immature woven bone. Osteoblasts and Fibrous dysplasia affects the temporal bone
osteoclasts are scarce or absent. Common finding very rarely. Its usual presentation is progressive
are hemorrhagic areas with associated hearing loss. Expanding mass over the postau-
inflammatory and giant-cell reactions. ricular region may be seen. Hearing loss is usu-
Three patterns of development of FD have ally conductive and is caused by external auditory
been described: (1) monostotic or involvement canal obliteration, although sensorineural deaf-
of a single bone (70 % of patients), (2) polyos- ness may develop if the lesion involves the inner
totic or involvement of multiple bones (30 %), ear structures [186]. The intracranial extension of
and (3) McCune–Albright syndrome, which is a the lesion causes symptoms, due to neural
rare variant of polyostotic disease, in which FD compression.
is associated with multiple endocrine dysfunc- The clinical evolution of FD is unpredictable:
tion and café-au-lait spots [216]. The skull and most of the lesions tend to stabilize with the onset
facial bones are affected in 10–30 % of patients of puberty, while other may show aggressive
with the monostotic pattern and in 50 % of growth and may even undergo malignant trans-
patients with polyostotic fibrous dysplasia formation. The management options vary respec-
[179, 190]. Most frequently affected bones are tively: from close observation to resection of the
the maxilla, the mandible, the frontal, and the lesions. Surgery is generally indicated in case of
sphenoid [179, 230]. cosmetic problems or compression of neural
Three CT variants of FD have been described. structures and is aimed at correcting or prevent-
Most frequently seen is the “ground glass” ing functional problems and achieving normal
appearance (56 %) or bone expansion with alter- facial esthetics [169, 198]. Depending on the par-
nating areas of radiolucency and radiopacity, fol- ticular site of origin, extension pattern, and clini-
lowed by the sclerotic or homogeneously dense cal presentation, the goal is to decompress the
pattern (23 %) and the pagetoid or cystic pattern cranial nerves or brain stem, to remove the bulk
(21 %) [184, 190]. The fibrous tissue in FD is of the lesion, and to resolve the esthetic problem
metabolically active and well vascularized. (see Case Presentation: Fibrous Dysplasia of the
Therefore, it enhances strongly and on MRI may Petrous Bone).
resemble soft tissue tumors [184]. On Radiotherapy is not recommended even in
T1-weighted images, it has low or intermediate case of incomplete removal because of the risk
signal intensity depending on the ratio of fibrous of malignant transformation [240]. Some
tissue to mineralized matrix [190]. On reports, however, suggest that even without
T2-weighted images, the signal intensity varies radiotherapy spontaneous malignant transfor-
from low to high. mation can occur [271].
Temporal Bone and Petrous Apex Lesions 835

Case Illustration 11.26 lower cranial nerves were also monitored and
Fibrous Dysplasia of the Petrous Bone the standard RS approach was performed.
This 17-year-old boy presented with a history Once the lesion in the CP angle was exposed,
of dizziness, vertigo, hearing loss, and tinnitus its capsule was coagulated and internal deb-
on the right side for more than 2 years. Several ulking was done. The lesion was very well
months later, progressive facial palsy appeared. vascularized and relatively firm with areas of
MRI was performed that demonstrated a con- calcification. Using CUSA and high-speed
trast-enhancing lesion in the petrous apex area diamond drill, the whole CP angle tumor part,
with small extension toward the CP angle as well as part of the petrous part, was
(a, b). Follow-up of the lesion with MRI was removed. The abducens nerve was identified
recommended by the consulting neurosurgeon. in the area of its exit at the brain stem and fol-
One month prior to presentation, double vision lowed to the Dorello’s canal. The lower cra-
to the right side appeared, and the new MRI nial nerves were severely compressed and
demonstrated further growth of the lesion. dislocated caudally but could be freed from
At examination the patient had peripheral the tumor and preserved. The continuity of
facial paresis (HB grade V), nonfunctional the facial and the cochlear nerves was also
hearing, and absent gag reflex on the right preserved. Complete removal of the lesion
side. CT showed an area of bone destruction without undue risk was considered impossi-
involving the medial part of the petrous bone, ble because of the encasement of the carotid
clivus, and sphenoid bone (c, d). The lesion artery’s petrous segment and the presence of
had the typical “ground glass” appearance: cavernous sinus extension. Thorough hemo-
bone expansion with alternating areas of stasis was achieved, and the tumor cavity was
increased and decreased density. On MRI the filled with fat pieces. The patient had an
lesion was heterogeneous, intensely enhanc- uneventful recovery with recovery of the
ing, and had smooth boundary to adjacent swallowing and improvement of the facial
structures (e–h). Compared to the initial nerve palsy to HB grade III in 6 months. (i, j:
images (a, b), a certain growth tendency was early postoperative CT control).
evident with predominant expansion in the CP The histopathological diagnosis was fibrous
angle, in the cavernous sinus, and in sphenoid dysplasia. Follow-up CT and MRI scans at 6
sinus areas. A presumptive diagnosis of sphe- months were recommended with the option of
nopetroclival fibrous dysplasia was made. second surgery via an endonasal approach in
The huge size and the extension of the case of growth of the remnant. Twelve months
lesion precluded its safe resection in one later, he sent his CT, which showed a large
stage. Considering the presence of brain stem tumor recurrence with obstructive hydroceph-
compression, the retrosigmoid approach was alus (k–n). A VP shunt was inserted elsewhere.
favored. Besides the facial nerve, EMG, The patient is scheduled for a new surgery at
BAEP, SEP, as well as the abducens and the our institute.
836 11 Other Cerebellopontine Angle Tumors

a b

c d

e f

(a, b) Initial MRI: T1-weighted MR images after T2-CISS sequence. (i, j) Postoperative CT, demon-
gadolinium application. CT scans (c, d) and MR strating the extent of removal of the lesion and the
images scans performed at presentation to our insti- decompression of the brain stem. (k–n) Last CT
tute. (e, f) T1-weighted sequences after gadolinium scans, showing a large tumor recurrence and obstruc-
administration. (g) T2-weighted sequence. (h) tive hydrocephalus
Temporal Bone and Petrous Apex Lesions 837

g h

i j
838 11 Other Cerebellopontine Angle Tumors

k l

m n
Temporal Bone and Petrous Apex Lesions 839

Osteomas ders, and genetic defects may contribute to


their etiology. Recently we presented the first
Osteomas are rare benign slow-growing cranial case of bilateral osteomas arising from the
tumors originating typically from the inner or IAC and extending to the CP angle:
outer table of the calvarium, the bones of the Fig. 11.23a–d [207].
face or mandible [178]. They are round to oval Osteomas are usually asymptomatic for many
sessile lesions, composed of compact bone. years and are discovered incidentally. They may
Skull-base osteomas in the fronto-ethmoid area cause symptoms due to local compressive effect,
have been described relatively frequently. The depending on their specific origin and extension:
pyramid is rarely affected and the CP angle and trigeminal neuralgia, sensorineural hearing loss,
IAC are extremely rare [237]. Less than 25 vertigo, and tinnitus or even symptoms/signs of
such cases have been reported in the literature brain stem compression [257, 264, 277].
[181, 257, 264]. It has been proposed that Clinically they may be undistinguishable from
trauma, inflammation, developmental disor- VS or CP meningiomas.

a b

IAC

c d

Fig. 11.23 CT (a) axial view, (b) coronal view, (c) sagittal view and MRI (d) of a patient with an osteoma in the area
of the porus of the IAC
840 11 Other Cerebellopontine Angle Tumors

Osteomas should be differentiated from osteo- compression of the cochlear and vestibular
chondroma, osteosarcoma, ossifying fibroma, nerves, however, surgery may be less beneficial.
osteoblastic metastasis, isolated eosinophilic
granuloma, Paget’s disease, giant-cell tumor,
osteoid osteoma, calcified meningioma and Pleomorphic Adenomas
monostotic fibrous dysplasia, as well as temporal
bone exostoses. Exostoses are usually multiple Pleomorphic adenoma is the most common tumor
broad-based bone elevations, characterized histo- of the parotid, submandibular, and minor salivary
logically by parallel, concentric layers of subpe- glands. It is composed of both glandular epithelial
riosteal bone [221]. Osteomas are usually solitary, and mesenchymal elements, hence the name mixed
pedunculated lesions, with typical internal struc- tumor. Intracranial invasion of the tumor is very
ture: they are composed of discrete fibrovascular rare [282]. Pleomorphic adenomas of the external
channels surrounded by irregularly oriented auditory canal are benign tumors of the ceruminal
lamellated bone [181, 277]. glands, arising from the lateral cartilaginous por-
Asymptomatic tumors should be observed and tion of the canal [177]. Ceruminal glands are
followed with regular CT examinations. The modified sweat glands of the skin of the external
treatment of choice of symptomatic or enlarging auditory meatus. These tumors are usually benign:
osteomas is surgical removal [181, 237]. wide local excision is the mainstay of treatment
Following surgery, symptoms usually completely [233]. They may, however, extend to the petrous
resolve or at least improve [196, 207]. Davis et al. bone and cause wide bone destruction with respec-
[196] analyzed 12 patients, 2 from the authors tive symptoms. Peters et al. [251] described a case
own experience and 10 from the literature. Three of a pleomorphic adenoma of the middle ear and
of the 8 operated patients showed improvement mastoid, causing extensive bony destruction and
of hearing loss, resolution of their dizziness extending into the posterior cranial fossa. Primary
occurred in 5 patients, and resolution of tinnitus posterior fossa or CP angle pleomorphic adenomas
occurred in 4 patients. In case of chronic are exceedingly rare [283].
Temporal Bone and Petrous Apex Lesions 841

Case Illustration 11.27 tumor, extending caudally to the level of the


Pleomorphic Adenoma of the Petrous Bone jugular foramen (a–d). The tumor, which
and CP Angle proved to be a pleomorphic adenoma, was
A 41-year-old male patient presented with a removed via the retrosigmoid route. After sur-
recurrent tumor of the left petrous bone and gery the patient recovered rapidly and had a
the left CP angle. He has been operated at slight (HB grade II) facial palsy at discharge.
another hospital for a pleomorphic adenoma 7 His gait improved considerably and in 6
years earlier via a cervical approach on the left months was normal.
side, followed by fractionated radiotherapy The patient presented 3 years later with
(50 Gy total dosage). CSF leak on the left side via a small perfora-
Seven years later, a MRI follow-up study tion of the tympanic membrane. His gait, as
showed a lesion in the left petrous bone with well as the facial nerve function, was normal.
large CP angle portion. An open transmastoid The trigeminal hypesthesia (V2,3), abducens
biopsy was done, confirming the diagnosis of nerve palsy, complete hearing loss, and tinni-
pleomorphic adenoma. The tumor was then tus on the left side, however, persisted. MRI
removed via a transmastoid transpetrous and CT revealed liquid collection in the mas-
approach. MRI study, performed 6 months toid and a suspicious bone defect in the lower
later, demonstrated tumor involvement of the medial part of the mastoid process. Considering
left cavernous sinus and of the adjacent skull- the clinical and radiological findings, a deci-
base area, which was treated with gamma sion for surgical treatment was taken: dural
knife radiosurgery. repair with fat pieces was performed trans-
At presentation to our department 3 years mastoidally and the Eustachian tube, as well
later, the patient had gait ataxia, trigeminal as the external ear canal, was closed. At 12
hypesthesia (V2, 3), abducens nerve palsy, months follow-up examination, the patient
complete hearing loss, and tinnitus on the left had no new complaints. The follow-up MRI is
side. MRI demonstrated a large CP angle shown on figures e–g.
842 11 Other Cerebellopontine Angle Tumors

a b

c d

MR examination, demonstrating the tumor in the left Postoperative MR images: (e, f) T1-weighted sequences
CP angle and the involvement of the petrous bone. (a, b) after gadolinium administration. (g) T2-weighted
T1-weighted sequences after gadolinium administra- sequence
tion. (c) T2-weighted sequence. (d) CISS sequence.
Temporal Bone and Petrous Apex Lesions 843

e f

g
844 11 Other Cerebellopontine Angle Tumors

Intralabyrinthine Schwannomas 4. Bonneville F, Savatovsky J, Chiras J (2007) Imaging


of cerebellopontine angle lesions: an update. Part 2:
intra-axial lesions, skull base lesions that may invade
Mayer has described for the first time in 1917. the CPA region, and non-enhancing extra-axial
Since then less than 60 cases have been presented lesions. Eur Radiol 17(11):2908–2920. doi:10.1007/
in the literature. These tumors originate from the s00330-007-0680-4
distant branches of the cochlear, superior vestibu- 5. Brackmann DE, Bartels LJ (1980) Rare tumors of the
cerebellopontine angle. Otolaryngol Head Neck Surg
lar, or inferior vestibular nerves. Previously, most 88(5):555–559
cases have been found at autopsy or incidentally 6. Chiechi MV, Smirniotopoulos JG, Jones RV (1995)
during labyrinthectomy for Meniere’s syndrome Intracranial subependymomas: CT and MR imaging
or intractable vertigo [208, 245]. Nowadays, gad- features in 24 cases. AJR Am J Roentgenol 165(5):
1245–1250
olinium-enhanced MRI allows for preoperative 7. Cosgrove GR, Villemure JG, Robitaille Y, Melanson
diagnosis of such tumors [200]. D (1985) Extraaxial ependymoma of the posterior
Patients with intralabyrinthine schwannomas fossa. Surg Neurol 24(4):433–436
present typically with hearing loss, vertigo, or 8. Cuthikonda B, Buckleair LJ, Goodman JC, Powell
SZ, Rose JE (2009) Ganglioglioma in the cerebello-
tinnitus [245]. Hearing loss might be of mixed pontine angle of an adult without seizures. J La State
type: a conductive component secondary to inter- Med Soc 161(3):143–146
ference with stapes movement might be present. 9. Donich D, Lee JH, Prayson R (1999) Giant extra-axial
Green and McKenzie [208] stressed that symp- cerebellopontine angle/cavernous sinus ependymoma:
case report. Neurosurgery 44(1):195–198
toms associated with intralabyrinthine schwan- 10. Enomoto H, Mizuno M, Katsumata T, Doi T (1991)
noma may be confused with Meniere’s disease Intracranial metastasis of a choroid plexus papilloma
(hearing loss, tinnitus, aural fullness, and epi- originating in the cerebellopontine angle region: a
sodic vertigo). Therefore, they recommend pre- case report. Surg Neurol 36(1):54–58
11. Forton G, Verlooy J, Cras P, Parizel P, Van de Heyning
operative high-resolution MRI in all patients with P (1992) Problems with flute playing: an otological
serviceable hearing and intractable vertigo, problem? Case report of a peculiar cerebellar
undergoing surgery for Meniere’s disease. astrocytoma. Acta Otorhinolaryngol Belg 46(4):
Patients with profound hearing loss are best 405–410
12. Fukui MB, Hogg JP, Martinez AJ (1997) Extraaxial
managed surgically. Surgical removal of such ependymoma of the posterior fossa. AJNR Am J
tumors leads to complete hearing loss. Therefore, Neuroradiol 18(6):1179–1181
in case of preserved functional hearing, initial 13. Furuya K, Sasaki T, Saito N, Atsuchi M, Kirino T
observation with serial audiograms and MRI (1995) Primary large choroid plexus papillomas in the
cerebellopontine angle: radiological manifestations
might be an alternative [200, 208]. and surgical management. Acta Neurochir (Wien)
135(3–4):144–149
14. Gilbertson RJ (2004) Medulloblastoma: signalling a
change in treatment. Lancet Oncol 5(4):209–218.
References doi:10.1016/S1470-2045(04)01424-X
15. Hoeffel C, Boukobza M, Polivka M, Lot G,
Intra-Axial Tumors Extending Guichard JP, Lafitte F, Reizine D, Merland JJ (1995)
MR manifestations of subependymomas. AJNR Am J
Secondarily to the Cerebellopontine Neuroradiol 16(10):2121–2129
Angle 16. House JL, Burt MR (1985) Primary CNS tumors
presenting as cerebellopontine angle tumors. Am J
1. Ahn MS, Jackler RK (1997) Exophytic brain tumors Otol (Suppl):147–153
mimicking primary lesions of the cerebellopontine 17. Ikezaki K, Matsushima T, Inoue T, Yokoyama N,
angle. Laryngoscope 107(4):466–471 Kaneko Y, Fukui M (1993) Correlation of microana-
2. Arnautovic KI, Husain MM, Linskey ME (2000) Cranial tomical localization with postoperative survival in
nerve root entry zone primary cerebellopontine angle posterior fossa ependymomas. Neurosurgery 32(1):
gliomas: a rare and poorly recognized subset of extra- 38–44
parenchymal tumors. J Neurooncol 49(3):205–212 18. Jallo G (2006) Brainstem gliomas. Childs Nerv Syst
3. Beutler AS, Hsiang JK, Moorhouse DF, Hansen LA, 22(1):1–2. doi:10.1007/s00381-005-1267-5
Alksne JF (1995) Pilocytic astrocytoma presenting as 19. Kariya S, Nishizaki K, Aoji K, Akagi H (1998)
an extra-axial tumor in the cerebellopontine angle: Primary malignant lymphoma in the cerebellopontine
case report. Neurosurgery 37(1):125–128 angle. J Laryngol Otol 112(5):476–479
References 845

20. Kasantikul V, Palmer JO, Netsky MG, Glasscock ME 34. Pollack IF, Gerszten PC, Martinez AJ, Lo KH,
3rd, Hays JW (1980) Glioma of the acoustic nerve. Shultz B, Albright AL, Janosky J, Deutsch M (1995)
Arch Otolaryngol 106(8):456–459 Intracranial ependymomas of childhood: long-term
21. Kieserman S, Linstrom C, McCormick S, Petschenik AJ outcome and prognostic factors. Neurosurgery
(1996) Choroid plexus papilloma of the cerebellopon- 37(4):655–666; discussion 666–657
tine angle. Am J Otol 17(1):119–122 35. Prayson RA, Suh JH (1999) Subependymomas:
22. Koral K, Kedzierski RM, Gimi B, Gomez A, clinicopathologic study of 14 tumors, including
Rollins NK (2008) Subependymoma of the cerebel- comparative MIB-1 immunohistochemical analysis
lopontine angle and prepontine cistern in a 15-year-old with other ependymal neoplasms. Arch Pathol Lab
adolescent boy. AJNR Am J Neuroradiol 29(1):190– Med 123(4):306–309, 10.1043/0003-9985(1999)
191, ajnr.A0821 [pii] 123<0306:S>2.0.CO;2
23. Kumar R, Achari G, Banerjee D, Chhabra DK (2001) 36. Raffel C (2004) Medulloblastoma: molecular genet-
Uncommon presentation of medulloblastoma. Childs ics and animal models. Neoplasia 6(4):310–322.
Nerv Syst 17(9):538–542; discussion 543 doi:10.1593/neo.03454
24. Kumar R, Achari G, Benerji D, Jain VK, Chhabra DK 37. Rea GL, Akerson RD, Rockswold GL, Smith SA
(2002) Choroid plexus papillomas of the cerebello- (1983) Subependymoma in a 2 1/2-year-old boy. Case
pontine angle. Neurol India 50(3):352–358 report. J Neurosurg 59(6):1088–1091. doi:10.3171/
25. Martin N, Pierot L, Sterkers O, Mompoint D, jns.1983.59.6.1088
Nahum H (1990) Primary choroid plexus papilloma 38. Rossi A, Caracciolo V, Russo G, Reiss K, Giordano A
of the cerebellopontine angle: MR imaging. (2008) Medulloblastoma: from molecular pathology
Neuroradiology 31(6):541–543 to therapy. Clin Cancer Res 14(4):971–976.
26. Massimino M, Giangaspero F, Garre ML, Genitori L, doi:10.1158/1078-0432.CCR-07-2072, 14/4/971 [pii]
Perilongo G, Collini P, Riva D, Valentini L, Scarzello 39. Rovit RL, Schechter MM, Chodroff P (1970) Choroid
G, Poggi G, Spreafico F, Peretta P, Mascarin M, plexus papillomas. Observations on radiographic
Modena P, Sozzi G, Bedini N, Biassoni V, Urgesi A, diagnosis. Am J Roentgenol Radium Ther Nucl Med
Balestrini MR, Finocchiaro G, Sandri A, Gandola L 110(3):608–617
(2006) Salvage treatment for childhood ependymoma 40. Rushing EJ, Cooper PB, Quezado M, Begnami M,
after surgery only: pitfalls of omitting “at once” adju- Crespo A, Smirniotopoulos JG, Ecklund J, Olsen C,
vant treatment. Int J Radiat Oncol Biol Phys Santi M (2007) Subependymoma revisited: clinico-
65(5):1440–1445, S0360-3016(06)00675-4 [pii] pathological evaluation of 83 cases. J Neurooncol
27. Mehta JS, Sharr MM (1998) An unusual cause of 85(3):297–305. doi:10.1007/s11060-007-9411-6
acute labyrinthine failure. J Laryngol Otol 112(2): 41. Sanford RA, Merchant TE, Zwienenberg-Lee M,
193–195 Kun LE, Boop FA (2009) Advances in surgical tech-
28. Merchant TE, Boop FA, Kun LE, Sanford RA (2008) niques for resection of childhood cerebellopontine
A retrospective study of surgery and reirradiation for angle ependymomas are key to survival. Childs Nerv
recurrent ependymoma. Int J Radiat Oncol Biol Phys Syst 25(10):1229–1240. doi:10.1007/s00381-009-
71(1):87–97, S0360-3016(07)04225-3 [pii] 0886-7
29. Meyers SP, Kemp SS, Tarr RW (1992) MR imaging 42. Springborg JB, Poulsgaard L, Thomsen J (2008)
features of medulloblastomas. AJR Am J Roentgenol Nonvestibular schwannoma tumors in the cerebello-
158(4):859–865 pontine angle: a structured approach and management
30. Milligan BD, Giannini C, Link MJ (2007) guidelines. Skull Base 18(4):217–227. doi:10.1055/s-
Ganglioglioma in the cerebellopontine angle in a 2007-1016959
child. Case report and review of the literature. J 43. Swaroop GR, Whittle IR (1997) Exophytic pon-
Neurosurg 107(4 Suppl):292–296. doi:10.3171/PED- tine glioblastoma mimicking acoustic neuroma.
07/10/292 J Neurosurg Sci 41(4):409–411
31. Mirone G, Schiabello L, Chibbaro S, Bouazza S, 44. Tacconi L, Delfini R, Cantore G (1996) Choroid
George B (2009) Pediatric primary pilocytic astrocy- plexus papillomas: consideration of a surgical series
toma of the cerebellopontine angle: a case report. of 33 cases. Acta Neurochir (Wien) 138(7):802–810
Childs Nerv Syst 25(2):247–251. doi:10.1007/ 45. Takada Y, Ohno K, Tamaki M, Hirakawa K (1999)
s00381-008-0690-9 Cerebellopontine angle pilocytic astrocytoma mim-
32. Nishimura T, Uchida Y, Fukuoka M, Ono Y, icking acoustic schwannoma. Neuroradiology 41(12):
Kurisaka M, Mori K (1998) Cerebellopontine angle 949–950
lymphoma: a case report and review of the literature. 46. Talacchi A, De Micheli E, Lombardo C, Turazzi S,
Surg Neurol 50(5):480–485, S0090-3019(97)00362-5 Bricolo A (1999) Choroid plexus papilloma of the
[pii]; discussion 485–486 cerebellopontine angle: a twelve patient series.
33. Pencalet P, Sainte-Rose C, Lellouch-Tubiana A, Kalifa C, Surg Neurol 51(6):621–629, S0090-3019(99)00024-5
Brunelle F, Sgouros S, Meyer P, Cinalli G, Zerah M, [pii]
Pierre-Kahn A, Renier D (1998) Papillomas and carcino- 47. Yamada S, Aiba T, Hara M (1993) Cerebellopontine
mas of the choroid plexus in children. J Neurosurg angle medulloblastoma: case report and literature
88(3):521–528. doi:10.3171/jns.1998.88.3.0521 review. Br J Neurosurg 7(1):91–94
846 11 Other Cerebellopontine Angle Tumors

48. Yang PJ, Seeger JF, Carmody RF, Mehta BA (1987) 62. Birch BD, McCormick PC (1996) High cervical split
Cerebellopontine angle lymphoma. AJNR Am J cord malformation and neurenteric cyst associated
Neuroradiol 8(2):368–369 with congenital mirror movements: case report.
49. Yoshimura J, Nishiyama K, Fukuda M, Watanabe M, Neurosurgery 38(4):813–815; discussion 815–816
Igarashi H, Fujii Y (2009) Adult cerebellopontine 63. Bonneville F, Sarrazin JL, Marsot-Dupuch K,
angle medulloblastoma originating in the pons mim- Iffenecker C, Cordoliani YS, Doyon D, Bonneville JF
icking focal brainstem tumor. J Neuroimaging (2001) Unusual lesions of the cerebellopontine angle:
19(4):385–387, JON298 [pii] a segmental approach. Radiographics 21(2):419–438
50. Yuh EL, Barkovich AJ, Gupta N (2009) Imaging of 64. Bonneville F, Savatovsky J, Chiras J (2007) Imaging
ependymomas: MRI and CT. Childs Nerv Syst of cerebellopontine angle lesions: an update. Part 1:
25(10):1203–1213. doi:10.1007/s00381-009-0878-7 enhancing extra-axial lesions. Eur Radiol 17(10):
2472–2482. doi:10.1007/s00330-007-0679-x
65. Bonneville F, Savatovsky J, Chiras J (2007) Imaging
of cerebellopontine angle lesions: an update. Part 2:
Rare Extra-axial Cerebellopontine intra-axial lesions, skull base lesions that may invade
Angle Lesions the CPA region, and non-enhancing extra-axial
lesions. Eur Radiol 17(11):2908–2920. doi:10.1007/
51. Abbott R (2004) The endoscopic management of s00330-007-0680-4
arachnoidal cysts. Neurosurg Clin N Am 15(1):9–17. 66. Brackmann DE, Bartels LJ (1980) Rare tumors of the
doi:10.1016/S1042-3680(03)00071-8 cerebellopontine angle. Otolaryngol Head Neck Surg
52. Abe K, Oyama K, Mori K, Ishimaru S, Eguchi M, 88(5):555–559
Maeda M (1999) Neurenteric cyst of the craniocervi- 67. Brodsky JR, Smith TW, Litofsky S, Lee DJ (2006)
cal junction – case report. Neurol Med Chir (Tokyo) Lipoma of the cerebellopontine angle. Am J Otolaryngol
39(12):875–880, JST.Journalarchive/nmc1959/39.875 27(4):271–274, S0196-0709(05)00218-8 [pii]
[pii] 68. Brooks ML, Mayer DP, Sataloff RT, Myers DL,
53. Agnoli AL, Laun A, Schonmayr R (1984) Entero- Spiegel JR, Ruiz S (1992) Intracanalicular arachnoid
genous intraspinal cysts. J Neurosurg 61(5):834–840. cyst mimicking acoustic neuroma: CT and MRI.
doi:10.3171/jns.1984.61.5.0834 Comput Med Imaging Graph 16(4):283–285
54. Aguiar PH, Agner C, Tavares FR, Yamaguchi N (2001) 69. Caldarelli M, Massimi L, Kondageski C, Di Rocco C
Unusual brain metastases from papillary thyroid carci- (2004) Intracranial midline dermoid and epidermoid
noma: case report. Neurosurgery 49(4):1008–1013 cysts in children. J Neurosurg 100(5 Suppl
55. Andaluz N, Pensak ML, Zuccarello M (2005) Pediatrics):473–480. doi:10.3171/ped.2004.100.5.0473
Multiple, peripheral aneurysms of the anterior inferior 70. Carvalho GA, Cervio A, Matthies C, Samii M (2000)
cerebellar artery. Acta Neurochir (Wien) 147(4):419– Subarachnoid fat dissemination after resection of a
422. doi:10.1007/s00701-004-0453-7; discussion 422 cerebellopontine angle dysontogenic cyst: case report
56. Arriaga MA, Lo WW, Brackmann DE (1995) and review of the literature. Neurosurgery 47(3):
Metastatic melanoma to the cerebellopontine angle. 760–763; discussion 763–764
Clinical and imaging characteristics. Arch Otolaryngol 71. Carvalho GA, Matthies C, Osorio E, Samii M (2003)
Head Neck Surg 121(9):1052–1056 Hamartomas of the internal auditory canal: report of
57. Babu R, Murali R (1991) Arachnoid cyst of the two cases. Neurosurgery 52(4):944–948; discussion
cerebellopontine angle manifesting as contralateral 948–949
trigeminal neuralgia: case report. Neurosurgery 28(6): 72. Celis MA, Mourier KL, Polivka M, Boissonnet H,
886–887 Kato T, Lot G, George B, Cophignon J (1992)
58. Barrera JE, Jenkins H, Said S (2004) Cavernous Cisternal cysticercosis of the cerebellopontine angle.
hemangioma of the internal auditory canal: a case A surgically-treated case and review of the literature.
report and review of the literature. Am J Otolaryngol Neurochirurgie 38(2):108–112
25(3):199–203, S0196070903001571 [pii] 73. Chang SD, Meisel JA, Hancock SL, Martin DP,
59. Bejjani GK, Wright DC, Schessel D, Sekhar LN McManus M, Adler JR Jr (1998) Treatment of heman-
(1998) Endodermal cysts of the posterior fossa. Report gioblastomas in von Hippel-Lindau disease with lin-
of three cases and review of the literature. J Neurosurg ear accelerator-based radiosurgery. Neurosurgery
89(2):326–335. doi:10.3171/jns.1998.89.2.0326 43(1):28–34; discussion 34–35
60. Bengochea FG, Blanco FL (1955) Arachnoidal cysts 74. Chaynes P, Thorn-Kany M, Sol JC, Arrue P,
of the cerebellopontine angle. J Neurosurg 12(1): Lagarrigue J, Manelfe C (1998) Imaging in neuren-
66–71. doi:10.3171/jns.1955.12.1.0066 teric cysts of the posterior cranial fossa. Neuroradiology
61. Bertalanffy H, Benes L, Miyazawa T, Alberti O, 40(6):374–376
Siegel AM, Sure U (2002) Cerebral cavernomas in the 75. Cheng JS, Cusick JF, Ho KC, Ulmer JL (2002)
adult. Review of the literature and analysis of 72 sur- Lateral supratentorial endodermal cyst: case report
gically treated patients. Neurosurg Rev 25(1–2):1–53; and review of literature. Neurosurgery 51(2):493–499;
discussion 54–55 discussion 499
References 847

76. Chihara Y, Iwasaki S, Ushio M, Sugasawa K, 92. Filling-Katz MR, Choyke PL, Patronas NJ,
Murofushi T (2009) Fusiform aneurysm of the basilar Gorin MB, Barba D, Chang R, Doppman JL,
artery presenting as a cerebellopontine angle mass. Seizinger B, Oldfield EH (1989) Radiologic screen-
Eur Arch Otorhinolaryngol 266(1):151–152. ing for von Hippel-Lindau disease: the role of
doi:10.1007/s00405-008-0635-7 Gd-DTPA enhanced MR imaging of the CNS. J
77. Chiong Y, Mulroy L, Fleetwood IG, Younis T (2009) Comput Assist Tomogr 13(5):743–755
Isolated metastasis to the cerebellopontine angle sec- 93. Geddes JF, Sutcliffe JC, King TT (1995) Mixed cra-
ondary to breast cancer. Can J Surg 52(5):E213–E214 nial nerve tumors in neurofibromatosis type 2. Clin
78. Constans JP, Meder F, Maiuri F, Donzelli R, Neuropathol 14(6):310–313
Spaziante R, de Divitiis E (1986) Posterior fossa 94. Geetha N, James FV, Ramachandran K,
hemangioblastomas. Surg Neurol 25(3):269–275 Ajithkumar TV, Nair MK (1999) Ovarian carcinoma
79. Conway JE, Chou D, Clatterbuck RE, Brem H, relapsing as a cerebellopontine angle tumour.
Long DM, Rigamonti D (2001) Hemangioblastomas Australas Radiol 43(3):388–390
of the central nervous system in von Hippel-Lindau 95. Gjuric M, Koester M, Paulus W (2000) Cavernous
syndrome and sporadic disease. Neurosurgery 48(1): hemangioma of the internal auditory canal arising
55–62; discussion 62–63 from the inferior vestibular nerve: case report and
80. Dalley RW, Robertson WD, Nugent RA, Durity FA review of the literature. Am J Otol 21(1):110–114
(1986) Computed tomography of anterior inferior 96. Gokcan MK, Batikhan H, Calguner M, Tataragasi
cerebellar artery aneurysm mimicking an acoustic AI (2006) Unilateral hearing loss as a presenting
neuroma. J Comput Assist Tomogr 10(5):881–884 manifestation of granulocytic sarcoma (chloroma).
81. Davis LE, Kornfeld M (1991) Neurocysticercosis: Otol Neurotol 27(1):106–109, 00129492-
neurologic, pathogenic, diagnostic and therapeutic 200601000-00019 [pii]
aspects. Eur Neurol 31(4):229–240 97. Gonul E, Yetiser S, Tasar M, Ongoru O (2006)
82. Debelius A, Rudolph J, Schrom T (2006) Lipoma of the Glioneural hamartoma of the VIIIth nerve. J Laryngol
cerebellopontineangle–acasereport.Laryngorhinootologie Otol 120(1):70–73, S0022215105009515 [pii]
85(2):128–131. doi:10.1055/s-2005-870116 98. Guidetti B, Gagliardi FM (1977) Epidermoid and
83. Depper MH, Carlow TJ, Crooks LA, Orrison WW Jr dermoid cysts. Clinical evaluation and late surgical
(1996) Intracranial dissemination of a pituitary adenoma: results. J Neurosurg 47(1):12–18. doi:10.3171/
presentation as an unusual mass in the cerebellopontine jns.1977.47.1.0012
angle. AJR Am J Roentgenol 166(6):1500–1501 99. Guilemany JM, Alobid I, Gaston F, Morrello A,
84. Deshmukh VR, Albuquerque FC, Zabramski JM, Bernal-Sprekelsen M (2005) Cerebellopontine angle
Spetzler RF (2003) Surgical management of cavern- and internal auditory canal metastasis from ductal
ous malformations involving the cranial nerves. carcinoma of the breast. Acta Otolaryngol 125(9):
Neurosurgery 53(2):352–357; discussion 357 1004–1007
85. Devkota UP, Lam JM, Ng H, Poon WS (1994) An 100. Hadley MN, Grahm TW, Daspit CP, Spetzler RF
anterior intradural neurenteric cyst of the cervical (1985) Otolaryngologic manifestations of posterior
spine: complete excision through central corpectomy fossa arachnoid cysts. Laryngoscope 95(6):
approach – case report. Neurosurgery 35(6): 678–681
1150–1153; discussion 1153–1154 101. Harris CP, Dias MS, Brockmeyer DL, Townsend JJ,
86. Drake CG, Friedman AH, Peerless SJ (1986) Willis BK, Apfelbaum RI (1991) Neurenteric cysts
Posterior fossa arteriovenous malformations. J Neuro- of the posterior fossa: recognition, management, and
surg 64(1):1–10. doi:10.3171/jns.1986.64.1.0001 embryogenesis. Neurosurgery 29(6):893–897; dis-
87. Duchene M, Benoudiba F, Iffenecker C, Hadj-Rabia cussion 897–898
M, Caldas J, Doyon D (1999) Neurocysticercosis. 102. Huang TW, Young YH (2002) Differentiation
J Radiol 80(12):1623–1627, MDOI-JR-12-1999-80- between cerebellopontine angle tumors in cancer
12-0221-0363-101019-ART2 [pii] patients. Otol Neurotol 23(6):975–979
88. Dutt SN, Mirza S, Chavda SV, Irving RM (2002) 103. Ikeda H, Deinsberger W, Boker DK (2000)
Radiologic differentiation of intracranial epidermoids Retroclival arachnoid cyst presenting with spontane-
from arachnoid cysts. Otol Neurotol 23(1):84–92 ous intracystic haemorrhage – case presentation.
89. Eynon-Lewis NJ, Kitchen N, Scaravilli F, Brookes Acta Neurochir (Wien) 142(11):1317–1318
GB (1998) Neurenteric cyst of the cerebellopontine 104. Ildan F, Gocer AI, Bagdatoglu H, Uzuneyupoglu
angle: case report. Neurosurgery 42(3):655–658 Z, Tuna M, Cetinalp E (1996) Isolated trigeminal
90. Falcioni M, Piccirillo E, Di Trapani G, Romano G, neuralgia secondary to distal anterior inferior cer-
Russo A (2004) Internal auditory canal metastasis. ebellar artery aneurysm. Neurosurg Rev 19(1):
Acta Otorhinolaryngol Ital 24(2):78–82 43–46
91. Filho FL, Tatagiba M, Carvalho GA, Weichhold W, 105. Iplikcioglu C, Bikmaz K, Gokduman CA, Bek S
Klekamp J, Samii M (2001) Neurenteric cyst of the (2006) Cerebellopontine angle lipoma with extracra-
craniocervical junction. Report of three cases. nial extension. J Clin Neurosci 13(10):1045–1047,
J Neurosurg 94(1 Suppl):129–132 S0967-5868(06)00471-1 [pii]
848 11 Other Cerebellopontine Angle Tumors

106. Izci Y, Secer HI, Gonul E, Onguru O (2007) 119. Link MJ, Driscoll CL, Giannini C (2002) Isolated,
Simultaneously occurring vestibular schwannoma giant cerebellopontine angle craniopharyngioma in a
and meningioma in the cerebellopontine angle: case patient with Gardner syndrome: case report.
report and literature review. Clin Neuropathol 26(5): Neurosurgery 51(1):221–225; discussion 225–226
219–223 120. Lüdemann W, Stan AC, Tatagiba M, Samii M (2000)
107. Jallo GI, Woo HH, Meshki C, Epstein FJ, Wisoff JH Sporadic unilateral vestibular schwannoma with
(1997) Arachnoid cysts of the cerebellopontine islets of meningioma: case report. Neurosurgery
angle: diagnosis and surgery. Neurosurgery 40(1): 47(2):451–452; discussion 452–454
31–37; discussion 37–38 121. Marin-Sanabria EA, Yamamoto H, Nagashima T,
108. Jamjoom AB, Jamjoom ZA, al-Fehaily M, Kohmura E (2007) Evaluation of the management of
el-Watidy S, al-Moallem M, Nain Ur R (1996) arachnoid cyst of the posterior fossa in pediatric pop-
Trigeminal neuralgia related to cerebellopontine ulation: experience over 27 years. Childs Nerv Syst
angle tumors. Neurosurg Rev 19(4):237–241 23(5):535–542. doi:10.1007/s00381-006-0284-3
109. Jawahar A, Kondziolka D, Garces YI, Flickinger JC, 122. Marton E, Basaldella L, Longatti PL (2009) Minimal
Pollock BE, Lunsford LD (2000) Stereotactic radio- surgery for a cerebellopontine angle lipoma. J Clin
surgery for hemangioblastomas of the brain. Acta Neurosci 16(1):129–132, S0967-5868(08)00185-9
Neurochir (Wien) 142(6):641–644; discussion [pii]
644–645 123. Mizushima H, Kobayashi N, Yoshiharu S, Kazuo H,
110. Kalavagunta S, Karkanevatos A, Swift AC (2006) Dohi K, Sasaki K, Matsumoto K (1999) Aneurysm
Giant vertebro-basilar aneurysm: an unusual cere- of the distal anterior inferior cerebellar artery at the
bello-pontine angle lesion. J Laryngol Otol medial branch: a case report and review of the litera-
120(2):e8 ture. Surg Neurol 52(2):137–142, S0090-
111. Kamitani H, Hirano N, Takigawa H, Yokota M, 3019(99)00042-7 [pii]
Miyata H, Ohama E, Watanabe T (2004) Attenuation 124. Moffat DA, O’Connor AF (1980) Bilateral internal
of vascularity by preoperative radiosurgery facili- carotid aneurysms in the petrous temporal bones.
tates total removal of a hypervascular hemangioblas- Arch Otolaryngol 106(3):172–175
toma at the cerebello-pontine angle: case report. 125. Moffat DA, Saunders JE, McElveen JT Jr, McFerran
Surg Neurol 62(3):238–243. doi:10.1016/j. DJ, Hardy DG (1993) Unusual cerebello-pontine
surneu.2003.09.030; discussion 243–234 angle tumours. J Laryngol Otol 107(12):1087–1098
112. Kawamata T, Kawamura H, Kubo O, Sasahara A, 126. Morris DP, Ballagh RH, Hong A, Moffat DA,
Yamazato M, Hori T (1999) Central nervous system Hardy DG (1995) Thrombosed posterior-inferior
metastasis from gallbladder carcinoma mimicking a cerebellar artery aneurysm: a rare cerebellopontine
meningioma. Case illustration. J Neurosurg 91(6): angle tumour. J Laryngol Otol 109(5):429–430
1059. doi:10.3171/jns.1999.91.6.1059 127. Muzumdar DP, Goel A, Desai KI (2001) Pontine
113. Kohan D, Downey LL, Lim J, Cohen NL, Elowitz E glioma and cerebellopontine angle epidermoid
(1997) Uncommon lesions presenting as tumors of tumour occurring as collision tumours. Br J Neuro-
the internal auditory canal and cerebellopontine surg 15(1):68–71
angle. Am J Otol 18(3):386–392 128. Neumann HP, Eggert HR, Weigel K, Friedburg H,
114. Komotar RJ, Connolly ES Jr, Khandji A, Wiestler OD, Schollmeyer P (1989) Hemangio-
Teitelbaum GP, Lavine SD (2005) Clinicoradiological blastomas of the central nervous system. A 10-year
review: symptomatic superior cerebellar artery study with special reference to von Hippel-Lindau
aneurysm imitating cerebellopontine angle tumor. syndrome. J Neurosurg 70(1):24–30. doi:10.3171/
Neurosurgery 56(4):836–840; discussion 836–840 jns.1989.70.1.0024
115. Krishnan KG, Schackert G (2006) Outcomes of 129. Noujaim SE, Rossi MD, Rao SK, Cacciarelli AA,
surgical resection of large solitary hemangioblasto- Mendonca RA, Wang AM, Coelho FH (1999) CT
mas of the craniocervical junction with limitations in and MR imaging of neurocysticercosis. AJR Am J
preoperative angiographic intervention: report of Roentgenol 173(6):1485–1490
three cases. Zentralbl Neurochir 67(3):137–143. 130. Nozaki K, Hashimoto N, Kikuta K, Takagi Y,
doi:10.1055/s-2006-933360 Kikuchi H (2006) Surgical applications to arterio-
116. Lakshmi M, Glastonbury CM (2009) Imaging of the venous malformations involving the brainstem.
cerebellopontine angle. Neuroimaging Clin N Am Neurosurgery 58(4 Suppl 2):ONS-270–ONS-278.
19(3):393–406, S1052-5149(09)00040-9 [pii] doi:10.1227/01.NEU.0000210001.75597.81; discus-
117. Lee SR, Sanches J, Mark AS, Dillon WP, Norman D, sion ONS-278–ONS-279
Newton TH (1989) Posterior fossa hemangioblasto- 131. Odake G, Yamaki T, Naruse S (1976) Neurenteric cyst
mas: MR imaging. Radiology 171(2):463–468 with meningomyelocele. Case report J Neurosurg
118. Leeman DJ, Chandrasekhar SS, Brackmann DE, 45(3):352–356. doi:10.3171/jns.1976.45.3.0352
Poletti BJ (1997) Collision tumors at the cerebello- 132. Omojola MF, al Hawashim NS, Zuwayed MA, al
pontine angle: case report with literature review. Ferayan A (1997) CT and MRI features of cavernous
Otolaryngol Head Neck Surg 117(6):S76–S80, haemangioma of internal auditory canal. Br J Radiol
S0194-5998(97)70063-9 [pii] 70(839):1184–1187
References 849

133. Orakcioglu B, Halatsch ME, Fortunati M, 147. Schuknecht HF, Gao YZ (1983) Arachnoid cyst in
Unterberg A, Yonekawa Y (2008) Intracranial der- the internal auditory canal. Ann Otol Rhinol
moid cysts: variations of radiological and clinical Laryngol 92(6 Pt 1):535–541
features. Acta Neurochir (Wien) 150(12): 148. Sepehrnia A, Rebolledo Godoy AP, Reusche E
1227–1234; discussion 1234 (2000) A cavernous hemangioma simulating an
134. Pappas DG, Schneiderman TS, Brackmann DE, intracanalicular acoustic neurinoma – a case report.
Simpson LC, Chandra-Sekar B, Sofferman RA Zentralbl Neurochir 61(4):194–197
(1989) Cavernous hemangiomas of the internal audi- 149. Shakudo M, Inoue Y, Ohata K, Tanaka S (2001)
tory canal. Otolaryngol Head Neck Surg 101(1): Neurenteric cyst with alteration of signal intensity
27–32 on follow-up MR images. AJNR Am J Neuroradiol
135. Pavesi G, Berlucchi S, Munari M, Manara R, 22(3):496–498
Scienza R, Opocher G (2010) Clinical and surgical 150. Shinogami M, Yamasoba T, Sasaki T (1998) Bilateral
features of lower brain stem hemangioblastomas in isolated metastases of malignant melanoma to the
von Hippel-Lindau disease. Acta Neurochir (Wien) cerebellopontine angle. Otolaryngol Head Neck
152(2):287–292. doi:10.1007/s00701-009-0512-1 Surg 118(2):276–279, S0194599898000539 [pii]
136. Redleaf MI, Bauer CA, Laurenzo JF, Gantz BJ 151. Solomon RA, Stein BM (1986) Management of arte-
(1994) Squamous cell carcinoma metastatic to the riovenous malformations of the brain stem. J Neurosurg
cerebellopontine angle. Ann Otol Rhinol Laryngol 64(6):857–864. doi:10.3171/jns.1986.64.6.0857
103(11):908–910 152. Spetzger U, Bertalanffy H, Huffmann B, Mayfrank
137. Rengachary SS, Watanabe I (1981) Ultrastructure L, Reul J, Gilsbach JM (1996) Hemangioblastomas
and pathogenesis of intracranial arachnoid cysts. of the spinal cord and the brainstem: diagnostic and
J Neuropathol Exp Neurol 40(1):61–83 therapeutic features. Neurosurg Rev 19(3):147–151
138. Revuelta R, Soto-Hernandez JL, Vales LO, 153. Sun Y, Wrede KH, Chen Z, Bao Y, Ling F (2009)
Gonzalez RH (2003) Cerebellopontine angle cystic- Ruptured intrameatal AICA aneurysms – a report of
ercus and concurrent vascular compression in a case two cases and review of the literature. Acta Neurochir
of trigeminal neuralgia. Clin Neurol Neurosurg (Wien) 151(11):1525–1530. doi:10.1007/s00701-
106(1):19–22, S0303846703000623 [pii] 009-0269-6
139. Richard S, David P, Marsot-Dupuch K, Giraud S, 154. Symon L, Murota T, Pell M, Bordi L (1993) Surgical
Beroud C, Resche F (2000) Central nervous system management of haemangioblastoma of the posterior
hemangioblastomas, endolymphatic sac tumors, and fossa. Acta Neurochir (Wien) 120(3–4):103–110
von Hippel-Lindau disease. Neurosurg Rev 23(1): 155. Takeuchi S, Tanaka R, Fujii Y, Abe H, Ito Y (2001)
1–22; discussion 23–24 Surgical treatment of hemangioblastomas with pre-
140. Rigamonti D, Johnson PC, Spetzler RF, Hadley MN, surgical endovascular embolization. Neurol Med
Drayer BP (1991) Cavernous malformations and Chir (Tokyo) 41(5):246–251, JST.JSTAGE/
capillary telangiectasia: a spectrum within a single nmc/41.246 [pii]; discussion 251–242
pathological entity. Neurosurgery 28(1):60–64 156. Tampieri D, Leblanc R, TerBrugge K (1993)
141. Rodriguez F, Scheithauer BW, Ockner DM, Preoperative embolization of brain and spinal
Giannini C (2004) Solitary fibrous tumor of the hemangioblastomas. Neurosurgery 33(3):502–505;
cerebellopontine angle with salivary gland heteroto- discussion 505
pia: a unique presentation. Am J Surg Pathol 28(1): 157. Tang GC, Piao YS, Zhao L, Lu DH (2007) Lung
139–142 adenocarcinoma metastasizing to cerebellopontine
142. Russo N, Domenicucci M, Beccaglia MR, Santoro A angle schwannoma (collision tumor). Acta Neurochir
(2008) Spontaneous reduction of intracranial arach- (Wien) 149(1):87–90. doi:10.1007/s00701-006-
noid cysts: a complete review. Br J Neurosurg 1049-1; discussion 90
22(5):626–629, 905633552 [pii] 158. Tankere F, Vitte E, Martin-Duverneuil N, Soudant J
143. Sahara Y, Nagasaka T, Takayasu M, Takagi T, Hata (2002) Cerebellopontine angle lipomas: report of
N, Yoshida J (2001) Recurrence of a neurenteric cyst four cases and review of the literature. Neurosurgery
with malignant transformation in the foramen mag- 50(3):626–631; discussion 631–622
num after total resection. Case report J Neurosurg 159. Tu AS, Wang H, Harris-McCorkle L, Saunders JR Jr
95(2):341–345. doi:10.3171/jns.2001.95.2.0341 (1994) Bilateral involvement of the cerebellopontine
144. Samii M, Carvalho GA, Schuhmann MU, Matthies C angles by malignant melanoma metastasis: a case
(1999) Arachnoid cysts of the posterior fossa. Surg report. Md Med J 43(11):967–970
Neurol 51(4):376–382, S0090-3019(98)00095-0 [pii] 160. Vazquez-Anon V, Botella C, Beltran A, Solera M,
145. Samii M, Nakamura M, Mirzai S, Vorkapic P, Cervio Piquer J (1997) Preoperative embolization of solid
A (2006) Cavernous angiomas within the internal cervicomedullary junction hemangioblastomas:
auditory canal. J Neurosurg 105(4):581–587. report of two cases. Neuroradiology 39(2):86–89
doi:10.3171/jns.2006.105.4.581 161. Warakaulle DR, Anslow P (2003) Differential diag-
146. Schuhmann MU, Ludeman WO, Schreiber H, Samii nosis of intracranial lesions with high signal on T1
M (1997) Cerebellopontine angle lipoma. Skull Base or low signal on T2-weighted MRI. Clin Radiol
7(4):199–205 58(12):922–933, S000992600300268X [pii]
850 11 Other Cerebellopontine Angle Tumors

162. Warren FM, Shelton C, Wiggins RH 3rd, Herrod 176. Austin-Seymour M, Munzenrider J, Goitein M,
HC, Harnsberger HR (2008) Imaging characteristics Verhey L, Urie M, Gentry R, Birnbaum S,
of metastatic lesions to the cerebellopontine angle. Ruotolo D, McManus P, Skates S et al (1989)
Otol Neurotol 29(6):835–838. doi:10.1097/ Fractionated proton radiation therapy of chordoma
MAO.0b013e31818133ae and low-grade chondrosarcoma of the base of the
163. Wilms GPC, Goossens L et al (1992) Differentiation skull. J Neurosurg 70(1):13–17. doi:10.3171/
of acoustic neurinoma and meningioma occurring jns.1989.70.1.0013
together in the cerebellopontine angle. Neurosurgery 177. Ayers LS, Depasquale K, Marlowe FI, Ghaderi M
30:443–445 (2010) Pleomorphic adenoma of the external audi-
164. Yang B, Jin HM, Sun LP, Cai W, Shi CR (2008) tory canal: a case report and review of the literature.
Posterior fossa abscesses secondary to dermal Ear Nose Throat J 89(3):E1–E3
sinus associated with dermoid cyst in children. 178. Bartlett JR (1971) Intracranial neurological
Neuropediatrics 39(1):39–42. doi:10.1055/s- complications of frontal and ethmoidal osteomas. Br
2008-1077086 J Surg 58(8):607–613
165. Yasargil MG, Abernathey CD, Sarioglu AC (1989) 179. Bibby K, McFadzean R (1994) Fibrous dysplasia of
Microneurosurgical treatment of intracranial der- the orbit. Br J Ophthalmol 78(4):266–270
moid and epidermoid tumors. Neurosurgery 24(4): 180. Bockmuhl U, Khalil HS, Draf W (2005)
561–567 Clinicoradiological and surgical considerations in
166. Yilmaz C, Altinors N, Sonmez E, Gulsen S, Caner H the treatment of cholesterol granuloma of the
(2010) Rare lesions of the cerebellopontine angle. petrous pyramid. Skull Base 15(4):263–267.
Turk Neurosurg 20(3):390–397. doi:10.5137/1019- doi:10.1055/s-2005-918618; discussion 267–268
5149.JTN.2961-10.0 181. Boedts M, Hermans R, Feenstra L (1997) Osteoma
167. Young YH, Lin CY, Lou PJ, Hsu MM (2001) of the internal auditory canal. Acta Otorhinolaryngol
Intracranial relapse of nasopharyngeal carcinoma Belg 51(3):191–193
manifested as sudden deafness. Otol Neurotol 182. Brackmann DE, Toh EH (2002) Surgical manage-
22(3):392–396 ment of petrous apex cholesterol granulomas. Otol
168. Yuh WT, Mayr-Yuh NA, Koci TM, Simon JH, Neurotol 23(4):529–533
Nelson KL, Zyroff J, Jinkins JR (1993) Metastatic 183. Brodkey JA, Robertson JH, Shea JJ 3rd, Gardner G
lesions involving the cerebellopontine angle. AJNR (1996) Cholesterol granulomas of the petrous
Am J Neuroradiol 14(1):99–106 apex: combined neurosurgical and otological man-
agement. J Neurosurg 85(4):625–633. doi:10.3171/
jns.1996.85.4.0625
184. Brown EW, Megerian CA, McKenna MJ, Weber A
Temporal Bone and Petrous Apex (1995) Fibrous dysplasia of the temporal bone:
Lesions imaging findings. AJR Am J Roentgenol 164(3):
679–682
169. Adada B, Al-Mefty O (2003) Fibrous dysplasia of the 185. Butman JA, Kim HJ, Baggenstos M, Ammerman JM,
clivus. Neurosurgery 52(2):318–322; discussion 323 Dambrosia J, Patsalides A, Patronas NJ, Oldfield
170. Al-Mefty O, Borba LA (1997) Skull base chordo- EH, Lonser RR (2007) Mechanisms of morbid
mas: a management challenge. J Neurosurg hearing loss associated with tumors of the endolym-
86(2):182–189. doi:10.3171/jns.1997.86.2.0182 phatic sac in von Hippel-Lindau disease. JAMA
171. Al-Mefty K, Pravdenkova S, Colli BO, Al-Mefty O, 298(1):41–48, 298/1/41 [pii]
Gokden M (2007) Chordoma and chondrosarcoma: 186. Chee GH, Chen JM (2002) Fibrous dysplasia of the
similar, but quite different, skull base tumors. Cancer temporal bone. Otol Neurotol 23(3):405–406
110(11):2457–2467. doi:10.1002/cncr.23073 187. Chen JM, Fisch U (1993) The transotic approach
172. Altschuler EM, Jungreis CA, Sekhar LN, Jannetta in acoustic neuroma surgery. J Otolaryngol 22(5):
PJ, Sheptak PE (1990) Operative treatment of intrac- 331–336
ranial epidermoid cysts and cholesterol granulomas: 188. Chole R (1985) Petrous apicitis: surgical anatomy.
report of 21 cases. Neurosurgery 26(4):606–613; Ann Otol Rhinol Laryngol 94:251–257
discussion 614 189. Chole RA, Donald PJ (1983) Petrous apicitis.
173. Amedee RG, Marks HW, Lyons GD (1987) Clinical considerations. Ann Otol Rhinol Laryngol
Cholesterol granuloma of the petrous apex. Am J 92(6 Pt 1):544–551
Otol 8(1):48–55 190. Chong VF, Khoo JB, Fan YF (2002) Fibrous
174. Amichetti M, Cianchetti M, Amelio D, Enrici RM, dysplasia involving the base of the skull. AJR Am J
Minniti G (2009) Proton therapy in chordoma of the Roentgenol 178(3):717–720
base of the skull: a systematic review. Neurosurg Rev 191. Choo D, Shotland L, Mastroianni M, Glenn G,
32(4):403–416. doi:10.1007/s10143-009-0194-4 van Waes C, Linehan WM, Oldfield EH (2004)
175. Arriaga MA, Brackmann DE (1991) Differential Endolymphatic sac tumors in von Hippel-Lindau
diagnosis of primary petrous apex lesions. Am J Otol disease. J Neurosurg 100(3):480–487. doi:10.3171/
12(6):470–474 jns.2004.100.3.0480
References 851

192. Colli BO, Al-Mefty O (2001) Chordomas of the 207. Gerganov VM, Samii A, Paterno V, Stan AC,
skull base: follow-up review and prognostic factors. Samii M (2008) Bilateral osteomas arising from the
Neurosurg Focus 10(3):E1, 100301 [pii] internal auditory canal: case report. Neurosurgery
193. Crockard HA, Cheeseman A, Steel T, Revesz T, 62(2):E528–E529. doi:10.1227/01.neu.0000316023.
Holton JL, Plowman N, Singh A, Crossman J (2001) 81786.b6; discussion E529
A multidisciplinary team approach to skull base 208. Green JD Jr, McKenzie JD (1999) Diagnosis and
chondrosarcomas. J Neurosurg 95(2):184–189. management of intralabyrinthine schwannomas.
doi:10.3171/jns.2001.95.2.0184 Laryngoscope 109(10):1626–1631. doi:10.1097/
194. Crockard HA, Steel T, Plowman N, Singh A, 00005537-199910000-00015
Crossman J, Revesz T, Holton JL, Cheeseman A 209. Griffith AJ, Terrell JE (1996) Transsphenoid endo-
(2001) A multidisciplinary team approach to skull scopic management of petrous apex cholesterol
base chordomas. J Neurosurg 95(2):175–183. granuloma. Otolaryngol Head Neck Surg 114(1):-
doi:10.3171/jns.2001.95.2.0175 91–94, S0194599896000174 [pii]
195. Dave AV, Diaz-Marchan PJ, Lee AG (1997) Clinical 210. Hatano M, De Donato G, Falcioni M, Sanna M
and magnetic resonance imaging features of (2011) Chondroblastoma of the temporal bone. Acta
Gradenigo syndrome. Am J Ophthalmol 124(4): Otolaryngol 131(8):890–895. doi:10.3109/0001648
568–570 9.2011.566579
196. Davis TC, Thedinger BA, Greene GM (2000) 211. Heffelfinger MJ, Dahlin DC, MacCarty CS,
Osteomas of the internal auditory canal: a report of Beabout JW (1973) Chordomas and cartilaginous
two cases. Am J Otol 21(6):852–856 tumors at the skull base. Cancer 32(2):410–420
197. Di Maio S, Temkin N, Ramanathan D, Sekhar LN 212. Heffner DK (1989) Low-grade adenocarcinoma of
(2011) Current comprehensive management of probable endolymphatic sac origin A clinicopatho-
cranial base chordomas: 10-year meta-analysis of logic study of 20 cases. Cancer 64(11):2292–2302
observational studies. J Neurosurg 115(6): 213. Heffner DK (1996) Are papillary adenomas endo-
1094–1105. doi:10.3171/2011.7.JNS11355 lymphatic sac tumors? Ann Otol Rhinol Laryngol
198. DiCaprio MR, Enneking WF (2005) Fibrous dyspla- 105(3):251–252
sia. Pathophysiology, evaluation, and treatment. 214. Hentschel S, Durity F (2002) Petrous apex granulo-
J Bone Joint Surg Am 87(8):1848–1864, 87/8/1848 mas: CT and MR imaging. Can J Neurol Sci
[pii] 29(2):169–170
199. Doucet V, Peretti-Viton P, Figarella-Branger D, 215. Howitz MF, Johansen C, Tos M, Charabi S, Olsen JH
Manera L, Salamon G (1997) MRI of intracranial (2000) Incidence of vestibular schwannoma in
chordomas. Extent of tumour and contrast enhance- Denmark, 1977–1995. Am J Otol 21(5):690–694
ment: criteria for differential diagnosis. 216. Itshayek E, Spector S, Gomori M, Segal R (2002)
Neuroradiology 39(8):571–576 Fibrous dysplasia in combination with aneurysmal
200. Doyle KJ, Brackmann DE (1994) Intralabyrinthine bone cyst of the occipital bone and the clivus: case
schwannomas. Otolaryngol Head Neck Surg 110(6): report and review of the literature. Neurosurgery
517–523, S0194-5998(94)70620-4 [pii] 51(3):815–817; discussion 817–818
201. Edgerton MT, Persing JA, Jane JA (1985) The 217. Jackler RK, Cho M (2003) A new theory to explain
surgical treatment of fibrous dysplasia. With empha- the genesis of petrous apex cholesterol granuloma.
sis on recent contributions from cranio-maxillo- Otol Neurotol 24(1):96–106; discussion 106
facial surgery. Ann Surg 202(4):459–479 218. Jackler RK, Parker DA (1992) Radiographic differ-
202. Eisenberg MB, Haddad G, Al-Mefty O (1997) ential diagnosis of petrous apex lesions. Am J Otol
Petrous apex cholesterol granulomas: evolution 13(6):561–574
and management. J Neurosurg 86(5):822–829. 219. Kaelin WG Jr (2002) Molecular basis of the VHL
doi:10.3171/jns.1997.86.5.0822 hereditary cancer syndrome. Nat Rev Cancer
203. Erdem E, Angtuaco EC, Van Hemert R, Park JS, 2(9):673–682. doi:10.1038/nrc885
Al-Mefty O (2003) Comprehensive review of 220. Kamrin RP, Potanos JN, Pool JL (1964) An evalua-
intracranial chordoma. Radiographics 23(4): tion of the diagnosis and treatment of chordoma.
995–1009 J Neurol Neurosurg Psychiatry 27:157–165
204. Feldman MD, Rao VM, Lowry LD, Kelly M (1986) 221. Kemink JL, Graham MD (1982) Osteomas and exo-
Fibrous dysplasia of the paranasal sinuses. stoses of the external auditory canal – medical and
Otolaryngol Head Neck Surg 95(2):222–225 surgical management. J Otolaryngol 11(2):101–106
205. Friedmann I (1959) Epidermoid cholesteatoma and 222. Korten AG, ter Berg HJ, Spincemaille GH, van der
cholesterol granuloma; experimental and human. Laan RT, Van de Wel AM (1998) Intracranial chond-
Ann Otol Rhinol Laryngol 68(1):57–79 rosarcoma: review of the literature and report of 15
206. Gay E, Sekhar LN, Rubinstein E, Wright DC, Sen C, cases. J Neurol Neurosurg Psychiatry 65(1):88–92
Janecka IP, Snyderman CH (1995) Chordomas and 223. Kurokawa R, Uchida K, Kawase T (2005) Surgical
chondrosarcomas of the cranial base: results and treatment of temporal bone chondroblastoma. Surg
follow-up of 60 patients. Neurosurgery 36(5):887– Neurol 63(3):265–268, S0090-3019(04)00514-2
896; discussion 896–897 [pii]; discussion 268
852 11 Other Cerebellopontine Angle Tumors

224. Kutz JW Jr, Verma S, Tan HT, Lo WW, Slattery WH 238. Moon IS, Kim J, Lee HK, Lee WS (2008) Surgical
3rd, Friedman RA (2007) Surgical management of treatment and outcomes of temporal bone chondro-
skull base chondroblastoma. Laryngoscope 117(5):- blastoma. Eur Arch Otorhinolaryngol 265(12):
848–853. doi:10.1097/MLG.0b013e3180337da6 1447–1454. doi:10.1007/s00405-008-0660-6
225. Lanzino G, Dumont AS, Lopes MB, Laws ER Jr 239. Moore KR, Harnsberger HR, Shelton C, Davidson HC
(2001) Skull base chordomas: overview of disease, (1998) ‘Leave me alone’ lesions of the petrous apex.
management options, and outcome. Neurosurg Focus AJNR Am J Neuroradiol 19(4):733–738
10(3):E12, 100312 [pii] 240. Mortensen A, Bojsen-Moller M, Rasmussen P
226. Li JC, Brackmann DE, Lo WW, Carberry JN, House (1989) Fibrous dysplasia of the skull with acromeg-
JW (1993) Reclassification of aggressive adenoma- aly and sarcomatous transformation. Two cases with
tous mastoid neoplasms as endolymphatic sac a review of the literature. J Neurooncol 7(1):25–29
tumors. Laryngoscope 103(12):1342–1348. 241. Mosnier I, Cyna-Gorse F, Grayeli AB, Fraysse B,
doi:10.1288/00005537-199312000-00004 Martin C, Robier A, Gardini B, Chelikh L, Sterkers
227. Lo WW, Applegate LJ, Carberry JN, Solti-Bohman O (2002) Management of cholesterol granulomas of
LG, House JW, Brackmann DE, Waluch V, Li JC the petrous apex based on clinical and radiologic
(1993) Endolymphatic sac tumors: radiologic evaluation. Otol Neurotol 23(4):522–528
appearance. Radiology 189(1):199–204 242. Muckle RP, De la Cruz A, Lo WM (1998) Petrous
228. Lo WW, Solti-Bohman LG, Brackmann DE, Gruskin P apex lesions. Am J Otol 19(2):219–225
(1984) Cholesterol granuloma of the petrous apex: CT 243. Mukherji SK, Albernaz VS, Lo WW, Gaffey MJ,
diagnosis. Radiology 153(3):705–711 Megerian CA, Feghali JG, Brook A, Lewin JS,
229. Lonser RR, Kim HJ, Butman JA, Vortmeyer AO, Lanzieri CF, Talbot JM, Meyer JR, Carmody RF,
Choo DI, Oldfield EH (2004) Tumors of the endo- Weissman JL, Smirniotopoulos JG, Rao VM, Jinkins
lymphatic sac in von Hippel-Lindau disease. N JR, Castillo M (1997) Papillary endolymphatic sac
Engl J Med 350(24):2481–2486. doi:10.1056/ tumors: CT, MR imaging, and angiographic findings
NEJMoa040666 in 20 patients. Radiology 202(3):801–808
230. Mallina S, Philip R, Chong AW, Gurdeep S (2007) 244. Neff B, Sataloff RT, Storey L, Hawkshaw M,
Fibrous dysplasia of the temporal bone. Med J Spiegel JR (2002) Chondrosarcoma of the skull
Malaysia 62(2):160–161 base. Laryngoscope 112(1):134–139. doi:10.1097/
231. Manasse P (1894) Uber granulations geschwulste 00005537-200201000-00023
mit fremdkorperriesen- zellen. Virchows Arch 245. Neff BA, Willcox TO Jr, Sataloff RT (2003)
136:245 Intralabyrinthine schwannomas. Otol Neurotol
232. Manski TJ, Heffner DK, Glenn GM, Patronas NJ, 24(2):299–307
Pikus AT, Katz D, Lebovics R, Sledjeski K, Choyke 246. Noel G, Feuvret L, Calugaru V, Dhermain F,
PL, Zbar B, Linehan WM, Oldfield EH (1997) Mammar H, Haie-Meder C, Ponvert D, Hasboun D,
Endolymphatic sac tumors. A source of morbid hear- Ferrand R, Nauraye C, Boisserie G, Beaudre A,
ing loss in von Hippel-Lindau disease. JAMA Gaboriaud G, Mazal A, Habrand JL, Mazeron JJ
277(18):1461–1466 (2005) Chordomas of the base of the skull and upper
233. Markou K, Karasmanis I, Vlachtsis K, Petridis D, cervical spine. One hundred patients irradiated by
Nikolaou A, Vital V (2008) Primary pleomorphic a 3D conformal technique combining photon and
adenoma of the external ear canal. Report of a case proton beams. Acta Oncol 44(7):700–708,
and literature review. Am J Otolaryngol 29(2):142– N47554P2G1676823 [pii]
146, S0196-0709(07)00047-6 [pii] 247. Noel G, Habrand JL, Jauffret E, de Crevoisier R,
234. Megerian CA, Haynes DS, Poe DS, Choo DI, Dederke S, Mammar H, Haie-Meder C, Pontvert D,
Keriakas TJ, Glasscock ME 3rd (2002) Hearing Hasboun D, Ferrand R, Boisserie G, Beaudre A,
preservation surgery for small endolymphatic sac Gaboriaud G, Guedea F, Petriz L, Mazeron JJ (2003)
tumors in patients with von Hippel-Lindau syn- Radiation therapy for chordoma and chondrosar-
drome. Otol Neurotol 23(3):378–387 coma of the skull base and the cervical spine.
235. Menezes AH, Gantz BJ, Traynelis VC, McCulloch Prognostic factors and patterns of failure. Strahlenther
TM (1997) Cranial base chordomas. Clin Neurosurg Onkol 179(4):241–248. doi:10.1007/s00066-003-
44:491–509 1065-5
236. Michaelson PG, Cable BB, Mair EA (2001) Image- 248. Oghalai JS, Buxbaum JL, Jackler RK, McDermott
guided transphenoidal drainage of a cholesterol MW (2005) Skull base chondrosarcoma originating
granuloma of the petrous apex in a child. Int J Pediatr from the petroclival junction. Otol Neurotol
Otorhinolaryngol 57(2):165–169, S0165-5876(00) 26(5):1052–1060, 00129492-200509000-00037
00456-0 [pii] [pii]
237. Moffat DA, Saunders JE, McElveen JT Jr, 249. Palacios E, Valvassori G (1999) Petrous apex lesions:
McFerran DJ, Hardy DG (1993) Unusual cerebello- cholesterol granuloma. Ear Nose Throat J 78(4):234
pontine angle tumours. J Laryngol Otol 107(12): 250. Pallini R, Maira G, Pierconti F, Falchetti ML, Alvino
1087–1098 E, Cimino-Reale G, Fernandez E, D’Ambrosio E,
References 853

Larocca LM (2003) Chordoma of the skull base: 264. Singh V, Annis JA, Todd GB (1992) Osteoma of the
predictors of tumor recurrence. J Neurosurg internal auditory canal presenting with sudden
98(4):812–822. doi:10.3171/jns.2003.98.4.0812 unilateral hearing loss. J Laryngol Otol 106(10):
251. Peters BR, Maddox HE 3rd, Batsakia JG (1988) 905–907
Pleomorphic adenoma of the middle ear and mastoid 265. Stuer C, Schramm J, Schaller C (2006) Skull
with posterior fossa extension. Arch Otolaryngol base chordomas: management and results. Neurol
Head Neck Surg 114(6):676–678 Med Chir (Tokyo) 46(3):118–124, JST.JSTAGE/
252. Presutti L, Villari D, Marchioni D (2006) Petrous nmc/46.118 [pii]; discussion 124–125
apex cholesterol granuloma: transsphenoid endo- 266. Takahashi S, Kawase T, Yoshida K, Hasegawa A,
scopic approach. J Laryngol Otol 120(6):e20, Mizoe JE (2009) Skull base chordomas: efficacy of
S0022215106009121 [pii] surgery followed by carbon ion radiotherapy. Acta
253. Richard S, David P, Marsot-Dupuch K, Giraud S, Neurochir (Wien) 151(7):759–769. doi:10.1007/
Beroud C, Resche F (2000) Central nervous system s00701-009-0383-5
hemangioblastomas, endolymphatic sac tumors, and 267. Terao T, Onoue H, Hashimoto T, Ishibashi T,
von Hippel-Lindau disease. Neurosurg Rev 23(1):1– Kogure T, Abe T (2001) Cholesterol granuloma in
22; discussion 23–24 the petrous apex: case report and review. Acta
254. Roche PH, Dufour H, Figarella-Branger D, Pellet W Neurochir (Wien) 143(9):947–952
(1998) Endolymphatic sac tumors: report of three 268. Thedinger BA, Nadol JB Jr, Montgomery WW,
cases. Neurosurgery 42(4):927–932 Thedinger BS, Greenberg JJ (1989) Radiographic
255. Rosenberg AE, Nielsen GP, Keel SB, Renard LG, diagnosis, surgical treatment, and long-term
Fitzek MM, Munzenrider JE, Liebsch NJ (1999) follow-up of cholesterol granulomas of the petrous
Chondrosarcoma of the base of the skull: a clinico- apex. Laryngoscope 99(9):896–907. doi:10.1288/
pathologic study of 200 cases with emphasis on its 00005537-198909000-00003
distinction from chordoma. Am J Surg Pathol 269. Thorne MC, Gebarski SS, Telian SA (2006) Rapid
23(11):1370–1378 expansion in a previously indolent cholesterol cyst: a
256. Royer MC, Pensak ML (2007) Cholesterol need for lifelong follow-up. Otol Neurotol 27(1):
granulomas. Curr Opin Otolaryngol Head 124–126, 00129492-200601000-00022 [pii]
Neck Surg 15(5):319–322. doi:10.1097/ 270. Tringali S, Linthicum FH Jr (2010) Cholesterol gran-
MOO.0b013e3282a08f74 uloma of the petrous apex. Otol Neurotol 31(9):-
257. Ruelle A, Datti R, Andrioli G (1994) Cerebellopontine 1518–1519. doi:10.1097/MAO.0b013e3181d279c3
angle osteoma causing trigeminal neuralgia: case 271. Tsai EC, Santoreneos S, Rutka JT (2002) Tumors of
report. Neurosurgery 35(6):1135–1137 the skull base in children: review of tumor types and
258. Sabin HI, Bordi LT, Symon L (1987) Epidermoid management strategies. Neurosurg Focus 12(5):e1,
cysts and cholesterol granulomas centered on the 120501 [pii]
posterior fossa: twenty years of diagnosis and man- 272. Tzortzidis F, Elahi F, Wright D, Natarajan SK,
agement. Neurosurgery 21(6):798–805 Sekhar LN (2006) Patient outcome at long-term fol-
259. Samii A, Gerganov V, Herold C, Gharabaghi A, low-up after aggressive microsurgical resection of
Hayashi N, Samii M (2009) Surgical treatment of skull cranial base chordomas. Neurosurgery 59(2):230–
base chondrosarcomas. Neurosurg Rev 32(1):67–75. 237. doi:10.1227/01.NEU.0000223441.51012.9D;
doi:10.1007/s10143-008-0170-4; discussion 75 discussion 230–237
260. Samii A, Gerganov VM, Herold C, Hayashi N, 273. Tzortzidis F, Elahi F, Wright DC, Temkin N,
Naka T, Mirzayan MJ, Ostertag H, Samii M (2007) Natarajan SK, Sekhar LN (2006) Patient outcome at
Chordomas of the skull base: surgical management long-term follow-up after aggressive microsurgical
and outcome. J Neurosurg 107(2):319–324. resection of cranial base chondrosarcomas.
doi:10.3171/JNS-07/08/0319 Neurosurgery 58(6):1090–1098. doi:10.1227/01.
261. Samii M, Faupel G (1977) Chondrosarcoma with NEU.0000215892.65663.54; discussion 1090–1098
intraspinal, intra- and extrathoracal extension (author’s 274. Volpe NJ, Liebsch NJ, Munzenrider JE, Lessell S
transl). Acta Neurochir (Wien) 39(1–2):71–80 (1993) Neuro-ophthalmologic findings in chordoma
262. Schulz-Ertner D, Karger CP, Feuerhake A, and chondrosarcoma of the skull base. Am J
Nikoghosyan A, Combs SE, Jakel O, Edler L, Ophthalmol 115(1):97–104
Scholz M, Debus J (2007) Effectiveness of carbon 275. Vortmeyer AO, Choo D, Pack S, Oldfield E,
ion radiotherapy in the treatment of skull-base Zhuang Z (2000) VHL gene inactivation in an
chordomas. Int J Radiat Oncol Biol Phys 68(2):- endolymphatic sac tumor associated with von
449–457, S0360-3016(06)03669-8 [pii] Hippel-Lindau disease. Neurology 55(3):460
263. Sen C, Triana AI, Berglind N, Godbold J, 276. Vortmeyer AO, Huang SC, Koch CA, Governale L,
Shrivastava RK (2010) Clival chordomas: clinical Dickerman RD, McKeever PE, Oldfield EH,
management, results, and complications in 71 Zhuang Z (2000) Somatic von Hippel-Lindau gene
patients. J Neurosurg 113(5):1059–1071. doi:10.3171 mutations detected in sporadic endolymphatic sac
/2009.9.JNS08596 tumors. Cancer Res 60(21):5963–5965
854 11 Other Cerebellopontine Angle Tumors

277. Vrabec JT, Lambert PR, Chaljub G (2000) Osteoma 281. Weber AL, Liebsch NJ, Sanchez R, Sweriduk ST Jr
of the internal auditory canal. Arch Otolaryngol (1994) Chordomas of the skull base. Radiologic and
Head Neck Surg 126(7):895–898, ocn90069 [pii] clinical evaluation. Neuroimaging Clin N Am
278. Wanebo JE, Bristol RE, Porter RR, Coons SW, 4(3):515–527
Spetzler RF (2006) Management of cranial base 282. Yamasaki T, Kikuchi H, Yamabe H, Yamashita J
chondrosarcomas. Neurosurgery 58(2):249–255. (1990) Multiple intracranial metastases following
doi:10.1227/01.NEU.0000194834.74873.FB; dis- malignant evolution in recurrent pleomorphic ade-
cussion 249–255 noma of the lacrimal gland – case report. Neurol
279. Warakaulle DR, Anslow P (2003) Differential diag- Med Chir (Tokyo) 30(13):1038–1042, JST.
nosis of intracranial lesions with high signal on T1 Journalarchive/nmc1959/30.1038 [pii]
or low signal on T2-weighted MRI. Clin Radiol 283. Yano H, Funakoshi T, Shinoda J, Sakai N,
58(12):922–933, S000992600300268X [pii] Kokuzawa G, Shimokawa K (1997) Primary pleo-
280. Weatherby RP, Dahlin DC, Ivins JC (1981) morphic adenoma in posterior cranial fossa. Brain
Postradiation sarcoma of bone: review of 78 Mayo Tumor Pathol 14(1):75–78
Clinic cases. Mayo Clin Proc 56(5):294–306
Management of Facial Palsy
12

Despite the major advancement in CP angle surgery, • Preparation of the stump; 1–2 mm of the
in certain cases it is impossible to preserve normal epineurium of the stump and of the graft has to
facial nerve function. Even if the nerve is pre- be resected with microscissors to create
served anatomically, complete and lasting paraly- smooth interface.
sis may ensue. Comprehensive management of • Tensionless anastomosis to prevent scar for-
patients with facial nerve paralysis aims at resto- mation and subsequent hindering of axonal
ration of function and appearance, as well as pre- regeneration.
vention of negative sequelae. • Suture, glue, or other alternative. The evalua-
tion of various neurorrhaphy techniques—
epineural vs. perineural sutures, laser
Restoration of Facial Nerve Function neurorrhaphy, tissue adhesives, tubulization,
and neurotrophic factors—revealed no major
Restoration of facial nerve function can be differences in regard to outcome [33]. In a pre-
performed by (1) nerve reconstruction, either vious study of the senior author, the degree of
directly or via an interposition graft, or (2) nerve reinnervation following nerve anastomosis
reanimation with donor nerves. Direct end-to-end with fibrin glue and with suture has been com-
neurorrhaphy yields the best results, followed by pared, and no significant differences were
reconstruction with interposition graft, and then found [31]. The use of fibrin glue is technically
reanimation with a donor nerve [19, 31]. easier and seems to lead to less scar formation.
The goal of any restorative procedure is to The facial nerve in its intracranial segment has
achieve normal facial appearance at rest, symme- no epineurium, and the anastomosis can be
try with movement, and restoration of muscle made with fibrin glue, instead of placing a
control [5]. Timing is a key factor with regard to suture through the substance of the nerve. In
functional outcome of facial nerve reconstruc- case of hypoglossal–facial anastomosis, we
tion: best results are achieved if the reconstruction apply two microsutures that are additionally
is performed within the first year after onset of secured with fibrin glue.
the palsy [26, 27].

Facial Nerve Reconstruction


Principles of Nerve Anastomosis
Indications
• Careful microsurgical technique of prepara- Direct reconstruction of the facial nerve leads to
tion of the stump and precise approximation optimal regeneration. It can be performed in case
of the anastomotic ends. the proximal nerve stump at the brain stem and

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 855


DOI 10.1007/978-3-642-35422-9_12, © Springer-Verlag Berlin Heidelberg 2013
856 12 Management of Facial Palsy

the distal stump are available. The minimum


length of the available proximal stump should be
3–5 mm. Importantly, the anastomosis should be
absolutely tension-free. An experimental study
performed by the senior author has shown that
optimal outcome is achieved in case of tension-
less end-to-end anastomosis, followed by recon-
struction using an interpositional neural graft.
The worse regeneration occurs in case the anasto-
mosis is performed under tension. If significant
gap between the proximal and distal ends exists,
interposition graft should be used. All these
techniques for primary facial reconstruction,
Fig. 12.1 The sural nerve graft is harvested using two
however, lead to better functional results than the short transverse skin incisions (arrows)
reanimation techniques, for example, the hypo-
glossal–facial anastomosis.
been removed). Both the sural nerve and the
End-to-End Anastomosis greater auricular nerve grafts are appropriate
If both the proximal and the distal nerve stumps in this regard [28].
are available within the CP angle, direct end- • The harvesting of the graft should not have
to- end anastomosis can be performed at the time undesirable cosmetic effect. Avoid donor site
of tumor surgery. It is feasible only in case of morbidity!
large VSs, in which the facial nerve has been • Graft nutrition!
elongated during tumor growth. In such cases, a
defect of up to 10–15 mm can be bridged without Sural Nerve Graft
creating an undue tension. The ends of the stumps The obtainable graft length is up to 40 cm. The
are laid on gelatin sponges, freshened, and fixed sural nerve is a sensory nerve formed from the
with fibrin glue. The benefits of the method, if medial sural cutaneous nerve of the tibial nerve
compared with the alternatives, are that sponta- and the peroneal communicating nerve of com-
neous facial movement can recover, the need of mon peroneal nerve. It supplies the skin of the
second surgery is obviated, and the time to recov- posterolateral aspect of the leg, lateral malleolus,
ery is shortened. and lateral side of the foot up to little toe. The
nerve runs between the two heads of the gastroc-
Reconstruction with an Interposition nemius muscle, then pierces the deep fascia, and
Graft runs distally lateral to the small saphenous vein
The functional restoration with an interposition between the lateral malleolus and the calcaneus.
graft is inferior to that of the direct tensionless Harvesting of sural nerve leads to mild sensory
end-to-end anastomosis but is still superior when deficit.
compared with the facial nerve reanimation Technique: The nerve is exposed first distally
techniques. by performing a skin incision (appr. 1 cm long)
at the junction of the middle and anterior thirds
Nerve Grafts of the line between the tip of calcaneus and lat-
• Length of the graft. The length of the graft and eral malleolus (Fig. 12.1). With blunt dissection,
the number of grafts do not affect the extent of the nerve is identified subcutaneously—it is
recovery [15, 27, 31]. usually slightly anterior to the small saphenous
• Diameter of the donor nerve. Optimally, the vein. A small hemostatic forceps is inserted
diameter of the donor nerve should match that beneath it, and slight traction is performed.
of the facial nerve (after the epineurium has Thus, with palpation or visually, the proximal
Restoration of Facial Nerve Function 857

course of the nerve can be identified. A second and can be followed to the junction of the upper
incision is placed accordingly to the expected and middle thirds of the posterior sternocleido-
location of the nerve 10–15 cm proximally. The mastoid border (Erb’s point).
nerve is dissected and—using the same tech-
nique of slight traction with a hemostatic Reconstruction Within the CP Angle
forceps—followed more proximally as long as with an Interposition Graft
necessary, and appropriate skin incisions are If both the proximal and the distal nerve stumps
made. The course of the nerve from distal to are available but cannot be coapted without undue
proximal is lateral to medial, and in the popliteal tension, an interposition graft with an appropriate
fossa, it lies approximately in the midline. length (usually 5–20 mm) may be used (Figs. 12.2,
Another important consideration is that the sural 12.3, 12.4, and 12.5).
nerve pierces the deep fascia in the junction area
between upper 2/3 and lower 1/3 of the posterior
leg. Large branches, usually from the middle
part of the nerve, should be cut via additional
skin incisions. The nerve is incised at the most
proximal level and stripped out by traction with
the forceps.

Great Auricular Nerve Graft


The maximal obtainable graft length is 7–10 cm.
Greater auricular nerve grafting, however, may
cause more disabling hypesthesias than sural
nerve resection.
Technique: The retrosigmoid skin incision has
to be extended inferior along the posterior border
of the sternocleidomastoid muscle as long as Fig. 12.2 Schematic illustration of a direct end-to-end
facial nerve reconstruction with an interposition graft
required. The nerve is identified under the skin

Fig. 12.3 MR images demonstrating large VS on the segment of the facial nerve was resected (priority was
right side. At surgery, the facial nerve was found to be given to complete tumor removal due to the relatively
intimately adherent to the tumor capsule over a length of young age of the patient). The technique used for recon-
2–3 cm. In order to remove the tumor completely, this struction of the nerves’ integrity is presented on Fig. 12.4
858 12 Management of Facial Palsy

a b c

d e f

Fig. 12.4 Reconstruction of the facial nerve in the CP of appropriate length is selected and approximated end-to-end
angle with a sural nerve interposition graft—intraopera- to the stumps. The diameter of the graft usually corre-
tive images. The tumor (see previous image) has been sponds well to the diameter of the facial nerve. Fibrin glue
removed completely. (a, b) Healthy stumps are prepared is used to seal the ends—in the CP angle (c) and in the
proximally and distally. A piece of Gelfoam is used as a IAC (d, e). The opened IAC is then filled with fat pieces to
platform to allow better adaptation. (c) Sural nerve graft prevent CSF leak (f)

The early experience of the senior author with was performed with Dr. Weigand; afterwards, all
facial nerve reconstruction in the CP angle was cases were performed with Dr. Draf) [11].
presented in 1994 [31]. The series included 16 The mastoid segment of the facial nerve is
patients with VSs and 1 with meningioma. identified and exposed by performing a mas-
Reinnervation was first noted in 4–12 months toidectomy (Figs. 12.6 and 12.7). It is transected
following the procedure. At follow-up, 24 % of below the lateral semicircular canal and trans-
the patients had HB grade III + facial nerve func- posed posterior towards the sinodural angle. The
tion, 47 % had HB grade III, and 29 % had HB sigmoid sinus is skeletonized, and the posterior
grade IV. surface of the pyramid at sinodural angle is
removed. The nerve graft—usually 30–50 mm
Intracranial–Intratemporal Technique long—is coapted to the proximal stump and
If only the proximal nerve stump is available, the brought out of the CP angle into the mastoid
nerve can be reconstructed by creating an anasto- through a presigmoid dural incision. Then it is
mosis to its distal segment within the temporal united to the distal facial nerve stump.
bone using the intracranial–intratemporal tech- As presented in the earlier cited study, 24 % of
nique developed by Samii in 1975 (the first case the patients after such reconstruction had HB
Restoration of Facial Nerve Function 859

a b

Fig. 12.5 (a) Intraoperative image of a direct facial nerve reconstruction in the CP angle with a nerve graft. (b–d)
Photos of the patient immediately after surgery (b), 9 months, (c) 15 months and (d) later

grade III+, 53 % had HB grade III, and 18 % had growth, an intracranial–extracranial reconstruction,
HB grade IV [29]. distal to the exit site of the nerve at stylomastoid
foramen, might be necessary (Fig. 12.8). An 80- to
Intracranial–Extracranial Technique 150-mm graft is required to reach this facial nerve
Occasionally, if the petrous segment of the facial segment [25, 28]. The technique was presented
nerve is destroyed by infection or neoplastic by Dott in 1958 [10], who performed the grafting
860 12 Management of Facial Palsy

in two stages: initially, the graft was united with grade III+, 60 % recovered to HB grade III, and
the proximal stump, brought out of the CP angle, 20 % to HB grade IV.
and tunneled subcutaneously between the ster-
nocleidomastoid and the splenius capitis muscles. Reinforcement Technique
The distal end was marked with a silver clip. In 3 A
months, the distal end of the graft was anastomo- A novel technique, introduced recently by the
sed to the peripheral stump of the facial nerve. senior author, is the end-to-end reinforcement of
The senior author has performed such recon- the facial nerve in the CP angle. It was applied in
structions in ten patients. In contrast to the origi- one case of large cystic VS. Tumor dissection led
nal description of Dott, the surgery was performed to partial loss of facial nerve fibers and, respec-
in one step. One patient (10 %) recovered to HB tively, to a marked reduction of response inten-
sity during proximal electrical stimulation.
Stimulation of the facial nerve in the IAC, how-
ever, elicited a normal response. A sural nerve
graft was used to reinforce the remaining func-
tional facial nerve fibers: it was coapted with
fibrin to the normally appearing proximal and
distal nerve ends (Figs. 12.9 and 12.10). The con-
tinuity of the facial nerve was thus not inter-
rupted. Eighteen months after surgery, good
recovery of facial function was recorded
(House–Brackmann grade III).

B
Second variant of the facial nerve reinforce-
ment technique is the end-to-side insertion of
an interposed sural nerve graft. It was used ini-
Fig. 12.6 Schematic illustration of an intracranial–
intratemporal facial nerve reconstruction. A proximal
tially in a case of large VS (extension grade
facial nerve stump, B nerve graft, C distal facial nerve T4b) and published in 2006 [30]. During the
stamp in mastoid, D trigeminal nerve dissection of the tumor capsule from the facial

a b

Fig. 12.7 Intracranial–intratemporal anastomosis. (a) which is presented schematically on figure (c). Photo of
CP angle part: the proximal facial nerve stamp and the the patient immediately after the surgery (d) and 12
graft are well visible. (b) Anastomosis in the mastoid, months later (e, f)
Restoration of Facial Nerve Function 861

c d

Fig. 12.7 (continued)

nerve, approximately 70 % of the nerve fibers continuity (Fig. 12.11). At 18 months follow-
were lost. The graft was interposed end-to-side up, the patient’s facial function had improved
between the normal proximal and distal parts of markedly to an HB grade III. The patient was
the facial nerve without interrupting its unfortunately lost to follow-up.
862 12 Management of Facial Palsy

Facial Nerve Reanimation with a Donor


Nerve (or Indirect Facial Nerve
Reconstruction)

Indications
Reanimation with donor nerve is indicated in
case of complete facial nerve loss or loss of its
proximal stump at the brain stem during tumor
surgery, as well as in case the anatomically intact
nerve shows no signs of recovery at follow-up. In
case of nerve loss, the procedure is performed
within 1 or 2 weeks after tumor removal. If a
complete facial palsy persists for 10–12 months
without signs of spontaneous functional or elec-
tromyographic recovery, even though the integ-
rity of the nerve has been preserved, nerve
Fig. 12.8 Schematic illustration of an intracranial–
extracranial facial nerve reconstruction reanimation procedure is also indicated. The

a b

Fig. 12.9 Preoperative MRI examination of a patient with large cystic VS grade T4B on the right side. See next figures
for operative details
Restoration of Facial Nerve Function 863

a b c

d e f

g h i

j k

Fig. 12.10 Intraoperative images illustrating the technique dissection of the nerve from the tumor capsule. (h) Removal
of facial nerve reinforcement in the CP angle. The patient of the last piece of tumor (Tu) from the nerve. (i) Part of the
had a 4.2-cm large cystic VS (T4b) (See previous figure). (a) facial nerve fibers were damaged. The stumps are marked
Initial view of the tumor (TS transverse sinus, SS sigmoid with arrows. Although the nerve responded to electrical
sinus, R retractor). (b) The IAC has been opened widely, stimulation, the response was rather weak. Therefore, a sural
exposing the intracanalicular tumor portion. (c) Partial nerve graft was prepared (j) and fixed with fibrin glue to the
removal of the intracanalicular tumor and exposure of the ends of the interrupted facial nerve fibers (k) (arrowheads:
distal facial nerve (7). (d) Identification of the medial segment anastomotic sites)
of the facial nerve, close to the brain stem. (e, g) Progressive
864 12 Management of Facial Palsy

skin incision is performed that is extended down


along the anterior border of the sternocleidomas-
toid muscle (Fig. 12.12). The facial nerve is
identified between the mastoid tip and the angle
of mandible as it courses distal to the stylomas-
toid foramen. Useful landmark for its location is
the “pointer” of the cartilaginous auditory canal.
Another helpful landmark is the digastric branch,
which can be dissected back to the main facial
trunk. Dissection is continued distally to the point
of nerves’ bifurcation.
The hypoglossal nerve is identified using the
posterior belly of the digastric muscle and the
anterior border of the sternocleidomastoid mus-
cle as landmarks. The nerve is traced distally as
far as possible, transected, and brought up in
front of the stylomastoid foramen medial to the
posterior belly of the digastric muscle. Its
Fig. 12.11 Schematic drawing of the end-to-side facial
nerve reinforcement technique (Reprinted from Samii descending ramus can be divided to acquire addi-
et al. [30]) tional length. The facial nerve is then transected
proximally—at the stylomastoid foramen—and
ipsilateral muscles can be partially reinnervated brought in proximity to the hypoglossal stump.
by collaterals from the contralateral facial nerve. Anastomosis is performed with two 9-0 perineu-
Therefore, the patient should be examined both ral sutures or with fibrin glue. In order to avoid
clinically and with EMG for signs of nerve recov- complete hemitongue denervation, the proximal
ery. The functional outcome is optimal if the sur- part of ansa cervicalis can be connected to the
gery is performed up to 12 months after the onset distal part of the hypoglossal nerve.
of the paralysis. Complete transection of the hypoglossal
Reanimation can be performed with various nerve, however, causes ipsilateral paralysis and
donor nerves: hypoglossal, accessory, phrenic hemitongue atrophy, which interferes with
nerves, or contralateral facial nerve. Best results mastication, speech, and swallowing [33].
are achieved with the hypoglossal nerve, which Hammerschlag [16] observed both speech and
could be explained by the close relations of the swallowing problems in 45 % of patients using
facial and hypoglossal cortical representations this technique. Conley and Baker [6] reported
and the existing physiologically close association severe tongue atrophy in 25 % of their series of
of facial and tongue movements. 137 cases. Although these problems may be tran-
sient, some patients show persistent difficulties
Hypoglossal–Facial Anastomosis that interfere with their daily lives [8, 18].
Classical hypoglossal–facial nerve anastomosis Variations of the classic techniques that allow
is performed end-to-end between the proximal for preservation of hypoglossal nerve function
part of the hypoglossal nerve and the distal seg- have been introduced, such as end-to-side anasto-
ment of the facial nerve at the stylomastoid mosis, jump interposition graft hypoglossal–
process. This method is the most reliable one to facial anastomosis, hypoglossal nerve longitudinal
promote reinnervation because it incorporates the split, and use of a descending hypoglossal branch
maximum number of axons. or of ansa cervicalis [1, 6, 7, 9, 17, 19].
May et al. [22] described the technique of
Technique indirect hypoglossal–facial anastomosis. The
The head is turned to the contralateral side. hypoglossal and facial nerves are anastomosed
A slightly curved linear retromastoid retroauricular with the interposition of a free nerve “jump” graft,
Restoration of Facial Nerve Function 865

a b c

d e f

Fig. 12.12 (a) A slightly curved linear retromastoid ret- was performed, and the facial nerve was identified in the
roauricular skin incision is performed. The digastric mus- fallopian canal (black arrow), approximately 5 mm proxi-
cle (DM) is identified. (M, mastoid). (b) The facial nerve mally to the stylomastoid foramen. (f, g) The distal part of
(arrow) was identified anterior to the stylomastoid fora- the facial nerve is mobile, coapted to the hypoglossal
men. (c) Identification of the hypoglossal nerve (double nerve (end-to-end) and fixed with fibrin glue
arrow) and transected distally (d). (e) Partial mastoidectomy

end-to-end to the peripheral facial nerve stump, The use of split hypoglossal nerve for facial
and end-to-side to the hypoglossal nerve. With reanimation is another alternative [1, 6, 9].
this method, postoperative tongue dysfunction Longitudinal splitting of the hypoglossal nerve is
and swallowing or speech problems can be com- performed, and one-half is routed cranially and
pletely avoided, as shown by Manni et al. [20] in connected in an end-to-end fashion with the
a series of 39 consecutive patients. However, as proximal stump of the facial nerve. The hypo-
with other crossover techniques, synkinesis can glossal nerve, however, is not a polyfascicular
still occur. Twenty-one percent of the patients in nerve, and the longitudinal incision may transect
this series recovered to HB grade II, 45 % recov- many interweaving axons. Therefore, although
ered to grade III, 24 % recovered to grade IV, and the technique leads to good facial reanimation, in
7 % to grade V, and in 3 % no recovery was noted. all patients, mild to moderate tongue atrophy is
The disadvantages of this technique are the exis- described. The degree of atrophy, however, is
tence of two suture lines, which may act as obsta- much less severe as that observed in some patients
cles to the regenerating axon sprouts, and the with classic hypoglossal–facial nerve anastomo-
donor site morbidity [13, 16, 18]. sis [1]. Another complication related to the
866 12 Management of Facial Palsy

technique is fibrosis involving the exposed nerve face recover but are possible only with concomi-
after being split [18]. To avoid such outcome, tant arm abduction. Furthermore, all patients
Cusimano and Sekhar [7] described a technique have weakness of the muscles innervated by the
of partial hypoglossal to facial anastomosis after accessory nerve.
partial division and limited splitting of the hypo-
glossal nerve. Facial to Facial Anastomosis
Following the experimental works of Viterbo (Cross-Face Graft)
[34, 35] on the end-to-side neurorrhaphy, a new Facial–facial nerve anastomosis is an option in
hypoglossal nerve-preserving technique was pro- patients in whom no other cranial nerve can be
moted: 30–50 % of its cross-sectional area of the sacrificed, such as those with lower cranial nerve
hypoglossal nerve are sectioned transversally, palsy, isolated hypoglossal nerve palsy, or those
and the facial nerve distal stump is coapted with profession for which speech is essential
directly to the sidewall of the hypoglossal nerve. (speech is dependent on intact hypoglossal
The fibers remaining intact enable good hemi- function!). It is used further to achieve some
tongue function. Alternatively, an epineural win- functional recovery after other reanimation tech-
dow is made. niques had failed or to augment partial recovery
Our favorite technique currently is the after hypoglossal–facial anastomosis in patients
side-to- end hypoglossal–facial anastomosis with incomplete facial paralysis showing only
(Fig. 12.13). A retromastoid retroauricular skin limited recovery over time.. The anatomical
incision is performed. The hypoglossal nerve premise for the procedure is the fact that cutting
is identified and dissected proximally. Then the of approximately 1/2 to 1/3 of the plexiform
facial nerve is found anterior to foramen stylo- facial nerve branches does not cause obvious
mastoideum. In order to obtain sufficient length functional deficit. Adjacent branches take over
of the facial nerve, its mastoid segment up to the function completely. The facial nerve of the
the geniculate ganglion is exposed following a healthy side is connected with nerve grafts to the
mastoidectomy. It is transected as proximally corresponding branches of the plegic side. In
as possible, dissected from the facial canal, and such cases, the muscle tone on the affected side is
reflected inferiorly. Two perineural sutures (9-0 partially preserved, whereas the graft enhances
or 8-0 monofilament nylon sutures) are sutured to the voluntary movements [14, 33].
unite the facial to a partially sectioned (approxi- Technique: The zygomatic branches are
mately 50 %) hypoglossal nerve [3, 4]. Fibrin exposed by performing vertical skin incisions
glue is applied as an additional support for the slightly anterior to the masseter muscles below the
anastomosis. Although theoretically fewer nerve zygomatic arch on both sides. These are the stron-
fibers are transferred to the facial muscles when gest facial branches, innervating the orbicularis
compared with the end-to-end technique, collat- oculi and the orbicularis oris muscles. Intraoperative
eral axonal sprouting provides sufficient func- electrostimulation is essential to estimate the role
tional motor reinnervation. of each branch because of their anatomical vari-
ability. Sural nerve graft is used as an end-to-end
Accessory Nerve anastomosis between the proximal part of the
Reanimation with the accessory nerve is recom- facial branch on the contralateral healthy side to
mended for patients with dysphagia or in case the the distal nerve part on the ipsilateral side. For the
potential alteration of the voice is a major con- subcutaneous tunneling of the graft, auxiliary skin
cern. With this donor nerve, however, the func- incisions in the nasolabial folds are helpful.
tional outcome is poorer, and disturbing Additional plastic surgeries are generally
concomitant movements of the shoulder can required to achieve good cosmetic effect because
occur. The experience of the senior author with the reinnervation appears slow and is generally
this technique is that facial symmetry at rest is weaker compared to the one achieved when the
very rarely achieved. Active movements of the hypoglossal nerve is used as a donor.
Restoration of Facial Nerve Function 867

a b c

d e f

g h i

j k l

Fig. 12.13 Intraoperative images illustrating the end-to- nerve (arrows). (i, j) Identification of the hypoglossal nerve
side hypoglossal–facial anastomosis. (a) Skin incision and (two small arrows). (k, m) Sectioning of appr. 50 % of its
exposure of the mastoid (M) and digastric muscle (DM); diameter (m, arrowheads). The mobilized facial nerve
asterisk—ear lobule. (b) Identification of the facial nerve (arrow) is attached to the sectioned part of the hypoglossal
distal to the stylomastoid foramen (FN). (c–f) Mastoidectomy nerve (double arrows) and fixed with two 9-0 monofilament
and identification of the facial nerve as close to the genicu- nylon sutures and fibrin glue
late ganglion, as possible. (g, h) Mobilization of the facial
868 12 Management of Facial Palsy

Table 12.1 Facial nerve functional outcome following various reconstructive procedures
Facial nerve function (House–Brackmann scale) (in %)
Type of reconstruction Grade II–III Grade III Grade IV Grade V–VI
Reconstruction in the CP angle 21 53 21 5
Intracranial–intratemporal reconstruction 25 44 19 6
Intracranial–extratemporal reconstruction 10 60 20 10
Hypoglossal–facial anastomosis 21 75 4
Facial–facial cross face reanimation 100
Modified from [25, 32]

Outcome increasing age is associated with a poorer outcome


The reinnervation in case of end-to-end facial [19, 23, 24]. The degree of recovery of facial func-
nerve reconstruction should become clinically tion was significantly better in patients without
obvious within 12–18 months. Recovery after facial nerve symptoms prior to tumor surgery [2].
this endpoint is unlikely [19]. In case of recon- Further determining factors are the degree of atro-
struction with an interposition graft, the endpoint phy of the facial nerve and mimetic muscles [36].
for recovery is approximately 24 months, while Falcioni et al. [12] reported that tumor type also
clinical improvement following hypoglossal– correlated to the outcome: the recovery rate
facial anastomosis can occur 2 years after nerve decreased progressively in iatrogenic lesions, VSs,
repair and continue even after the third year. In petrous bone cholesteatomas, and facial nerve
the series of Malik et al. [19], best results, defined tumors. However, this relationship was also
as HB grades £ III, were achieved with the pri- influenced by the duration of the preoperative
mary end-to-side coaptation (85 %), followed by facial nerve dysfunction in every group. In our
reconstruction with a cable graft (56 %), and experience, as well as according to other authors
hypoglossal–facial anastomosis (25 %). [19], tumor type does not influence outcome.
Arriaga and Brackmann [2] reviewed the out- Improvement of facial function more than HB
come of facial nerve repair in cases of CP angle grades II to III is unattainable, whatever tech-
tumors using end-to-end coaptation and cable nerve nique is applied. The recovery of facial nerve
graft interposition. They found that HB grade IV function is a gradual process that has to be sup-
was achieved in 7/13 cases for end-to- end coapta- plemented by consequential physiotherapy.
tion and 4/8 cases of cable nerve graft and con- Throughout the period, the patient should receive
cluded that good results are obtained by immediately psychological support.
reestablishing anatomic continuity of the nerve with Following prolonged facial paralysis, the regen-
direct reanastomosis or with interposition graft. erative process is often associated with misdirected
The senior author’s experience with various axonal regrowth. As a result, synkinesis, epiphora,
types of facial nerve reconstruction is presented and varying degrees of hemifacial contracture can
on Table 12.1. Reconstruction in the CP angle occur, which are difficult to manage. A potential
led to satisfactory reinnervation (HB grade III or effective therapy in case of involuntary blepharos-
better) in 69–74 % (reconstruction within the CP pasm is botulinum A toxin injection. Generally,
angle vs. intracranial–intratemporal anastomosis). synkinesis tends to decrease after several years.
The functional outcome after hypoglossal–facial
anastomosis was even better: House–Brackmann
grade III had 75 %, and function between grades
II and III had 21 %. Plastic Surgical Procedures
Timing of surgery is the factor most closely
related to outcome. Late surgery—more than Additional plastic surgical correction should be con-
1 year after the onset of facial dysfunction—leads sidered if the recovery is insufficient and can be per-
to poorer reinnervation. According to some studies, formed without any time limit. Muscle transposition
Eye Care 869

techniques are used when the distal neuromuscular becomes possible. 5) In case of a weak mouth
unit is absent or nonviable due to fibrosis [21]. Most angle, the healthy side is pushed toward the midline
and fixed with the fingertips. The sick mouth angle
effective are the temporalis and masseter muscles can then be trained more easily. 6) Any electrical
transpositions. Facial slings with fascia lata or allo- stimulation of nerves or muscles must be avoided
plasts, such as expanded polytetrafluoroethylene, to minimize the development of contractures.
can improve facial symmetry [33].

Eye Care
Physiotherapy
In patients with facial nerve palsy, eye care is the
Transcutaneous electrical stimulation may most important concern. Maintenance of mois-
adversely affect recovery by promoting the devel- ture and protection from mechanical injury are
opment of synkinesis. Physiotherapy is of key critical to avoid complications, such as blurred
role to prevent muscle atrophy or synkinesis and vision, exposure keratopathy, corneal abrasions,
to promote functional recovery. The specific type and even blindness. Dry eye is caused by a com-
of physiotherapy is related to the degree of rein- bination of factors: reduced lacrimation, dimin-
nervation and risk of synkinesis. ished corneal reflex, or inability to close the eye
The authors’ concept for the supportive completely (lagophthalmos) and occasionally by
physiotherapy has been described in a previous eyelid malposition (paralytic ectropion). Although
publication [32]: the intermediate nerve function correlates with
1) In case of complete paralysis, the patient needs the degree of facial nerve dysfunction, diminished
to participate in a physiotherapy program for at lacrimation can occur even in case of normal or
least 3 years. The program consists of daily indi- nearly normal function of the facial nerve.
vidual exercises that are performed six times for 5 The management is directed at maintaining a nor-
to 10 minutes. During the first 1 to 2 years, the
patient needs to be accompanied by a specialized mal corneal epithelium, which provides comfort and
physiotherapist for two to three weekly sessions. preserves visual acuity. Patients with severe muscle
2) Each exercise consists of the trial of an active atrophy, as well as those with trigeminal nerve deficits,
movement and then a massage in the requested need special protection. The coexistence of corneal
direction, as follows: a) lifting eyebrows without
any help and striking eyebrows up with the anesthesia and decreased tear production or lago-
fingertips, b) frowning and pushing eyebrows phthalmos accelerates the corneal degeneration:
together, c) closing eyes and sliding down eyelids, • Topical eye lubricants such as artificial tears.
d) wrinkling the nose and pushing cheeks together, They should be applied frequently enough to
e) blowing the cheeks and holding the mouth angles
tight, f) forming a small “o” with the lips and push- keep the eye moist, for example, every 3–4 h.
ing the mouth angles together, g) smiling and pull- • Gels are thicker than artificial tears and pro-
ing mouth angles apart, and h) smiling strongly and vide more lubrication than eyedrops. However,
pushing the healthy side toward the midline to they are thick and can blur vision.
facilitate some action on the sick side. 3) If eye clo-
sure is especially weak, it is combined with strong • Moisture chamber or hourglass dressing. It cre-
biting until it becomes complete. Then the patient ates a moist ocular environment and lessen tear
tries very slowly to relax the masseter muscles evaporation but allows better vision than a patch.
while imagining closed eyes; after complete relax- • Nighttime protection. Lubricants (gels and
ation, the eyes are opened. This whole sequence is
repeated three to five times. The same exercise is ointments) combined with a patch provide the
helpful in reducing synkinesis between eye closure optimal protection.
and mouth angle. 4) If eye closure is weak in hypo- • Punctal plug insertion or partial blocking the
glossal–facial combination, a similar exercise is lacrimal duct. A relatively simple procedure
performed. The eye is closed and supported by
pressing the tongue against the teeth. The tongue is that might be necessary if dryness of the cor-
then very slowly loosened while the eye is imag- nea is a persistent problem.
ined to be closed. Within weeks to months, inde- • Tarsorrhaphy. In case a prolonged period of
pendent eye closure without any tongue movement recovery of facial nerve function is antici-
870 12 Management of Facial Palsy

pated, the outer or inner corner of the lids can nerve. J Neurosurg 90(1):27–34. doi:10.3171/
be sutured (lateral or medial tarsorrhaphy). jns.1999.90.1.0027
9. Dellon A (1992) Restoration of facial nerve function:
Generally, a lateral tarsorrhaphy of <5 mm is an approach for the twenty-first century. Neurosurg Q
not noticeable and is cosmetically acceptable. 2:199–222
Medial tarsorrhaphies should be avoided 10. Dott NM (1958) Facial paralysis; restitution by extra-
because of their disfiguring effect. petrous nerve graft. Proc R Soc Med 51(11):900–902
11. Draf W, Samii M (1980) Intracranial-intratemporal
• If a 5-mm tarsorrhaphy is not sufficient to anastomosis of the facial nerve (author’s transl).
manage the exposure keratitis, a reanimation Laryngol Rhinol Otol (Stuttg) 59(5):282–287
procedure should be considered. Skin-tone 12. Falcioni M, Taibah A, Russo A, Piccirillo E, Sanna M
external eyelid weights or gold eyelid weights (2003) Facial nerve grafting. Otol Neurotol
24(3):486–489
can be used. The weights are attached to the 13. Flores LP (2007) Surgical results of the Hypoglossal-
upper lid and improve mechanical blink. Facial nerve Jump Graft technique. Acta Neurochir
• Botulinum toxin. It is injected at the upper (Wien) 149(12):1205–1210. doi:10.1007/s00701-
border of the tarsus and produces complete 007-1412-x; discussion 1210
14. Frey M, Giovanoli P, Michaelidou M (2006)
ptosis to protect the cornea. Functional upgrading of partially recovered facial
• Ophthalmic follow-up with slit-lamp examina- palsy by cross-face nerve grafting with distal end-to-
tion and fluorescein staining are advised to assess side neurorrhaphy. Plast Reconstr Surg 117(2):597–
corneal and visual status after any lid procedure. 608. doi:10.1097/01.prs.0000197136.56749.c6
15. Gidley PW, Gantz BJ, Rubinstein JT (1999) Facial
nerve grafts: from cerebellopontine angle and beyond.
Am J Otol 20(6):781–788
References 16. Hammerschlag PE (1999) Facial reanimation with
jump interpositional graft hypoglossal facial anasto-
1. Arai H, Sato K, Yanai A (1995) Hemihypoglossal- mosis and hypoglossal facial anastomosis: evolution
facial nerve anastomosis in treating unilateral facial in management of facial paralysis. Laryngoscope
palsy after acoustic neurinoma resection. J Neurosurg 109(2 Pt 2 Suppl 90):1–23
82(1):51–54. doi:10.3171/jns.1995.82.1.0051 17. Kessler LA, Moldaver J, Pool JL (1959) Hypoglossal-
2. Arriaga MA, Brackmann DE (1992) Facial nerve facial anastomosis for treatment of facial paralysis.
repair techniques in cerebellopontine angle tumor sur- Neurology 9(2):118–125
gery. Am J Otol 13(4):356–359 18. Magliulo G, D’Amico R, Forino M (2001) Results
3. Asaoka K, Sawamura Y (1999) Hypoglossal-facial and complications of facial reanimation following
nerve side-to-end anastomosis. J Neurosurg cerebellopontine angle surgery. Eur Arch
91(1):163–164 Otorhinolaryngol 258(1):45–48
4. Campero A, Socolovsky M (2007) Facial reanimation 19. Malik TH, Kelly G, Ahmed A, Saeed SR, Ramsden
by means of the hypoglossal nerve: anatomic com- RT (2005) A comparison of surgical techniques used
parison of different techniques. Neurosurgery 61(3 in dynamic reanimation of the paralyzed face. Otol
Suppl):41–49. doi:10.1227/01. Neurotol 26(2):284–291. doi:00129492-200503000-
neu.0000289710.95426.19; discussion 49–50 00028 [pii]
5. Cheney ML, McKenna MJ, Nath R, Healy C, Bartlett 20. Manni JJ, Beurskens CH, van de Velde C, Stokroos RJ
SP (1999) Facial nerve reconstruction and facial (2001) Reanimation of the paralyzed face by indirect
reanimation following oncologic surgery. Head hypoglossal-facial nerve anastomosis. Am J Surg
Neck 21(3):276–284. doi:10.1002/(SICI)1097- 182(3):268–273. doi:S0002-9610(01)00715-2 [pii]
0347(199905)21:3<276::AID-HED15>3.0.CO;2-K 21. May M (1984) Muscle transposition for facial reani-
[pii] mation. Indications and results. Arch Otolaryngol
6. Conley J, Baker DC (1979) Hypoglossal-facial nerve 110(3):184–189
anastomosis for reinnervation of the paralyzed face. 22. May M, Sobol SM, Mester SJ (1991) Hypoglossal-
Plast Reconstr Surg 63(1):63–72 facial nerve interpositional-jump graft for facial reani-
7. Cusimano MD, Sekhar L (1994) Partial hypoglossal mation without tongue atrophy. Otolaryngol Head
to facial nerve anastomosis for reinnervation of the Neck Surg 104(6):818–825
paralyzed face in patients with lower cranial nerve 23. Pitty LF, Tator CH (1992) Hypoglossal-facial nerve
palsies: technical note. Neurosurgery 35(3):532–533; anastomosis for facial nerve palsy following surgery
discussion 533–534 for cerebellopontine angle tumors. J Neurosurg
8. Darrouzet V, Guerin J, Bebear JP (1999) New tech- 77(5):724–731. doi:10.3171/jns.1992.77.5.0724
nique of side-to-end hypoglossal-facial nerve attach- 24. Sabin HI, Bordi LT, Symon L, Compton JS (1990)
ment with translocation of the infratemporal facial Facio-hypoglossal anastomosis for the treatment
References 871

of facial palsy after acoustic neuroma resection. Br J 32. Samii M, Matthies C (1997) Management of 1000
Neurosurg 4(4):313–317 vestibular schwannomas (acoustic neuromas): the
25. Samii M (1981) Preservation and reconstruction of facial nerve–preservation and restitution of function.
the facial nerve in the cerebellopontine angle. In: Neurosurgery 40(4):684–694; discussion 694–685
Samii M, Janetta P (eds) The cranial nerves. Springer, 33. Tate JR, Tollefson TT (2006) Advances in facial rean-
Berlin, pp 438–450 imation. Curr Opin Otolaryngol Head Neck Surg
26. Samii M (1984) Facial nerve grafting in acoustic 14(4):242–248. doi:10.1097/01.moo.0000233594.
neurinoma. Clin Plast Surg 11(1):221–225 84175.a0
27. Samii M (1986) Surgery of facial nerve paralysis. No 34. Viterbo F, Trindade JC, Hoshino K, Mazzoni A (1994)
Shinkei Geka 14(8):943–955 Two end-to-side neurorrhaphies and nerve graft with
28. Samii M, Draf W (1989) Surgery of the skull base. removal of the epineural sheath: experimental study in
Springer, Heidelberg rats. Br J Plast Surg 47(2):75–80
29. Samii M, Jannetta P (eds) (1981) The cranial nerves: 35. Viterbo F, Trindade JC, Hoshino K, Mazzoni Neto A
anatomy, pathology, pathophysiology, diagnosis, (1994) End-to-side neurorrhaphy with removal of the
treatment. Springer, New York epineural sheath: an experimental study in rats. Plast
30. Samii M, Koerbel A, Safavi-Abbasi S, Di Rocco F, Reconstr Surg 94(7):1038–1047
Samii A, Gharabaghi A (2006) Using an end-to-side 36. Ylikoski J, Brackmann DE, Savolainen S (1982)
interposed sural nerve graft for facial nerve reinforce- Facial nerve abnormalities after acoustic tumor
ment after vestibular schwannoma resection. Technical removal. Morphological and clinical study of seven
note. J Neurosurg 105(6):920–923. doi:10.3171/ patients with postoperative facial paralysis. Arch
jns.2006.105.6.920 Otolaryngol 108(12):795–800
31. Samii M, Matthies C (1994) Indication, technique and
results of facial nerve reconstruction. Acta Neurochir
(Wien) 130(1–4):125–139
Index

A Arteriovenous malformations (AVMs), 105–108, 778


AAO-HNS hearing classification, 255 Aspergillosis, 86–88
Abducens nerve schwannoma, 581–583 Aspergillus ssp., 87
Accessory nerve, 10, 41, 55, 60–61 Atypical choroid plexus papillomas, 101
Accessory nerve schwannoma, 637, 649–651 Atypical meningiomas, 79
Acidophilic adenoma, 93–95 Audiometric examinations, 165
Acoustic neuromas Auditory brain stem implants (ABI)
anatomy, 34 cochlear implantation, 371
arterial relationships, 43, 45, 48 complications, 368, 370
brain stem relationships, 39 factors, 370
history of, 1 indications, 364
middle neurovascular complex, 34 intraoperative images, 364, 368–370
retrosigmoid approach, 35, 37–39 lip-reading enhancement, 370
venous relationships, 44–47 MRI, large left-sided VS, 364, 368
Acute bacterial sive purulent meningitis, 84–86 penetrating electrodes, 370–371
Adamantinomatous craniopharyngioma, 97–98 prosthesis
Adenohypophysis, spindle cell oncocytoma, 96 magnetless ABI, 364, 367
Aggressive papillary middle ear tumors. removal of right-sided tumor, 364–366
See Endolymphatic sac tumors (ELST) rehabilitation, 370
AICA. See Anteroinferior cerebellar artery (AICA) Auditory midbrain implant (AMI), 371
Anaplastic ependymoma, 100, 724–725 Autonomous nervous system pathology, 77
Anaplastic hemangiopericytoma, 81–82 Autosomal dominant polycystic kidney disease, 108
Anaplastic meningiomas, 81 AVMs. See Arteriovenous malformations (AVMs)
Anastomosis
facial-facial nerve, 866
hypoglossal-facial, 864–867 B
Ancient schwannomas, 74–75 Bacterial meningitis
Aneurysmal bone cyst, 90, 91 acute bacterial sive purulent meningitis, 84–85
Angiomatous meningiomas, 78 sarcoidosis, 86, 87
Anteroinferior cerebellar artery (AICA) tuberculous meningitis, 85–87
anterior view, 15 Basophilic adenoma, 95
cerebellopontine angle, 11, 15–16 Besnier–Boeck–Schaumann disease, 86, 87
CNs VII and VIII in, 15 Bilateral cerebellopontine (CP) angle tumors
compression, 26 bilateral osteomas, 354–358
Antineutrophil cytoplasmic antibody (ANCA)-mediated extensive skull base meningioma, 352, 354
inflammations, 112 foramina of Luschka, 352–353
Arachnoid cysts melanoma metastases, 359–363
complete removal, 736–739 metastatic lesions in cerebellar hemispheres, 352
CT and MR appearance, 731 Bilateral vestibular schwannomas, 74
description, 731 Bill’s bar, 37
medial cyst wall, incomplete resection of, 733–735 Blood vessels and circulation pathology
patient management, 731 arteriovenous malformation, 105–108
surgical options, 731–732 hypertensive angiopathy and hemorrhage,
symptoms, 732 109–111
Arterial relationships ischemic brain infarct, 109–110
middle neurovascular complex, 43, 45, 48 saccular aneurysm, 107–109
upper neurovascular complex, 26–29 vasculitis, 110–112

M. Samii, V. Gerganov, Surgery of Cerebellopontine Lesions, 873


DOI 10.1007/978-3-642-35422-9, © Springer-Verlag Berlin Heidelberg 2013
874 Index

Brain stem Chemodectoma, 77


acoustic neuromas, 39 Cholesterol granuloma of petrous apex
middle neurovascular complex, 39 classic obstruction-vacuum theory, 789
Brain stem gliomas clinical presentation, 790
description, 703 CT and MRI, 789
low-grade exophytic astrocytoma, 705, 708–709 description, 788–789
low-grade gliomas, 706, 710–713 differential diagnosis, 789
MR imaging, 704 incidence, 788
pilocytic astrocytoma, 703–705 management options, 790–791
surgery, 705 recurrence rate, 791
tumor removal steps, 705, 707 retrosigmoid intradural inframeatal
Breast carcinoma metastases, 766 approach, 792–797
Chondroblastomas, 90, 91, 818–821
Chondrosarcomas (CSA)
C clinical presentation, 807, 809
Cavernomas grades, 807
clinical presentation, 778, 781 histological types, 807
in IAC, 783, 785–787 in jugular foramen area, 815–817
incidence, 778 neuroimaging, 807
intracanalicular, 781, 782 origin, 807
recurrence risk, 784 pathology, 90, 91
retrosigmoid approach, 784–787 removal, anterior petrosal route, 810–811
signs/symptoms, 781 retrosigmoid suprameatal approach, 812–814
total microsurgical resection, 784 Chordoid meningiomas, 79
Cell-mediated inflammations, 112 Chordomas
Cellular ependymoma, 100 chondroid, 803–804
Cellular schwannomas, 74–75 endonasal-combined microsurgical removal, 799,
Cerebellar arteries, 16–18 801–803
Cerebellar brain stem fissures, 14–17 iatrogenic injury, 800
Cerebellar hemispheres management, 798–800, 803–804
edema in, 777 neuroimaging, 798
metastatic lesions in, 352 origin, 798
Cerebellar surfaces, 11, 14–17 pathology, 91–92
Cerebellomedullary fissure, 16 retrosigmoid approach, 805–806
Cerebellomesencephalic fissure, 14, 16 symptoms/signs, 798
Cerebellopontine angle transethmoid transsphenoidal approach, 799–800
anteroinferior cerebellar artery, 11, 15–16 Choriocarcinoma, 104
cleavage plane, 12, 13 Choroid plexus
dura, 12 AVM, 107
epidermoids (see Epidermoids) carcinoma, 101
flocculus and choroid plexus, 12 flocculus and, 12, 58
meatal dura, 12, 13 and foramen of Luschka, 44
meningiomas (see Meningiomas) papillomas, 100–101, 714–717
middle neurovascular complex, 36 Chromophobic adenoma, 95, 96
posteroinferior cerebellar artery, 11, 17 Clear cell ependymoma, 100
superior cerebellar artery, 11, 14 Clear cell meningiomas, 79
superior petrosal vein, 12 Clival chordoma, 799, 801–803
surgical management Clival meningiomas, 380–382
hearing preservation, 6 Cochlear nerves, 37
microsurgical technique, 5–6 Cochlear schwannomas, 605
middle fossa approach, 5 Collision tumors, 767–772
radiosurgery, 6 Communicating hydrocephalus, 275, 277–278
surgical improvements, 3–4 Cranial nerves
surgical microscope, 5–6 VS, 297
translabyrinthine approach, 4–5 posterior cranial fossa, 9–11
transpetrosal approach, 5 Craniopharyngioma, 97–98
vestibular schwannomas, 2 Cryptococcosis, 88–89
vestibulocochlear nerve, 13 Cryptococcus ssp., 88
Cerebellopontine fissure, 16 CSA. See Chondrosarcomas (CSA)
Index 875

CSF leak, 300–302 Ependymomas


Cysticercosis, 88, 89 description, 718
Cystic jugular foramen schwannomas, 614–615, 637, differential diagnosis, 718
652–655 pathology, 99–100
Cystic tumors, 252 patient care, 718
Cystic VS prognostic factors, 719
capsule, 234–235 radiotherapy, 719
MR imaging, 233, 236–237, 239–240 recurrences, 719
nerve adhesion, 238 Epidermoid cyst, 105, 107
proteolytic enzymes, 232 Epidermoids
tumor debulking, 234 angled endoscope usage, 515
case study
endoscopic inspection, 524, 529, 535, 539, 540
D patients clinical history, 519, 524, 530, 535, 540
Deep noncortical AVM, 107 postoperative periods, 519, 523, 530, 531, 534,
Delayed facial nerve palsy, 254 535, 540, 541
Dermoid cyst, 105, 107 preoperative images, 524–526, 530–532,
Dermoids 536–537, 540, 541
case illustration, 742–743 RS approach, 526–528, 530, 532–534, 537–538,
complications, 740–741 541–544
CT and MRI, 740, 741 CISS MR sequence, 509, 511
description, 740 CT and MRI images, 509, 510, 512–514
vs. epidermoids, 741 diffusion-weighted imaging, 511, 516–517
signs and symptoms, 740 features, 509
Developmental venous anomalies (DVAs), 773, 775–777 FLAIR sequence, 509, 511
Diffuse melanocytosis, 83 frontotemporal approach, 519
Dumbbell tumors, 559–568, 575–576 location/extension, 509, 515
management, 511–512, 514–515
MR cisternography, 511
E petrous bone, 515, 518
Echinococcosis, 88, 89 retrosigmoid approach, 511, 523
Echinococcus granulosus, 89 signs/symptoms, 511
Ehlers–Danlos syndrome type IV, 108 supra and infratentorial extension, 519–521
ELST. See Endolymphatic sac tumors (ELST) tumor capsule dissection, 515
Embryonal carcinoma, 104 Erdheim–Chester disease, 103
Endolymphatic sac tumor, 92–93 Extended MF approach, 118–119
Endolymphatic sac tumors (ELST) Extra-axial tumors
case illustration aneurysms, 773, 774
sporadic cystic ELST, 831–833 arachnoid cysts
sporadic ELST removal, 823–826 CT and MR appearance, 731
VHL syndrome-associated ELST, 827–830 description, 731
incidence, 818 medial cyst wall, incomplete
management, 822 resection of, 733–735
neuroimaging, 821 patient management, 731
origin, 818 removal, 736–739
symptoms, 822 surgical options, 731–732
unilateral hearing loss, 821–822 symptoms, 732
Endonasal-combined microsurgical technique, 799, arteriovenous malformations, 778
801–803 cavernomas
Endoscopy case illustration, 783, 785–787
clival chordoma removal, 799, 801–803 clinical presentation, 778, 781
microsurgical technique, 720–723 incidence, 778
transcervical tumor removal, 618–624 intracanalicular, 781, 782
End-to-end facial nerve reinforcement technique, 860, recurrence risk, 784
862, 863 RS approach, 784–787
End-to-side facial nerve insertion technique, 860–861, signs/symptoms, 781
864 total microsurgical resection, 784
Eosinophilic adenoma, 95, 96 collision tumors, 767–772
Eosinophilic granuloma, 102 dermoids, 740–743
876 Index

Extra-axial tumors (cont.) reoperations, 252


developmental venous anomalies, 773, 775–777 taste disturbance, 255
hamartomas, 744–747 tumor characteristics, 250–252
hemangioblastomas Facial nerve palsy, 253–254
case illustration, 760–763 Facial nerve reconstruction
cerebral angiography, 757–758 end-to-end anastomosis, 856
description, 757 end-to-end reinforcement, 860, 862, 863
MRI, 757–758 end-to-side insertion technique, 860–861, 864
operative strategy considerations, 759 eye care, 869–870
preoperative endovascular functional outcome, 868
embolization, 758–759 goal, 855
radiotherapy, 759 indications, 855–856
staged multimodal management, 758 with interposition graft
surgical removal, 758 in CP angle, 857–859
lipomas, 748–749 great auricular nerve graft, 857
metastasis requirements, 856
case illustration, 766 sural nerve graft, 856–857
clinical factors, 764 intracranial-extracranial technique, 859–860, 862
management, 765 intracranial-intratemporal technique, 858–861
primary malignancies, 764 nerve anastomosis, principles of, 855
radiologic appearance, 764 physiotherapy, 869
neurenteric cysts plastic surgical procedures, 868–869
case illustration, 752–756 with reanimation techniques
clinical presentation, 749–750 accessory nerve, 866
endodermal origin, 749 facial-facial nerve anastomosis, 866
management, 750 hypoglossal-facial anastomosis, 864–867
MR characteristics, 749–751 indications, 862, 864
neuroimaging, 749 timing, 855, 868
spinal, 749 Facial schwannomas
vascular lesions, 773 case illustration, 590–593, 596, 599–308
Extrameatal tumor removal complete tumor removal with resection, 596–598
tumor debulking/decompression, 187, 191–192 ganglion geniculi, 584, 588–589
tumor variables, 190–191 in mastoid segment, 584–587
ultrasonic aspirator, 188, 190 middle cranial fossa, 584
radiological patterns, 594–595
Fibrous dysplasia (FD)
F description, 834
Facial-facial nerve anastomosis, 866 development patterns, 834
Facial motion classification, 251 pathology, 90
Facial nerve of petrous bone, 835–838
arterial compression, 58–59 radiological differential diagnosis, 834
infrafloccular exposure, 56–57 Fibrous meningiomas, 78
meatal relationships, 37, 40–42 Flocculus, 12
operative techniques development Fluid-attenuated inversion recovery imaging
translabyrinthine approach, 4–5 (FLAIR), 509, 511
transpetrosal approach, 5 Foramen jugular chondrosarcoma, 807, 808
vestibular schwannomas Foramen magnum meningiomas
cystic tumors, 252 clinical presentation, 482
delayed facial nerve palsy, 254 complete tumor removal, 489–491
facial nerve palsy, 253–254 facial and vestibulocochlear nerve
grading systems, 250 complex, 489, 492–493
intermediate nerve dysfunction, 254–255 factors, 483
intraoperative monitoring, 252 headache and vertigo, 496–498
NF-2, 252 iatrogenic injury avoidance, 487
operative approach, 253 lateral suboccipital approach, 487–489
patient age, 252 limits, 482
preoperative facial palsy, 252–253 lower clivus extendsion, 483, 486
preservation, 250 occipital headache and gait instability, 499–500
radiosurgery, 252 outcome, 489, 493
Index 877

preoperative neuroradiological Hearing and VS


examinations, 482–483 classification, 255–256
rupture contralateral vertebral artery, 489, 494–495 preservation rate, 262, 266–267
size of craniotomy and dentate ligament, 483–485 risk factors, 256
Foramen of Luschka, 10, 39, 44 tumor removal and preservation, 257–271
Fourth ventricle Hearing loss, 201
anatomy, 18–19 Hearing preservation, history of, 6
choroid plexus pathology, 100–101 Hemangioblastomas
subependymoma, 727–729 case illustration, 760–763
telovelar approach to, 64–65, 70 cerebral angiography, 757–758
Functioning adenomas, 94 description, 757
Fungal meningitis MRI, 757–758
aspergillosis, 86–88 operative strategy considerations, 759
cryptococcosis, 88–89 preoperative endovascular embolization, 758–759
radiotherapy, 759
staged multimodal management, 758
G
surgical removal, 758
Gardner–Robertso scale, 255
Hemangiopericytoma, 81–82
Gardner-type NF-2, 315
Hematopoietic system pathology
Germ cells pathology
Langerhans cell histiocytosis, 102–103
dysontogenetic cysts, 105–107
non-Langerhans cell histiocytosis, 103
tumors, 104–106
primary CNS lymphomas, 102
Germinoma, 104
Hemorrhagic complications, 201
Giant partially thrombosed aneurysm, 773, 774
Hereditary hemorrhagic telangiectasia, 109
Giant (extension grade T5) VS
High jugular bulb
bone-window MRI, 207–210
classification, 173, 174
hearing loss, 201
grade III, 181, 182
hemorrhagic complications, 201
intraoperative images, 177–179
neurological examination, 207
postoperative imaging, 180
operated seven times, 245–246
preoperative imaging, 176
preoperative MRI, 202
House–Brackmann grading system, 250
RS approach, 204–205
Hydrocephalus associated VS
Glomus jugulare tumors
clinical management, 273, 282
acute lower cranial nerve dysfunction, 668
communicating, 275, 277–278
autonomous nervous system, 77
MRI, 274–275
clinical presentation, 666, 668
obstructive, 275
features, 664
retrosigmoid approach, 280
Fisch classification, 665
Hypertensive angiopathy and brain hemorrhage, 109–111
Glasscock–Jackson classification, 665
Hypoglossal-facial anastomosis, 864–867
neuroimaging, 665–667
Hypoglossal nerve, 59
pathogenesis, 664–665
Hypoglossal nerve schwannomas, 664
surgery of
carotid artery, jugular vein, lower cranial
nerves, 670–673
I
embolization, 670, 680–685
Immature teratoma, 105, 106
neuronavigation screenshots, 670, 676
Immune-complex-mediated inflammations, 112
radiotherapy/radiosurgery, 670, 673
Indirect facial nerve reconstruction. See Reanimation
transmastoid infralabyrinthine approach, 670,
techniques, facial nerve reconstruction
674–675, 677–679
Inframeatal meningiomas
watchful waiting, 668
case illustration, 407–408
Glomus tympanicum tumor, 77
foramen magnum level, 401
Glossopharyngeal nerve, 59, 61–63, 646, 664
pattern of dislocation, 376, 379, 380
Granular cell tumor, 97
Internal acoustic meatus, 38, 41
Great auricular nerve graft, n facial nerve
Intra-axial tumors
reconstruction, 857
brain stem gliomas
description, 703
H low-grade exophytic astrocytoma, 705, 708–709
Hamartomas, 744–747 low-grade gliomas, 706, 710–713
Hand–Schüller–Christian disease, 102, 103 MR imaging, 704
878 Index

Intra-axial tumors (cont.) type C, T1-weighted and T2-weighted


pilocytic astrocytoma, 703–705 MRI, 629–630
surgery, 705 type D, dumbbell-shaped tumor, 629, 631
tumor removal steps, 705, 707 origin, 627
choroid plexus papillomas, 714–717 retrosigmoid endoscope-assisted approach, 637,
clinical manifestation, 703 656–659
ependymomas staged removal in two steps, 637, 660–663
anaplastic, 724–725 type A schwannomas
description, 718 with intracranial growth, 632–636
differential diagnosis, 718 pure intracranial location, 637, 646–648
patient care, 718 RS approach and opening of JF, 637, 640–644
prognostic factors, 719 type D, 637–639
radiotherapy, 719 type II cervical and extradural transmastoid
recurrences, 719 infralabyrinthine approach, 637, 645
lymphomas, 730 vagus nerve schwannoma, 627–628
medulloblastomas, 730 Jugular foramen (JF) tumors
subependymomas, 726–729 clinical presentation and diagnosis, 610–611
Intracanalicular meningiomas, 416–418 craniectomy, mastoidectomy, 616
Intracanalicular tumor grading, 152 electrophysiological monitoring, 612
Intracanalicular (extension grade 1) VS endoscope-assisted tumor removal, 617–624
adhesions to nerves, 219–221 extended endoscopic endonasal approach, 625
capsule and adhesion, 217–218 infratemporal approach, 612
CT and MRI, 195 intradural exposure, 616
hearing loss, 195 lateral suboccipital approach, 612–615
microsurgery, 193–197 lower cranial nerves dysfunction, 625
retrosigmoid approach, 193–194 management, 611
stereotactic radiosurgery, 193 neck dissection and exposure of cervical region,
Intracranial-extracranial facial nerve vessels, and nerves, 616
reconstruction, 859–860, 862 neuroimaging
Intracranial-intratemporal facial nerve digital subtraction angiography, 610
reconstruction, 858–861 MR signal characteristics, 610
Intralabyrinthine schwannomas, 844 native and contrast-enhanced MRI, 609
Intraosseous meningiomas, 419–421 typical radiological characteristics, 609–610
Ischemic brain infarct, 109–110 pars nervosa, 611
postoperative care
aspiration pneumonia, 625–626
J CSF leak, 626
Jugular foramen (JF) meningiomas speech-language therapy, 626
clinical manifestation, 691 primary bone tumors, 609
facial nerve, 691 radiosurgery and stereotactic radiotherapy, 626–627
features, 686 reconstruction of dura and skull base, 616–617
neuroimaging structures, 609
en plaque appearance, 686–688 tumor removal, 616–617
foramen magnum area, 686, 688 Jugulotympanic paraganglioma, 77
primary meningioma, 686, 689–690 Juvenile xanthogranuloma, 103
operative treatment
with large CP angle part, 693–697
parameters, 691 L
outcome, 692 Langerhans cell histiocytosis (LCH), 102–103
recurrence rate, 691–692 Lateral suboccipital approach, 710–713
Jugular foramen (JF) schwannomas Leptomeningeal melanocytic lesions, 83, 84
accessory nerve schwannoma, 637, 649–651 Lipomas, 748–749
classifications, 627 Lower cranial nerve schwannomas. See Jugular foramen
clinical presentation, 628–629 (JF) tumors
neuroimaging Lower neurovascular complex
cystic schwannomas, 637, 652–655 glossopharyngeal nerve, 61–63
type A with large intracranial part and neural relationships, 59–61
smaller JF part, 629 vascular compression, 63–64
type B, axial and coronal T1-weighted vascular relationships, 62–63
MRI, 629–630 Low-grade exophytic astrocytoma, 705, 708–709
Index 879

Lymphomas, 730 hormonal therapy and chemotherapy, 435


Lymphoplasmacyte-rich meningiomas, 78 IAC involvement
case illustration, 411, 413–415
high-speed drill, 411–412
M neuroimaging, 410–411
Malignant melanoma, 83 variable extension patterns, 409–410
Malignant peripheral nerve sheath tumor inframeatal, 376, 379, 380
(MPNST), 76–77 internal decompression, 394
Marfan’s syndrome, 108 intracanalicular, 416–418
Mature teratoma, 105, 106 intraosseous, 419–421
Meatal dura, 12, 13, 36–38 jugular foramen
Meckel’s cave, 21, 32, 40 clinical manifestation, 691
Medial VS facial nerve, 691
caudal tumor extension, 229, 231 features, 686
cranial nerve dissection, 229 neuroimaging, 686–690
preoperative imaging, 228–229 operative treatment, 691, 693–697
RS approach, 230 outcome, 692
Medulloblastomas, 730 recurrence rate, 691–692
Melanocytoma, 83 lateral retromeatal
Melanocytosis, 83 case illustration, 398–400
Melanomatosis, 83 CISS and T2-weighted sequences, 396–397
Melanotic schwannomas, 76 sigmoid sinus involvement, 396
Meningeal hemangiopericytoma, 81, 82 management
Meninges pathology adhesive characteristics, 393–394
acute bacterial sive purulent meningitis, 84–85 hearing preservation, 393
aspergillosis, 86–88 hydrocephalus, 394
cryptococcosis, 88–89 treatment options, 392
cysticercosis, 88, 89 multiple, 424–431
echinococcosis, 88, 89 neuroimaging
hemangioblastoma, 82–84 cavernous sinus and retrosellar growth, 382,
hemangiopericytoma, 81–82 384–385
leptomeningeal melanocytic lesions, 83 CSF signal characteristics, 390
meningioma, 78–81 dural tail sign, 382, 386
meningiosis and solid metastatic tumors, 83, 84 intratumoral calcification, 380, 382
sarcoidosis, 86, 87 mushrooming effect, 383, 387
tuberculous meningitis, 85–87 petroclival meningioma with perifocal edema,
Meningiomas 383, 388–389
atypical and anaplastic, 423–424 secondary intracanalicular extension,
centered at IAC, 376, 379 382, 385–386
clinical presentation, 390–392 tumor-feeding arteries, 390
CP angle definitions, 376 outcome, 422–423, 472
epidemiology, 375 pathogenesis and pathology, 375–376
examination, 392 pathology
foramen magnum age distribution, 78
clinical presentation, 482 anaplastic, 81
complete tumor removal, 489–491 angiomatous, 78, 79
facial and vestibulocochlear nerve complex, atypical, 79, 81
489, 492–493 chordoid, 79, 81
factors, 483 classification, 376
headache and vertigo, 496–498 clear cell, 79, 80
iatrogenic injury avoidance, 487 epidemiology, 78
lateral suboccipital approach, 487–489 fibrous, 78, 79
limits, 482 grading, 78
lower clivus extension, 483, 486 histology, 78
occipital headache and gait instability, 499–500 lymphoplasmacyte-rich, 78, 80
outcome, 489, 493 meningothelial, 78, 79
preoperative neuroradiological examinations, metaplastic, 78–80
482–483 microcystic, 78, 80
rupture contralateral vertebral artery, 489, 494–495 NF-2 mutations, 375–376
size of craniotomy and dentate ligament, 483–485 papillary, 81
880 Index

Meningiomas (cont.) fourth ventricle, 18–19


psammomatous, 78, 79 lower neurovascular complex
rhabdoid, 81 glossopharyngeal nerve, 61–63
secretory, 78, 80 neural relationships, 59–61
sex distribution, 78 vascular compression, 63–64
transitional, 78, 79 vascular relationships, 62–63
perioperative management, 392 middle neurovascular complex
petroclival acoustic neuromas, 34
case illustration, 438, 440–442 arterial relationships, 43, 45, 48
cavernous sinus, 435–436 brain stem relationships, 39
endoscopeassisted removal, 452, 477–481 cerebellopontine angle, 36
features, 435–437 meatal relationships, 36–38
jugular foramen and foramen magnum, 472–476 neural relationships, 39, 42–44
with large supratentorial part, 451–453 retrosigmoid approach, 35, 37–39
located medial to trigeminal nerve, 435–436 vascular compression, 49–57
management of, 437 venous relationships, 44–47
massive venous infarction, 447, 449–450 posterior cranial fossa, 9, 10
Meckel’s cave, 435–436 telovelar approach, to fourth ventricle,
pattern of dislocation, 376, 379 64–65, 70
presigmoid approach, 444–447 tumor removal
RSMA (see Retrosigmoid suprameatal approach) distorted nerves, 65, 69
small petrous apex, 438–439 dural attachment, 65, 67
surgical management, 443 meningioma attachment, 65, 66
two-staged removal, 453–458 nerve displacement, 65, 67
venous phase of DSA, 447–448 routes, 65
postoperative management, 422 upper neurovascular complex
premeatal, 376, 378, 380 arterial relationships, 26–29
pure clival, 380–382 suprameatal extension, 32–33
recurrence, 423 trigeminal root anatomy, 20–24
removal, retrosigmoid approach, 65, 66 vascular compression, 24–26
retromeatal, 376, 378, 380 venous relationships, 29–32
retrosigmoid approach, 394–395 Microsurgical technique, history of, 5–6
sphenopetroclival, 376–377 Middle fossa (MF) approach
suprameatal, 376, 378, 380 advantages and drawbacks, 115
tentorial, 432–434 extended, 118–119
Meningiosis and solid metastatic tumors, 83–84 history of, 5
Meningitis, 302 IAC, 115
Meningothelial meningiomas, 78 standard, 115–118
Metaplastic meningiomas, 78 Middle neurovascular complex
Metastasis acoustic neuromas, 34
breast carcinoma metastases, 766 arterial relationships, 43, 45, 48
case illustration, 766 brain stem relationships, 39
clinical factors, 764 cerebellopontine angle, 36
management, 765 meatal relationships, 36–38
primary malignancies, 764 neural relationships, 39, 42–44
radiologic appearance, 764 retrosigmoid approach, 35, 37–39
Microcystic meningiomas, 78 vascular compression, 49–57
Microsurgical anatomy, of CA acoustic neuromas removal, 53
cerebellar arteries, 16–18 cochlear and vestibular nerve
cerebellar brain stem fissures, 14–17 compression, 53–54, 56
cerebellar surfaces, 11, 14–17 geniculate neuralgia, 56, 57, 62
cerebellopontine angle hemifacial spasm, 51
anteroinferior cerebellar artery, 11, 15–16 venous relationships, 44–47
cleavage plane, 12, 13 Monostotic fibrous dysplasia (MFD), 90
dura, 12 Multiple endocrine neoplasia type 1
flocculus and choroid plexus, 12 (MEN 1), 93
meatal dura, 12, 13 Multiple meningiomas
posteroinferior cerebellar artery, 11, 17 differential diagnosis, 344
superior cerebellar artery, 11, 14 MRI findings, 426–432
superior petrosal vein, 12 in NF-2 patients, 424
vestibulocochlear nerve, 13 treatment, 425
Index 881

N hemangiopericytoma, 81–82
Neural relationships leptomeningeal melanocytic lesions, 83
lower neurovascular complex, 59–61 meningioma, 78–81
middle neurovascular complex, 39, 42–44 meningiosis and solid metastatic tumors, 83–84
Neurenteric cysts sarcoidosis, 86, 87
case illustration, 752–756 tuberculous meningitis, 85–87
clinical presentation, 749–750 osseous skull base
endodermal origin, 749 aneurysmal bone cyst, 90, 91
management, 750 chondroblastoma, 90, 91
MR characteristics, 749–751 chondrosarcoma, 90, 91
neuroimaging, 749 chordoma, 91–92
spinal, 749 endolymphatic sac tumor, 92–93
Neurocysticercosis, 788 fibrous dysplasia, 90
Neurofibromatosis type 2 (NF-2) osteochondroma, 90, 91
classification, 315 peripheral nervous system
clinical presentation MPNST, 76–77
bilateral VS, 316–317 schwannomas, 74–76
case illustration, 317–320 regional glial tumors
manifestation, 316 ependymoma, 99–100
multiple spinal schwannomas, 321–323 pilocytic astrocytoma, 99
differential diagnosis, 343 pilomyxoid astrocytoma, 99–100
facial nerve and VS, 252 sellar region
Gardner-type, 315 craniopharyngioma, 97–98
genetics, 315–316 granular cell tumor, 97
large VSs, 328–332 pituicytoma, 96
management, 321, 323–328 pituitary adenoma, 93–96
modified NIH criteria, 315–316 pituitary carcinoma, 95–97
multiple meningiomas, 424 spindle cell oncocytoma, 96
mutations, 375–376 xanthogranuloma, 98–99
personal series, 337–342, 345–351 New Hannover hearing classification, 256
revised clinical criteria, 315–316 NF-2. See Neurofibromatosis type 2 (NF-2)
smaller VS, 328, 332–335 Nonfunctioning adenomas, 94
surgery on hearing side, 332–333, 336 Non-langerhans cell histiocytosis, 103
Neurohypophysis, granular cell tumor, 97 Nonvestibular schwannomas
Neuropathology abducens nerve, 581–583
autonomous nervous system, 77 cochlear, 605
blood vessels and circulation facial
arteriovenous malformation, 105–108 case illustration, 590–593, 596, 599–308
hypertensive angiopathy and brain complete tumor removal with resection,
hemorrhage, 109–111 596–598
ischemic brain infarct, 109–110 ganglion geniculi, 584, 588–589
saccular aneurysm, 107–109 in mastoid segment, 584–587
vasculitis, 110–112 middle cranial fossa, 584
choroid plexus radiological patterns, 594–595
carcinoma, 101 lower cranial nerve (see Jugular foramen (JF) tumors)
papilloma, 100–101 oculomotor nerve, 578
germ cells trigeminal
tumorlike dysontogenetic cysts, 105, 107 classification, 547, 548
tumors, 104–106 clinical presentation, 549
hematopoietic system dumbbell tumors, 559–568, 575–576
langerhans cell histiocytosis, 102–103 infratemporal extracranial location, 569–574
non-langerhans cell histiocytosis, 103 management, 549–550
primary CNS lymphomas, 102 neuroimaging, 547–549
meninges operative technique, 569, 577
acute bacterial sive purulent meningitis, outcome, 577
84–86 posterior fossa location, 553–558
aspergillosis, 86–88 predominant location in middle cranial
cryptococcosis, 88–89 fossa, 550–553
cysticercosis, 88, 89 radiotherapy, 577–578
echinococcosis, 88, 89 surgical treatment, 550
hemangioblastoma, 82–83 trochlear, 578–580
882 Index

O incidence, 788
Obstructive hydrocephalus, 275 management options, 790–791
Oculomotor nerve schwannoma, 578 recurrence rate, 791
Osseous skull base pathology removal of, 792–797
aneurysmal bone cyst, 90, 91 description, 788
chondroblastoma, 90, 91 mucoceles, 788
chondrosarcoma, 90, 91 Petrous bone epidermoids, 515, 518
chordoma, 91–92 Pilocytic astrocytoma, 99, 703–705
endolymphatic sac tumor, 92–93 Pilomyxoid astrocytoma, 99–100
fibrous dysplasia, 90 Pituicytoma, 96
osteochondroma, 90, 91 Pituitary adenoma
Osteoblastic metaplasia, 80 classification, 93–94
Osteochondroma, 90, 91 ectopic, 93
Osteomas, 839–840 functioning, 94
microadenomas, 93
nonfunctioning, 94
P Pituitary carcinoma, 95–97
Papillary craniopharyngioma, 98 Pleomorphic adenoma, 840–843
Papillary ependymoma, 100 Plexiform schwannoma, 76
Papillary meningiomas, 81 Pneumocephalus, 300
Parasitic meningitis, 88, 89 Polyostotic fibrous dysplasia (PFD), 90
Pars nervosa, 611 Posterior cranial fossa
Partial translabyrinthine approach, 121 anatomy, 9
Peripheral aneurysms, 773 cranial nerves, 9–11
Peripheral nervous system pathology neurovascular complexes, 20
MPNST, 76–77 Posteroinferior cerebellar artery (PICA)
schwannoma cerebellopontine angle, 11, 17–18
bilateral vestibular, 74, 76 CNs X and XI, 17
cellular, 74–75 Postoperative hydrocephalus, 302–304
histology, 74 Premeatal meningiomas
melanotic, 76 case illustration, 401, 404–406
nonvestibular, 74 complete removal of, 401, 403
Petroclival meningiomas limited growth into IAC, 401–402
case illustration, 438, 440–442 pattern of dislocation, 376, 378, 380
cavernous sinus, 435–436 Primary CNS lymphomas (PCNSL), 102
endoscopeassisted removal, 452, 477–481 Proteolytic enzymes, 232
features, 435–437 Psammomatous meningiomas, 78
jugular foramen and foramen magnum, 472–476
with large supratentorial part, 451–453
located medial to trigeminal nerve, 435–436 R
management of, 437 Radiosurgery, history of, 6
massive venous infarction, 447, 449–450 Reanimation techniques, facial nerve reconstruction
Meckel’s cave, 435–436 accessory nerve, 866
pattern of dislocation, 376, 379 facial-facial nerve anastomosis, 866
presigmoid approach, 444–447 hypoglossal-facial anastomosis, 864–867
RSMA (see Retrosigmoid suprameatal approach) indications, 862, 864
small petrous apex, 438–439 Regional glial tumors pathology
sporadic unilateral VS and, 768–772 ependymomas, 100
surgical management, 443 pilocytic astrocytoma, 99
two-staged removal, 453, 456–458 pilomyxoid astrocytoma, 99, 100
venous phase of DSA, 447–448 Retrolabyrinthine approach, 121
Petrosal veins, 29, 48, 49 Retromeatal meningiomas
Petrous apex case illustration, 398–400
cholesterol granuloma CISS and T2-weighted sequences, 396–397
classic obstruction-vacuum theory, 789 pattern of dislocation, 376, 378, 380f
clinical presentation, 790 sigmoid sinus involvement, 396
CT and MRI, 789 Retrosigmoid approach
description, 788–789 anesthesia considerations, 123, 126
differential diagnosis, 789 benefits of, 122–123
Index 883

chordomas, 805–806 Sinus thrombosis, 299–300


drawbacks Sphenopetroclival meningiomas, 376–377
limited exposure of the fundus, 134–135, 138 Spinal neurenteric cysts, 749
need of cerebellar retraction, 136 Spindle cell oncocytoma, 96
pneumocephalus and tension Sporadic unilateral vestibular schwannomas, 768–772
pneumocephalus, 137–140 Standard MF approach, 115–118
postoperative headache, 136–137 Subependymomas, 726–729
suboptimal operative technique, 134, 137 Suboccipital approach, 487–489, 612–615, 710–713
dura incision, 131–132 Suboccipital headache, 300
hemostasis, dural closure and mastoid Superficial cortical AVM, 107
occlusion, 132–134 Superior cerebellar artery (SCA)
microinstrument, 128, 129 cerebellopontine angle, 11, 14, 16, 18
patient positioning, 123–125 compression, 26
posterior skull base reconstruction, 132–134 Superior petrosal vein, 12, 29
semi-sitting position, 126–128 Suprameatal extension
sporadic ELST removal, 823–826 retrosigmoid approach
Retrosigmoid intradural inframeatal approach, 792–797 cerebellum, 33
Retrosigmoid suprameatal approach (RSMA) intraosseous structures, 32
approach/Samii approach neural structures, 32
CP angle levels, 453 petrosal vein, 33
extensive brain stem edema, 455, 459–460 tubercle removal, 32
hearing loss on right side, 454, 463–469 tumor removal, 34
intradural resection of petrous apex, 451–452 upper neurovascular complex, 32–33
Meckel’s cave and posterior cavernous Suprameatal meningiomas, 376, 378, 380
sinus, 455, 461–462 Sural nerve grafts, in facial nerve
slight hearing loss on right side, 454, 470–471 reconstruction, 856–857
Rhabdoid meningiomas, 81
Rosai–Dorfman disease, 103
T
Tanycytic ependymoma, 100
S Telovelar approach, to fourth ventricle, 64–65, 70
Saccular aneurysm, 107–109 Temporal bone, 40, 41
Samii approach/RSMA Teratomas, 105, 106
CP angle levels, 453 Thrombosed aneurysms, 773, 774
extensive brain stem edema, 455, 459–460 Tinnitus, 158, 298
hearing loss on right side, 454, 463–469 Torulosis, 88–89
intradural resection of petrous apex, 451–452 Transcochlear approach, 121
Meckel’s cave and posterior cavernous Transethmoid transsphenoidal approach, 799–800
sinus, 455, 461–462 Transitional meningiomas, 78
slight hearing loss on right side, 454, 470–471 Translabyrinthine (TL) approach
Sarcoidosis, 86, 87 advantages, 119
Schwannoma anatomical variations, 119
bilateral vestibular, 74, 76 history of, 4–5
cellular, 74–75 labyrinthectomy, 120
histology, 74 mastoidectomy, 119–120
jugular foramen (See Jugular foramen (JF) operative technique, 119
schwannomas) partial approach, 121
melanotic, 76 vs. retrolabyrinthine approach, 121
nonvestibular, 74 surgical outcome, 120–121
Schwannomatosis, 344 TL-retrosigmoid approach, 121–122
Secretory meningiomas, 78 TL-transtentorial approach, 122
Sellar region pathology transcochlear, 121
craniopharyngioma, 97–98 Transmastoid approach, 823–826
granular cell tumor, 97 Transpetrosal approach, history of, 5
pituicytoma, 96 Trigeminal nerve
pituitary adenoma, 93–95 arterial compression sites, 27–29
pituitary carcinoma, 95–97 retrosigmoid approach, 25
spindle cell oncocytoma, 96 and SCA, 26
xanthogranuloma, 98–99 venous compression sites, 30–31
884 Index

Trigeminal root anatomy meatal relationships, 37


anastomoses, 23 retrosigmoid exposure, 13
Meckel’s cave, 21 superior, 37, 41
motor rootlets, 21 vestibular schwannomas, 150
nerve hook, 23 Vestibular schwannomas (VS)
pons, 22, 23 audiometric and audiological
sensory root and motor rootlets, 22 examinations, 165
upper neurovascular complex, 20–24 bilateral, 74, 76
Trigeminal schwannomas brain stem, 214
classification, 547, 548 classification, 152–156
clinical presentation, 549 clinical presentation
dumbbell tumors, 559–568, 575–576 cisternal stage, 158–159
infratemporal extracranial location, 569–574 intracanalicular, 157–158
management, 549–550 MR imaging, 157, 158
neuroimaging, 547–549 tinnitus, 158
operative technique, 569, 577 conservative management, 162–163
outcome, 577 cystic VS
posterior fossa location, 553–558 capsule, 234–235
predominant location in middle cranial MR imaging, 233, 236–237, 239–240
fossa, 550–553 nerve adhesion, 238
radiotherapy, 577–578 proteolytic enzymes, 232
surgical treatment, 550 tumor debulking, 234
Trochlear schwannomas, 578–580 electronystagmography, 165
Tuberculous meningitis, 85–87 epidemiology, 147–148
Tumorlike dysontogenetic cysts, 105, 107 facial nerve
Tumor removal, multiple neurovascular complexes cystic tumors, 252
distorted nerves, 65, 69 delayed facial nerve palsy, 254
dural attachment, 65, 67 facial nerve palsy, 253–254
meningioma attachment, 65, 66 grading systems, 250
nerve displacement, 65, 67 intermediate nerve dysfunction, 254–255
routes, 65 intraoperative monitoring, 252
Type A trigeminal schwannomas, 550–553 NF-2, 252
Type B trigeminal schwannomas, 553–558 operative approach, 253
Type C trigeminal schwannomas, 559–568, 575–576 patient age, 252
Type D trigeminal schwannomas, 569–574 preoperative facial palsy, 252–253
preservation, 250
radiosurgery, 252
U reoperations, 252
Upper neurovascular complex taste disturbance, 255
arterial relationships, 26–29 tumor characteristics, 250–252
suprameatal extension, 32–33 giant (extension grade T5) VS
trigeminal root anatomy, 20–24 bone-window MRI, 207–210
vascular compression, 24–26 hearing loss, 201
venous relationships, 29–32 hemorrhagic complications, 201
neurological examination, 207
operated seven times, 245–246
V preoperative MRI, 202
Vascular compression RS approach, 204–205
lower neurovascular complex, 63–64 hearing
middle neurovascular complex, 49–57 classification, 255–256
upper neurovascular complex, 24–26 preservation rate, 262, 266–267
Vasculitis, 110–112 VS removal and preservation, 257–271
Venous relationships risk factors, 256
middle neurovascular complex, 44–47 history, of removal, 1–3
upper neurovascular complex, 29–32 hydrocephalus associated
Ventricular roof, 18 clinical management, 273, 282
Vestibular nerve communicating, 275, 277–278
cerebellopontine angle, 12 MRI, 274–275
compression syndromes, 53–54, 56 obstructive, 275
inferior, 37, 41 retrosigmoid approach, 280
Index 885

IAC opening retrosigmoid approach, 165


canal opening, 168 extrameatal tumor removal, 187–192
CT imaging, 168–169 high jugular bulb, 172–174
dura incision, 168 IAC opening, 168–173
MR imaging, 170 intracanalicular tumor removal, 187–189
posterior wall, 171 intracanalicular (extension grade 1)
intracanalicular (extension grade 1), 193–197 VS, 193–194
with large petrous bone involvement vascular considerations, 174–175, 183–187
brain stem, 214 surgical management
CT scan, 214 following previous radiosurgery, 287–291
debulking, 213 following previous surgery and
facial nerve, 213 radiosurgery, 292–296
massive destruction, 211–212 neurological symptoms, 284
large VS removal with primary debulking, 166–167 tumor removal, 163–165
of lateral IAC tumor adhesion/capsule
postoperative period, 198 incomplete tumor removal, 224–227
preoperative MRI, 198–200 intracanalicular VS, 217–221
medial VSs microsurgery, 215
caudal tumor extension, 229, 231 preoperative MRI, 215–216
cranial nerve dissection, 229 without obvious tumor capsule, 222–223
preoperative imaging, 228–229 Vestibulocochlear nerve
RS approach, 230 cerebellopontine angle, 13
molecular genetics, 148 foramen of Luschka, 44
neuroimaging, 156 infrafloccular exposure, 56
NF2 protein, 148–149 left cerebellopontine angle, 36
operative complications and management neural relationships, 42
cerebellar symptoms, 297–298 in petrosal surface, 10, 12
cranial nerves, 297 retrosigmoid exposure, 13
CSF leak, 300–302 von Hippel-Lindau syndrome (VHL)
meningitis, 302 with endolymphatic sac tumor, 93, 818, 827–830
mortality, 304 with hemangioblastoma, 83, 757
pneumocephalus, 300 VS. See Vestibular schwannomas (VS)
postoperative hydrocephalus, 302–304
sinus thrombosis, 299–300
suboccipital headache, postoperative, 300 W
tinnitus, 298 Waterhouse-Friderichsen syndrome, 85
vascular complications, 298–299 Willis’ circle, atherosclerosis of, 109
vestibular symptoms, 298 Wishart-type NF-2, 315
pathogenesis
growth pattern and rate, 150–152
nerve, 150 X
origin, 149–150 Xanthogranuloma, 98–99
site, 150 Xanthomatous metaplasia, 80
preoperative examinations, 165
radiosurgical management, 282–283
recurrent VS Y
after middle fossa removal, 248–249 Yolk sac tumor, 104
clinical management, 247
MRI, 241–242
preoperative MRI, 243–244 Z
prevalence, 240 Zellballen, 77

You might also like