Ventricular Tumors

Published on 26/03/2015 by admin

Filed under Neurosurgery

Last modified 26/03/2015

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 4 (1 votes)

This article have been viewed 6879 times

CHAPTER 138 Ventricular Tumors

Tumors of the ventricular system comprise a wide variety of both benign and malignant mass lesions located within the ventricular cavity or arising from neural structures forming the ventricular system. They can be grossly classified as intra-axial and extra-axial lesions. The clinical findings are dominated by obstructive hydrocephalus as a result of obliteration of the cerebrospinal fluid (CSF) pathway. Rarely, however, these tumors can metastasize within the CSF pathway.

Although there were some first descriptions of ventricular neoplasms by Shaw in 1854, it was Walter Dandy who had the most impact on descriptions of and treatment options for ventricular pathologies. During his career, he published more than 30 articles on these tumors and pioneered several surgical approaches to them that are still used today. In 1933, he dedicated a book to this subject, Benign Tumors in the Third Ventricle of the Brain: Diagnosis and Treatment.1

More publications on ventricular lesions followed, and with ongoing technical progress, interest in surgical approaches to the ventricular system was growing. As a result, many studies and books were published on this topic in the following years, including milestones such as Surgery of the 3rd Ventricle by Michael L. J. Apuzzo.2

The goal of this chapter is to present an overview of the pathology and classification of intraventricular tumors and describe the principles of diagnostic and therapeutic modalities. We focus on surgical indications and various operative approaches and corridors to the ventricular system and discuss tips and pitfalls of different surgical methods. The preoperative and postoperative medical management of patients with these tumors is highlighted, as well as the introduction of adjuvant therapies.

Pathology

Tumors of the ventricular system account for less than 1% of intracranial lesions,3,4 most of which are benign and slow growing. For this reason, symptoms develop at a late stage in many instances. According to Casotto and coworkers,5 these lesions can be categorized as primary or secondary. Primary, or true, ventricular tumors are those originating from the ventricular wall and extending strictly into the ventricular system. Paraventricular, or secondary, tumors are those originating from structures adjacent to the ventricular system. The major part of such neoplasms is located within the ventricular cavity, but a portion of the tumor is outside the ventricular system.5 Among the most common primary tumors are colloid cysts, choroid plexus papillomas, ependymomas, epidermoid and dermoid cysts, and craniopharyngiomas. Secondary tumors include meningiomas, gliomas, pituitary adenomas, and arachnoid cysts.6 According to Apuzzo,2 only around 10% of these masses are confined to the ventricular system.

Table 138-1 lists the variety of pathologic lesions encountered at our institution in a series of 143 patients who were systematically monitored after surgery. In addition to this patient population, the senior author (H.B.) has surgically treated a considerable number of individuals suffering from various ventricular tumors while working at other institutions. Because this additional patient group has not yet been monitored systematically in the same fashion as the population just mentioned, only a few cases from this second patient group have been included in this chapter.

TABLE 138-1 Histologic Diagnoses of Ventricular Tumors in a Single-Institution Consecutive Series of 143 Patients

HISTOLOGY n
Colloid cyst 19
Craniopharyngioma 16
Pilocytic astrocytoma 15
Pineocytoma (WHO grade I) 9
Cavernous malformation 8
Medulloblastoma 8
Metastatic tumor 7
Glioblastoma 6
Pineal cyst 6
Subependymoma 5
Fibrillary astrocytoma 4
Anaplastic ependymoma 3
Meningioma 4
Central neurocytoma 3
Pineocytoma (WHO grade II) 3
Anaplastic astrocytoma 2
Dermoid cyst 2
Ependymoma 2
Epidermoid cyst 2
Gliosis 2
Hemangioblastoma 2
Lymphoma 2
Neurenteric cyst 2
Pineoblastoma 2
Pituitary adenoma 2
Chordoid glioma 1
Gliosarcoma 1
Granular cell tumor 1
Plexus papilloma 1
Anaplastic plexus papilloma 1
Germinoma 1
Teratoma 1

We have divided the lesions in our follow-up series into five groups according to tumor location: 22% of tumors were located within both lateral ventricles, 18.5% occurred in the third ventricle, 19% were tumors of the hypothalamic and suprasellar region, 14% were located within the fourth ventricle, and 26% were found in the pineal and mesencephalic tectal region. Overall, tumors varied from a few millimeters to 60 mm in maximum diameter, but in 40% of patients, the range was 11 to 20 mm.

Clinical Features

Because of the deep location of the ventricular system within the brain and its proximity to important neural and vascular structures, ventricular tumors may cause a great variety of clinical symptoms. Grossly, symptoms may be divided into two types: symptoms caused by CSF obstruction and those caused by compression of certain neuronal structures. Because many ventricular tumors are benign and slow growing, they may reach considerable size before causing nonspecific symptoms.7 Common clinical symptoms include headache, vertigo, visual disturbances, difficulty concentrating, changes in personality, cognitive deficits, motor weakness, and epileptic seizures.3 Acute hydrocephalus causes headache, nausea, and vomiting. Additionally, memory deficits and gait disturbances may occur.

In some instances, special neurological symptoms such as hemianopia, hemiparesis, and hemihypoesthesia can develop. For instance, craniopharyngiomas may cause endocrine dysfunction with stunted growth, hypothyroidism, and hypogonadism. In adults, visual disturbances (caused by compression of the chiasm) and mental disturbances can frequently be observed.

Colloid cysts may cause chronic, acute, or intermittent hydrocephalus that becomes clinically manifested as paroxysmal posture with headache, nausea, vomiting, and disturbed consciousness.8 Some authors have described diplopia, psycho-organic syndrome, psychomotor retardation, and short-term memory deficits in patients with these lesions.9 Rarely, colloid cysts may even cause sudden death as a consequence of acute obstructive hydrocephalus.10

Tumors that infiltrate the midbrain may result in motor and sensory dysfunction, as well as Parinaud’s syndrome with vertical gaze palsy and nystagmus. This syndrome is caused by compression of the tectal region of the midbrain by tumors of the pineal region and tectal plate. Tumors of the fourth ventricle, such as medulloblastomas, most frequently cause cerebellar ataxia.

In our series, the duration of symptoms from onset to therapy varied from a few hours to 15 years. In almost half the study population, the duration of symptoms ranged from 2 to 8 weeks.

Imaging Studies

Presently, magnetic resonance imaging (MRI) is the “gold standard” imaging study for showing the tumor’s exact location, size, extent, vascularity, and relationship to adjacent structures, as well as the shape of the ventricles and surrounding brain and the amount of perifocal edema. However, many patients may undergo computed tomography (CT) as first-line diagnostic imaging. Colloid cysts appear on CT as isodense or slightly hyperdense lesions within the foramen of Monro. The structure is typically homogeneous, with the presence of calcifications being an exception. On MRI, a colloid cyst shows homogeneous hyperintensity on T1-weighted images and appears as a homogeneous area of hypointensity surrounded by a hyperintense rim on T2-weighted images.

Ependymomas commonly appear as homogeneous lesions on CT with contrast enhancement. Intratumoral cysts may be apparent, and approximately half of the tumors show calcifications. On MRI, ependymomas may have a variety of appearances. Frequently, they are seen as hypointense or isointense lesions with cystic portions. Intratumorally, necrotic areas, blood vessels, hemorrhage, or hemosiderin may be present. Contrast enhancement is heterogeneous.2 Subependymomas are well-circumscribed lesions that appear isodense or hyperdense on CT. On MRI they appear as lobulated structures, hypointense or isointense on T1-weighted images and hyperintense on T2-weighted ones. Contrast enhancement is weak or absent.11

In up to 75% of cases, choroid plexus papillomas appear as isodense or hyperdense well-circumscribed lesions on CT. Contrast enhancement is high, and calcifications or bleeding can frequently be observed. MRI studies show these lesions to be isointense on T1- and T2-weighted images, and contrast enhancement is homogeneous. MRI does not readily permit differentiation of choroid plexus carcinomas from plexus papillomas.2

In up to 90% of craniopharyngiomas, cystic and solid portions can be seen. On CT, the solid portions show contrast enhancement. On MRI, they appear as partially cystic and partially solid lesions with heterogeneous signal intensity. Contrast enhancement is high.4

Meningiomas are well-circumscribed lesions that appear hyperdense on CT. On MRI, they are isointense on T1-weighted images, with homogeneous contrast enhancement.

Low-grade astrocytomas appear as hypodense structures that show calcification in up to 15% of cases. Contrast enhancement is low or absent. On MRI, they appear as hypointense structures on T1-weighted images and as hyperintense lesions on T2-weighted images. Contrast enhancement is almost absent.

Anaplastic astrocytomas are heterogeneously contrast-enhancing lesions that only rarely show calcifications. On MRI, they appear as hypointense lesions on T1-weighted images and as hyperintense lesions on T2-weighted images, with variable contrast enhancement. Glioblastomas typically show irregular contrast enhancement on the periphery with necrotic areas in the center. On MRI, they appear as heterogeneous hypointense lesions on T1-weighted images and as hyperintense lesions on T2-weighted images.

Surgical Anatomy

Lateral Ventricle

The lateral ventricle is a paired C-shaped structure that lies deep within the cerebral hemispheres and wraps around the thalamus. It is divided into the anterior (frontal) horn, the body (cella media), the atrium (trigone), and the occipital and temporal horns.

Both lateral ventricles communicate with the third ventricle via the foramina of Monro and are surrounded by the following structures: septum pellucidum, thalamus, caudate nucleus, corpus callosum, and fornix (Fig. 138-1). The largest structure in connection with the lateral ventricle is the corpus callosum. It is the largest commissural fiber tract of the brain and is divided into four different parts: the rostrum, genu (anteriorly), body, and splenium (posteriorly).

The caudate nucleus, a part of the corpus striatum, is divided into a head, body, and tail. It covers the lateral border of the anterior horn and the body of the lateral ventricle and is part of the roof of the temporal horn. In close association, the stria terminalis runs between the caudate nucleus and thalamus and from the temporal horn to the body.

The fornix connects the hippocampus with the hypothalamus and various other structures. It originates in the alveus and runs as the fimbria along the medial aspect of the hippocampus on the floor of the temporal horn. Then it arches as the crus around the thalamus under the splenium and proceeds rostrally over the thalamus to join the crus on the opposite side, where it forms the body of the fornix lying beneath the corpus callosum. At the level of the foramen of Monro they separate again, curve ventrally around the foramen to form the columns that run posterior to the anterior commissure and into the hypothalamus, and finally reach the mamillary bodies.

The frontal horn is the part of the lateral ventricle lying anterior to the foramen of Monro. The medial wall is formed by the septum pellucidum and the lateral wall, by the head of the caudate nucleus. The roof and floor of the anterior horn are formed by the genu and rostrum of the corpus callosum.

The body of the lateral ventricle extends from the foramen of Monro backward to the junction of the corpus callosum, with the fornix located at the posterior end of the septum pellucidum. The lateral wall is formed by the body of the caudate nucleus, the medial wall by the septum pellucidum and the fornix, the roof by the body of the corpus callosum, and the floor by the thalamus.

The atrium lies between the body of the lateral ventricle and the temporal and occipital horns. The medial, superior, and lateral walls are formed by fibers of the corpus callosum, called the forceps major and tapetum. The tapetum itself is part of the sagittal stratum that also contains the optic radiation. The floor is formed by the collateral eminence (trigone) and the calcar avis, which represent bulges over the posterior end of the collateral sulcus and the calcarine sulcus, respectively.

The occipital horn extends posteriorly to the atrium. As in the atrium, the roof and the lateral wall are formed by commissural fibers of the corpus callosum (tapetum). The floor is formed by the collateral trigone and the medial wall by the bulge of the splenium and the calcar avis.

The temporal horn ends behind the amygdala. The roof is formed medially by the stria terminalis and laterally by the tail of the caudate nucleus and tapetum. In the floor, one finds the collateral eminence and the hippocampal formation. On the medial side above the hippocampus are the fimbria fornicis and the choroidal fissure.

The choroidal fissure, to which the choroidal plexus is attached, is a small cleft between the thalamus and fornix. It extends from the foramen of Monro backward along the body, atrium, and temporal horn of the lateral ventricle. The arterial supply of the choroid plexus is derived from the anterior and posterior choroidal arteries through the choroidal fissure. The ventricular veins drain into the internal cerebral veins or the basal veins. Important veins are the anterior septal and caudate veins, the thalamostriate vein, the superior choroidal vein, and the medial atrial vein, which drain into the internal cerebral vein. The inferior choroidal vein, the inferior ventricular vein, the veins of the amygdala and hippocampus, and the lateral atrial vein drain into the basal vein of Rosenthal.

Tumors of the lateral ventricle can originate from the periventricular white matter extending into the ventricle, from structures within the ventricle (such as the ependyma or the choroid plexus), or from ectopic tissue that has been trapped within the ventricle. The tumors commonly found here are astrocytomas, ependymomas, choroid plexus papillomas, neurocytomas, tuberous sclerosis tumors, and meningiomas. Their vascular supply varies according to the exact location, but their main feeder vessels are the anterior choroidal and posterior lateral choroidal arteries.

Third Ventricle

The third ventricle is located in the middle of the brain; it is connected to the lateral ventricles by the foramina of Monro and to the fourth ventricle by the aqueduct of Sylvius.

The lateral walls are formed by the thalamus and hypothalamus and separated by the hypothalamic sulcus (Fig. 138-2). In approximately 75% of patients, a connection between the lateral walls can be found in the upper part of the third ventricle and is referred to as the massa intermedia.

The floor of the third ventricle, which extends from the optic chiasm to the aqueduct of Sylvius, includes the infundibulum, the tuber cinereum, the mamillary bodies, and the posterior perforated substance. The roof of the third ventricle extends from the foramen of Monro to the suprapineal recess and is formed by five layers: the fornix (superior layer), the two layers of the tela choroidea (which envelope the vascular layer [velum interpositum]), and the choroid plexus layer. The vascular layer contains the internal cerebral veins and the medial posterior choroidal arteries. Because of its U shape, the junction of the thalamostriate vein with the anterior septal vein to form the internal cerebral vein at the posterior margin of the foramen of Monro is referred to as a venous angle and can be well identified on cerebral angiograms or with CT/MR angiography. In 30% of cases, this junction is located 3 to 7 mm behind the posterior border of the foramen of Monro.12 If such a posteriorly located venous junction is present, posterior enlargement of the foramen of Monro along the choroidal fissure can open a direct trajectory into the third ventricle. The two internal cerebral veins run close to each other up to the pineal recess, where they deviate from the midline and proceed along the superolateral surface of the pineal body to the deepest point of the splenium to form the vein of Galen. The plexus of the third ventricle is attached to the lower layer of the tela choroidea.

The anterior border of the third ventricle extends from the optic chiasm to the foramen of Monro and is formed by the optic chiasm, the lamina terminalis, the anterior commissure, and the column of the fornix.

The posterior border of the third ventricle extends from the aqueduct of Sylvius to the suprapineal recess. Between these structures are the posterior commissure, the pineal body (with its recess), and the habenular commissure.

Anatomically, tumors of the third ventricle can originate from three different regions: (1) from the periventricular, mainly sellar or suprasellar region, with expansion into the ventricle (e.g., craniopharyngiomas, pituitary adenomas, and optic gliomas); (2) from the ventricular wall (e.g., hypothalamic or thalamic gliomas, tumors of the pineal region, and ependymomas); and (3) less frequently from within the ventricle (e.g., choroid plexus tumors and meningiomas).

Fourth Ventricle

The fourth ventricle is located between the cerebellum posteriorly, the brainstem anteriorly, and the middle cerebellar peduncles. In the craniocaudal direction, the fourth ventricle extends from the aqueduct of Sylvius to the obex. This ventricle has a ventral floor formed by the dorsal surface of the lower midbrain (the pons and medulla inferiorly); a lateral wall formed by the superior, medial, and inferior cerebellar peduncles; and the lateral recess, in which the fourth ventricle communicates with the cerebellopontine angle. The superior part of the roof of the fourth ventricle is formed by the lingula, the superior medullary velum, and the fastigium, whereas the inferior part of the roof is formed by the tela choroidea, the choroid plexus, the inferior medullary velum, and the uvula and nodulus of the vermis.

To obtain a wide view into the fourth ventricle up to the aqueduct, the lower vermis must be elevated and retracted dorsally and superiorly. Retraction, however, must be gentle to avoid damage to the vermis. For this purpose, arachnoid dissection and sectioning of the tela choroidea are necessary. In practice, the so-called telovelar approach provides the best access inferiorly to the fourth ventricle. Lesions located within the fourth ventricle originate either from the part of the brainstem that forms the floor of the fourth ventricle, from the tela choroidea and choroid plexus, or from various parts of the cerebellum. A number of pathologies that do not primarily originate within the boundaries of the fourth ventricle may nevertheless extend into the ventricle: tumors of the dorsal medulla (foramen magnum tumors), tectal masses that expand inferiorly, or primary tumors of the cerebellar hemispheres that extend into the fourth ventricle.

The posterior inferior cerebellar artery (PICA) is the most important artery related to the fourth ventricle. Its course can be divided into five segments: the anterior medullary segment, the lateral medullary segment, the tonsillomedullary or posterior medullary segment, the telovelotonsillar or supratonsillar segment, and the distal segment. Primarily, the first three segments are the origin of the arterial supply to the lower brainstem and vermis, but these segments may also supply tumors located in the fourth ventricle. Therefore, it is particularly important to study the individual course of the PICA and to distinguish between branches of the PICA that exclusively supply the tumor and those that supply the brainstem and cerebellum. It is also important to analyze the vascular supply of the tela choroidea and choroid plexus because tumor-supplying branches may also emerge from these vessels. Tumors that extend within the fourth ventricle may not necessarily invade the brainstem as preoperative MRI may sometimes suggest, although on preoperative MRI the precise relationship between tumor and brainstem may not always be clear. Thus, some tumors with exophytic growth may consist of a limited tumoral and vascular pedicle and a large portion that compresses the brainstem but is not completely attached to the floor of the fourth ventricle. For instance, this is the case in ependymomas. The lateral extension of the neoplasm may be confined to the fourth ventricle or may extend beyond it. This is the case when a medulloblastoma, ependymoma, or glioma is expanding through the foramen of Luschka into the cerebellopontine cistern, where it may encase the rootlets of the caudal cranial nerves. Large tumors may also extend caudally beyond the level of the obex and fill the space dorsal to the superior cervical cord with or without invading the neuraxis.

Preparation

Most ventricular tumors are a surgical challenge because of their relatively rare occurrence, deep location, proximity to vital structures, and narrowness of the surgical field. For this reason, the procedure warrants meticulous preparation. Patients with tumors that have caused significant perilesional edema should first be treated with high-dose steroids for 1 or 2 days. Although surgery for most tumors of the lateral and anterior third ventricle can be performed with the patient placed in the supine or lateral position, tumors of the dorsal third ventricle and those located within the fourth ventricle are often removed more safely with the patient in the sitting position. For the latter patient group, it is advisable to exclude the presence of an open foramen ovale before surgery by performing a cardiac ultrasound examination. MRI data for neuronavigation should be available in most cases because this tool may greatly facilitate orientation during surgery. Likewise, an endoscope should be prepared for use during the microsurgical procedure as an assisting tool because it can easily visualize hidden corners of the ventricular system during the procedure. In patients with severe and acute CSF obstruction, a right external ventricular drain (bilateral in the case of obstruction of the foramen of Monro) should be placed just before the microsurgical procedure. If indicated and possible, endoscopic third ventriculostomy can be applied for the same reason. Surgical procedures for ventricular tumors should be performed by an experienced microneurosurgeon who is familiar with the complex ventricular anatomy and anatomic variations.

Surgical Removal of Lateral Ventricle Tumors

General Aspects

Because of their deep location, the lateral ventricles are not readily accessible for surgery. Either the cerebral hemisphere or the corpus callosum must be traversed to reach the ventricular cavity. To minimize parenchymal traumatization by surgical manipulation, natural pathways such as the cerebral sulci, fissures, and cisterns should be used. It was Yasargil and Abdulrauf who introduced this philosophic shift from transcerebral to transcisternal planning of surgical corridors.13

The key is to avoid damage to cortical or subcortical structures that control eloquent brain function (e.g., the motor and speech area or the visual system and their connecting fiber systems, including the optic radiation, pyramidal tract, and arcuate and superior longitudinal fasciculi, all of which could potentially lead to procedure-related morbidity).

There are two generally accepted avenues to the lateral ventricle: the transcortical and interhemispheric pathways. The decision to approach transcortically or via an interhemispheric route depends on the location and size of the tumor and varies on a case-by-case basis.1315 Currently, extirpation of lesions located in the lateral ventricle is in the domain of microsurgery, but an increasing number of studies have reported successful removal with endoscopic techniques.1620

Depending on the location of the tumor within the lateral ventricle, the lesion can be accessed from either an anterior, anterolateral, or posterior direction.3,1315,21,22 The surgical approach varies according to the exact location of the tumor within the ventricular cavity, the relationship between the tumor and intraventricular or surrounding anatomic structures, and the tumor’s size and pattern of vascular supply.

The craniotomy should provide sufficient working space and allow adequate visualization in both the anteroposterior and mediolateral directions. When using the interhemispheric approach, the pericallosal and callosomarginal arteries, as well as veins draining toward the superior sagittal sinus, must be preserved. Local ischemia and contusions caused by inappropriate retraction must be avoided. The cortical and callosal incisions should be kept to a minimum but, nonetheless, must be large enough to allow complete exposure of the pathology and visualization of the ventricular cavity.

The choice between the transcortical and transcallosal routes is controversial.2,3,23,24 Several authors believe that the frequency of postoperative seizures is higher when using the transcortical approach.15,21 More recent studies, however, have shown that this is not necessarily the case.2527

Approaches to the Lateral Ventricles

The neurosurgical pioneer Walter Dandy was the first to introduce the two elementary concepts of transcortical and interhemispheric approaches for removal of ventricular tumors.1,28 With growing knowledge of neuroanatomy and technical improvements, variations of these trajectories with different entry points have been developed and constantly modified.2123,2931 In the beginning, the transcortical approach was preferred for removal of tumors located within the lateral cavities, but recently there has been a shift toward the transcallosal route.

Recently, growing interest in the anatomy of the human white matter with the use of Joseph Klinger’s dissecting technique has improved understanding of the complex fiber system.12,32,33 Moreover, diffusion tensor imaging has made in vivo visualization of association, commissural, and projection fibers possible and has influenced neurosurgical decision making in approaching intraventricular tumors.3440 Direct visualization of the white matter fiber tracts gives the opportunity to choose the ideal trajectory and passage through the corpus callosum with minimal damage to neuronal structures on an individual patient basis. Preoperative planning can be further enhanced by using the Dextroscope technique, which offers fusion of the imaging studies to form a three-dimensional model. This tool can be used to explore a patient’s individual anatomy and provides the opportunity to digitally simulate various approaches before surgery.4145

The frontal horn of the lateral ventricle can best be accessed by the anterior interhemispheric transcallosal, by the anterior transcortical, or by the superior frontal sulcus approach. The body of the lateral ventricle is best accessed with the anterior interhemispheric transcallosal or the transcortical approach. The temporal horn of the lateral ventricle can be reached by the transsylvian and occipitotemporal sulcus approaches. Access to the atrium is best gained by the posterior interhemispheric transcingular and the intraparietal sulcus approaches. The occipital horn of the lateral ventricle can be reached with the posterior interhemispheric transcingular approach. The anterior and posterior transcortical and transcallosal approaches are suitable for access not only to the lateral ventricles but also to the third ventricle.

The following is a description of these approaches as used for accessing the lateral cavity. The specific aspects of third ventricular exposure with these approaches are discussed in Chapter 139.

The Anterior Transcallosal Interhemispheric Approach

This approach provides excellent exposure of the frontal horns and bodies of the lateral cavities and even gives access to the anterior part of the third ventricle. The standard position of the patient is supine with elevation and flexion of the head. The incision is made behind the hairline, usually in a bicoronal fashion. For the craniotomy, two bur holes are drilled on the contralateral side close to the sagittal sinus. Preoperative imaging studies with MR or CT venography and intraoperative neuronavigation are recommended for planning placement of the bur holes to avoid damage not only to the sinus but also to irregular bridging veins.46 In general, a craniotomy diameter of 4 to 5 cm is sufficient for tumor removal; however, it may be necessary to vary the size of the craniotomy based on the extent of the tumor and the depth at which it has to be resected. In the best case, the chosen trajectory preserves the bridging veins. Mobilization of the draining veins is important for gaining space, and their sacrifice should be avoided.

Brain relaxation can be achieved by osmotherapy and, most importantly, by release of CSF, mainly from the sulcal and callosal cisterns. This strategy is preferred over the use of rigid retractors so as to protect neural structures from pressure and tearing damage. Entrance to and advance within the interhemispheric fissure are achieved by blunt dissection with tailed cotton strips and balls.

When the corpus callosum is reached, an entrance of 10 to 15 mm is generally enough for removal of most lesions located in the frontal and middle parts of the lateral ventricles. The smoothest way of opening is to dissect along the plane of the fibers with fine-tipped bipolar forceps and small tailed cotton strips. When the ventricular cavity is entered, drainage of CSF leads to additional relaxation and can be augmented by incision of the septum pellucidum.

For better orientation in the presence of large tumor masses with severely distorted ventricles, the surgeon will find the thalamostriate vein located on the right side of the plexus in the right lateral ventricle and on the left side of the plexus in the left lateral ventricle. The further course of the plexus and vein leads to the internal cerebral vein, which serves as a guide for localization of the fornix and thalamus and thus acts as an important landmark.

Caution needs to be taken regarding the genu of the internal capsule, which reaches the surface of the ventricle lateral to the foramen of Monro where the thalamostriate vein turns medially toward the internal cerebral vein.4749 Recent publications show that complications related to this approach include mainly seizures, sensory-motor deficits, and visual and cognitive impairment.14,23,50,51

The Intraparietal Sulcus Approach

The intraparietal sulcus approach has been described for lesions of the medial and lateral portion of the trigone.29,51,54,55 The ventricle is explored by opening the intraparietal sulcus and then dissecting the parietal white matter toward the ventricle. Although this approach leads to direct access to the trigone, one should be aware that it poses a risk for possible neurological complications, such as visual field defects, apraxia, and acalculia.

The Transsylvian Approach to the Temporal Horn

The proximal transsylvian approach was developed by Yasargil in 1967 for the treatment of vascular and neoplastic lesions in the mesial temporal area. Later, this approach was used for selective amygdalohippocampectomy.56 The approach consists of pterional craniotomy and opening of the proximal part of the sylvian fissure with an incision about 3.0 to 5.0 cm long. Through a pial incision lateral to the M1 segment of the middle cerebral artery between the origin of the anterior temporal and temporopolar arteries, one can gain access to the temporal horn. In particular, limbic tumors of the amygdala, hippocampus, and parahippocampal area extending into the temporal horn can be removed with this approach without injuring the adjacent neocortex of the superior, middle, and inferior temporal gyrus and lateral temporo-occipital gyrus.

The Occipitotemporal Sulcus Approach

The occipitotemporal sulcus approach can be used to access lesions in the posterior part of the temporal horn.51 The occipitotemporal sulcus can be opened with a subtemporal craniotomy to allow access to the temporal horn. Placing a lumbar drain before surgery for release of CSF before opening the dura reduces the need for brain retraction. As is the case with other lateral transcortical approaches, there is a risk for postoperative visual field deficits.

Authors’ Surgical Technique

In principle, our technique does not significantly differ from descriptions of the approaches just discussed. Our most important access route to the lateral ventricles is the interhemispheric transcallosal approach. The skin incision is bicoronal, usually at the level of or slightly behind the coronal suture. We place two or three bur holes directly above the superior sagittal sinus. The craniotomy is generally performed on the right side but extends slightly to the contralateral side to completely expose the superior sagittal sinus. Small bur holes are placed obliquely in the skull at the margin of the craniotomy so that the dural flap (with its pedicle toward the sinus) can be sutured to the bone. This allows a straight-line view into the interhemispheric fissure directly at the level of the falx cerebri (Fig. 138-3).

We prefer wide dissection of the interhemispheric fissure in the anteroposterior direction. In many instances, this allows working in the depth of the lateral ventricle without the use of self-retaining retractors. Care is taken to penetrate the corpus callosum more laterally to avoid entrance into the ventricle on the wrong side of the septum pellucidum, which may be displaced by a large tumor. We use a probe of the ultrasonic aspirator that is sufficiently long and suitable for working in a deep and narrow surgical field. It must be borne in mind that intralesional bleeding can lead to a significant increase in tumor volume as a result of congestion and that the secondary increase in CSF pressure from the local obstruction may provoke a considerable increase in intracranial pressure that may additionally narrow the surgical field. This complication can be avoided by subsequent rapid tumor debulking with the aid of the ultrasonic aspirator, particularly if the tumor has a high tendency to bleed during the initial stage of the resection.

Common Tumors of the Lateral Ventricles

Ependymomas

The first description of ependymomas was made by Bailey and Cushing in 1926.57 Ependymomas account for about 2% to 9% of intracranial tumors, and they are more frequent in the pediatric population, with an incidence of 6% to 12% and a peak in the first 3 years of life. They are equally likely to occur in boys and girls.58 Ependymomas arise from ependymal cells, and thus they are found in the entire ventricular system, especially in the infratentorial region.5963 These tumors usually tend to grow slowly and show perivascular pseudorosettes and ependymal rosettes as key features on histologic examination. Most ependymomas are classified as grade II tumors according to the World Health Organization (WHO) classification of 2007, but there also exists an anaplastic variant, and progression to it from lower grades has been described.58,63,64 Therefore, histopathologic examination is recommended in any case. CT images of ependymomas appear isodense, with heterogeneous contrast enhancement. On MRI, they are well circumscribed, isointense to hypointense, and enhance heterogeneously.

Clinical symptoms arise from obstructive hydrocephalus, which leads to intracranial hypertension. In addition, cognitive changes are seen frequently, especially memory and attention deficits.65

Surgery plays a key role in the treatment of ependymomas because gross-total resection (Fig. 138-4) is associated with improved prognosis.66 If pathologic examination confirms malignant features and MRI studies show tumor remnants, radiotherapy should be discussed (Fig. 138-5).67

Subependymomas

Subependymomas were first described by Scheinker in 1945.68 Subependymomas are rare and account for just 0.2% to 0.7% of intracerebral tumors.69,70 They are benign, slow-growing tumors, often attached to the ventricular wall, but they do not invade surrounding structures. Therefore, this pathology is classified as WHO grade I. Although they most commonly arise within the fourth ventricle, they are located within the lateral ventricles in up to 40% of patients.58,71,72 Subependymomas show a male preponderance, with a male-to-female ratio of 2.3 : 1. Histologically, this tumor shows isomorphic nuclei in clustered patterns, a fibrillary matrix, and small cysts.58 On CT these lesions appear as hypodense to isodense lobular masses with cystic components that do not usually show contrast enhancement. On MRI, this pathology appears as a solid to cystic tumor.

Depending on location and progression, subependymomas become clinically evident by obstruction of CSF pathways, with signs of increased intracranial pressure typically causing headache, nausea and vomiting, gait disturbances, or vertigo (Fig. 138-6). Patients most commonly become symptomatic between the ages of 40 and 60. Because of the benign natural history of these tumors, they remain clinically silent in many cases and are often detected only during autopsy studies.69,72 At present, with easy access to cerebral imaging studies, a growing number of subependymomas are diagnosed incidentally.70,7274

image

FIGURE 138-6 Preoperative axial (A and B), coronal (C and D), and sagittal (E and F) magnetic resonance images (MRIs) of a 32-year-old woman (the patient shown in Fig. 138-3). The patient had severe headache, vomiting, and memory deficit. The solid, highly vascularized tumor, a subependymoma, was completely removed as shown on postoperative coronal (G) and sagittal (H) MRIs. There were no neurological or other deficits after the microsurgical procedure.

Management of patients in whom an incidental finding is diagnosed is still under debate. If clinical signs are absent, routine follow-up with imaging studies is recommended. If dynamic growth during follow-up or the development of clinical symptoms occurs, microsurgical removal of the mass is the treatment of choice. Because of the benign origin of this entity, radical extirpation defines cure. If imaging studies reveal residual tumor, further treatment options depend on the patient’s condition. If no further deficits have occurred, surveillance with MRI is justified. In patients with existing neurological deficits caused by the remnant mass, operative resection (or eventually radiotherapy) should be discussed.70,72,74

Central Neurocytomas

These rare tumors were first described by Hassoun and colleagues in 1982.75 This emphasizes the fact that this lesion was just recently determined to be distinct from other intraventricular pathologies and therefore has often been mistaken for another entity.75 Overall, central neurocytomas represent only 0.1% to 0.5% of all intracranial tumors.76,77 They generally occur in the lateral ventricles, with a predilection for the left frontal horn. Typically, they are detected between the second and fourth decades of life, with equal distribution between the sexes.7779 Histologic examination of central neurocytomas shows uniform round cells, perivascular pseudorosettes, and a honeycomb appearance, thus indicating their tendency to mimic the architectural patterns of intracerebral gliomas.77,80 They are classified as WHO grade II tumors.58

On CT and MRI, central neurocytomas appear hyperdense and hyperintense (with moderate contrast enhancement), respectively. The presence of necrosis, cysts, and calcifications in these tumors sometimes causes a heterogeneous appearance.77,81 Differential diagnosis includes oligodendroglioma, ependymoma, and papilloma arising from the choroid plexus.77,82

In most cases, central neurocytomas are slow growing and become clinically evident as a result of increased intracranial pressure from obstructive hydrocephalus that causes symptoms of headache, nausea, vomiting, gait disturbance, and cognitive deficits. Surgical removal remains the treatment of choice and defines cure (Fig. 138-7). Nevertheless, although central neurocytomas are generally considered to be benign, malignant courses with progression of remnant tumor and cerebral and spinal dissemination have been reported and associated with unfavorable clinical outcomes.8386 If radical removal cannot be achieved, adjuvant therapies such as radiation therapy and chemotherapy are options, but their benefits are still under debate. Reduction of tumor mass has been reported with both therapies.58,84,85,87,88

Low-Grade Gliomas

Fibrillary astrocytomas and subependymal giant cell astrocytomas (SEGAs) can occur frequently within the lateral ventricles. The fibrillary component is the most frequent variant of common astrocytomas. On histopathologic examination, nuclear atypia is present, including enlarged, irregular hyperchromatic nuclei. Additionally, mucoid cysts can occur within the matrix. This tumor shows diffuse and infiltrative growth; giant cell astrocytomas are found in patients suffering from tuberous sclerosis. This coexistence is well documented in the literature, and the neoplasm is typically located in the vicinity of the foramen of Monro and can extend to the cavity of the lateral ventricle.8992 Unfortunately, astrocytomas are closely associated with morbidity and mortality in this particular setting.91,93,94 SEGA is considered a benign and thus slow-growing tumor originating from the wall of the lateral ventricles. The lesion is structurally based on spindle and gemistocyte-like cells. In some cases, additional ganglion-like cells can be found. Clusters, perivascular pseudopalisades, and calcification are typical histologic characteristics. Consequently, SEGA was categorized as WHO grade I in 2007.58,89 The tumor typically occurs in patients younger than 20 years.

On CT, this lesion appears hypodense and well circumscribed and shows contrast enhancement. On T1- and T2-weighted MRI, the tumor appears as a hypointense mass but in some cases can have high signal intensity on T2-weighted images (Fig. 138-8A). In all cases the tumor shows enhancement when contrast material is administered (Fig. 138-8B), with a somewhat heterogeneous appearance caused by the irregular occurrence of calcification.91,92

Neurological symptoms are caused mainly by obstruction of CSF pathways, which leads to typical clinical symptoms related to the increased intracranial pressure; however, astrocytomas can bleed, which can result in intraventricular hemorrhage, depending on their location, and lead to sudden deterioration. If symptomatic lesions are diagnosed, surgical resection is the treatment of choice. With regard to incidental findings of ventricular gliomas, treatment is still controversial. In patients with small nonsymptomatic tumors, clinical follow-up with regular outpatient consultations and imaging studies is justified. If the lesion displays dynamic growth and can potentially displace CSF pathways, surgical removal in the absence of actual neurological deficits should be considered.89,91,92,95,96

High-Grade Gliomas

The tendency toward malignant change in astrocytomas correlates with the patient’s age. High-grade gliomas of the lateral ventricle may frequently arise from the corpus callosum, the septum pellucidum, or the thalamus and show a predilection for the anterior horn of the lateral ventricle.97

Anaplastic astrocytomas are malignant lesions (WHO grade III) with typical diffuse and infiltrating growth patterns (Fig. 138-9). The mean age at diagnosis peaks between 45 and 51 years. There is a slight male preponderance, with a male-to-female ratio that varies from 1.1 : 1 to 1.6 : 1.98,99 Histologic features include increased cellularity, nuclear atypia, and mitotic activity. When anaplastic transformation is in progress, the nuclei vary in number, size, and shape. Additionally, abnormal mitoses can be detected.

Glioblastoma is the most malignant tumor known in the central nervous system. This tumor accounts for 12% to 15% of all intracranial tumors and shows astrocytic differentiation in 60% to 75% of cases. The mean age at diagnosis is 45 to 75 years, with a male-to-female ratio of 1.28 : 1. Histopathologic study reveals the typical malignant characteristics, such as nuclear atypia, cellular pleomorphism, a high mitotic rate, and necrosis. These are WHO grade IV tumors.58,100

MRI may show the typical aspect of high-grade glioma, a central necrotic area surrounded by ring contrast enhancement. Whether to surgically resect a glioblastoma and to what extent depends on the patient’s neurological and neuropsychological status and the size, location, and extension of the tumor within the adjacent anatomic structures. In particular, if the fornices have been invaded, biopsy is preferred instead of extensive surgery.

Choroid Plexus Papillomas

Choroid plexus papillomas account for 0.5% to 0.6% of intracranial lesions in adults and for 2% to 5% of intracranial neoplastic lesions in children. They are among the most common tumors in children younger than 2 years. When these lesions are located in the lateral ventricle, the median age at diagnosis is 18 months, and there is no predilection to occur in either sex.58 In around 20% of pediatric cases, these tumors show malignant transformation; approximately half of them are located within the atrium. They are similar in aspect to the glomus of the choroid plexus, being formed by a single cell layer of cuboid or cylindrical cells surrounded by a thin fibrovascular structure. In the latest WHO classification, choroid plexus papillomas are classified as grade I. In imaging studies, these tumors frequently contain calcifications, hemorrhagic elements, or cysts. In some cases, choroid plexus papillomas may fill the ventricular cavity, whereas in others, the ventricle may be markedly dilated ipsilaterally.

Surgical removal is the treatment of choice (Fig. 138-10). It is helpful to expose and interrupt the proximal arterial supply from the choroidal artery to devascularize the tumor at an early stage of the procedure. Debulking is then continued with the aid of an ultrasonic aspirator. At the end of the procedure, the remaining portions of the apparently healthy choroid plexus are resected as well to ensure complete tumor removal.101104

Meningiomas

Meningiomas of the lateral ventricle originate from the stroma of the choroid plexus and arise at the tela choroidea. Ventricular meningiomas account only for 1% to 5% of all ventricular tumors and are located within the atrium in most cases.105,106 They may show calcifications and, occasionally, cystic degeneration.107 Usually, these tumors are highly vascularized (Fig. 138-11). The arterial supply stems from branches of the choroidal arteries, and they drain into the deep ventricular veins. Their histopathologic features and natural behavior are the same as those in any other location in the neural axis. Therefore, most intraventricular meningiomas are benign, slow-growing lesions (WHO grade I). Reports exist of a malignant course in some meningiomas, with recurrence and progression to atypical (WHO grade II) or anaplastic (WHO grade III) variants with the occurrence of metastases.108110

Buy Membership for Neurosurgery Category to continue reading. Learn more here