Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

J Neurooncol (2016) 130:351–366

DOI 10.1007/s11060-016-2138-5

TOPIC REVIEW

Microsurgical resection of pineal region tumors


Adam M. Sonabend1 • Stephen Bowden1 • Jeffrey N. Bruce1

Received: 9 February 2016 / Accepted: 2 May 2016 / Published online: 19 May 2016
Ó Springer Science+Business Media New York 2016

Abstract The extensive variety of possible histologic Background


subtypes makes it imperative to establish a tissue diagnosis
in patients with pineal region tumors. Management deci- The role of surgical resection in the treatment of pineal
sions regarding adjuvant therapy, prognosis, and follow-up region tumors has grown markedly over the last few dec-
strategies vary with the histologic diagnosis. Specialized ades due to enlightened appreciation of the value of
surgical and stereotactic techniques have evolved to pro- accurate tissue diagnosis combined with improved micro-
vide the neurosurgeon with an array of safe and effective surgical techniques. The pineal region is among the most
options for obtaining a tissue diagnosis. Advanced micro- complex areas of the brain with regard to pathology,
surgical techniques combined with improved preoperative necessitating a histologic diagnosis for optimal treatment
management and postoperative critical care methods have planning. The spectrum of distinct tumor types that may
made aggressive surgical resection a mainstay of man- arise in this location are divided into four categories,
agement. Aggressive surgical resection has resulted in including germ cell tumors, pineal parenchymal cell
excellent long-term prognoses for nearly all patients with tumors, glial cell tumors, as well as a wide variety of
benign tumors and a large percentage of patients with miscellaneous tumors and cysts [1]. All types exist on a
malignant tumors. However, pineal region surgery remains continuum from benign to malignant and mixed tumors of
fraught with potential pitfalls, and these favorable results more than one cell type may also occur [2–4]. In addition
are dependent on an advanced level of surgical expertise. to neoplastic disease, vascular lesions can also occur,
including cavernous malformations, arteriovenous malfor-
Keywords Pineal region tumor  Supracerebellar- mations, and vein of Galen malformations [5, 6].
infratentorial approach  Occipital-transtentorial approach  Research in pineal region tumors has been historically
Transcallosal-interhemispheric approach  Microsurgery  limited due to a relatively low incidence [only 1.2 % of all
Operative technique Central Nervous System (CNS) tumors] [7], the diversity
of pathology challenging the characterization of outcome
for patients by histological type, and differences in man-
agement paradigms. Conservative non-surgical manage-
ment with empiric radiation was historically favored
because of the highly specialized surgical demands in the
pre-microsurgical era and exquisite radiosensitivity of
Electronic supplementary material The online version of this some tumor types [8–13]. Unfortunately, this strategy of
article (doi:10.1007/s11060-016-2138-5) contains supplementary
material, which is available to authorized users. ‘‘blind radiation’’ led to unnecessary and potentially
harmful radiation exposure in a large percentage of patients
& Jeffrey N. Bruce with benign or radiation-resistant tumors [14–16].
jnb2@cumc.columbia.edu
Management of pineal region tumors now incorporates
1
Department of Neurological Surgery, Columbia University surgical resection in most cases. Evidence suggests resec-
Medical Center, New York, NY, USA tion is associated with better outcomes. Further, the greater

123
352 J Neurooncol (2016) 130:351–366

sophistication and diversification in treatment strategies Management


necessitates tumor tissue for proper tailoring of adjuvant
therapy, determining prognosis, and establishing a follow- Several adaptable approaches are available to patients with
up plan [1, 17–27]. pineal region lesions to confirm diagnosis, manage asso-
The first attempts at resecting pineal region lesions ciated symptoms such as hydrocephalus or mass effect, and
were attempted by Horsley, Brummer and Schloffer [28, facilitate cytoreduction. These management strategies
29], but were associated with considerable morbidity in should be tailored to the clinical presentation, with special
the absence of microsurgical techniques. Outcomes soon attention to associated clinical conditions such as hydro-
improved, with Oppenheim and Krause [30] performing cephalus, elevated tumor markers, metastatic disease,
the first successful removal of a tumor from the pineal advanced age, and general medical status of the patient
region in 1913. In 1921, Dandy [31] outlined a transcal- (Fig. 1).
losal approach to pineal tumors after developing the
technique in dogs. The technical repertoire continued to Ruling out pineal cyst
expand when, in 1926, Krause [32] reported modest
success with an infratentorial approach. Stein [25] suc- It is important to rule out the presence of a pineal cyst, par-
cessfully modified Krause’s infratentorial approach and ticularly for pineal region lesions found incidentally, as cysts
incorporated microsurgical techniques, ushering in the have been found in 1.0–4.0 % of patients undergoing brain
modern era of pineal surgery in 1971. Additional supra- MRI [42]. Pineal cysts may mimic pilocytic astrocytomas
tentorial approaches have since been refined and added to radiographically, though tumors can be distinguished clini-
the surgical repertoire [1, 33–38]. The largest series of cally by their progressive and symptomatic nature. Cysts are
pineal region surgery published over the last 15 years generally\2 cm in greatest dimension [42], with a contrast-
have demonstrated that resection of these lesions is pos- enhancing rim representing compressed pineal gland tissue.
sible, with morbidity and mortality rates ranging between They are benign, normal variants of the gland which are
1–20 and 0–10 %, respectively, and rates of gross total generally asymptomatic and do not require treatment unless
resection between 49 and 91 % (Table 1) [39–41]. they are causing aqueductal obstruction.

Table 1 Literature overview of microsurgical series for pineal region tumors in the last 20 years (modified with permission from [39])
Authors Year n Approach Age Pathology GTR Mortality Major Permanent
group (%) (%) morbidity (%) minor
morbidity (%)

Bruce and Stein [71] 1995 160 ITSC A and P All 45 4 3 19


TCIH
OTT
Chandy and Damaraju [112] 1998 48 ITSC A and P ‘‘Benign 55 0 NA NA
OTT lesions’’
Kang et al. [113] 1998 16 OTT A and P All 37.5 0 0 19
ITSC
TCIH
Shin et al. [114] 1998 21 OTT A and P All 54.5 0 0 5
Konovalov and Pitskhelauri [4] 2003 201 OTT A and P All 58 10 NA [20
ITSC
Hernesniemi et al. [40] 2008 119 ITSC A and P All 88 0 1 4.9
OTT
Bruce [39] 2011 128 ITSC A and P All 49 2 1 NA
TCIH
OTT
Qi et al. [41] 2014 143 OTT A and P All 91.6 0.7 3.5 5.6
TCIH transcallosal interhemispheric, ITSC infratentorial supracerebellar, OTT occipital transtentorial, TT transcortical transventricular, NA not
available, A Adult, P pediatric

123
J Neurooncol (2016) 130:351–366 353

Fig. 1 A treatment algorithm


for the approach to the patient
with a pineal region lesion,
including consideration of the
presence of hydrocephalus and
germ cell markers
(Abbreviations: XRT/Chemo =
external radiotherapy and
chemotherapy, STR = subtotal
resection, GTR = gross total
resection)

Management of hydrocephalus pathognomonic for malignant germ cell elements and


precludes the need for surgical resection [45–47]. CSF
Obstructive hydrocephalus, present in most patients on levels are more sensitive than serum levels in detection [2,
presentation, can be managed in several ways. Symp- 45, 47]. a-Fetoprotein indicates the presence of fetal yolk
tomatic patients are best managed with a stereotactic-gui- sac elements and is associated with endodermal sinus
ded endoscopic third ventriculostomy to gradually reduce tumors, embryonal cell carcinomas, or immature teratomas
intracranial pressure and resolve symptoms prior to tumor [45, 47–52], whereas b-human chorionic gonadotropin,
resection [43]. While consideration may be given to ven- produced by trophoblastic elements, indicates the presence
triculoperitoneal shunting, endoscopic third ventricu- of choriocarcinomas, embryonal cell carcinomas, or ger-
lostomy is preferable as it eliminates potential minomas [45, 47–49, 51, 53–56]. Though the presence of
complications such as infection, overshunting, and peri- germ cell markers is confirmatory for a malignant germ cell
toneal seeding of malignant cells. Mildly symptomatic tumor, the absence of germ cell markers should be inter-
patients in whom gross total resection is anticipated will preted cautiously because it does not rule out the presence
benefit from a ventricular drain placed at the time of sur- of a germinoma or embryonal cell carcinoma. Similarly, if
gery [20, 44]. Mild asymptomatic hydrocephalus fre- a-fetoprotein is elevated in the presence of a germinoma, it
quently resolves without the CSF diversion following is likely that an embryonal cell carcinoma or endodermal
resection of a pineal mass, as the third ventricle is com- sinus tumor is present as part of a mixed tumor [57].
municated with the quadrigeminal cistern or aqueductal The clinical utility of these tumor markers to the neu-
compression is relieved. rosurgeon is twofold. They are useful in establishing a
baseline post-operatively to monitor treatment response or
Tumor markers and non-operative diagnosis detect recurrence. More importantly, they reliably indicate
the presence of a germ cell tumor, allowing those patients
The presence of a-fetoprotein or b-human chorionic to forego surgery entirely in favor of radiation and
gonadotropin, measureable in either serum or CSF, is chemotherapy for these particularly sensitive tumor types.

123
354 J Neurooncol (2016) 130:351–366

Surgery applicable. There are, however, some scenarios in which


one method is strongly preferred. Patients with known
Anatomical considerations primary systemic tumors, multiple lesions, or other
comorbidities that increase surgical risk are good candi-
The pineal gland is an encapsulated structure that resides in dates for stereotactic biopsy [44, 45]. Radiographic evi-
a deep position near the geometric center of the brain. The dence of brainstem invasion might favor stereotactic
pineal gland is essentially an extra-axial structure, a feature biopsy, but the degree of invasion seen radiographically is
that facilitates the establishment of a surgical plane unreliable and may not reflect a dissectible tumor capsule
between adjacent structures during resection. The pineal at surgery. Open resection has the advantage of facilitating
gland is bordered by the posterior commissure ventrally, the collection of larger amounts of tissue. Extensive tissue
the habenular commissure dorsally, and the corpus callo- sampling is particularly desirable for pineal region lesions
sum superiorly (Fig. 2) [58]. The velum interpositum, given their frequent heterogeneity and mixed cell popula-
which incorporates the internal cerebral veins and choroid tions, as histologic differences may be subtle even to
plexus, lies in close proximity to the gland. The basal veins experienced neuropathologists. The clinical benefit that
of Rosenthal join the internal cerebral veins to form the comes with reduction of the tumor burden is another
vein of Galen before draining into the straight sinus. The advantage of open resection. Resection is usually complete
orientation of the deep venous system relative to the gland and curative for the third of tumors that are benign
is variable and has consequences for surgical planning (Table 2) [44, 61, 71]. The clinical advantage gained by
(Fig. 3). The more common superior and dorsal orientation debulking is less apparent with malignant tumors; however,
is most conducive to the infratentorial supracerebellar anecdotal evidence favors more radical resection when
approach. The less common inferior and ventral location is possible in order to improve the response to adjuvant
more conducive to a supratentorial approach. The gland therapy [34, 44, 71–73]. Patients with mild hydrocephalus
receives its blood supply from branches of the medial and gain a particular benefit with open surgery, as total resec-
lateral choroidal arteries through anastomoses to the peri- tion may preclude the need for shunting [20, 44].
callosal, posterior cerebral, superior cerebellar, and Two options are available if the decision to pursue
quadrigeminal arteries [46, 59]. biopsy is made. Stereotactic biopsy offers the advantages
Most masses in the pineal region originate infratentori- of relative ease of performance, minimal anesthesia, and
ally and expand into the posterior third ventricle. With minimal risk of complications [19, 23, 74–76]. There
further progression, they may extend into the thalamus or remains a risk of hemorrhage via several mechanisms,
posteriorly over the dorsal surface of the quadrigeminal including bleeding in highly vascular tumors, damage to
plate. Malignant tumors in this region, particularly when the deep venous system, and bleeding into the ventricle
they are of glial origin, can invade the midbrain and tha- [20, 44, 75, 77]. However, several large series have vali-
lamus, ultimately determining the tumor’s resectability. dated the relative safety of this technique despite these
lesions’ hazardous location [19, 74]. Neuroendoscopic
Resection versus biopsy biopsy has become an alternative to stereotactic biopsy
recently, taking advantage of flexible endoscopes to
Given the pathologic diversity found in this region, a tissue simultaneously perform a third ventriculostomy and biopsy
diagnosis is necessary to optimize a treatment plan through the same burr hole. This approach carries similar
including choice of adjuvant therapy, postoperative bleeding risks and, perhaps more significantly, is also
workup, and estimating prognosis [4, 44, 60–63]. CSF subject to inadvertent sampling bias.
cytology and radiographic characteristics may provide
insight into these decisions, but are not sufficiently sensi- Craniotomy techniques
tive to supplant tissue diagnosis [64–69]. The only excep-
tion to the rule of tissue diagnosis is the presence of Several variations exist for approaches to the pineal region,
malignant germ cell markers which obviate the need for generally categorized as either supratentorial or infraten-
chemotherapy and radiation therapy without histologic torial [38, 44, 61]. Supratentorial approaches include the
confirmation [44, 69, 70]. transcallosal interhemispheric, occipital transtentorial, and
Tissue diagnosis can be achieved by either biopsy or the less common transcortical transventricular [34, 44, 78,
open surgical resection. Careful consideration of the 79]. The infratentorial approach most commonly employed
patient’s clinical presentation, the tumor’s radiographic is the infratentorial supracerebellar [25, 80]. The choice
characteristics, and the surgeon’s experience with each between these depends mainly on the surgeon’s experience
method should be considered, as neither is universally and comfort, as many of these approaches are

123
J Neurooncol (2016) 130:351–366 355

Fig. 2 Sagittal (a) and dorsal (b) drawings of pineal region anatomy (modified with permission from Diane Abeloff; from [90])

interchangeable. There are several caveats: large tumors convergence of the vein of Galen and internal cerebral
that extend above the tentorium or laterally to the trigone veins, where they can interfere with tumor removal.
of the lateral ventricle are generally most amenable to the Most pineal tumors lie infratentorially and in the mid-
field of view from a supratentorial approach [44]. These line, giving the infratentorial supracerebellar approach
greater exposures come at the cost of working around the several natural advantages [44, 80]. One key benefit comes

123
356 J Neurooncol (2016) 130:351–366

with the sitting position, whereby gravity allows both the


cerebellum and the tumor to drop downward, naturally
enlarging the exposure while minimizing the need for
retraction. Further, this facilitates dissection of the tumor
off the deep venous system and velum interpositum, which
is often the most technically challenging portion of tumor
dissection. Second, this approach allows for a midline
trajectory. Lastly, the deep venous system lies superior to
the mass, thus shielding it through the majority of the
dissection. While offering many advantages, including the
possibility of removal of lesions extending anteriorly into
the third ventricle, the infratentorial supracerebellar
approach is less optimal for lesions with significant
supratentorial or lateral extension.

Patient positioning

Numerous patient positions can be employed, including the


sitting, lateral, and prone positions, with each having its
respective advantages and disadvantages (Fig. 4).

Sitting position

The sitting position is the preferred set-up for the


infratentorial supracerebellar approach (Fig. 4a) [44, 78].
Gravity minimizes the pooling of blood in the operative
field and facilitates dissection of the tumor from the deep
venous system. The risks inherent to this position include
air embolism, pneumocephalus, and subdural hematoma
associated with cortical collapse. These can be anticipated
and managed with proper precautions [44, 81]. End tidal
CO2 is a sensitive indicator of air emboli. Precordial
Doppler monitoring is another method of detecting these
Fig. 3 Magnetic resonance images demonstrating the variable rela-
tionship between the deep venous system and pineal region tumors.
small amounts of air entering the venous system and should
a The deep venous system is superior and dorsal to the tumor. This is be used. A central venous catheter can be used to remove
the most common configuration and is more conducive to the entrapped air.
infratentorial supracerebellar approach. b The deep venous system is The patient is brought to a sitting position from supine
inferior and ventral. This is less common but more conducive to a
supratentorial approach (from [115])
by manipulating the table while an assistant stabilizes the
patient’s head. A reverse table capable of being lowered
close to the floor is ideal. The head is flexed such that the
Table 2 Extent of resection for 181 Consecutive Pineal Region tentorium is approximately parallel to the floor. The
Surgeries at the New York Neurological Institute (1990–2014) (rep-
patient’s legs should be elevated to assist venous return.
rinted with permission from [39])
The surgeon should ensure at least two fingerbreadths of
Biopsy Subtotal Radical subtotal space between the patient’s chin and sternum to avoid
resection resection/gross total
resection airway compromise and impairment of venous return. A
three-pin vise-type head holder or skull clamp immobilizes
Benign 2 5 70 the head. A Greenberg self-retaining retractor or analogous
Malignant 12 29 63 system is attached to assist with cerebellar retraction and
Total 14 (6 %) 34 (19 %) 133 (73 %) hold cottonoids. The microscope should be balanced in a
Radical subtotal resection = no visible tumor at surgery or on post- position to allow direct horizontal view of the operative
operative MRI but tumor was not well encapsulated field.

123
J Neurooncol (2016) 130:351–366 357

Fig. 4 Patient positioning in preparation for pineal region tumor surgery. a Sitting position, b three-quarter prone position, c prone position,
d concorde position (a–c from [79]; d from [116])

Lateral and three-quarter prone positions elevated to facilitate venous return. This position better
suits the more posterior approaches, such as the occipital
The lateral decubitus position is generally used with the transtentorial, rather than the more vertical transcallosal
patient lying on their right side, such that the dependent, approach, as the nondominant hemisphere is largely
nondominant right hemisphere is relaxed down away from retracted by gravity and the horizontal operative plane
the falx [82]. For most approaches, the head should be reduces surgeon fatigue.
positioned approximately 30° above horizontal in the
midsagittal plane. In contrast, the occipital transtentorial Prone position
approach requires that the head should be positioned with
the patient’s nose rotated 30° towards the floor. The prone position is a simple and safe option for supra-
A more accommodating variant of the lateral position is tentorial approaches and comes with several advantages
the three-quarter prone position (Fig. 4b) [83]. The legs are (Fig. 4c) [44, 80]. First, it is generally a comfortable posi-
flexed and pressure points are minimized with pillows, an tion for the surgeon’s hands, though the elevated operative
axillary roll under the patient’s right axilla, and a roll under field makes it difficult to sit. Second, it is particularly
the left thorax. A three-pin Mayfield head-holder supports useful for two surgeons to work together, as it allows for
the head, which is slightly extended and rotated to the left placement of an operative microscope with a bridge setup
at a 45° oblique angle, again such that the nondominant and thus simultaneous binocular vision. Rotating the head
hemisphere is dependent. The patient is secured with straps 15° away from the craniotomy side yields a common
so that the table can be rotated during the operation to variation known as the Concorde position (Fig. 4d) [84].
improve exposure when necessary. The legs and feet are The prone position is less practical for the infratentorial

123
358 J Neurooncol (2016) 130:351–366

supracerebellar approach due to the steep angle of the Fig. 5 Infratentorial supracerebellar approach. a Sagittal diagram c
tentorium, but nonetheless remains a viable option for the showing a the initial trajectory in line with the vein of Galen and b the
adjustment downward in line with the central axis of the tumor after
surgeon pursuing a supratentorial route to resection. opening the arachnoid over the quadrigeminal plate. b Operative
photograph showing the dorsal surface of the tumor after opening of
Approaches the arachnoid and division of the precentral cerebellar vein.
c Diagram of the exposure seen in b. d Operative photograph with
a view into the third ventricle after excision of the tumor. e Diagram of
Infratentorial supracerebellar approach the tumor bed seen in d (from [116])

The infratentorial supracerebellar approach (Fig. 5, Video 1)


is usually performed with the patient in the sitting position
[44, 61, 80]. If necessary, a ventricular drain can be placed cerebellum more inferiorly and posteriorly. Deeper adhe-
in the trigone of the lateral ventricle through a Frazier burr sions and bridging veins can be divided when they become
hole. Exposure is begun with a midline incision extending visible near the anterior vermis with further cerebellar
from just above the torcula and external occipital protuber- retraction. With the retractor in place, the opalescent
ance down to the level of the C4 spinous process. The cut is arachnoid covering the pineal region can be seen.
brought through the nuchal ligament of the suboccipital Because of the range of angles and depths encountered
musculature. The craniotomy is centered just below the along the operative trajectory, a microscope with a variable
torcular and must be extensive enough to provide room for objective is preferred to facilitate the use of long instru-
the surgical instruments and adequate light from the oper- ments. A freestanding armrest assists the surgeon and
ating microscope. Craniotomy with replacement of the bone prevents fatigue.
flap reduces the incidence of postoperative aseptic menin- The arachnoid overlying the quadrigeminal plate is
gitis, fluid collections, and discomfort, and is thus preferred sharply opened. This is generally an avascular plane
over craniectomy. Slots are drilled over the sagittal sinus, requiring minimal cautery. The precentral cerebellar vein,
above the torcula, and over the lateral sinuses bilaterally. A identified as it courses from the anterior vermis to the vein
final slot is made 1 or 2 cm above the foramen magnum in of Galen, should be dissected, cauterized, and divided.
the midline. The slots are connected with a craniotome, Although this vein can be taken without substantial risk,
allowing the bone flap to be elevated. Care must be taken to the sacrifice of any other veins of the deep venous system is
remove sufficient bone above the transverse sinus to ensure not advisable. The retractor is then adjusted to visualize the
a clear view along the tentorium. Bone edges are waxed and inferior portion of the tumor. The trajectory of the micro-
all venous bleeding is controlled to avoid air emboli. scope is re-oriented to be in line with the central axis of the
A curvilinear dural opening is done between the lateral tumor, away from the initial plane parallel to the tentorium,
aspects of the craniotomy with a concavity directed supe- where it would otherwise lead to direct encounter with the
riorly. The dural flap is reflected upward and anchored with vein of Galen (Fig. 5a). The retractor can often be removed
slight tension. Excess retraction obstructs the sinuses and following removal of CSF once the quadrigeminal cistern
should be avoided. The inferior dura acts to support the is widely opened.
cerebellar hemispheres. If the posterior fossa appears tight, Once the posterior surface of the tumor is exposed, the
fluid can be removed through a ventricular drain or by central portion is cauterized and opened with a long-han-
opening the cisterna magna. dled knife or bayonet scissors (Fig. 5b, c). Specimens can
Opening of the infratentorial corridor is followed by be taken from within the capsule and sent for frozen
cautery and careful division of the arachnoidal adhesions intraoperative pathology consultation for diagnosis. The
and midline bridging veins between the dorsal surface of accuracy of frozen tissue diagnosis is low due to hetero-
the cerebellum and the underside of the tentorium and geneity and mixed cell tumors, however, and this should be
straight sinus using microsurgical techniques. The presence taken into consideration during intraoperative decision
of extensive collateral circulation minimizes complications making. Following initial sampling, the tumor is internally
from venous sacrifice; however, to minimize risk, it is debulked using a variety of instruments including suction,
desirable to spare any veins found laterally [85]. Cauter- cautery, tumor forceps, or an ultrasonic aspiration device if
izing the bridging veins and dividing them midway can necessary. Most tumors are soft and are amenable to
minimize bleeding from the sinus. Following division of debulking with a large-bore Japanese-style suction with
these attachments, the cerebellum drops away from the variable control. As the tumor is decompressed, the capsule
tentorium, providing an excellent corridor with minimal can be separated more easily from the surrounding thala-
brain retraction. The exposed dorsal surface of the cere- mus and midbrain. Most of the vessels along the wall of the
bellum can be protected with padding such as Telfa, and a capsule are choroidal vessels and need not be preserved.
brain retractor can be temporarily used to bring the Capsule dissection continues until the third ventricle is

123
J Neurooncol (2016) 130:351–366 359

123
360 J Neurooncol (2016) 130:351–366

encountered. The tumor is then carefully dissected inferi- where the tumor resides in the third ventricle [79, 89, 90];
orly off the brainstem. This is often the most difficult however, a wide craniotomy, provides flexibility in deter-
portion of the tumor dissection but can be facilitated by mining the corridor and avoiding bridging veins. The
retracting the tumor superiorly and dissecting it bluntly off craniotomy is generally centered over the vertex to avoid
the brainstem under direct vision. Last, the superior aspect manipulation of the occipital lobe. Burr holes are made
of the tumor is removed after separating the attachments over the sagittal sinus, both anteriorly and posteriorly, and
along the velum interpositum and the deep venous system. a craniotome is used to turn a generous craniotomy. The
These can be carefully cauterized and sharply dissected, craniotomy should extend 1–2 cm to the left of the sagittal
although injury to the deep venous system can be difficult sinus. Bleeding encountered from the sagittal sinus can be
to control and must be avoided. controlled with hemostatic agents.
The degree of tumor invasion dictates the intraoperative The dura is opened in a U-shaped fashion and reflected
decision regarding the extent of resection. Direct brainstem medially toward the sagittal sinus. The bridging veins are
invasion is the extreme example in limiting resection, and inspected, and an approach is chosen that will sacrifice the
the extent pursued is determined by the surgeon’s judg- minimum number of veins. Sufficient exposure can rarely
ment. Analyses of retrospective series have found an be achieved without sacrifice of at least one bridging vein,
association between extent of resection and survival for although sacrifice of more than one should be avoided if
pineal cell tumors including pineoblastoma, but other possible. Because these tumors are deeply seated, even a
smaller studies have failed to confirm this association [86– small opening provides a wide angle of deep exposure. The
88]. Our experience has shown that gross total resection exposed hemisphere is covered with Bicol or Telfa and a
also decreases the risk for postoperative hemorrhage and retractor system such as the Greenberg retractor is used to
thus, should be pursued in the absence of contraindications. frame the opening. Two retractors are placed to draw the
Following complete tumor resection, the surgeon should parietal lobe back in a gentle arc. These retractors can be
have a comprehensive view into the third ventricle used along the falx, which may be divided inferiorly to
(Fig. 5d, e). Flexible mirrors are used to examine the provide further retraction. This is generally a nonvascular
inferior portion of the tumor bed in order to verify the corridor and contains few adhesions between the falx and
extent of resection and to avoid leaving any blood clots. the cingulate gyrus.
Careful attention must be given to hemostasis [44, 81] with The corpus callosum is identified with the operating
direct but careful cautery preferred over the use of hemo- microscope by its stark white appearance. The pericallosal
static agents which can float into the ventricle and cause arteries are identified as a paired structure running over the
CSF obstruction. If absolutely necessary, long strips of corpus callosum. These arteries can be retracted either
Surgical draped over the surface of the cerebellum and together to one side or with separate retractors to each side.
covering the tumor bed can provide hemostasis with small The opening into the corpus callosum, centered over the
risk of migration and subsequent obstruction. maximal bulge of the tumor, is generally about 2 cm (see
Once hemostasis is obtained the dura is closed in as Fig. 6a). Whereas posterior openings in the splenium have
watertight a manner as possible. The bone flap is plated been performed routinely without reported deficits, we
into place to reduce postoperative pain and inflammation. have generally avoided this approach to minimize the risk
The patient should be extubated with a reasonable degree of a disconnection syndrome. The corpus callosum is
of head elevation to avoid shifting the decompressed brain generally thin and is opened by gentle suction and cautery.
within the cranial vault. The lateral extent of this opening is a balance between
exposing the tumor and avoiding damage to the pericallosal
arteries. If necessary, the tentorium and falx can be divided
Transcallosal interhemispheric approach to provide additional exposure (Fig. 6b).
Once through the corpus callosum, the dorsal surface of
The transcallosal interhemispheric approach between the the tumor can be seen, and early identification of the veins
falx and hemisphere of the brain involves a corridor along of the deep venous system is necessary to prevent injury.
the parieto-occipital junction. Dandy’s early work recog- The importance of the deep venous system and the degree
nized the importance of the deep venous system and the of venous collaterals is a topic of anecdotal speculation, as
cortical bridging veins between the hemisphere and the consequences of venous sacrifice in the posterior fossa are
sinus. Any of the aforementioned patient positions can be not well-reported in the literature. Even so, there is some
used for this approach, although the prone or sitting posi- evidence that one vein can be sacrificed safely in several
tion is generally preferred. instances, but interruption of two would likely greatly
A linear incision is used on the scalp. Subsequent increase the risk of a devastating infarction [91]. Once the
positioning of the bone flap is variable and depends on tumor is exposed, it is debulked and then dissected as

123
J Neurooncol (2016) 130:351–366 361

Fig. 6 Transcallosal interhemispheric approach. a Overview of the corpus callosum opening. b Magnified view of the opening into the corpus
callosum. The falx and tentorium may be divided for further exposure (a from [79]; b from [117])

described previously. Leaving a ventricular drain in place retract the ipsilateral hemisphere. It can be removed as the
for 1 or 2 days is optional. operation progresses and CSF is gradually drained.
Under the operating microscope, the straight sinus is
Occipital transtentorial approach identified so that the tentorium can be divided adjacent to it
(Fig. 7a). Intraoperative Doppler ultrasound can be useful
The occipital transtentorial approach is a variation of the for localizing the straight sinus, and avoiding it during the
supratentorial approaches (Fig. 7, Video 2) [35, 92]. A tentorial opening. A retractor can be placed over the falx
three-quarter prone or lateral position is generally pre- for exposure. The falx can be divided to facilitate further
ferred. This approach to the pineal region uses an oblique retraction if necessary. At this point, the arachnoid over-
trajectory for lesions that are essentially midline and may lying the tumor and the quadrigeminal cisterns can be seen
therefore be disorienting to surgeons who are not familiar (Fig. 7b). Tumor removal proceeds as described earlier
with it. However, division of the tentorium provides while taking care to avoid injury to the deep venous sys-
excellent exposure of the quadrigeminal plate, thus making tem. Closure and hemostasis proceed similarly to methods
it particularly advantageous for tumors that extend described above.
inferiorly.
A linear scalp incision is used. A burr hole is placed in Transcortical transventricular approach
the midline over the sagittal sinus just above the torcula,
with a second burr hole 6–10 cm above this. A craniotome The transcortical transventricular approach uses a trajec-
is used to turn a generous craniotomy extending 1–2 cm to tory through the right lateral ventricle via a transcortical
the left of midline. Alternatively, it is feasible to place the incision [78]. This approach is rarely preferred because the
burr holes adjacent to the sagittal sinus without crossing the exposure is limited and the need for a cortical incision is
midline. If this option is chosen, it is important to get as undesirable. Obviously, an entry point should be chosen in
close as possible to the sagittal sinus to avoid a bony noneloquent cortex. Stereotactic guidance is often useful
overhang that limits the operative view. with this approach and may be desirable for a tumor that
Retraction of the nondominant occipital lobe is aided by extends into the lateral ventricle.
gravity, owing to the three-quarter prone position, and
further facilitated by the lack of bridging veins near the
occipital pole. Mannitol and ventricular drainage are useful Postoperative care
for relaxing the brain and minimizing the risk for hemi-
anopia from excessive occipital lobe retraction. Additional High potency corticosteroids should be maintained for the
brain relaxation is obtained by reverse Trendelenburg bed first few days, then tapered with improvement of the
positioning. A brain retractor is initially placed to gently patient’s condition [81, 89]. Additionally, seizure

123
362 J Neurooncol (2016) 130:351–366

Fig. 7 Occipital transtentorial approach. a Dividing the tentorium and falx can increase exposure of the pineal region, b tumor exposure after
dividing the tentorium, c operative photo of the diagram in b. d Operative photo after tumor removal (from [117])

prophylaxis is recommended during early recovery for outcomes. A drain placed at the time of surgery should be
supratentorial approaches, but is not necessary long-term. removed or converted to a shunt in these first 72 h to
Lethargy and mild cognitive impairment are common, minimize infection risk. Lastly, MRI with gadolinium
making it difficult to evaluate neurological status in the contrast should be performed within 48 h to determine the
immediate postoperative period, particularly in patients extent of resection and guide future management decisions
with extensive subdural air as a result of the sitting posi- [93].
tion. In the first 72 h post-operatively, careful and frequent
neurological examinations are necessary. Any changes
should be investigated with appropriate radiographic Complications
imaging to rule out the possibility of hydrocephalus,
hemorrhage, or residual air. Shunt malfunction caused by Patients frequently experience ocular symptoms in the
air, blood, or operative debris is a frequent problem arising postoperative period, most notably impaired extraocular
during this timeframe. This is particularly worrisome movements, including limited upgaze and convergence
because deterioration and major morbidity can occur [37, 71, 81], and pupillary dysfunction, such as difficulty
rapidly. Early mobilization and ambulation, including focusing. These problems are often transient, though they
physical therapy and rehabilitation consults, improve can persist for several months. A residual mild limitation of

123
J Neurooncol (2016) 130:351–366 363

upgaze is not unusual but bears little clinical significance. techniques. Large case series in the last 20 years report
Otherwise, permanent impairment is rare. As with most operative mortality rates between 0 and 4 % and permanent
neurological deficits, their persistence and magnitude are morbidity in 0–5.6 % of patients (Table 1) [39–41]. Long-
proportional to the degree to which they were present term outcomes remain largely reliant on tumor histology
preoperatively [81, 94]. Similarly, ataxia is frequently and responsiveness to adjuvant therapy. Outcomes are
present but resolves within a few days after surgery. particularly variable with malignant tumors, but complete
More severe morbidity is rare but can be a sequela of surgical removal, excellent long-term follow-up, and
overzealous brainstem manipulation. This can lead to cog- probable cure can be expected for benign tumors [44, 45,
nitive impairment or, in its extreme form, to akinetic mutism. 61, 98–100].
Complications are more common in previously irradiated
patients, patients with invasive tumors, and those who were
progressively symptomatic preoperatively [81, 94].
One of the most devastating complications is hemor- Postoperative work-up
rhage into an incompletely resected tumor bed. Patients
with highly vascular, invasive tumors such as malignant Imaging for follow-up planning should include the afore-
pineal parenchymal tumors are at greatest risk [19, 81, 95]. mentioned brain MRI with gadolinium contrast within 72 h
Small hemorrhages can be managed conservatively, but a of surgery as well as a spinal MRI to look for spinal
large hemorrhage may require immediate evacuation. Such seeding in patients with pineal cell tumors, malignant germ
decisions must consider the possibility of obstructive cell tumors, and ependymomas [1, 44, 101, 102]. Spinal
hydrocephalus. Another potential vascular complication is imaging may be difficult to interpret early in the postop-
venous infarct, which can extend into the midbrain with erative period because blood clots or operative debris may
devastating consequences. This rare and unpre- mimic spinal metastasis. If the images are equivocal, serial
dictable complication is thought to be the result of venous images should be obtained before instituting spinal irradi-
insufficiency in a small subset of patients who cannot tol- ation. CSF cytology should be evaluated by lumbar punc-
erate the sacrifice of bridging veins in the cerebellum. ture postoperatively, although this has not been particularly
There are several approach-specific complications to helpful in predicting the metastatic potential of these
consider as well. Brain retraction and sacrifice of bridging tumors [11, 44, 51, 103–105]. Overall, the incidence of
veins in supratentorial approaches may lead to hemiparesis spinal seeding is low, and prophylactic spinal irradiation is
[81, 96]. In particular, retraction of the parietal lobe may not recommended unless there is clear radiographic evi-
cause sensory or stereognostic deficits on the opposite side dence of metastasis [20, 44, 48, 106–110]. The one pos-
[89]. Occipital lobe retraction during the transtentorial sible exception is in patients with highly malignant
approach can cause visual field defects [34, 89, 97]. pineoblastomas [44, 71, 111]. In addition to these imaging
Although disconnection syndromes have been reported studies, tumor markers, when present preoperatively,
with corpus callosum incisions, this has been rare in our should be measured in the postoperative period to serve as
experience, even when the splenium is divided [79, 89]. a baseline for detecting early recurrence or for monitoring
Complications related to the sitting position include response to treatment.
subdural hematoma, hygroma, and ventricular collapse [61,
71, 81]. These conditions are also commonly self-limited.
Air embolism is rarely a problem but can be anticipated Conclusion
intraoperatively by a drop in end-tidal carbon dioxide
levels or with Doppler monitoring. The extensive variety of possible histologic subtypes
Overall, pineal tumor patients are generally young and makes it imperative to establish a tissue diagnosis in
have relatively few medical problems. Consequently, the patients with pineal region tumors. Management decisions
incidence of medical complications such as cardiac or regarding adjuvant therapy, prognosis, and follow-up
respiratory problems is low. strategies vary with the histologic diagnosis. Specialized
surgical and stereotactic techniques have evolved to pro-
vide the neurosurgeon with an array of safe and effective
Surgical outcomes options for obtaining a tissue diagnosis.
Advanced microsurgical techniques combined with
Surgery in the pineal region is among the most arduous of improved preoperative management and postoperative
microsurgical challenges and good outcomes rely on the critical care methods have made aggressive surgical
expertise of individual surgeons. Outcomes have improved resection a mainstay of management. Aggressive surgical
dramatically with the advent of modern microsurgical resection has resulted in excellent long-term prognoses for

123
364 J Neurooncol (2016) 130:351–366

nearly all patients with benign tumors and a large per- 21. Hoffman HJ, Yoshida M, Becker LE, Hendrick EB, Humphreys
centage of patients with malignant tumors. RP (1994) Pineal region tumors in childhood. Experience at the
Hospital for Sick Children, 1983. Pediatr Neurosurg 21:91–103
(discussion 104)
22. Neuwelt EA (1985) An update on the surgical treatment of
malignant pineal region tumors. Clin Neurosurg 32:397–428
23. Pluchino F, Broggi G, Fornari M, Franzini A, Solero CL,
References Allegranza A (1989) Surgical approach to pineal tumours. Acta
Neurochir (Wien) 96:26–31
1. Bruce J (1993) Management of pineal region tumors. Neurosurg 24. Rout D, Sharma A, Radhakrishnan VV, Rao VR (1984)
Quart 3(2):103–119 Exploration of the pineal region: observations and results. Surg
2. Sano K (1983) Pineal region tumors: problems in pathology and Neurol 21:135–140
treatment. Clin Neurosurg 30:59–91 25. Stein BM (1971) The infratentorial supracerebellar approach to
3. Horowitz MB, Hall WA (1991) Central nervous system germi- pineal lesions. J Neurosurg 35:197–202. doi:10.3171/jns.1971.
nomas: a review. Arch Neurol 48:652–657 35.2.0197
4. Konovalov AN, Pitskhelauri DI (2003) Principles of treatment 26. Suzuki J, Iwabuchi T (1965) Surgical removal of pineal tumors
of the pineal region tumors. Surg Neurol 59(4):250–268 (Pinealomas and Teratomas). Experience in a series of 19 cases.
5. Fukui M, Matsuoka S, Hasuo K, Numaguchi Y, Kitamura K J Neurosurg 23:565–571. doi:10.3171/jns.1965.23.6.0565
(1983) Cavernous hemangioma in the pineal region. Surg 27. Hoffman HJ, Yoshida M, Becker LE, Hendrick EB, Humphreys
Neurol 20:209–215 RP (1984) Experience with pineal region tumours in childhood.
6. Fetell MR, Bruce JN, Burke AM, Cross DT, Torres RA, Powers Neurol Res 6:107–112
JM, Stein BM (1991) Non-neoplastic pineal cysts. Neurology 28. Zulch KJ (1981) Reflections on the surgery of the pineal gland
41:1034–1040 (a glimpse into the past). Gleanings from medical history.
7. Dolecek TA, Propp JM, Stroup NE, Kruchko C (2012) CBTRUS Neurosurg Rev 4:159–163
statistical report: primary brain and central nervous system 29. Borit A (1981) History of tumors of the pineal region. Am J
tumors diagnosed in the United States in 2005–2009. Neuro Surg Pathol 5:613–620
Oncol 14(Suppl 5):v1–v49. doi:10.1093/neuonc/nos218 30. Oppenheim H, Krause F (1913) Operative Erfolge bei
8. Abay EO 2nd, Laws ER Jr, Grado GL, Bruckman JE, Forbes Geschwulsten der sehhugel- und. Vierhugelgegend. Berl Klin
GS, Gomez MR, Scott M (1981) Pineal tumors in children and Wochenshr 50:2316–2322
adolescents. Treatment by CSF shunting and radiotherapy. 31. Dandy W (1921) An operation for the removal of pineal tumors.
J Neurosurg 55:889–895. doi:10.3171/jns.1981.55.6.0889 Surg Gynecol Obstet 33:113–119
9. Cummins FM, Taveras JM, Schlesinger EB (1960) Treatment of 32. Krause F (1926) Operative Frielegung der Vierhugel nebst
gliomas of the third ventricle and pinealomas; with special Beobachtungen uber Hirndruck und Dekompression. Zentrabl
reference to the value of radiotherapy. Neurology 10:1031–1036 Chir 53:2812–2819
10. Donat JF, Okazaki H, Gomez MR, Reagan TJ, Baker HL Jr, 33. Bruce J, Fetell M, Stein B (1996) Surgical approaches to pineal
Laws ER Jr (1978) Pineal tumors. A 53-year experience. Arch tumors. In: Wilkins RH, Rengachary SS (eds) Neurosurgery.
Neurol 35:736–740 McGraw-Hill, New York
11. DeGirolami U, Schmidek H (1973) Clinicopathological study of 34. Lapras C, Patet JD, Mottolese C, Lapras C Jr (1987) Direct
53 tumors of the pineal region. J Neurosurg 39:455–462. doi:10. surgery for pineal tumors: occipital-transtentorial approach.
3171/jns.1973.39.4.0455 Prog Exp Tumor Res 30:268–280
12. Wagenen V (1931) A surgical approach for the removal of 35. Poppen JL (1966) The right occipital approach to a pinealoma.
certain pineal tumors. Surg Gynecol Obstet 37:216–220 J Neurosurg 25:706–710. doi:10.3171/jns.1966.25.6.0706
13. Dandy W (1936) Operative experience in cases of pineal tumor. 36. Sano K (1987) Pineal region and posterior third ventricular
Arch Surg 33:19–46 tumors: a surgical overview. Williams & Wilkins, Baltimore
14. Duffner PK, Cohen ME, Thomas PR, Lansky SB (1985) The 37. Little KM, Friedman AH, Fukushima T (2001) Surgical
long-term effects of cranial irradiation on the central nervous approaches to pineal region tumors. J Neurooncol 54:287–299
system. Cancer 56:1841–1846 38. Reid WS, Clark WK (1978) Comparison of the infratentorial
15. Jenkin D, Berry M, Chan H, Greenberg M, Hendrick B, Hoff- and transtentorial approaches to the pineal region. Neurosurgery
man H, Humphreys R, Sonley M, Weitzman S (1990) Pineal 3:1–8
region germinomas in childhood treatment considerations. Int J 39. Bruce J (2011) Youman’s neurological surgery: pineal tumors.
Radiat Oncol Biol Phys 18:541–545 WB Saunders, Philadelphia
16. Sakai N, Yamada H, Andoh T, Hirata T, Shimizu K, Shinoda J 40. Hernesniemi J, Romani R, Albayrak BS, Lehto H, Dashti R,
(1988) Primary intracranial germ-cell tumors. A retrospective Ramsey C 3rd, Karatas A, Cardia A, Navratil O, Piippo A,
analysis with special reference to long-term results of treatment Fujiki M, Toninelli S, Niemela M (2008) Microsurgical man-
and the behavior of rare types of tumors. Acta Oncol 27:43–50 agement of pineal region lesions: personal experience with 119
17. Casentini L, Colombo F, Pozza F, Benedetti A (1990) Combined patients. Surg Neurol 70:576–583. doi:10.1016/j.surneu.2008.
radiosurgery and external radiotherapy of intracranial germino- 07.019
mas. Surg Neurol 34:79–86 41. Qi S, Fan J, Zhang XA, Zhang H, Qiu B, Fang L (2014) Radical
18. Chapman PH, Linggood RM (1980) The management of pineal resection of nongerminomatous pineal region tumors via the
area tumors: a recent reappraisal. Cancer 46:1253–1257 occipital transtentorial approach based on arachnoidal consid-
19. Dempsey PK, Kondziolka D, Lunsford LD (1992) Stereotactic eration: experience on a series of 143 patients. Acta Neurochir
diagnosis and treatment of pineal region tumours and vascular (Wien) 156:2253–2262. doi:10.1007/s00701-014-2224-4
malformations. Acta Neurochir (Wien) 116:14–22 42. Al-Holou WN, Terman SW, Kilburg C, Garton HJ, Muraszko
20. Edwards MS, Hudgins RJ, Wilson CB, Levin VA, Wara WM KM, Chandler WF, Ibrahim M, Maher CO (2011) Prevalence
(1988) Pineal region tumors in children. J Neurosurg 68: and natural history of pineal cysts in adults. J Neurosurg
689–697 115:1106–1114. doi:10.3171/2011.6.JNS11506

123
J Neurooncol (2016) 130:351–366 365

43. Goodman RR (1993) Magnetic resonance imaging-directed Neurol 65:1004–1011. doi:10.1097/01.jnen.0000240462.80263.


stereotactic endoscopic third ventriculostomy. Neurosurgery 13
32:1043–1047 (discussion 1047) 63. Calaminus G, Bamberg M, Jurgens H, Kortmann RD, Sorensen
44. Bruce JN, Ogden AT (2004) Surgical strategies for treating N, Wiestler OD, Gobel U (2004) Impact of surgery,
patients with pineal region tumors. J Neurooncol 69:221–236 chemotherapy and irradiation on long term outcome of
45. Connolly ES, Bruce J, Sonabend AM (2011) Pineal cell and intracranial malignant non-germinomatous germ cell tumors:
germ cell tumors. In: Kaye AH, Laws ER (eds) Brain tumors, results of the German Cooperative Trial MAKEI 89. Klin
3rd edn. Elsevier, New York Padiatr 216:141–149. doi:10.1055/s-2004-822626
46. Yamamoto I, Kageyama N (1980) Microsurgical anatomy of the 64. Satoh H, Uozumi T, Kiya K, Kurisu K, Arita K, Sumida M,
pineal region. J Neurosurg 53:205–221. doi:10.3171/jns.1980. Ikawa F (1995) MRI of pineal region tumours: relationship
53.2.0205 between tumours and adjacent structures. Neuroradiology
47. Allen JC, Nisselbaum J, Epstein F, Rosen G, Schwartz MK 37:624–630
(1979) Alphafetoprotein and human chorionic gonadotropin 65. Zee CS, Segall H, Apuzzo M, Destian S, Colletti P, Ahmadi J,
determination in cerebrospinal fluid. An aid to the diagnosis and Clark C (1991) MR imaging of pineal region neoplasms.
management of intracranial germ-cell tumors. J Neurosurg J Comput Assist Tomogr 15:56–63
51:368–374. doi:10.3171/jns.1979.51.3.0368 66. Fujimaki T, Matsutani M, Funada N, Kirino T, Takakura K,
48. Jennings MT, Gelman R, Hochberg F (1985) Intracranial germ- Nakamura O, Tamura A, Sano K (1994) CT and MRI features of
cell tumors: natural history and pathogenesis. J Neurosurg intracranial germ cell tumors. J Neurooncol 19:217–226
63:155–167. doi:10.3171/jns.1985.63.2.0155 67. Liang L, Korogi Y, Sugahara T, Ikushima I, Shigematsu Y,
49. Arita N, Ushio Y, Hayakawa T, Uozumi T, Watanabe M, Mori Okuda T, Takahashi M, Kochi M, Ushio Y (2002) MRI of
T, Mogami H (1980) Serum levels of alpha-fetoprotein, human intracranial germ-cell tumours. Neuroradiology 44:382–388.
chorionic gonadotropin and carcinoembryonic antigen in doi:10.1007/s00234-001-0752-0
patients with primary intracranial germ cell tumors. Oncodev 68. D’Andrea AD, Packer RJ, Rorke LB, Bilaniuk LT, Sutton LN,
Biol Med 1:235–240 Bruce DA, Schut L (1987) Pineocytomas of childhood. A
50. Bamberg M, Metz K, Alberti W, Heckemann R, Schulz U reappraisal of natural history and response to therapy. Cancer
(1984) Endodermal sinus tumor of the pineal region. Metastases 59:1353–1357
through a ventriculoperitoneal shunt. Cancer 54:903–906 69. Kersh CR, Constable WC, Eisert DR, Spaulding CA, Hahn SS,
51. Jooma R, Kendall BE (1983) Diagnosis and management of Jenrette JM 3rd, Marks RD Jr (1988) Primary central nervous
pineal tumors. J Neurosurg 58:654–665. doi:10.3171/jns.1983. system germ cell tumors. Effect of histologic confirmation on
58.5.0654 radiotherapy. Cancer 61:2148–2152
52. Wilson ER, Takei Y, Bikoff WT, O’Brien MS, Tindall GT, 70. Choi JU, Kim DS, Chung SS, Kim TS (1998) Treatment of germ
Boehm WM (1979) Abdominal metastases of primary cell tumors in the pineal region. Childs Nerv Syst 14:41–48
intracranial yolk sac tumors through ventriculoperitoneal shunts: 71. Bruce JN, Stein BM (1995) Surgical management of pineal
report of three cases. Neurosurgery 5:356–364 region tumors. Acta Neurochir (Wien) 134:130–135
53. Haase J, Borgaard-Pedersen B (1979) Alpha-feto-protein (AFP) 72. Sawamura Y, de Tribolet N, Ishii N, Abe H (1997) Management
and human chorionic gonadotropin (HCG) as biochemical of primary intracranial germinomas: diagnostic surgery or rad-
markers of intracranial germ-cell tumours. Acta Neurochir ical resection? J Neurosurg 87:262–266. doi:10.3171/jns.1997.
(Wien) 50:67–69 87.2.0262
54. Neuwelt EA, Smith RG (1979) Presence of lymphocyte mem- 73. Schild SE, Scheithauer BW, Haddock MG, Wong WW, Lyons
brane surface markers on ‘‘small cells’’ in a pineal germinoma. MK, Marks LB, Norman MG, Burger PC (1996) Histologically
Ann Neurol 6:133–136. doi:10.1002/ana.410060211 confirmed pineal tumors and other germ cell tumors of the brain.
55. Page R, Doshi B, Sharr MM (1986) Primary intracranial Cancer 78:2564–2571
choriocarcinoma. J Neurol Neurosurg Psychiatry 49:93–95 74. Kreth FW, Schatz CR, Pagenstecher A, Faist M, Volk B,
56. Takeuchi J, Handa H, Nagata I (1978) Suprasellar germinoma. Ostertag CB (1996) Stereotactic management of lesions of
J Neurosurg 49:41–48. doi:10.3171/jns.1978.49.1.0041 the pineal region. Neurosurgery 39:280–289 (discussion
57. Russell DS, Rubinstein LJ (1989) Tumours and tumour-like 289–291)
lesions of maldevelopmental origin. In: Russell DS, Rubinstein 75. Pecker J, Scarabin JM, Vallee B, Brucher JM (1979) Treatment
LJ (eds) Pathology of tumours of the nervous system. Williams in tumours of the pineal region: value of sterotaxic biopsy. Surg
& Wilkins, Baltimore, p 664 Neurol 12:341–348
58. Erlich SS, Apuzzo ML (1985) The pineal gland: anatomy, 76. Regis J, Bouillot P, Rouby-Volot F, Figarella-Branger D,
physiology, and clinical significance. J Neurosurg 63:321–341. Dufour H, Peragut JC (1996) Pineal region tumors and the role
doi:10.3171/jns.1985.63.3.0321 of stereotactic biopsy: review of the mortality, morbidity, and
59. Quest DO, Kleriga E (1980) Microsurgical anatomy of the diagnostic rates in 370 cases. Neurosurgery 39:907–912 (dis-
pineal region. Neurosurgery 6:385–390 cussion 912–904)
60. Villano JL, Propp JM, Porter KR, Stewart AK, Valyi-Nagy T, Li 77. Chandrasoma PT, Smith MM, Apuzzo ML (1989) Stereotactic
X, Engelhard HH, McCarthy BJ (2008) Malignant pineal germ- biopsy in the diagnosis of brain masses: comparison of results of
cell tumors: an analysis of cases from three tumor registries. biopsy and resected surgical specimen. Neurosurgery
Neuro Oncol 10:121–130. doi:10.1215/15228517-2007-054 24:160–165
61. Stein BM, Bruce JN (1992) Surgical management of pineal 78. Van Wagenen WP (1931) A surgical approach for the removal
region tumors (honored guest lecture). Clin Neurosurg of certain pineal tumors. Surg Genecol Obstet 37:216–220
39:509–532 79. Bruce JN (2000) Posterior third ventricle tumors. In: Kaye AH,
62. Fevre-Montange M, Hasselblatt M, Figarella-Branger D, Black PM (eds) Operative neurosurgery. Churchill Livingstone,
Chauveinc L, Champier J, Saint-Pierre G, Taillandier L, Coulon London, pp 769–775
A, Paulus W, Fauchon F, Jouvet A (2006) Prognosis and 80. Bruce JN (2012) Management of pineal region tumors. In:
histopathologic features in papillary tumors of the pineal region: Quinones-Hinojosa A (ed) Schmidek and sweet: operative
a retrospective multicenter study of 31 cases. J Neuropathol Exp neurosurgical techniques, 6th edn. Elsevier, New York

123
366 J Neurooncol (2016) 130:351–366

81. Bruce JN, Stein BM (1993) Complications of surgery for pineal 99. Vaquero J, Ramiro J, Martinez R, Coca S, Bravo G (1990)
region tumors. In: Posto KD, Friedman ED, McCormick PC Clinicopathological experience with pineocytomas: report of
(eds) Postoperative complications in intracranial surgery. five surgically treated cases. Neurosurgery 27:612–618 (dis-
Thieme, New York cussion 618–619)
82. McComb JG, Apuzzo MLJ (1988) The lateral decubitus position 100. Deshmukh VR, Smith KA, Rekate HL, Coons S, Spetzler RF
for the surgical approach to the pineal location tumors. Concepts (2004) Diagnosis and management of pineocytomas. Neuro-
Pediatr Neurosurg 8:186 surgery 55:349–355 (discussion 355–347)
83. Ausman JI, Malik GM, Dujovny M, Mann R (1988) Three- 101. Chang SM, Lillis-Hearne PK, Larson DA, Wara WM, Bollen
quarter prone approach to the pineal-tentorial region. Surg AW, Prados MD (1995) Pineoblastoma in adults. Neurosurgery
Neurol 29:298–306 37:383–390 (discussion 390–381)
84. Kobayashi S, Sugita K, Tanaka Y, Kyoshima K (1983) 102. Rippe DJ, Boyko OB, Friedman HS, Oakes WJ, Schold SC Jr,
Infratentorial approach to the pineal region in the prone position: DeLong GR, Meisler WJ (1990) Gd-DTPA-enhanced MR
concorde position. Technical note. J Neurosurg 58:141–143. imaging of leptomeningeal spread of primary intracranial CNS
doi:10.3171/jns.1983.58.1.0141 tumor in children. AJNR Am J Neuroradiol 11:329–332
85. Ueyama T, Al-Mefty O, Tamaki N (1998) Bridging veins on the 103. Waga S, Handa H, Yamashita J (1979) Intracranial germinomas:
tentorial surface of the cerebellum: a microsurgical anatomic treatment and results. Surg Neurol 11:167–172
study and operative considerations. Neurosurgery 43:1137–1145 104. Shibamoto Y, Abe M, Yamashita J, Takahashi M, Hiraoka M,
86. Yu T, Sun X, Wang J, Ren X, Lin N, Lin S (2016) Twenty-seven Ono K, Tsutsui K (1988) Treatment results of intracranial ger-
cases of pineal parenchymal tumours of intermediate differen- minoma as a function of the irradiated volume. Int J Radiat
tiation: mitotic count, Ki-67 labelling index and extent of Oncol Biol Phys 15:285–290
resection predict prognosis. J Neurol Neurosurg Psychiatry 105. Ueki K, Tanaka R (1980) Treatments and prognoses of pineal
87:386–395. doi:10.1136/jnnp-2014-309805 tumors—experience of 110 cases. Neurol Med Chir (Tokyo)
87. Farnia B, Allen PK, Brown PD, Khatua S, Levine NB, Li J, 20:1–26
Penas-Prado M, Mahajan A, Ghia AJ (2014) Clinical outcomes 106. Dattoli MJ, Newall J (1990) Radiation therapy for intracranial
and patterns of failure in pineoblastoma: a 30-year, single-in- germinoma: the case for limited volume treatment. Int J Radiat
stitution retrospective review. World Neurosurg 82:1232–1241. Oncol Biol Phys 19:429–433
doi:10.1016/j.wneu.2014.07.010 107. Disclafani A, Hudgins RJ, Edwards MS, Wara W, Wilson CB,
88. Tate M, Sughrue ME, Rutkowski MJ, Kane AJ, Aranda D, Levin VA (1989) Pineocytomas. Cancer 63:302–304
McClinton L, McClinton L, Barani IJ, Parsa AT (2012) The 108. Linstadt D, Wara WM, Edwards MS, Hudgins RJ, Sheline GE
long-term postsurgical prognosis of patients with pineoblastoma. (1988) Radiotherapy of primary intracranial germinomas: the
Cancer 118:173–179. doi:10.1002/cncr.26300 case against routine craniospinal irradiation. Int J Radiat Oncol
89. Apuzzo M, Tung H (1993) Supratentorial approaches to the Biol Phys 15:291–297
pineal region. In: Apuzzo M (ed) Brain surgery: complication 109. Wolden SL, Wara WM, Larson DA, Prados MD, Edwards MS,
avoidance and management. Churchill-Livingstone, New York, Sneed PK (1995) Radiation therapy for primary intracranial
p 486 germ-cell tumors. Int J Radiat Oncol Biol Phys 32:943–949
90. McComb J, Levy M, Apuzzo M (1998) The posterior intra- 110. Fuller B, Kapp D, Cox RJ (1993) Radiation therapy of pineal
hemispheric retrocallosal and transcallosal approaches to the region tumors: 25 new cases and a review of 208 previously
third ventricle region. In: Apuzzo M (ed) Surgery of the third reported cases. Int J Radiat Oncol Biol Phys 28:229–245
ventricle. Williams & Wilkins, Baltimore, pp 743–777 111. Schild SE, Scheithauer BW, Schomberg PJ, Hook CC, Kelly PJ,
91. Chaynes P (2003) Microsurgical anatomy of the great cerebral Frick L, Robinow JS, Buskirk SJ (1993) Pineal parenchymal
vein of Galen and its tributaries. J Neurosurg 99:1028–1038. tumors. Clinical, pathologic, and therapeutic aspects. Cancer
doi:10.3171/jns.2003.99.6.1028 72:870–880
92. Horrax G (1937) Extirpation of a huge pinealoma from a patient 112. Chandy MJ, Damaraju SC (1998) Benign tumours of the pineal
with pubertas praecox. Arch Neurol Psychiatry 37:385–397 region: a prospective study from 1983 to 1997. Br J Neurosurg
93. Bruce J (2006) Pineal region masses: clinical features and 12:228–233
management. In: Schmidek H (ed) Operative neurosurgical 113. Kang JK, Jeun SS, Hong YK, Park CK, Son BC, Lee IW, Kim
techniques, 5th edn. Saunders, Philadelphia, pp 786–797 MC (1998) Experience with pineal region tumors. Childs Nerv
94. Lapras C, Patet JD (1987) Controversies, techniques and Syst 14:63–68
strategies for pineal tumor surgery. In: Apuzzo M (ed) Surgery 114. Shin HJ, Cho BK, Jung HW, Wang KC (1998) Pediatric pineal
of the third ventricle. Williams & Wilkins, Baltimore, p 649 tumors: need for a direct surgical approach and complications of
95. Herrick MK, Rubinstein LJ (1979) The cytological differenti- the occipital transtentorial approach. Childs Nerv Syst
ating potential of pineal parenchymal neoplasms (true pinealo- 14:174–178
mas). A clinicopathological study of 28 tumours. Brain 115. Bruce J (2016) Pineal tumors. In: Winn HR (ed) Youman &
102:289–320 Winn’s neurological surgery, 7th edn. WB Saunders,
96. Apuzzo M, Litofsky NS (1993) Surgery in and around the Philadelphia
anterior third ventricle. In: Apuzzo M (ed) Brain surgery: 116. Bruce J, Stein B (2000) Supracerebellar infratentorial approach.
complication avoidance and management. Churchill Living- In: Kaye A, Black P (eds) Operative neurosurgery, vol 1.
stone, New York, p 541 Churchill Livingstone, London, pp 815–824
97. Nazzaro JM, Shults WT, Neuwelt EA (1992) Neuro-ophthal- 117. Rosenfeld J (2000) Supratentorial approaches to pineal tumors.
mological function of patients with pineal region tumors In: Kaye A, Black P (eds) Operative neurosurgery, vol 1.
approached transtentorially in the semisitting position. J Neuro- Churchill Livingstone, London, pp 825–840
surg 76:746–751. doi:10.3171/jns.1992.76.5.0746
98. Rubinstein LJ (1981) Cytogenesis and differentiation of pineal
neoplasms. Hum Pathol 12:441–448

123

You might also like