clinical indications for gamma knife radiosurgery a review...

166
Clinical Indications for Gamma Knife Radiosurgery A Review of the Published Clinical Evidence through 2014 Bodo Lippitz, MD June 2015

Upload: others

Post on 09-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

Clinical Indications for Gamma Knife Radiosurgery A Review of the Published Clinical Evidence through 2014

Bodo Lippitz, MD June 2015

Page 2: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

2

Table of Contents Introduction 4 Gamma Knife Treatment Indications: Overview 5 Technical Accuracy 6 Brain Metastases: General Considerations 8 Quality of Life and Treatment of Brain Metastases 9 Prognostic Factors in Brain Metastases 10 Treatment Safety in Competing Technologies 11 Comparative Effectiveness: Stereotactic Radiosurgery vs. Open Surgery 11 Stereotactic Radiosurgery for Brain Metastases in Elderly Patients 12 Stereotactic Radiosurgery in Larger Metastases 12 Stereotactic Radiosurgery of the Surgical Resection Cavity 13 Salvage Gamma Knife Surgery after Failed WBRT 14 Cognitive Function after Gamma Knife 14 Short-Term Adverse Effects 14 Imaging Follow-up 15 Adverse Radiation Effects 15 Cystic Metastases 15 Bevacizumab for Refractory Adverse Radiation Effects 16 Leptomeningeal Seeding 17 Stereotactic Radiosurgery of Multiple Brain Metastases 17 Brain Metastases from Breast Cancer 20 Local Tumor Control 20 Tumor Subtypes 21 Brain Metastases from Lung Cancer 23 Local Tumor Control (Lung Cancer) 23 Tumor subtypes (Lung Cancer) 23 SCLC 24 Local Control and Survival in Brain Metastases from NSCLC 25 Additional Chemotherapy (Temozolomide and Erlotinib) 27 Chemotherapy in NSCLC 27 Brain Metastases from Renal Cancer 28 Brain Metastases from Malignant Melanoma: Local Control and Survival 28 Chemotherapy (Malignant Melanoma) 30 Brain Metastases from Colorectal Carcinoma 30 Meningiomas 32 Treatment Doses (Meningiomas) 32 Tumor Control in Meningiomas in General 33 Long-Term Outcome (Meningiomas) 34 Large Meningioma 35 Specific Anatomical Meningioma Locations 37 Cavernous Sinus and Skull Base Meningiomas 37 Sellar and Parasellar Meningiomas 38 Petroclival Meningiomas 39 Convexity Meningiomas 39 Posterior Fossa Meningioma 40 Tentorial Meningioma 40 Cerebellopontine Angle Meningiomas 40 Olfactory Groove Meningioma 40 Parasagittal and Parafalcine Meningiomas 41 Combination Surgery/Radiosurgery 42 Malignant and Atypical Meningioma 42 Vestibular Schwannoma 44 Conservative Management of Vestibular Schwannoma 45 Tumor Control (Vestibular Schwannoma) 47 Long-term Outcomes (Vestibular Schwannoma) 48

Page 3: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

3

Quality of Life (Vestibular Schwannoma) 49 Intracanalicular Acoustic Neurinoma 50 Hearing Preservation (Vestibular Schwannoma) 51 Threshold Doses for the Preservation of Hearing 54 Malignant Transformation 55 Facial Nerve Palsy 57 Comparison Gamma Knife vs. Surgery 57 Combination with Subtotal Resection 58 Pituitary Adenomas 60 Non-Secreting Pituitary Adenomas 61 Side effects: Visual Dysfunction 64 Threshold Doses 64 Primary Management of Non-Secreting Pituitary Adenomas 66 Acromegaly 66 Stereotactic radiosurgery of Acromegaly 67 Adenoma Subtypes 68 Latency of Endocrinological Normalisation 69 Radiosurgical Salvage Treatment after Failed Conventional Radiotherapy 69 Radiation Doses (Pituitary Adenomas) 70 Relevance of Pre-operative Hormonal Levels 70 Radioprotective Effect of Anti-Secretory Therapy 71 Side Effects in Radiosurgery of Pituitary Adenoma 71 Prolactinoma 73 Gamma Knife Radiosurgery for Prolactinoma 73 Cushing’s Disease 76 Nelson's Syndrome 78 Whole-Sellar Stereotactic Radiosurgery 78 Second Intervention after Failed Resection in Cushing’s Disease 78 Gamma Knife Radiosurgery for Malignant Gliomas and Glioblastomas 80 Clinical Outcome Criteria 82 Side Effects after Radiosurgery of Recurrent Glioblastoma 82 Outcome after Radiosurgical Salvage Treatment of Recurrent Glioblastoma 83 Arteriovenous Malformations (AVM) 88 Treatment Doses (AVM) 90 Clinical Relevance of Technical Development 91 General Obliteration Rate in AVM 92 Side Effects (AVM) 94 Gamma Knife Treatment of AVMs Spetzler-Martin Grade III 96 Gamma Knife treatment for AVMs Spetzler-Martin Grade I or II 97 Dural Arteriovenous Fistulas 97 Effect of Gamma Knife on Seizure Activity in AVM Patients 98 Pediatric AVM 99 Thalamic/Basal Ganglia and Brainstem AVM 101 Volume-staged Gamma Knife Radiosurgery in Large AVM 103 Retreatment after Failed Obliteration 104 Gamma Knife Thalamotomy for Tremor 105 Target Definition (VIM) 108 Trigeminal Neuralgia 111 Clinical Outcome of Gamma Knife Radiosurgery for Trigeminal Neuralgia 113 Trigeminal Neuralgia Related to Multiple Sclerosis 118 Treatment Doses (Trigeminal Neuralgia) 119 Target Location (Trigeminal Neuralgia) 119 Retreatment with Gamma Knife (Trigeminal Neuralgia) 120 Comparative effectiveness: Gamma Knife vs. Microvascular Decompression 121 References 124

Page 4: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

4

Introduction

The principle of Gamma Knife radiosurgery implies concentrating radiation within a target in the

brain while avoiding radiation of the surrounding healthy tissue. The Gamma Knife achieves this by

the mechanical focusing of 192 radiation sources, which allows shaping of an extremely defined

irradiated volume in the brain. With modern planning techniques and multiple isocenters virtually

any shape of tumor can be matched with high precision radiation. Radiosurgical techniques and

particularly the Gamma Knife provide a very steep radiation dose fall-off. The irradiation hits the

pathological cells with a locally high concentration of energy while at the same time the neighboring

areas of the brain are spared from being affected by the irradiation. This specific gradient of

radiation in all three dimensions guarantees that the surrounding brain tissue is exposed to only

very little radiation and thereby “protected” against undesired radiation effects.

Radiosurgery is generally carried out in one session under local anesthesia resulting in very low

physical stress for the patient. The necessary precision of target localization requires a stereotactic

MRI or CT study before radiosurgery and a stereotactic frame fixation (for Linac and Gamma Knife).

Radiation doses are expressed as “prescription doses” or “minimum doses” reflecting the dose

applied to the tumor periphery. Generally, prescription doses of 15-25 Gy are applied in

radiosurgery of brain tumors. In Gamma Knife treatments this minimum dose commonly

corresponds to 50% of the maximally applied dose (50% isodose) resulting in an inhomogeneous

dose distribution within the tumor often ranging between 36 and 50 Gy. The lack of dose

homogeneity is irrelevant for the outcome, because the high dose area lies within the tumor tissue.

The stereotactic head frame guarantees the precision needed to protect healthy brain tissue and

directs the radiation focus into the target. So far more than a million patients have been treated in

worldwide 269 Gamma Knifes centers. The number of annually treated patients has doubled in the

last decade and is increasing steadily.

The American College of Radiology and American Society for Radiation Oncology has issued practice

guideline for the Performance of Stereotactic Radiosurgery as multidisciplinary approach and

defines organization, appropriate staffing, and defines the qualifications and responsibilities of all

the involved personnel, including the radiation oncologist, neurosurgeon, and qualified medical

physicist including quality assurance issues for the treatment unit, stereotactic accessories, medical

imaging, and treatment-planning system 1.

Page 5: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

5

Gamma Knife Treatment Indications: Overview

The Gamma Knife technology has enabled neurosurgeons to treat many conditions that are

otherwise inaccessible for therapy. The goal of radiosurgery is to maintain quality of life.

In some indications Gamma Knife replaces the open tumor operation. That comprises tumors that

are either surgically inaccessible or tumors that had been treated unsuccessfully by conventional

surgery, chemotherapy and/or radiation therapy. In other indications Gamma Knife is used as an

additive therapy, which enables the conventional surgeon to operate less radically and thereby with

a lower complication rate.

It is important to note that Gamma Knife radiosurgery is highly effective even for tumors that are

relatively resistant to traditional external beam radiation therapy. Numerous scientific studies have

documented the efficacy of radiosurgery against various brain tumors. The majority of brain

metastases can be treated successfully using the Gamma Knife with excellent tumor control rates

and low associated morbidity. The method was shown to be highly effective even against multiple

metastases. Recent results even show a beneficial effect of Gamma Knife treatment in the therapy

of gliomas and pediatric and less frequently occurring brain tumors such as pilocytic astrocytoma

and ependymoma.

Gamma Knife radiosurgery stops the growth of benign brain tumors such as meningioma. In this

indication Gamma Knife radiosurgery is generally used as a complement to open neurosurgical

resection in order to allow less invasive operations hereby reducing the otherwise high rate of

surgery-related morbidity, intensive care and other ancillary treatments.

A further important group treated with Gamma Knife comprises patients with vestibular

Schwannoma (Acoustic Neurinoma), another benign tumor located close to the brain stem. These

patients are otherwise treated with highly sophisticated and invasive open surgery. Radiosurgery

provides highly efficient local control of these tumors avoiding many of the complications that are

associated with open surgery.

Gamma Knife treatments have a very long track record in the treatment of postoperatively recurring

both endocrinologically active and inactive pituitary adenoma. In patients with arterio-venous

malformations (AVM) of the brain the Gamma Knife is used as an alternative to open surgery.

Recent results documented that Gamma Knife radiosurgery provides excellent treatment option for

patients with trigeminal neuralgia who fail to respond to medical therapy. Gamma Knife provides a

Page 6: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

6

highly efficient and cost effective alternative in the treatment of Parkinson tremor otherwise

treated with highly costly deep brain stimulation.

Summary Treatment Indications

Brain Metastases

Vestibular Schwannoma (Acoustic Neurinoma)

Meningioma

Glioma and other rarer brain tumors

Pituitary Adenoma

Arterio-venous malformations (AVM)

Trigeminal Neuralgia

Functional Indications (Thalamotomy for tremor etc.)

Technical Accuracy

The accuracy of the applied radiation is an inherent part of comparative safety. In the recent Gamma

Knife model (Perfexion), the accuracy of the mechanical versus radiation isocenter was found to be

0.05 mm 2. Linac planning is negatively affected by a loss of imaging precision due to the

requirement of CT based planning and phantom studies with a stereotactic Linac have shown that

a CT localized target can be irradiated with a positional accuracy of 0.8 mm in any direction with

95% confidence 3.

This technical accuracy is superior to alternative radiosurgery techniques 4-6 .The closer the

prescription dose is matched to the treated target and the steeper the dose gradient around the

target, the less normal tissue is irradiated. This matching of the dose to a 3-dimensional target is

generally quantified using the conformity index, which reflects the amount of normal tissue within

the prescription isodose. A simplified conformity index is the ratio of prescription volume to target

volume, which would ideally be 1, if no healthy tissue were irradiated. Polymer gel measurements

verified the optimal dose distribution in the most recent Gamma Knife models (Perfexion) with a

conformity of 1.17 7. A very recent study examined the radiation dose distributions for treating

multiple brain metastases created by Gamma Knife Perfexion in comparison to alternative

techniques (Linac single-isocenter volumetric modulated arc radiosurgery) showing that the even

the advanced modern stereotactic Linac consistently irradiated a higher amount of normal brain

Page 7: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

7

tissues. These increases were largest for lower isodose levels, with the volumes of normal brain that

received 20%-50% and 60%-90% of the prescription dose showing average increases of 403% and

227%, respectively. Radiosurgery quality metrics indicated that the Gamma Knife plan was superior

in each case 8. Extracranial doses vary depending on the technology used. Due to effective shielding,

Gamma Knife doses decreased steeply with distance from the treated brain, delivering only 13 mSv

at the lower pelvis in comparison to 117 and 132 mSv during CyberKnife treatments. Further re-

duction of extracranial doses has been shown in the recent Gamma Knife model Perfexion 9,10.

In a comparison of three most prominent systems for stereotactic radiosurgery (the Cyberknife

system, the Gamma Knife Perfexion and the Novalis system) it was shown that the Gamma Knife

showed significantly steeper gradients compared with the Novalis and the Cyberknife system

(Gamma Knife vs. Novalis, p = 0.014; Gamma Knife vs. Cyberknife, p = 0.002) 11. The investigation of

inter-target dose interplay effects demonstrated that for multi-target SRS, Gamma Knife PFX spares

the normal brain tissue best when compared to CyberKnife, Novalis linear accelerator equipped

with a 3.0-mm multi-leaf collimator, and the Varian Truebeam flattening-filter-free linear

accelerator. The volumes of normal brain receiving 4 and 12 Gy were higher, and increased more

swiftly per target, for Linac-based SRS platforms than Gamma Knife Perfexion 12. Linac Rapid Arc

plans were compared to Gamma Knife plans and dose-related analysis was performed. Rapid Arc

plans showed an inferior isotropic falloff and a shallower dose gradient. Both Rapid Arc and Gamma

Knife plans resulted in equivalent conformality while plans from GK showed sharper dose gradients

13 resulting in a lower radiation exposure of healthy target surrounding tissue.

The results of prior investigations had shown that the CyberKnife delivered significantly higher

peripheral doses to normal tissue than comparable model C Gamma Knife. Chuang, Petti and

colleagues investigated the peripheral dose reduction for CyberKnife radiosurgery treatments after

the installation of a Linac shielding upgrade and demonstrated that the additional shielding

decreased the peripheral dose, expressed as a percentage of the delivered monitor units (MU), by

a maximum of 59%. For distances between 30 and 70 cm from the field edge, the additional

shielding reduced the peripheral dose by between 20% and 55%, but the CyberKnife peripheral dose

still remains higher than doses measured in the center’s previous study for the model C Gamma

Knife 14.

Page 8: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

8

The Leksell Gamma Knife Perfexion (Elekta Instruments AB, Stockholm, Sweden) exceeds the

capabilities of previous gamma knife models in terms of treatment efficiency, conformity, and

radiation protection. The automated collimator arrangement in the Perfexion makes it possible to

produce more complex treatment volumes than with previous models of the gamma knife. This

results from the enhanced ability to shape isodose volumes, even for single isocenters. The

collimator arrangement and the patient positioning system also allow shorter patient transit times,

reducing unwanted radiation exposure during movement between isocenters 2.

In conclusion, the geometrical accuracy, dose accuracy and the dose gradient around the target

and the protection of extra-cranial tissue are superior in the Gamma Knife.

Brain Metastases: General Considerations

More than 185,000 treatments for brain metastases have so far been carried with Gamma Knife

radiosurgery. When Pubmed is searched for ‘Gamma Knife and brain metastases 1784 publications

appear, 101 publications appear for the corresponding search for stereotactic Linac and 37 for

CyberKnife. A wide range of retrospective and a prospective studies applying Gamma Knife

radiosurgery with more than 6000 patients with brain metastases provide consistent and

reproducible results with an average local tumor control between 84% and 97 %.

Multivariate analysis indicated that longer freedom from progression was significantly associated

with higher radiosurgical dose; local tumor control was superior for metastases treated at minimum

doses of 18 Gy or higher. Brain metastases tend to shrink gradually after radiosurgery; the tumor

associated perifocal edema disappears after several weeks. Stereotactic radiosurgery is effective

even against brain metastases from melanoma or renal cancer that are resistant to fractionated

external beam radiation. The tumor volume is the most important limiting factor in radiosurgery.

The volume of the irradiated healthy brain in the penumbra of the metastasis increases when larger

metastases are treated. This can result in formation of a local edema around the irradiated target,

typically 6-9 months after radiosurgery. This effect is generally transient, but may require a steroid

medication. Adverse radiation effects are generally seen in larger metastases or non-conformal

treatments and are very rare when metastases smaller than 2.5 cm are treated.

While WBRT has been applied for decades as standard therapy in the treatment of brain metastases,

recent scientific evidence including multiple randomized studies have now demonstrated a low and

insufficient response rate of brain metastases after WBRT 15-21, which is reflected by randomized

Page 9: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

9

data showing no life extending effect of WBRT 22-29. New metastases develop despite previous WBRT

in 23%- 50% and in this situation WBRT cannot be repeated 30-32. Recent randomized studies have

shown that WBRT has a negative impact on cognition, verbal memory and quality of life 33-36.

Based on these recent data WBRT does not appear to be sufficiently locally effective, does not

prolong the life of cancer patients and is potentially harmful for cognitive functions and quality of

life in patients with brain metastases. As a result, WBRT can no longer be considered as safe and

effective to provide a reliable and reproducible treatment in patients with brain metastases.

Quality of Life and Treatment of Brain Metastases

Patients receiving stereotactic radiosurgery alone were at significantly lower risk of a decline in

learning and memory function by 4 months when compared to patients undergoing additional

WBRT 33 .The conclusion from this study was interpreted as level I evidence to support the use of

stereotactic radiosurgery alone. Another recent prospective randomized study showed that

fractionated cranial irradiation causes a negative impact on health-related quality of life scales

particularly due to fatigue and hair loss 36 and causes cognitive dysfunction immediately after the

beginning of radiotherapy. Sub-acute radiation effects on verbal memory function are observed,

both after therapeutic and prophylactic cranial irradiation 35. These effects were more pronounced

in patients with above-average performance at baseline 35. A recently published prospective

randomized EORTC phase III trial with 359 patients showed that WBRT after surgery or radiosurgery

of brain metastases negatively impacted the health-related quality-of-life 34. Delayed significant CNS

toxicity after fractionated radiotherapy is a known phenomenon 37. It had previously been shown in

a smaller prospective study after Gamma Knife treatment for brain metastases that quality of life

parameters remained either unchanged or improved in patients who had no evidence of intracranial

or extracranial tumor progression 38. Quality of life was also analyzed as part of the prospective

randomized analysis RTOG 95-08 (2004). At 6 months follow-up a statistically significant improve-

ment in clinical performance and decreased steroid use was found in the stereotactic radiosurgery

boost treatment group when compared with the patients who had been treated by fractionated

radiotherapy only 39.

Prognostic Factors in Brain Metastases

Page 10: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

10

A multi-institutional retrospective (1985 to 2007) database of 3,940 patients with newly diagnosed brain

metastases was studied using univariate and multivariate analyses of prognostic factors associated with

outcomes by primary site and treatment. Significant prognostic factors varied by diagnosis. For lung cancer,

prognostic factors were Karnofsky performance score, age, presence of extracranial metastases, and number

of brain metastases, confirming the original Lung-GPA. For melanoma and renal cell cancer, prognostic

factors were Karnofsky performance score and the number of brain metastases. For breast cancer, prognostic

factors were tumor subtype, Karnofsky performance score, and age. For GI cancer, the only prognostic factor

was the Karnofsky performance score 40. Another study analyzed data from 1508 consecutive patients

treated with Gamma Knife without upfront whole-brain radiation therapy with 1-10 newly diagnosed brain

metastases with a volume of < 10 cm3 and a Karnofsky Performance Scale score > 70. Multivariate analysis

revealed significant prognostic factors for overall survival to be sex (poor prognostic factor: male, p < 0.0001),

recursive partitioning analysis class (Class I vs. Class II and Class II vs. III, both p < 0.0001), primary site (lung

vs. breast, p = 0.0047), and number of tumors (Group A vs. Group B, p < 0.0001). However, no statistically

difference was detected between patients with 2-4 tumors (577 cases) and 5-10 tumors (366 cases)(p =

0.1027) 41 .

Others confirmed that there was no relationship between number of brain metastases and

survival after excluding patients with single brain metastases. On multivariate analysis, favorable

prognostic factors included age <50, smaller total target volume, and longer interval from WBRT to

SRS in breast cancer patients; smaller number of brain metastases, KPS >60, and controlled primary

in non-small-cell lung cancer patients; and smaller total target volume in melanoma patients 42. The

prognostic factors of 400 patients with CNS metastases of breast cancer were reviewed Seoul

National University Hospital. The patients with good performance status, non-visceral extracranial

metastases, with longer interval from the date of primary breast cancer to the date of CNS

metastasis, chemotherapy after WBRT and gamma-knife surgery, had better outcomes in univariate

analyses. In multivariate analysis, good performance status, gamma-knife surgery and systemic

chemotherapy after WBRT were independent prognostic factors for overall survival after CNS

metastases 43.

Treatment safety in competing technologies

The results of prior investigations had shown that the CyberKnife delivered significantly higher

peripheral doses to normal tissue than comparable model C Gamma Knife. Chuang, Petti and

Page 11: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

11

colleagues investigated the peripheral dose reduction for CyberKnife radiosurgery treatments after

the installation of a Linac shielding upgrade and demonstrated that the additional shielding

decreased the peripheral dose, expressed as a percentage of the delivered monitor units (MU), by

a maximum of 59%. For distances between 30 and 70 cm from the field edge, the additional

shielding reduced the peripheral dose by between 20% and 55%, but the CyberKnife peripheral dose

still remains higher than doses measured in the center’s previous study for the model C Gamma

Knife 14.

The Leksell Gamma Knife Perfexion (Elekta Instruments AB, Stockholm, Sweden) exceeds the

capabilities of previous gamma knife models in terms of treatment efficiency, conformity, and

radiation protection. The automated collimator arrangement in the Perfexion makes it possible to

produce more complex treatment volumes than with previous models of the gamma knife. This

results from the enhanced ability to shape isodose volumes, even for single isocenters. The

collimator arrangement and the patient positioning system also allow shorter patient transit times,

reducing unwanted radiation exposure during movement between isocenters 2.

Comparative Effectiveness: Stereotactic Radiosurgery vs. Open Surgery

Three older retrospective 44-46 and one recent prospective randomized study 47compared the

efficacy of radiosurgery of brain metastases with open microsurgery (plus WBRT) which is

significantly more invasive that stereotactic radiosurgery. Treatment results did not differ in terms

of survival, neurological death rates, and freedom from local recurrence. Radiosurgery was

associated with a shorter hospital stay, less frequent and shorter steroid application, and lower

frequency of toxicities. The randomized study showed that improved scores for quality of life were

seen 6 weeks after radiosurgery 47. As stated in a recent systematic review and evidence-based

clinical practice guideline, for smaller metastases the therapeutic effect of Gamma Knife radiosur-

gery can be considered as equivalent to resection+WBRT (level 3 recommendation) 48.

Stereotactic Radiosurgery for Brain Metastases in Elderly Patients

It was recently shown that SRS for patients aged >/=75 years with brain metastases is an effective

and safe treatment modality that appears to improve survival, with outcomes that compare

favorably with those reported for younger patients 49.

Page 12: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

12

Similarly Watanabe and colleagues showed in a retrospective case-matched study, that patients

with brain metastases who are 80 years of age or older (n=165), are not unfavorable candidates for

SRS. In this age group the post-SRS median survival time was shorter than in the cohort aged 65-79

years (5.3 vs. 6.9 months), but this difference was not statistically significant 50.

Stereotactic Radiosurgery in Larger Metastases

Larger brain metastases with a clinically relevant mass effect should be removed surgically. Single

session radiosurgery in larger metastases should be avoided, since the volume of the irradiated

healthy brain in the penumbra of the metastasis increases when larger metastases are treated. As

a result, the tumor volume is the primary limiting factor in radiosurgery. A consecutive series of 109

patients with 119 large intracranial metastatic lesions were treated with Gamma Knife surgery

(median tumor volume 16.8 cm3; range 6.0-74.8 cm3). Peri-tumoral brain edema as defined on T2-

weighted MRI sequences had decreased in 66.4% of lesions was stable in 17.6%, but had progressed

in 16.0% 51.

In an attempt to reduce side effects, hypo-fractionated two-session Gamma Knife radiosurgery

using 20-30 Gy given in two fractions 3-4 weeks apart was applied for large metastatic brain tumors.

(Median tumor volumes 17.8 cm(3) at first GKS and 9.7 cm(3) at second GKS.). The local control rate

was 85 % at 6 months and 61 % at 12 months. The overall survival rate after GKS was 63 % at 6

months and 45 % at 12 months 52,53. Alternatively, hypo-fractionated Radiosurgery using 21 Gy in

3 fractions (67%), 24 Gy in 4 fractions (14%) and 30 Gy in 5 fractions (12%) was given in another

series resulting in Kaplan-Meier estimates of 1 year local control of 61% . Symptomatic radiation

necrosis occurred in 7% 54. In Linac SRS of large brain metastases (>3 cm) a median dose of 24 Gy

was applied in 2 to 5 fractions for a median treatment volume of 15.6 mL (range, 10 to 82.7 mL),

yielding an actuarial 6-month and 1-year overall survival rates were 22% and 13%, respectively. No

patients in this cohort experienced acute or late complications secondary to SRS 55.

Stereotactic Radiosurgery of the Surgical Resection Cavity

The risk for local recurrence is high after surgical resection and fractionated radiotherapy is often

recommended in order to improve postoperative local control. In order to reduce the known risks

of WBRT, however, stereotactic radiosurgery has been applied to the resection cavity. The efficacy

of postoperative Gamma Knife surgery to the tumor cavity following gross-total resection of a brain

Page 13: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

13

metastasis has been evaluated in a retrospective review. Patients with confirmed metastatic

disease underwent Gamma Knife surgery to the postoperative resection cavity following gross-

total resection of the tumor (n=47) (lung cancer: 40%, melanoma: 21%, renal cell carcinoma: 15%,

breast cancer:15%, gastrointestinal malignancies: 9%). The mean duration between resection and

radiosurgery was 15 days (range 2-115 days). The mean volume of the treated cavity was 10.5 cm3

(range 1.75-35.45 cm3), and the mean dose to the cavity margin was 19 Gy. Local tumor control at

the site of the surgical cavity was achieved in 94% 56. Another retrospective analysis of 110 patients

who had received Gamma Knife stereotactic radiosurgery to the tumor bed of resected brain

metastasis showed actuarial local control of the cavity at 12 months in 73%. ; Symptomatic

radiation-induced enhancement occurred in 6% of patients and leptomeningeal dissemination in

11% 57. After stereotactic radiosurgery to the cavity using a lower median dose of 16 Gy delivered

to the 50% isodose line median treatment volume 9.2 cm3), a 1-year actuarial local control of 81%

was achieved. Leptomeningeal carcinomatosis developed in 14% and was associated with breast

histology and infratentorial cavities 58

In another group where Gamma knife stereotactic radiosurgery without whole brain radiotherapy

was used to treat the tumor cavity following resection of solitary brain metastasis (mean marginal

dose 17.1 Gy), only 8.9% had local recurrence in the immediate vicinity of the resection cavity,

qualifying as "local failures" 59.

Salvage Gamma Knife Surgery after Failed WBRT

The efficacy of Gamma Knife for treating recurrent or new brain metastases after whole-brain

radiotherapy (WBRT) in patients with small-cell lung cancer (SCLC) was evaluated as the first-line

radiation therapy (n=44). The median duration between WBRT and first GKS was 8.8 months and

the tumor control rate was 95.8 % showing that GKS may be an effective option for controlling SCLC-

associated brain metastases after WBRT and for preventing neurological death in patients without

carcinomatous meningitis 60. Others showed comparatively low 1-year local control rates of 57%61

in patients with small-cell lung cancer.

A miscellaneous cohort of 310 patients with brain metastases of various tumor types was treated

with Gamma Knife for brain metastases after prior WBRT and showed a median survival of 8.4

Page 14: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

14

months 42. This series underlines the need for radiosurgical salvage treatment. Similarly, a median

overall survival time of 8.2 months and 1- year local control rates of 76.6% were shown after

stereotactic radiosurgery salvage treatment for 77 patients with recurrence of brain metastases

after whole brain radiotherapy (WBRT). Prescribed dose (>/=20 Gy) was an independent predictive

factors for local tumor control 62.

Cognitive Function after Gamma Knife

Previous randomized studies have shown significant improvement in clinical performance and

decreased steroid use in a stereotactic radiosurgery boost cohort when compared with the patients

who had been treated by fractionated radiotherapy 39 and that WBRT–induced cognitive

dysfunction was avoided when SRS was used 33 A study by Nakazaki and Kano evaluated the mini-

mental status examination (MMSE) scores of patients with brain metastases after gamma knife

radiosurgery without whole-brain radiation therapy in 119 consecutive patients. In 43.2 % with pre-

GKS MMSE scores </=27, the MMSE scores improved by >/=3 points, whereas 15 of all patients

19.7 % experienced deteriorations of >/=3 points. Mental deterioration of patients with large

symptomatic metastatic tumors tended to improve after gamma knife 63.

Short-Term Adverse Effects

A prospective study demonstrated that Gamma Knife is well tolerated with few patients developing

major acute effects. The study determined acute complications of intracranial stereotactic

radiosurgery, specifically Gamma Knife in 76 eligible patients. At 1 week, 24% complained of

minimal scalp numbness, but only 13% had minimal scalp numbness at 1 month and 2% at 2 months,

while 13% developed pin site pain at 1 week with a median intensity level of 2 out of 10. By one

month, only 3% had pin site pain with a median intensity level of 3 out of 10. There was no pin-site

infection at 1 and 2 months. A total of 84% of treated patients returned to work a median of 4 days

after SRS 64.

Imaging Follow-up

DWI/ADC appears to be a practical method for studying the efficacy of radiosurgery and predicting

early metastases response progression. A decrease signal on DWI and increased ADC (Apparent

diffusion coefficient) values are indicators of good tumor control. DWI (Tumor/white matter) ratios

Page 15: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

15

between the metastases and the normal, contralateral frontal white matter at each time point was

determined in addition to the ADC values for metastases. When using the initial ADCT values of

metastases to predict tumor response, sensitivity and specificity were 85.5 and 72.7%, respectively

65.

Adverse Radiation Effects

Yamamoto and associates studied the actual incidence and predictors of delayed complications

after Stereotactic Radiosurgery for brain metastases in 167 patients showing 10.2% delayed

complications (mass lesions with or without cyst in 8, cyst alone in 8, edema in 2) occurring 24.0-

121.0 months (median, 57.5 months) after GKRS. The actuarial incidences of delayed complications

estimated by competing risk analysis were 4.2% at the 60th month after GKRS. Among various pre-

GKRS clinical factors, univariate analysis demonstrated tumor volume-related factors: largest tumor

volume and tumor volume </=10 cc vs >10 cc to be the only significant predictors of delayed

complications. Univariate analysis revealed no correlations between delayed complications and

radiosurgical parameters (ie, radiosurgical doses, conformity and gradient indexes, and brain

volumes receiving >5 Gy and >12 Gy) 66.

Cystic Metastases

In 111 tumors with cystic components treated with Gamma Knife (Mean target volume 3.3 mL;

range, 0.1-23 mL; median prescription dose 21 Gy; range, 15-24 Gy) local control rates were 91%,

63%, and 37% at 6, 12, and 18 months, respectively. Local control was improved in lung primary and

worse in patients with prior WBRT 67. In order to reduce the volume of irradiated tissue in the

treatment of large cystic metastases, Higuchi and colleagues applied the technique of stereotactic

aspiration of the cyst with Gamma Knife treatment immediately thereafter. Following aspiration,

tumor volume, including the cystic component, decreased from a mean of 20.3 cm(3) to a mean of

10.3 cm(3). At least 20 Gy was delivered to the entire lesion in 24 of 25 cases. Good tumor control

was obtained in 16 of 21 cases that could be evaluated during a median follow-up period of 11

months (range 1-27 months); however, reaccumulation of cyst contents was observed in 2 patients

who required Ommaya reservoir placement 68.

Page 16: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

16

Alternatively a ”Donut's shape" radiosurgical treatment planning with a chain-like application of

multiple, small-sized isocenters has been used in large cystic intracranial metastases, after which

the majority of treated lesions showed significant shrinkage after radiosurgery while no major

complications were described 69.

In 2015 Lee, Kim and colleagues retrospectively reviewed the data of 37 cystic brain metastases of

28 patients who were treated with Gamma Knife. Cyst drainage was performed in 8 large lesions

before GKRS to decrease the target volume. The mean target volume was 4.8 cm3 (range, 0.3-15.8

cm3) at the time of GKRS, and the mean prescription dose was 16.6 Gy (range, 13-22 Gy).The

actuarial median survival time was 17.7 ± 10.2 months, and the primary tumor status was a

significant prognostic factor for survival. The actuarial local tumor control rate at 6 and 12 months

was 93.1% and 82.3%, respectively. Among the various factors, only prescription dose (>15 Gy)

was a significant factor related to local tumor control after multivariate analysis (p = 0.049) 70 .

Bevacizumab for refractory adverse radiation effects

Bevacizumab reduces both symptoms and reactive imaging changes in patients with refractory

adverse radiation effects (ARE), which are otherwise unresponsive to corticosteroids. In the therapy

of treatment refractory ARE Bevacizumab was applied in 29 patients with brain tumors or vascular

malformations who had developed clinical and/or imaging evidence of ARE after radiosurgery

(average dose of 7.4 mg/kg over a mean of 5.7 weeks at a median of 16 months following SRS).

Ninety percent (18/20) with clinically symptomatic ARE had neurological improvement after

bevacizumab therapy. Twenty-six patients had a decrease of 62 % of T2/FLAIR volumes and a 50 %

decrease in magnetic resonance imaging intravenous contrast enhancement volumes. Symptoms

recurred in 11 of the 20 patients after discontinuing therapy 71 .

Others showed in a Linac series that bevacizumab treatment offered symptomatic relief in adverse

radiation effects, a reduction in steroid requirement, and a dramatic radiographic response. All

patients initially receiving steroids had a reduction in steroid requirement, and the vast majority

having an improvement in or stability of ARE-associated symptoms. Bevacizumab as a treatment

for radiation necrosis of brain metastases post stereotactic radiosurgery 72 .

Page 17: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

17

Leptomeningeal seeding

When fractionated whole brain radiotherapy is avoided in the treatment of brain metastases, the

risk for leptomeningeal seeding could possibly increase. Among 827 patients that underwent

Gamma Knife radiosurgery for brain metastases, the overall incidence of leptomeningeal seeding

was 5.3 %. The actuarial rates for leptomeningeal seeding at 6 and 12 months were 3.1 and 5.8,

respectively. In addition, the risk of leptomeningeal seeding following Gamma Knife may be

associated with multiple lesions and breast cancer 73 . After Gamma Knife treatment of non-small

cell lung cancer brain metastases, 3.3% developed a leptomeningeal disease 74.

Stereotactic Radiosurgery of Multiple Brain Metastases

In most patients with multiple metastases, tumors tend to be small and do not affect the patient via

local mass effect. Rather than the number of metastases, the total tumor volume appears to have

an important influence on the prognosis 75,76. In general there is a tendency to treat a larger number

of brain metastases in a single radiosurgical session, since numerous studies document high local

control after radiosurgical treatment of more than 3 brain metastases. For example, Gamma Knife

treatment in patients with 3-6 brain metastases provided actuarial 2-year control rates of 74.3% 77.

In patients with ten or more cerebral metastatic lesions who had undergone Gamma Knife surgery,

local control was achieved in 79.5% of the lesions until 6 months after GKS. New lesions appeared

in 26.9% patients during the same period 78. The number of lesions was not a significant predictor

of overall survival in 53 patients with ≥ 10 brain metastases treated with Gamma Knife79 and in

another series of 96 patients with 5-15 brain metastases who had been treated using Gamma Knife

surgery. Actuarial tumor control was calculated to be 92.4% at 6 months, 84.8% at 12 months, and

74.9% at 24 months post-GKS. The median overall survival was found to be 4.73 months (range 0.4-

41.8 months)80. Similarly, the median survival following Gamma Knife radiosurgery for 10 or more

brain metastases was 4 months in another series with 61 patients, with improved survival in patients

with fewer than 14 brain metastases 81. Similarly, some series indicate a less favorable median

survival for patients with more than three or more than seven metastases 82, but there is no

simplistic correlation between the number of metastases and the patient’s prognosis. Hence, an

increased number of brain metastases can be an unfavorable factor for longer survival, but the

impact on survival time appears to be rather limited. There is increasing evidence that other factors

such as the subtype of the primary cancer 83-86 and the state of the systemic disease 87 are major

Page 18: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

18

contributing factors for the prognosis.

The outcome after stereotactic radiosurgery without WBRT in patients with five to ten brain

metastases is non-inferior to that in patients with two to four brain metastases as demonstrated

in 2014 in a prospective observational study comprising 1194 patients 88. Yamamoto and

colleagues recently (2013) published a retrospective case-matched study of patients who had

undergone stereotactic radiosurgery without WBRT and demonstrated that the median survival

time after radiosurgery was 7.9 months in patients with 1-4 brain metastases (n=548), and 7 months

in patients with more than 4 metastases (n=548) (p = 0.01) 89 . This retrospective study suggests that

increased tumor number is an unfavorable factor for longer survival. However, the small post-SRS

median survival time difference, 0.9 months, between the two groups was not considered clinically

meaningful. Similarly, the same group showed that post-SRS median survival times did not differ

significantly between patients with with 2-9 tumors (1254 patients) and patients with 10 or more

tumors (6.8 vs 6.0 months; p = 0.10) 90.

In line with the previously cited findings, Serizawa and Yamamoto published a combined series of

1508 consecutive cases and reported no difference in survival between patients with 2-4 and 5-10

brain metastases 41. The treatment outcomes of a consecutive series of 1,676 patients who

underwent Gamma Knife radiosurgery for brain metastases was studied and 14 pairs of groups,

based on tumor numbers, were compared : 1 vs. 2, 2 vs. 3, 3 vs. 4, - - -, and 14 vs. 15. Among the 14

pairs, only the 1 vs. 2 pair showed a significant median survival time difference (p = 0.0002); no

significant differences were detected for the other 13 pairs. Although tumor number was

demonstrated to have a significant impact on the duration of survival, approximately 85% of

patients with brain metastasis died of causes other than brain disease progression, regardless of

tumor number 91. In an earlier series of 521 patients treated with Gamma Knife, a similar outcome

in terms of overall and neurological survival was found for patients with few (≤3) and many (4-10)

brain metastases 92. Serizawa evaluated the results of gamma knife radiosurgery alone for selected

patients with 1-10 brain metastases without prophylactic whole-brain radiation therapy in 778

consecutive cases meeting the JLGK0901 study inclusion. On multivariate analysis, significant poor

prognostic factors for overall survival were active systemic disease, poor (<70) initial KPS, and male

gender. Neurological survival and qualitative survival at 1 year were 92.7% and 88.2%, respectively.

This study revealed that brain lesion number has no effect on overall survival neurological survival

Page 19: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

19

or qualitative survival, which is anticipated to be confirmed by the JLGK0901 study 93. Sustained

local tumor control after Gamma Knife treatment in patients with 10 or more brain metastases was

achieved in 81% of patients 81.

Previously smaller retrospective studies had shown that number of brain metastases did not have a

direct significant impact on survival 94-97, which was confirmed after Gamma Knife treatment of pa-

tients with breast cancer 98 and renal cancer 99. In a cohort of 251 patients who had initially been

treated with Gamma Knife alone at the MD Anderson Cancer Center it was shown that the number

of brain metastases was not predictive of distant brain failure, local control, and overall survival 100.

Similarly, in a retrospective analysis total of 310 patients who had been treated with Gamma Knife

radiosurgery after prior WBRT at the Stanford University School of Medicine there was no

relationship between the number of brain metastases and survival after excluding patients with

single metastases 42. The prediction of outcome based on the number of lesions was not feasible

in another Gamma Knife series with 330 patients 101.

Patients who had undergone Gamma Knife radiosurgery at the Yonsei University College of

Medicine, Seoul (n=323) were divided into groups according to the number of lesions visible on MR

images: Median survival was 10 months for patients with 1-5 metastases; 10 months for 6-10 lesions,

13 months for 11-15 lesions and 8 months for patients with more than 15 brain metastases 102 The

difference was not statistically significant.

Similarly, patients with more than 4 brain metastases do not appear to have an inferior outcome in

terms of neurological death, repeat SRS, maintenance of good neurological state and radiosurgery-

related complications 103, but several treatment recommendations and practice guidelines limit the

number of brain metastases that can be treated with radiosurgery to a virtually arbitrary number of

three or four without consideration of tumor volume or diagnostic method. These

recommendations are primarily based on level I evidence and hence on the available three

randomized studies, where the analysis was limited to patients with less than 3-4 metastases

according to the studies’ selection criteria: patients with a larger number of metastases had been

excluded from evaluation 39,104,105.

Page 20: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

20

In summary, there is no documented evidence for a lack of radiosurgical efficacy in multiple

metastases or for the existence of another superior treatment for this condition.

On the other hand, the total brain metastasis volume was a strong and independent predictor for

overall survival in 250 patients with 1-14 brain metastases treated with Gamma Knife surgery with

a total tumor volume cutoff value of ≥ 2 cm3 (p = 0.008) being a stronger predictor of overall survival

than the number of brain metastases (p = 0.098) 106. Local tumor control at 1 year was 97% for

lesions < 2 cm3 compared with 75% for lesions ≥ 2 cm3 (p < 0.001). Median overall survival was 7.1

months and the 1-year local control rate was 91.5%. On univariate analysis an increasing total tumor

volume was significantly associated with worse survival (p = 0.031) whereas the number of brain

metastases, analyzed as a continuous variable, was not (p = 0.082) 106.

Brain Metastases from Breast Cancer

Chemotherapy of breast cancer has made dramatic improvements over the last decade resulting in

significantly prolonged patient survival, unfortunately without impact on brain metastases. A

correspondingly effective treatment of brain metastases from breast cancer is paramount, but the

standard fractionated whole brain radiotherapy is insufficient to allow a reliable control over brain

metastases. The recently improved systemic therapeutic approaches for breast cancer patients

emphasize the necessity for an equally effective radiosurgical treatment of the brain metastases.

Local Tumor Control

A Pubmed search provided a total of 9 clinical series after Gamma Knife radiosurgery radiosurgery

treatment of breast cancer brain metastases with a total of 1137 published patients describing a

local tumor control ranging between 71%-97% with a median local control of more than 85% in

5/9 studies in a total of 74%% of all published patients (846/1137) 98,107-114 .

Kondziolka, Lunsford and colleagues reported that sustained local tumor control was achieved in

90% of 350 consecutive female patients who underwent Gamma Knife radiosurgery for 1535 brain

metastases patients. Symptomatic adverse radiation effects occurred in 6% of patients. Factors

associated with longer progression-free survival included a better RPA class, fewer brain

metastases, a smaller total tumor volume per patient, and a higher tumor margin dose 112. Another

study after Gamma Knife surgery for brain metastases in 101 patients reported local tumor control

Page 21: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

21

of 97%, and a tumor response rate of 82.3% 111. In a cohort of 62 patients with brain metastases

from breast cancer treated by Gamma Knife surgery actuarial local control rate was 89.5±4.5% and

70.5±6.9% at 6 and 12 months after GKS, respectively. No prognostic factors were found to affect

local control rate. Median survival after Gamma Knife was 73.0 weeks 109. A treated tumor volume

>5000 mm3 was significantly correlated with poor local tumor control according to a multivariate

analysis (hazard risk=7.091, p=0.01) 107.

The median survival time after Gamma Knife surgery was 16.0 months for 95 newly diagnosed

patients brain metastases from breast cancer and 11.7 months for 81 patients with recurrent brain

metastases 113.

Tumor subtypes

After analysis of a large multi-institutional retrospective database of 865 patients, Sperduto and

colleagues concluded that tumor subtypes are an important prognostic factor for survival in patients

with breast cancer. Patients with Basal (triple negative), HER2 (HER2 positive/ER/PR negative),

Luminal B (triple positive) and Luminal A (ER/PR positive/HER2 negative) tumor subtypes had a

median survival from primary diagnosis of 39.6, 66.4, 90.3 and 72.7 months (p < 0.01) and median

survival from diagnosis of brain metastases of 7.3, 17.9, 22.9 and 10.0 months (p < 0.01),

respectively. Patients with Basal and HER2 tumor subtypes have short time interval from primary

diagnosis to development of brain metastases 115. The incidence of brain metastasis in patients with

metastatic breast cancer ranges from 14 to 16%. Patients classified as triple-negative presented

more frequently with brain metastasis as the first site (26%) than patients in the human epidermal

growth factor receptor 2 (HER2)-positive (6%) or luminal (12%) subtypes 116. The analysis by

Sperduto, Mehta and associates of a multi-institutional retrospective database of 400 breast cancer

patients treated for newly diagnosed brain metastases showed that significant prognostic factors

were Karnofsky performance status, HER2, ER/PR status, and the interaction between ER/PR and

HER2. Among HER2-negative patients, being ER/PR positive improved median survival time from 6.4

to 9.7 months, whereas in HER2-positive patients, being ER/PR positive improved median survival

time from 17.9 to 20.7 months 117.

In 154 breast cancer patients who had been treated with Gamma Knife surgery for brain metastases,

multivariate analysis revealed that HER2+ status independently predicted improved survival (p =

Page 22: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

22

0.007), while women with basal subtype had high rates of distant brain failure and worsened

survival. Breast cancer subtype did not predict time to local failure (p = 0.554), but did predict

distant brain failure rate (76, 47, 47, 36 % at 1 year for Basal, Luminal A/B, HER2, and Luminal

HER2 respectively, p < 0.001) 118. Survival benefit was associated with non- triple-negative

phenotype status and lower recursive partitioning analysis class at the initial SRS in 103 breast

cancer patients with brain metastases (multivariate analysis) 119.

Advances in chemotherapy for breast cancer have prolonged overall survival, especially for patients

with human epidermal growth factor receptor-2 (HER2) positive cancer. Patients with HER2-

overexpressing tumors were found to be a distinct subgroup for which a longer survival time. In 80

consecutive patients who underwent Gamma Knife radiosurgery, the median survival time in HER2-

positive and -negative patients were 16.6 and 7.1 months, respectively (P = 0.001) Survival benefit

was associated with non- triple-negative phenotype status and lower recursive partitioning analysis

class at the initial SRS in 103 breast cancer patients with brain metastases (multivariate analysis)120

and similarly (in 2014) median overall survival was 22 months for the HER2-positive group

compared with 12 months for the HER2-negative group (p = 0.053) in another series of

radiosurgically treated breast cancer patients with brain metastases 121.

In 40 patients with HER2-overexpression, treatment results were compared between two sub-

groups: lapatinib-based therapy (24 patients) versus non-lapatinib-based therapy (16 patients).

The rates of 1- and 2-year overall survival after gamma knife SRS were 50 and 26 %, respectively.

Interestingly, in sub-analysis of patients with HER2-overexpression, lapatinib-based

chemotherapy was also associated with better local tumor control (86% vs. 69 %, P < 0.001) 120.

Brain Metastases from Lung Cancer

Patients with a complete response after multimodality therapy for locally advanced non-small cell

lung cancer (NSCLC) are at high risk for the subsequent development of brain metastases. Chen

found that 55% developed brain metastases. The 5-year estimates of brain metastasis-free survival

for patients with squamous and non-squamous cancers were 57% and 34%, respectively 122.

Page 23: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

23

Local Tumor Control

A total of 18 clinical series after Gamma Knife radiosurgery radiosurgery treatment of NSCLC brain

metastases with a total of 2660 published patients (10 series published after 2007) describing a local

tumor control ranging between 74%-98% with a median local control of > 85% in 13/18 studies

comprising 82% of published patients (2183/2660) 123,74,124-136 .

Tumor subtypes

Histological subtype of lung cancer appears to predict outcomes. Multivariate analysis confirmed

small cell carcinoma histology as a significant predictor of worsened local control (hazard ratio [HR]:

6.46, P = .025) and distant failure (HR: 3.32, P = .0027) in 271 patients with brain metastases from

primary lung cancer treated with Gamma Knife surgery. For NSCLC histologies, squamous cell

carcinoma predicted for worsened overall survival (HR: 1.77, P < .0121) 137.

EGFR tyrosine kinase mutation and ALK translocation results in improved response to targeted

therapies but that mutation status itself does not predict survival and local control in patients with

brain metastases from NSCLC: In 89 262 NSCLC patients treated with gamma knife radiosurgery for

brain metastases genotyping was available: This cohort was selected to determine outcome

predictors. Median overall survival rates for the EGFR, KRAS, echinoderm microtubule-associated

protein-like 4 (EML4)-ALK mutated, and wild-type cohorts were 17, 7, 27, and 12 months,

respectively (P = .019). Targeted therapy was a strong predictor of increased overall survival on

univariate (P = .037) and multivariate (P = .022) analysis. Overall survival for targeted versus non-

targeted therapy was 21 and 11 months, respectively (P = .071) 138.

On multivariate analysis, positive EGFR mutational status was a highly significant predictor for

decreased survival (hazard ratio: 8.2; 95% CI: 2.0-33.7; P = .003). However, when EGFR-mutant

cases were re-categorized based on whether they received tyrosine kinase inhibitor, overall

survival was no longer significantly shorter (hazard ratio: 1.5; P = .471). Local failure rates were

not different between tumor subtypes 138 .

Non-small cell lung cancer includes a spectrum of radiosensitive and radioresistant tumors. Others

found that EGFR mutation, ALK translocation, and metastasis size were independent predictors

Page 24: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

24

for superior local control after gamma knife treatment, since the predicted median in-field local

control was significantly longer for EGFR-mutant and ALK-translocated tumors compared with

other patients with NSCLC (P < 0.001), whereas distant brain recurrence time was equivalent (P =

0.97)139.

SCLC

The standard treatment of small cell lung cancer (SCLC) is fractionated whole brain radiotherapy,

but WBRT does not protect from recurrences or new metastases. This situation is particularly

difficult since WBRT cannot be repeated and patients generally have to remain untreated. Gamma

Knife surgery appears to be as effective in treating brain metastases from SCLC as for those from

NSCLC.

In a larger series of 245 patients with radiosurgically (Gamma Knife) treated brain metastases from

lung cancer Serizawa et al. found that there was no significant difference in survival between

patients with SCLC or NSCLC; this finding was verified by multivariate analysis 130. In a salvage

situation Gamma Knife stereotactic radiosurgery has been successfully applied for new metastases

in 51 patients with SCLC and previous WBRT. In this difficult clinical situation the 1-year local control

rate was 57% with concurrent chemotherapy improving local control (HR = 89; p = 0.006) 61. This

local control rate is comparatively low and at a higher median prescribed dose of 20.0 Gy, others

found a tumor control rate of 95.8 % at a rather short median follow-up period of 4 months in 44

patients who were treated with Gamma Knife for recurrent or new SCLC-associated brain

metastases after WBRT 60 .

Another series reported a 1-year actuarial local control of 86% after Gamma Knife treatment of

brain metastases from small-cell lung cancer. The overall survival was 9 months after Gamma Knife

and new brain metastases outside of the treated area developed in 61% of patients at a median

time of 7 months; 81% of these patients had received previous WBRT 134.

Local Control and Survival in Brain Metastases from NSCLC

In 2015 Bowden, Lunsford and associates reviewed 720 patients with NSCLC brain metastasis who

Page 25: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

25

underwent Gamma Knife radiosurgery 140. The overall local tumor control rate in the entire group

was 92.8%. The median survival time after diagnosis of brain metastasis from NSCLC was 12.6

months, and the median survival after SRS was 8.5 months. The 1-, 2-, and 5-year survival rates after

SRS were 39%, 21%, and 10%, respectively140. Post-radiosurgery survival was decreased in patients

treated with prior whole-brain radiation therapy compared with SRS alone (p = 0.003). A total of

24% underwent repeat SRS for new or resistant metastatic deposits 140. In patients with NSCLC

(n=373), who had undergone Gamma Knife radiosurgery for eight or fewer brain metastases,

Motta and colleagues reported a mean overall survival 14.2 months. The prescription dose was

relative high (22.5 Gy). RPA class and overall tumor volume >5 cc were the only two covariates

independently predictive of overall survival in patients who died of cerebral progression 128.

Previously Sheehan had published a retrospective review of 273 patients who had undergone

Gamma Knife surgery (GKS) to treat a total of 627 NSCLC metastases. The overall median patient

survival time was 10 months from GKS treatment in those patients with adenocarcinoma and 7

months for those with other histological tumor types. In patients with no active extracranial disease

at the time of GKS, the median survival time was 16 months. Gamma knife surgery for NSCLC

metastases offered effective local tumor control in approximately 84% of patients. In multivariate

analyses, factors significantly affecting survival included: 1) female sex (p = 0.014); 2) preoperative

Karnofsky Performance Scale score (p < 0.0001); 3) adenocarcinoma histological subtype (p =

0.0028); 4) active systemic disease (p = 0.0001); and 5) time from lung cancer diagnosis to the

development of brain metastasis (p = 0.0074). Prior tumor resection or whole-brain radiation

therapy did not correlate with extended patient survival time 133.

Serizawa showed in 245 patients with brain metastases from lung cancer a 1-year tumor control

rate of 94.5% in the SCLC group and 98% in the NSCLC group. The median survival time was 9.1

months in the SCLC group and 8.6 months in the NSCLC group. The 1-year survival rates in the SCLC

group were 86.5% for neurological survival and 68.9% for qualitative survival; those in the NSCLC

group were 87.9% for neurological and 78.9% for qualitative survival 130. Similarly, 100 patients

underwent Gamma knife radiosurgery treatment for 184 brain metastases from NSCLC at the

Marmara University Hospital, Istanbul, Turkey and local tumor control was achieved in 95% of the

metastases. In multivariate analysis, adenocarcinoma histology, Karnofsky performance status (KPS)

Page 26: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

26

score > 80, 1-3 metastases and tumor diameter <2 cm were related to longer survival. Addition of

WBRT did not have any effect on overall survival 136.

A study of 42 patients with synchronous, solitary brain metastasis from NSCLC with a median KPS of

90 who were treated with Gamma Knife radiosurgery showed a rather positive prognosis with a

median overall survival of 18 months and a 1-year actuarial OS rates of 71.3%. For patients who

underwent definitive thoracic therapy, the median OS was 26.4 months compared with 13.1 months

for those who had non-definitive therapy, and the 5-year actuarial OS was 34.6% vs. 0% (p < 0.0001).

The prognostic factors significant on multivariate analysis were definitive thoracic therapy (p =

0.020) and KPS (p = 0.001). The 5-year OS of 21% demonstrates the potential for long-term survival

in patients treated with gamma knife stereotactic radiosurgery. The authors argued that patients

with good KPS should be considered for definitive thoracic therapy 141.

The overall survival was 70.8% at 1 year and the actuarial local control rate was 91.5% at 1 year and

85.5% at 2 years in 89 patients who underwent Gamma Knife SRS for 1-4 metastases, at Lille

University Hospital (KPS score of 60 or greater; mean radiation dose 23.41 Gy; range 18-26Gy; mean

tumor volume 2.47cm3; range 0.3-6.7cm3). The overall complication rate was 10.1% 74

In 228 patients with non-small cell lung cancer treated with Gamma Knife radiosurgery (median

prescription dose 20 Gy; range, 14-24 Gy), median overall survival from time of SRS was 12.1 months

and estimated local control at 12 months was 74%. The adjusted Kaplan-Meier 1-year local control

for conformality index ≥ 1.75 was 84% versus 69% for conformality index <1.75 123.

In 26 patients with incidentally-identified NSCLC brain metastases (KPS 90-100) the median survival

was 8.2 months from diagnosis of brain metastases135.

A very small but one of the few studies that compared the effectiveness of Gamma Knife

radiosurgery for the treatment of multiple brain metastases from lung cancer with that of

fractionated whole brain radiation therapy (WBRT) (n= 33) found that the 1-year overall survival

rates were 47.7% in the Gamma Knife radiosurgery group and 10.5% in the WBRT group. The

overall survival time in the GKS group was significantly longer than in the WBRT group (p=0.04)

142.

Page 27: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

27

Additional Chemotherapy (Temozolomide and Erlotinib)

Because temozolomide (TMZ) and erlotinib (ETN) cross the blood-brain barrier and have

documented activity in NSCLC, a phase 3 study was designed to test whether these drugs would

improve the overall survival associated with WBRT + SRS. Sperduto, Mehta and associated showed

in a randomized study with 126 NSCLC patients (n=126) harboring 1 to 3 brain metastases that the

addition of TMZ or ETN to WBRT + SRS in NSCLC patients with 1 to 3 brain metastases did not

improve survival and possibly had a deleterious effect 143.

Chemotherapy in NSCLC

In a Korean randomized controlled trial of 105 patients with one to four asymptomatic brain

metastases of non-small-cell lung cancer, patients were randomly assigned to receive stereotactic

radiosurgery followed by chemotherapy or upfront chemotherapy. Symptomatic progression of

brain metastases was observed more frequently in the upfront chemotherapy group (26.5%) than

the SRS group (18.4%) but without statistical significance. The median OS time was 14.6 months in

the SRS group and 15.3 months for the upfront chemotherapy group (P = 0.418). Although this

study included smaller sample size than initially anticipated due to early termination, SRS followed

by chemotherapy did not improve OS in oligo-brain metastases NSCLC patients compared with

upfront chemotherapy. The survival rate in this study was unexpectedly high. When the study was

designed in 2008, the OS rate of 35% at 1 year (6–7 months of the median OS) was anticipated

while in the current study, the 1-year survival rate was 57% in the GKS group and 58% in the

observation group, and may be attributed to the relatively homogenous study population with

better prognostic factors such as high adenocarcinoma histology (80%) and activated EGFR

mutation (30%) 144.

Brain Metastases from Renal Cancer

Brain metastases of renal cell carcinoma are considered radioresistant. While brain metastases of

renal cell carcinoma are considered unresponsive to fractionated WBRT with a reported survival of

only 3.0-4.4 months 145, Gamma Knife treatment provided consistent local control rates of 74% -

96% in 9 clinical series after Gamma Knife radiosurgery of renal cell cancer brain metastases with a

total of 583 published patients describing a local tumor control ranging between 74% - 96% with a

Page 28: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

28

median local control > 85% in 7/9 studies comprising a total of 78% of published patients (453/583)

146-154 . For RPA classes I median survival was between 18 and 24 months 149,155.

The addition of WBRT does not prevent the development of new remote tumors in patients with

renal cell cancer metastases 99,146.

In brain metastases from renal cell carcinoma treated with gamma knife radiosurgery from 158

consecutive patients, sustained local tumor control was achieved in 92% of patients. Symptomatic

adverse radiation effects occurred in 7%. Fifty-seven patients had undergone prior WBRT. The

overall survival after SRS was 38% at 12 months, with a median survival of 8.2 months. Factors

associated with longer survival included younger age, longer interval between primary diagnosis

and brain metastases, lower recursive partitioning analysis class, higher Karnofsky performance

status, smaller number of brain metastases, and no prior WBRT 154.

Brain Metastases from Malignant Melanoma: Local Control and Survival

Based on the limited local tumor control rate after WBRT, brain metastases of malignant melanoma

are considered radioresistant. This restriction does not apply to radiosurgery, since various series

after Gamma Knife radiosurgery of malignant melanoma report reproducible local tumor control

rates between 63-90% in a total of 13 clinical series after Gamma Knife radiosurgery radiosurgery

treatment of otherwise radioresistant melanoma brain metastases with a total of 1316 published

patients with a median local control of more than 85% in 6 out of 13 studies and 47 % of published

patients (614/1316) 75,147,151,156-165. Gamma knife radiosurgery for malignant melanoma brain

metastases is safe and effective and provides a high rate of durable local control.

Mathieu and colleagues had reviewed 245 patients with malignant melanoma brain metastases

treated with Gamma knife radiosurgery. The median tumor volume was 4.4 cm3. The median

margin doses were 18 Gy. Sustained local control was achieved in 86.2% of tumors and median

survival was 5.3 months after radiosurgery. Patients survived a median of 7.8 months from the

diagnosis of brain metastases and 44.9 months after the diagnosis of the primary tumor.

Symptomatic radiation changes occurred in 6.6% 159. Gamma-Knife radiosurgery without whole-

brain radiotherapy was used in the management of 106 patients with brain metastases of malignant

melanoma with a local control rate of 83.7% at a median survival from the time of GKR of 5.09

months 158. Virtually identical results were achieved in 2014, when a total of 129 consecutive

Page 29: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

29

patients were summarized after Gamma Knife radiosurgery for brain metastases from melanoma.

The median survival time was 6.7 months. Local tumor control was 81 % at 12 months 166.

In 2011 Liew, Lunsford and colleagues reviewed 333 consecutive patients with melanoma who

underwent SRS for 1570 brain metastases from cutaneous and mucosal/acral melanoma. Sustained

local tumor control was achieved in 73% of the patients and actuarial survival rates were 25% at

12 months. There were 6% asymptomatic and 7% symptomatic radiation effects. Patients with ≤ 8

brain metastases, no prior WBRT, and the recursive partitioning analysis Class I had extended

survivals (median 54.3 months) 156. In 77 patients who had been treated with Gamma knife

radiosurgery for brain melanoma metastases, growth control was achieved in 89.1% of the tumors.

The median survival was 7 months (range 0-73 months) after GKS and 67 months (range 4-327

months) from the time of diagnosis. A total tumor volume of less than 5 cc was associated with

longer survival (P = 0.041). Survival was significantly longer in recursive partitioning analysis in RPA

class 1 (22 months) than RPA class 2 (7 months) and RPA class 3 (3 months; P = 0.008) 75.

Powell and colleagues had previously reviewed the records of 50 patients with melanoma, who had

undergone Gamma Knife treatment. The actuarial 12-month rate for freedom from local

progression for melanoma was 63.0%. The percentage of coverage of the prescribed dose to target

volume was the only treatment-related variable associated with local control: 12-month actuarial

rate of freedom from local progression was 71.4% for lesions receiving >or= 90% coverage versus

0.0% for lesions receiving < 90% (p = 0.00048)147.

Chemotherapy

The survival of patients with melanoma brain metastases managed with ipilimumab and definitive

radiosurgery can exceed the commonly anticipated 4-6 months. Using ipilimumab in a supportive

treatment paradigm of radiosurgery for brain oligometastases was associated with an increased

median survival from 4.9 to 21.3 months, with a 2-year survival rate of 19.7% versus 47.2% 167.

Another study demonstrated in 2015 that survival of patients with melanoma brain metastases

treated with ipilimumab combined with SRS may be comparable to patients without brain

metastases with median survivals of 29.3 months from date of stage IV for melanoma brain

metastases treated with ipilimumab and 33.1 months for patients without brain metastases,

Page 30: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

30

respectively (HR = 0.93, P = 0.896) 168. In another retrospective study, administration of ipilimumab

did not improve intracerebral disease control or survival in patients with limited brain metastases

who received Gamma Knife SRS. The 6-month OS was 56% in patients who received ipilimumab and

45% in those who received SRS alone (P =0.18) 169

A small study suggested that melanoma patients with brain metastases harboring BRAF mutation

appear to be a distinct sub-group with a favorable response to vemurafenib and radiation therapy

and acceptable morbidity 170. While interrupting BRAF inhibitors treatment for melanoma patients

is for still advised for other radiation therapies, interruption does not appear necessary in

combination with stereotactic radiosurgery. Among 53 GKRS carried out in 30 patients who ever

received BRAF inhibitors and Gamma-Knife radiosurgery, 33 Gamma-Knife radiosurgery were

carried out in 24 patients while under BRAF inhibitors treatment, and no increased side effects were

observed 171.

Brain Metastases from Colo-rectal Carcinoma

A total of 6 clinical series after Gamma Knife radiosurgery treatment of colorectal cancer brain

metastases with a total of 373 published patients describing a local tumor control ranging between

71% - 96% with a median local control > 85% in 3/6 studies comprising 61% of published patients

(227/373) 172-177. The addition of WBRT to radiosurgery did not improve survival and local tumor

control rates 176,177.

There are few reports on the effect of Gamma Knife surgery for brain metastases from colorectal

cancer. Skeie, Pedersen and colleagues from the Haukeland University Hospital, Bergen, Norway

studied 80 patients with 140 metastases with mean tumor volume of 6.13 cm3 (0.01-35.5); the

prescription dose was 21.1 Gy. Growth control was achieved in 76.9% and was better if a high

prescription dose of 25 Gy was used, 88.4% vs. 71.4% (P = 0.017), or if tumor volume was <5 cm3

(86.4%). The median survival was 6 months (range 0-75) after GKS. Radiation-induced edema was

seen in 22.1%, but was asymptomatic in 93.8% 178.

Matsunaga, Fujino and associates analyzed the outcomes in 152 patients with 616 brain metastases

from colorectal cancer who had been treated with Gamma Knife at a mean prescription dose of 18.5

Gy (mean tumor volume 2.0 cm3). The local tumor growth control rate was 91.2%. The significant

factors for unfavorable local tumor growth control, based on multivariate analysis, were larger

Page 31: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

31

tumor volume (p = 0.001) and lower margin dose (p = 0.016). The median overall survival time was

6 months. The cause of death was systemic disease in 112 patients and neurological disease in 13

patients 179. A virtually identical tumor control rate was seen by others: Da Silva, Sheehan and

colleagues from the University of Virginia, Charlottesville performed a retrospective review of 40

patients who had undergone Gamma Knife treatment for a total of 118 metastases from

gastrointestinal cancers demonstrating a tumor control rate of 91% and a median survival time of

6.7 months 172.

In a rare comparison of outcome between Gamma Knife and fractionated whole brain

radiotherapy, Park and colleagues reviewed 56 patients with brain metastases from advanced

gastric cancer, treated with GKR or WBRT to assess prognostic factors affecting survival. Fifteen

patients with brain metastases from advanced gastric cancer were treated with Gamma Knife and

the median survival after treatment was 40.0 weeks [95% confidence interval (CI) 44.9-132.1 weeks]

and median survival after WBRT was 9.0 weeks. The use of Gamma Knife was correlated with

prolonged survival in univariate and multivariate analyses 180.

Meningiomas

Meningiomas are considered a classical indication for open microsurgery, but the surgical resection

has to be complete to avoid local tumor recurrences. Microsurgery has been developed

considerably within the recent decade but reaches its natural limitations in many anatomical

locations in the brain. In frequent situations a complete resection of a meningioma is still difficult

to achieve without significant side effects. Complete surgical removal of meningiomas in functional

anatomical locations can be associated with a significant risk for postoperative neurological deficits.

Consequently, the close involvement of eloquent or sensitive structures makes complete tumor

resections virtually impossible when the neurological function is supposed to be preserved. Critical

Page 32: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

32

anatomical regions are skull base, particularly the cavernous sinus and the petroclival region, but

even complete resections of parafalcine meningioma can be complicated when the sagittal sinus is

infiltrated.

Hence, when relevant and sensible structure are involved, the meningioma is often resected

incompletely in order to preserve neurological function, but long term follow up studies have

demonstrated that incompletely resected meningiomas carry a significant risk for symptomatic

recurrences. Stereotactic radiosurgery has gained an important role in these situations and also in

cases of smaller meningioma, which do not necessitate a surgical resection. Microsurgery can be

completed or even replaced by Gamma Knife radiosurgery, which has changed the therapeutic

spectrum in a significant way.

Treatment Doses

The necessary radiation doses to achieve tumor control are an important issue. While the majority

of Gamma Knife centers apply doses of 15-16Gy to the tumor margin, Iwai studied 108 patients with

benign cranial base meningiomas after low-dose Gamma Knife radiosurgery at a median

prescription dose of12 Gy (median tumor volume 8.1 cm3). After a mean follow-up of 86.1 months

the actuarial progression-free survival rate, including malignant transformation and outside

recurrence, was 93% at 5 years and 83% at 10 years and permanent radiation injury occurred in 6%

181. On the other hand, a recent multicenter study of 763 patients with benign sellar and parasellar

meningiomas showed that a tumor margin dose < 13 Gy significantly increased the likelihood of

tumor progression after Gamma Knife treatment 182.

Tumor Control in Meningiomas in General

Few data are available concerning the natural history of meningiomas and the necessity to treat

postoperative remnants has sometimes been questioned, but it has been shown that benign

meningiomas grow slowly at highly variable growth rates with median doubling times ranging

between 415 days and 8 years 183,184.

For the evaluation of the natural history of petroclival meningiomas, Van Havenbergh, Samii and

colleagus followed patients at a median of 85 months with regular clinical and radiological

Page 33: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

33

monitoring. After a median follow-up of 85 months (range 48 to 120 months), radiological tumor

growth was observed in 76% of the cases with associated functional deterioration in 63% of the

growing tumors 185 .

As a result, recent years have seen an essential modification in clinical neurosurgical approaches of

meningioma, a shift away from aggressive surgical solutions toward a disease management with

lower invasiveness. The current priority emphasizes the patient’s quality of life and avoidance of

postoperative defects. Gamma Knife treatment allows a less aggressive upfront surgical approach

with lower morbidity and lower complication rates. The aim of Gamma Knife treatments of

meningioma is to achieve a growth arrest, in other words: local tumor control. The Gamma Knife

treatment however, has a clear limitation in the tumor volume. In larger tumors the neurosurgical

tumor removal is essential, but the more “sensible” parts of the tumor can be left in place and later

be treated with the Gamma knife.

In summary Gamma Knife radiosurgery allows less aggressive or minimal invasive tumor resections

and helps to reduce treatment induced side effects. Most large series after Gamma Knife

radiosurgery demonstrate 5 year-actuarial tumor control rates for patients with grade I

meningiomas in excess of 90%.

The 5-year-actuarial tumor control after Gamma Knife treatment of meningioma was reported

as ranging between 91% and 99% of patients in 13 published Gamma Knife studies comprising

6268 meningioma patients 181,186-197.

Under the auspices of the European Gamma Knife Society (EGKS) a retrospective observational

analysis of Gamma Knife radiosurgery for meningioma was carried out at 15 participating centers

comprising 4565 consecutive patients harboring 5300 benign meningiomas. Detailed results from

3768 meningiomas were analyzed and published. The 5- and 10-year progression-free survival rates

were 95.2% and 88.6%, respectively. Tumor control was higher for imaging defined tumors vs grade

I meningiomas (P < .001), for female vs male patients (P < .001), for sporadic vs multiple

meningiomas (P < .001), and for skull base vs convexity tumors (P < .001). Permanent morbidity rate

was 6.6% at the last follow-up 191.

Similarly, Kondziolka, Flickinger and colleagues had studied a cohort of 972 patients with 1045

intracranial meningiomas treated with Gamma Knife radiosurgery as definitive management during

Page 34: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

34

an 18-year period. The overall control rate for patients with benign meningiomas (WHO Grade I)

was 93%. For patients with WHO Grade II and III tumors, tumor control was 50 and 17%,

respectively. Delayed resection after radiosurgery was necessary in only 5% at a mean of 35 months.

After 10 years, Grade 1 tumors were still controlled in 91%. The overall morbidity rate was 7.7%.

Symptomatic peritumoral imaging changes developed in 4% of the patients at a mean of 8 months

198.

Similar results with 5-year actuarial control rates of 87% for typical meningiomas, 49% for atypical

tumors and 0% for malignant lesions has also been shown previously by Malik, Kemeny and

colleagues in a systematic review of a consecutive series of 309 meningiomas treated with gamma

knife stereotactic radiosurgery. Complications from radiosurgery were rare, occurring in 3% of

tumors, and were most frequently trigeminal and eye movement disturbances treating cavernous

sinus meningiomas 199.

Long-Term Outcome

In an attempt to determine whether long-term outcomes remain stable over time, Kondziolka,

Lunsford and colleagues studied 290 consecutive patients who had undergone Gamma Knife

radiosurgery for a meningioma (median tumor margin dose 15 Gy; median tumor volume 5.5 mL).

The overall tumor control rate was 91%. And the 10- and 20-year actuarial rates of freedom from

tumor progression of the targeted tumor were 87.7%±2.5% and 87.2%±4.2%. Of 234 patients who

had symptoms before SRS, 26% improved, 54% had no change in symptoms and 20% gradually

worsened. Adverse radiation effects were detected in 3.1% of patients 200.

In a retrospective review of 251 patients having SRS for imaging-defined intracranial meningiomas

Pollock found a 3- and 10-year local control rate of 99.4% with 3.2% of patients having temporary

and 9.2% permanent symptomatic radiation-related complications. Radiation-related complications

were associated with convexity/falx tumors and increasing tumor volume 201 .

Large Meningioma

Starke, Sheehan and associates reviewed 75 patients with tumors greater than 8 cm3 in volume,

Page 35: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

35

which was defined as a large tumor. After a mean follow-up of 6.5 years (range 0.5-21 years), the

tumor volume was unchanged or decreased in 84%. Actuarial rates of progression-free survival at

5 and 10 years were 88.6%, and 77.2%, respectively 202.

In Cox multivariable analysis, covariates associated with tumor progression were history of

radiotherapy (p=0.006), and tumor volume greater than 14 cm3 (p=0.066). Neurological function

was deteriorated in 17%. Tumor progression was present in 64% of patients with new or worsening

neurological decline 202.

actuarial tumor control rate at 5

year median follow-up Number of patients Reference

91 % 72 months 168

Flannery TJ, Kano H, Lunsford LD, Sirin S, Tormenti

M, Niranjan A, et al: Long-term control of

petroclival meningiomas through radiosurgery.

Journal of neurosurgery 112:957-964, 2010

92.8% 30.5 months 92

Roche PH, Regis J, Dufour H, Fournier HD, Delsanti

C, Pellet W, et al: Gamma knife radiosurgery in the

management of cavernous sinus meningiomas.

Journal of neurosurgery 93 Suppl 3:68-73, 2000

93 % 964

Lee JY, Kondziolka D, Flickinger JC, Lunsford LD:

Radiosurgery for intracranial meningiomas. Prog

Neurol Surg 20:142-149, 2007

93 % 86.1 months 108

Iwai Y, Yamanaka K, Ikeda H: Gamma Knife

radiosurgery for skull base meningioma: long-term

results of low-dose treatment. Journal of

neurosurgery 109:804-810, 2008

93.2 % 29 months 219

Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD:

Gamma knife radiosurgery of imaging-diagnosed

intracranial meningioma. International journal of

radiation oncology, biology, physics 56:801-806,

2003

94.1 % 47 months 40

Maruyama K, Shin M, Kurita H, Kawahara N,

Morita A, Kirino T: Proposed treatment strategy

for cavernous sinus meningiomas: a prospective

study. Neurosurgery 55:1068-1075, 2004

95.2 % 63 months 3768

Santacroce A, Walier M, Regis J, Liscak R, Motti E,

Lindquist C, et al: Long-term tumor control of

benign intracranial meningiomas after

radiosurgery in a series of 4565 patients.

Neurosurgery 70:32-39; discussion 39, 2012

96 % 48.2 months 138

Nicolato A, Foroni R, Alessandrini F, Maluta S,

Bricolo A, Gerosa M: The role of Gamma Knife

radiosurgery in the management of cavernous

sinus meningiomas. International journal of

radiation oncology, biology, physics 53:992-1000,

2002

Page 36: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

36

97.5 %

(at 4 years) 48 months (mean) 46

Aichholzer M, Bertalanffy A, Dietrich W, Roessler K,

Pfisterer W, Ungersboeck K, et al: Gamma knife

radiosurgery of skull base meningiomas. Acta

neurochirurgica 142:647-652; discussion 652-643,

2000

87% 309 Malik et al. Br J Neurosurg. 2005 199

97.9 % 60 months 368

Kollova A, Liscak R, Novotny J, Jr., Vladyka V,

Simonova G, Janouskova L: Gamma Knife surgery

for benign meningioma. Journal of neurosurgery

107:325-336, 2007

99 %

(overall control in 48 mo) 46 months 66

Hayashi M, Chernov M, Tamura N, Izawa M,

Muragaki Y, Iseki H, et al: Gamma knife robotic

microradiosurgery for benign skull base

meningiomas: tumor shrinkage may depend on the

amount of radiation energy delivered per lesion

volume (unit energy). Stereotactic and functional

neurosurgery 89:6-16, 2011

94 % 103 months (mean) 36

Zachenhofer I, Wolfsberger S, Aichholzer M,

Bertalanffy A, Roessler K, Kitz K, et al: Gamma-knife

radiosurgery for cranial base meningiomas:

experience of tumor control, clinical course, and

morbidity in a follow-up of more than 8 years.

Neurosurgery 58:28-36; discussion 28-36, 2006

96 % 78 months 255

Starke RM, Williams BJ, Hiles C, Nguyen JH,

Elsharkawy MY, Sheehan JP: Gamma knife surgery

for skull base meningiomas. Journal of

neurosurgery 116:588-597, 2012

Reported 5 year-actuarial tumor control after Gamma Knife treatment of meningioma

Specific Anatomical Meningioma Locations

Cavernous Sinus and Skull Base Meningiomas

Starke and Sheehan and colleagues reviewed the outcomes in 255 patients with skull base

meningiomas treated at the University of Virginia and found that actuarial progression-free survival

at 5, and 10 years was 96%, and 79%, respectively. In Cox multivariate analysis, tumor progression

included age greater then 65 years decreasing dose to tumor margin. At most recent clinical follow-

up 10% of patients had deteriorated in their neurological condition. In multivariate analysis, the

factors predictive of new or worsening symptoms were increasing duration of follow-up, tumor

Page 37: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

37

progression, decreasing maximum dose, and petrous or clival location versus parasellar, petroclival,

and cerebellopontine angle location 197.

Pollock, Foote and colleaues studied 115 patients with cavernous sinus meningiomas with 5%

showing tumor progression (in field, n = 3; marginal, n = 3) at a median of 74 months (range 42-145

months) after SRS. The local tumor control rate was 99% at 5 years and 93% at 10 years after SRS.

Fourteen patients (12%) had permanent complications at a median onset of 23 months. The

complication rate for patients with a treatment volume of 9.3 cm3 or less was 3% compared with

21% for patients with a treatment volume greater than 9.4 cm3 203.

Skeie analyzed 100 patients with meningiomas involving the cavernous sinus who had received

Gamma Knife radiosurgery at Haukeland University Hospital, Bergen, Norway and who were

followed for a mean of 82.0 months showing tumor growth control in 90.4% of benign meningiomas.

The 5- and 10-year actuarial tumor growth control rates were 94.2%, and 91.6%, respectively. The

complication rate was 6 %. At last follow-up, 88.0% of patients were able to live independent lives

204 .

Similarly, 98 patients with skull base meningiomas were treated with Gamma Knife radiosurgery at

Teikyo University School of Medicine, Tokyo, Japan and were followed a median period of 53.2

months (12.2-204.4 mo), showing local tumor control rates were of 86.9% and 78.9% at 5 years

and 10 years, respectively. Tumors with volume of 4 cm3 or smaller (97.5% vs. 76.1% at 5 years, p =

0.001) and tumors completely included within the isodose line of 14 Gy or more (p = 0.0006) showed

higher local control rates. Cranial nerve deficits deteriorated in 3% 205.

A series of 192 patients with skull base meningiomas treated with Gamma knife radiosurgery at the

Na Homolce Hospital, Prague had been published in 2004. This series is interesting since contact

with the chiasma or optic tract was present in 32% and was not regarded as a contraindication for

gamma knife radiosurgery. Neurological deficit improved in 63% of patients, temporary morbidity

occurred in 11% and persistent morbidity remained in 4.5%. The risk for the development of a

radiation –induced edema was lower in patients with no history of edema prior to radiosurgery and

in meningiomas of the posterior skull base and at prescription ≤ 14 Gy 206 .

Page 38: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

38

A series of 101 patients with skull-base meningiomas were treated with Gamma Knife at Nakamura

Memorial Hospital, Sapporo, Hakkaido, Japan with a mean follow-up period of 51.9 months showing

a tumor control rates of 95.5% in cavernous sinus meningiomas and 98.4% in posterior fossa and

skull-base meningiomas 207.

In a series of 63 patients with skull base meningiomas treated with Gamma Knife radiosurgery who

had been followed for more than 48 months, the actuarial 5-year tumor control rate was 90.2%

(mean tumor volume: 6.5 cm(3): range 0.5-18.4 cm(3), mean marginal dose :12.6 Gy: range, 7.0-

20.0 Gy) 208. Similarly a retrospective review of 136 patients with meningiomas who received

gamma knife radiosurgery progression-free survival rates at 5, and 10 years were 95% and 85%,

respectively 209.

Sellar and Parasellar Meningiomas

Due to their involvement of neurovascular and endocrine structures, complete resection of

parasellar and sellar meningiomas can be associated with significant morbidity, and incomplete

resections are common. At 10 centers a multicenter study of patients with benign sellar and

parasellar meningiomas was conducted through the North American Gamma Knife Consortium. and

763 patients were identified with 50.7% having undergone at least one resection before Gamma

Knife. The median follow-up after GKRS was 66.7 months (range 6-216 months). At the last follow-

up, tumor volumes remained stable or decreased in 90.2% of patients and actuarial progression-

free survival rates at 5 and 8 years were 95% and 88%, respectively. A tumor margin dose < 13 Gy

significantly increased the likelihood of tumor progression after GKRS. At the last clinical follow-

up, 13.8% of patients had experienced symptom progression 182.

The group University of Virginia, Charlottesville had separately analyzed the outcomes of Gamma

Knife surgery used in 138 patients with parasellar meningiomas at a median follow-up duration 75.5

months (range 24-216 months) and showed that in 86% the tumor volume was unchanged or had

decreased at last follow-up while 10% developed new cranial nerve palsies. Kaplan-Meier analysis

demonstrated radiographic progression-free survival at 5 and 10 years to be 95.4% and 69%,

respectively 210.

Page 39: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

39

Petroclival Meningiomas

A multicenter study of 254 patients with a benign petroclival meningioma was conducted through

the North American Gamma Knife Consortium. One hundred and forty patients were treated with

upfront radiosurgery, and 114 following surgery. At mean follow up of 71 months (range 6-252),

tumor volumes increased in 9 % of tumors, remained stable in 52 %, and decreased in 39 %. Kaplan-

Meier actuarial progression free survival rates at 10 years were 84 %. At last clinical follow-up,

93.6 % of patients demonstrated no change or improvement in their neurological condition whereas

6.4 % of patients experienced progression of symptoms 211.

Petroclival meningiomas were treated with Gamma Knife radiosurgery in 168 patients and analyzed

by Flannery, Kondziolka and colleagues after a median follow-up of 72 months (median tumor

volume: 6.1 cm3; range 0.3-32.5 cm3, median prescription dose: 13 Gy;range 9-18 Gy) showing

overall 5- and 10-year progression-free survival rates of 91 and 86%, respectively. Significant risk

factors for tumor progression were a tumor volume > or = 8 cm3 (p = 0.001) and male sex (p =

0.02) 212.

Convexity Meningiomas

The same group also analyzed 125 patients with convexity meningiomas managed using Gamma

Knife radiosurgery. For patients with benign tumors (Grade I) and those without prior surgery, the

actuarial tumor control rate was 95.3+/-2.3% and 85.8+/-9.3%, respectively. The overall morbidity

rate was 9.6%. Symptomatic peritumoral imaging changes compatible with edema or adverse

radiation effects developed in 5%, at a mean of 8 months 213.

Posterior Fossa Meningioma

At 7 medical centers participating in the North American Gamma Knife Consortium, 675 patients

undergoing SRS for a posterior fossa meningioma were identified (mean tumor volume 6.5 cm3,

median margin dose of 13.6 Gy; range 8-40 Gy). At a mean follow-up of 60.1 months, tumor control

was achieved in 91.2% of cases. Actuarial tumor control was 81% at 10 years after radiosurgery.

Patients with a smaller tumor volume and no prior radiation therapy were more likely to have a

favorable response after radiosurgery 214.

Tentorial Meningioma

Page 40: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

40

Thirty-nine patients with tentorial meningiomas underwent SRS using various Gamma Knife

technologies (median tumor volume 4.6 cm3; range 0.5-36.6 cm3 ; median prescription dose 14 Gy;

range 8.9-18 Gy). The median follow-up period was 41 months (range 6-183 months). At the last

imaging follow-up, tumor volumes decreased in 22 patients (57 %), remained stable in 13 patients

(33 %), and increased in 4 patients (10 %). At the last clinical follow-up, 8 % demonstrated worsening

symptoms 215 .

Cerebellopontine Angle Meningiomas

From 7 institutions participating in the North American Gamma Knife Consortium, 177 patients with

benign cerebellopontine angle meningiomas treated with GKRS were included for analysis (median

tumor volume 3.6 cc; median prescription dose 13 Gy). The mean radiologic follow-up was 47

months. The actuarial rates of progression-free survival at 5 and 10 years were 93% and 77%,

respectively. Male sex (P = .014), prior fractionated radiation therapy (P = .010) were independent

predictors of tumor progression. Symptomatic adverse radiation effects and permanent

neurological deterioration were observed in 1.1% and 9% of patients, respectively 216.

Olfactory Groove Meningioma

In the treatment of olfactory groove meningioma stereotactic radiosurgery provided both long-term

tumor control and preservation of olfaction.

Gamma Knife stereotactic radiosurgery of olfactory groove meningioma was studied in 41 patients

with median clinical and imaging follow-up of 76 and 65 months, respectively (median tumor

volume 8.5 cm3; range 0.6-56.1; mean prescription dose 13 Gy; range 10-20; median estimated dose

to the olfactory nerve was 5.1 Gy; range 1.1-18.1). No patient reported deterioration in olfaction

after radiosurgery. The progression-free tumor control rates were 95% at 2, 10 and 20 years.

Symptomatic adverse radiation effects occurred 7% 217.

Parasagittal and Parafalcine Meningiomas

Hasegawa studied 46 patients harbouring convexity, parasagittal, or falcine meningiomas

undergoing Gamma Knife radiosurgery (median tumor volume: 8 cm3 , median prescription doses:

16 Gy) with actuarial 5- and 10-year local tumor control rates of 87% and 71%, respectively. The

actuarial symptomatic radiation-induced edema rate was 7%. Out of 6 patients with pre-

Page 41: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

41

radiosurgical edema, 4 developed severe panhemispheric edema after GKS . The authors

concluded that radiation-induced edema is more common in convexity, parasagittal, and falcine

meningiomas than skull base meningiomas 218.

A virtually identical outcome was reported by Ding, Sheehan and associates who retrospectively

reviewed 65 patients with 90 WHO Grade I parasagittal (59%) and parafalcine (41%) meningiomas

after a mean MRI follow-up of 56.6 months (median treatment volumes 3.7 cm3, median

prescription dose 15 Gy). The actuarial tumor control rate was 85% at 3 years and 70% at 5 years.

Symptomatic postradiosurgery peritumoral edema was observed in 8.2%. Two patients (4.1%) died

of tumor progression 219.

Zada, Apuzzo and colleagues reviewed in 116 patients who underwent Gamma Knife treatments for

benign intracranial meningiomas (n=116; median tumor volume: 3.4 cm3, median prescription

dose:16 Gy) finding overall tumor control in 94% and an overall 5-year and 10-year actuarial tumor

control rate of 98.9% and 84% with complications appearing in 8%, generally in larger meningiomas

(11 vs 5.7 cm3, P = .003). Tumor progression occurred at a mean time of 7.5 years after GKRS,

reinforcing the need for long-term surveillance despite initial tumor control. Treatment failure was

related to undercoverage of lesions in the majority of cases, with the remainder demonstrating

evidence of abnormal tumor biology 220 .

Combination Surgery/Radiosurgery

Meningiomas with growth onto or into the major venous sinuses, that is, venous meningiomas,

provide management problems regarding their radical removal and preservation of venous drainage.

The relationship to venous structures often precludes radical surgery; the risk of recurrence and

aggressive histology is greater for parasagittal meningiomas than in other locations.

The surgical approach to inresectable tumor remnants would be a ’wait-and-see’ attitude with a

potential removal only if the regrowing tumor has reached a local clinically relevant mass effect.

Gamma Knife radiosurgery of remnant tumors is an alternative option. Treatment, histopathology,

Page 42: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

42

and follow-up data of 100 consecutive patients undergoing surgery for venous meningiomas were

prospectively collected by Mathiesen, Ulfarsson and colleagues at the Karolinska Hospital,

Stockholm. Patients with Simpson grade 1 surgery had a recurrence rate of 10%. Gamma-knife

surgery was considered as a direct postsurgical adjunct or as an adjunct after a period of radiological

follow-up. Patients with deliberate nonradical surgery (Simpson grade IV) had a tumor recurrence

rate of 72%, whereas a combined treatment of direct gamma-knife radiosurgery after a tailored

microsurgical resection (Simpson IV gamma) allowed return to a low recurrence rate of 10% 221.

Malignant and Atypical Meningioma

Atypical or malignant meningiomas are associated with high levels of recurrence and mortality and

a significantly higher local recurrence rate is reported after radiosurgery of these aggressive

meningiomas.

Out of 228 patients with microsurgically treated atypical meningiomas at the Barrow Neurological

Institute, gross total resections were achieved in 58%. Overall 22% demonstrated tumor recurrence

at a median of 20.2 months postoperatively. The recurrence rate for patients receiving SRS was 25%

and for IMRT was 18% 222.

Even after stereotactic radiosurgery atypical or malignant meningiomas have a high risk for local

recurrence: Tumor recurrence after Gamma Knife radiosurgery was identified in 31.4% of patients

with atypical and malignant meningiomas (n=35). Three-year overall survival and progression-free

survival rates were 78.0% and 65.0%, respectively. WHO grade II 3-year OS and progression-free

survival were 83.4% and 70.1%, respectively, while WHO grade III 3-year OS and progression-free

survival were 33.3% and 0%. Larger tumor size predicted poor survival, while nuclear atypia,

necrosis, and increased mitotic rate are risk factors for recurrence 223.

Similarly, another series with atypical meningiomas (n=9) and anaplastic meningiomas (n=7) (mean

tumor volume 7.1 cm3; mean marginal doses 18.8 Gy; mean length of follow-up 37.1 months)

showed stabilization of tumor growth during after Gamma Knife radiosurgery in 29% , which was

significantly associated with small tumor volume, and greater marginal irradiation doses. In this

series 31 % died because of tumor progression within the average time period of 16.8 months after

treatment 224.

Page 43: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

43

A virtually identical tumor control of 34% at 3 years was seen in another series of 30 patients with

(WHO) grade II or grade III meningiomas who had undergone Gamma Knife surgery in Pittsburgh 225,

or in In 22 patients with atypical meningioma who underwent Gamma Knife at the University of

Tokyo Hospital with local control rates of 39%, and 16% at 2 and 5 years, respectively. Hanakita,

Saito and colleagues stated that a margin dose higher than 18 Gy (p = 0.02), and a Karnofsky

Performance Scale (KPS) score of 90 or more (p = 0.02) were factors associated with a longer

duration of tumor control in the univariate analysis 226.

Using a multivariate analysis, Kim, Jung and colleagues found that the mitotic count (≤8 per 10 high-

power fields; HPF) and the MIB-1 proliferation marker labeling index (LI; ≤8%) were significant

favorable prognostic factors for the radiosurgical outcomes of patients with non-benign

meningiomas. The actuarial local tumor control rates for 35 patients with atypical meningiomas at

2 and 3 years after radiosurgery were 53 and 36%, respectively, whereas those for anaplastic

meningiomas were 35% at 1 year and 10% at 2 years 227.

Acoustic Neuroma

Acoustic Neuromas (vestibular schwannomas) are generally slow-growing, intracranial extra-axial

benign tumors that usually develop from the vestibular portion of the eighth nerve. A progressive

unilateral hearing decline is the most common symptom that leads to the diagnosis of a vestibular

schwannoma. Vestibular schwannomas (Acoustic Neuroma) are accounting for approximately 10%

of newly diagnosed intracranial tumors occurring at a frequency of 2000 to 2500 new cases per year

in the United States 228 . Acoustic Neuroma is a benign tumor with anatomically close association

with the vestibular, the cochlear and the facial nerves. With increasing volume acoustic neuromas

can exert pressure on the brainstem. The typical tumor location adjacent to the brainstem in

connection with the close involvement of the described cranial nerves explains the need for tumor

Page 44: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

44

control, but also the problems that arise when these benign tumors are surgically removed, because

generally after open tumor resection hearing cannot be preserved and even the facial nerve can be

endangered by surgery.

As to therapeutic options for acoustic neuromas, the controversies have been fierce and not rarely

polemic. Therapeutic options include microsurgical removal, radiosurgery, or possibly fractionated

radiotherapy and even monitoring without treatment. Previously acoustic neuromas were the

domain of surgery alone. Surgical resection however, is associated with a significant risk for side

effects: loss of hearing, potential facial nerve damage and general morbidity.

A questionnaire concerning postoperative symptoms and the Short Form 36 (SF-36) QOL instrument

was mailed to 97 consecutive patients who had undergone acoustic neuroma surgery via the

translabyrinthine approach. The respondents’ QOL was rated significantly below published norms

and their work capacity was reportedly reduced. The majority (63%) of respondents reported facial

nerve dysfunction postoperatively. Balance problems were exacerbated by movement in 74% of

patients, and 28% believed that their balance problem was incapacitating. Of those who had

experienced recovery of facial nerve function, 63% reported that it had taken longer than 9 months.

32% of respondents reported a change in employment as a result of their acoustic neuroma 229 .

Gamma Knife is highly effective in stopping potential tumor growth while preserving normal

postoperative facial function and hearing with low treatment associated side effects. The goal of

every acoustic neuroma management is to provide the best outcome with the lowest possible risk

level. The long-term results after radiosurgical treatment are comparable and often even superior

to the best results with open microsurgical techniques. Tumor control was achieved in 87-96% of

1179 published patients in 5 studies published between 2002 and 2005 230-234.

Myrseth and colleagues conducted a prospective, open, non- randomized study of treatment-

associated morbidity in patients undergoing either microsurgery or Gamma Knife radiosurgery for

vestibular schwannomas in 91 patients. None of the patients who underwent surgery had

serviceable hearing postoperatively, while 68% patients preserved hearing after Gamma Knife

treatment. Quality of life was significantly better in the Gamma Knife group at 2 years. The mean

hospital stay was 2.5 days (range, 2–5 days) in the Gamma Knife group and 12.5 days (range, 10–30

days) in the surgery group (P< 0.0009, Student’s t test) 235.

Page 45: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

45

Conservative Management of Vestibular Schwannoma

Natural history and outcome was studied following the conservative management of 72 patients

with unilateral vestibular schwannomas. The mean duration of follow-up was 80 months (range 52-

242 months). Hearing deterioration with pure tone averages and speech discrimination scores

occurred irrespective of tumor growth. The mean tumor growth rate for the entire group at the

second review was 1 mm/year (range -0.84-9.65 mm/year). There was significant tumor growth

seen in 38.9%, no or insignificant growth in 41.7% 236. In order to determine the outcomes and risks

of conservative management of acoustic neuromas, gamma-knife radiosurgery, and microsurgery

Yamakami, Yamaura and colleagues performed a systematic review and analyzed data from 903

patients with conservative management, 1475 with Gamma Knife radiosurgery, and 5005 with

microsurgery from 38 studies identified in MEDLINE searches. Conservative management over a

3.1-year period resulted in tumor growth in 51% of acoustic neuromas with an average growth rate

of 1.87 mm per year, and 20% of acoustic neuromas ultimately required surgical intervention. After

conservative management a third of the patients lost useful hearing. Gamma Knife radiosurgery

significantly reduced the percentage of acoustic neuromas that enlarged to 8%, and reduced the

percentage that underwent microsurgery to 4.6% over a 3.8-year period. Microsurgery resulted in

tumor recurrence in 1.8%, in mortality in 0.63%, and major disability in 2.9%, respectively 237.

Due to the invasiveness and associated morbidity of surgery and due to the certainty of

postoperative hearing loss the traditional surgical policy In intracanalicular vestibular schwannoma

has previously been to monitor the tumor and to treat only in case of significant growth or a

significant mass effect. Regis and colleagues tested this ‘wait-and-see strategy’ in 47 patients versus

a control group of 34 patients who had been treated with proactive Gamma Knife surgery in the

treatment of small intracanalicular vestibular schwannomas in 47 patients and who were followed

prospectively. In the wait-and-see group, there was tumor growth in 77%. At 3, 4, and 5 years, the

useful hearing preservation rates were 75%, 52%, and 41% in the wait-and-see group and 77%,

70%, and 64% in the Gamma Knife control group, respectively. The authors concluded that the

chances of maintaining functional hearing and avoiding further intervention were higher in cases

treated by GKS (79% and 60% at 2 and 5 years, respectively) than in cases managed by the wait-

and-see strategy (43% and 14% at 2 and 5 years, respectively). As a result, proactive Gamma Knife

Page 46: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

46

radiosurgery was recommended at the time of diagnosis when hearing is still useful 238.

Similarly Breivik, Lund-Johansen and associates showed that Gamma Knife radiosurgery reduced

the tumor growth rate and thereby the incidence rate of new treatment about tenfold. Hence,

the need for treatment following initial Gamma Knife or conservative management differed at

highly significant levels (log-rank test, P < .001). Prospectively data of patients with unilateral

vestibular schwannoma extending outside the internal acoustic canal had been collected and the

outcome after Gamma Knife treatment (n=113) was compared with patients following conservative

management (n=124). Mean follow-up time was 55.0 months. Serviceable hearing was lost in 76%

after conservative management and in 64% after Gamma Knife. Hearing is lost at similar rates in

both groups. Symptoms and quality of life seem not to be significantly affected by Gamma Knife 239 .

Patients with ≤3 cm vestibular schwannoma who underwent primary microsurgery, gamma knife

surgery, or observation at 2 independent hospitals were identified and were surveyed via mail after

a mean time of 7.7 years after initial treatment, and a total of 539 respondents were analyzed. More

than 75% of all patients had nonserviceable hearing at the last clinical follow-up. Patients in the

surgical series had the greatest hearing loss. In this series active treatment with microsurgery or

Gamma Knife surgery did not appear to be protective, because patients who were observed had the

greatest probability of durable hearing 240.

Tumor Control

In 2004 Regis and colleagues had initially reviewed the outcome in the first 1000 patients with

cerebello-pontine angle schwannomas that were consecutively treated by Gamma Knife at the

LaTimone University Hospital , Marseille. Tumor control at last follow-up was 97%. Globally, a

clinical trigeminal injury was observed in 0.6% of the patients and a facial palsy in 1.3%. There was

clearly a decrease of the incidence of neuropathies with time; no facial palsy being reported among

the last 258 patients. The rate of functional hearing preservation (Gardner) for patients initially in

class I was 77.8% (47.6% for class II) at 3 Years. This rate of functional preservation reached 95%

among patients with tinnitus as a first symptom 233.

Page 47: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

47

In another large retrospective analysis Lunsford, Kondziolka and colleagues analyzed the outcome

after Gamma Knife treatment of vestibular schwannomas in 829 patients with VSs who had been

treated between 1987 and 2002 at the University of Pittsburgh (average tumor volume 2.5 cm3;

median prescription dose 13 Gy, range 10-20 Gy). Tumor control rates without need for additional

treatment at 10 years were 97%. Unchanged hearing preservation was achieved in 50 to 77% of

patients (up to 90% in those with intracanalicular tumors). Facial neuropathy risks were less than

1%. Trigeminal symptoms were detected in less than 3% of patients whose tumors reached the

level of the trigeminal nerve. No patient sustained significant perioperative morbidity. The average

duration of hospital stay was less than 1 day 228.

Murphy, Suh and colleagues analyzed the long-term outcome of 117 patients with vestibular

schwannoma who had been treated with Gamma Knife radiosurgery at the Cleveland Clinic (mean

tumor volume 1.95 ± 2.42 ml; prescription dose 13Gy in 82%, 13Gy in 14%). Imaging-documented

tumor progression was present in 7.8%, but in 3/8 of these the lesion eventually stabilized. Only 5

patients required a neurosurgical intervention. The estimated 1-, 3-, and 5-year rates of progression

to surgery were 1, 4.6, and 8.9%, respectively. One patient (1%) developed trigeminal neuropathy,

5% developed permanent facial neuropathy, 4% reported vertigo, and 18% had new gait imbalance

following Gamma Knife treatment 241.

The high local control rate was confirmed by Delbrouck, Massager and colleagues from the Erasmus

Hospital of Brussels who demonstrated after Gamma Knife radiosurgery in 415 patients that

unilateral vestibular schwannoma were stable or declining in size in 90.44% of cases. At minimum

one-year audiological follow-up, serviceable hearing was preserved in 65.97% 242 .

Chung, Pan and colleagues performed a retrospective study of 195 patients with acoustic neuroma

who had been treated with Gamma Knife radiosurgery (mean tumor volume 4.1 cm3; range 0.04-

23.1 cm3; prescription dose: 11-18.2 Gy). During a median follow-up period of 31 months resection

was avoided in 96.8% of cases. Twelve of 20 patients retained serviceable hearing. Two patients

experienced a temporary facial palsy. Two patients developed a new trigeminal neuralgia. There

was no treatment-related death 231.

Long-term Outcomes

Page 48: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

48

The recording of long-term outcomes is essential in the treatment of slowly growing benign tumors.

Hasegawa, Yoshida and colleagues followed 73 patients harbouring acoustic neuromas over median

of 135 months after Gamma Knife treatment (mean tumor volume 6.3 cm3, mean prescription

doses 14.6 Gy). The actuarial 10-year progression-free survival rate was 87% overall, and 93% in

patients with tumor volumes less than 10 cm3. No patient experienced malignant transformation

232.

Sun and Liu from the Beijing Neurosurgical Institute assessed the long-term clinical outcomes in 200

patients who underwent Gamma Knife surgery (median tumor margin dose 13.0 Gy; range 6.0-14.4

Gy; median tumor volume 3.6 cm3 ; range 0.3-27.3 cm3). After a median duration of follow-up of

109 months (range 8-195 months), the tumor control rate was 89.5%. Using the Kaplan-Meier

method, the authors found the estimated 5-, 10-, and 15-year tumor control rates to be 93%, 86%,

and 79%, respectively; and the estimated 3-, 5-, and 10-year hearing preservation rates were 96%,

92%, and 70%, respectively. For unknown reasons, the rate of side effects in this study was

comparatively high, since 13.7% exhibited transient mild facial palsy or facial spasm was seen and

1.1% persistent mild facial palsy, while 2.6% suffered from persistent mild facial numbness 243.

Long-term data were reviewed from a consecutive cohort of 128 patients with vestibular

schwannoma treated with Gamma knife radiosurgery at Karolinska University Hospital with a

median follow-up time of 104 months (range 11-165) and radiological median follow-up of 86

months (range 5-170). Tumor control was achieved in 92 % of patients after a single Gamma Knife

treatment. Neither pre-treatment growth of the vestibular schwannoma, or a large tumor size (Koos

grade 3 & 4) was correlated with a higher degree of treatment failure 244.

Quality of Life

The impact of Gamma Knife radiosurgery on the quality of life 97 of patients with a sporadic

vestibular schwannoma was analyzed using the medical outcome study short form 36 (SF-36) and

Glasgow benefit inventory (mean tumor size was 17 mm; range 6-39 mm; mean marginal dose 11.1

Gy; range 9.3-12.5 Gy). While there was a wide range in individual quality of life results, this study

showed that GKRS for sporadic vestibular schwannoma has little impact on the general quality of

Page 49: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

49

life of the vestibular schwannoma patient 245.

A similar study monitored 59 patients with vestibular schwannoma after Gamma Knife treatment

with a median follow-up of 15 months in a prospective follow-up algorithm including 36-item Short

Form Health Survey (SF-36), Hearing Handicap Inventory, Dizziness Handicap Inventory, Tinnitus

Handicap Inventory, pure-tone average, and speech discrimination hearing scores (Gardner-

Robertson and American Academy of Otolaryngology), performed before and after Gamma Knife

radiosurgery at 1-, 3-, 6-, 12-, and 18-month post-treatment intervals. Approximately 47% of

patients with baseline serviceable hearing maintained serviceable hearing at 12 months. Although

it can be criticized that the study had a relatively short follow-up and approximately 50% survey

completion, it was demonstrated that no significant decline in global quality of life occurred after

Gamma Knife radiosurgery 246.

Long-term data were reviewed from a consecutive cohort of 128 patients with vestibular

schwannoma treated with Gamma knife radiosurgery. Median follow-up time was 104 months

(range 11-165). Data on quality of life (EQ-5D score) were obtained in 90 % of all cases at the end

of follow-up, showing low morbidity and a high quality of life with median index of 0.91 (max. score

1.0) 244 .

Intracanalicular Acoustic Neurinoma

In general, patients with intracanalicular vestibular schwannoma have limited symptoms, while a

surgical approach invariably causes a loss of hearing and other potential side effects. Hence the

‘surgical tradition’ in these tumors has been a wait-and-see strategy until a clinically relevant mass

effect is reached. Since radiosurgery has a considerably lower risk profile, this policy has been

questioned with regard to functional preservation. Regis, Roche and colleagues have followed 47

patients harboring an intracanalicular vestibular schwannomas prospectively over a mean period of

43.8 ± 40 months (range 9-222 months). And have compared this population with a control group

of 34 patients harboring a unilateral intracanalicular vestibular schwannomas who were

consecutively treated by Gamma Knife surgery and had functional hearing at the time of

radiosurgery. In the wait-and-see group, there was tumor growth in 77% . Of the 47 patients in the

Page 50: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

50

wait-and-see group, treatment failure (tumor growth requiring treatment) was observed in 74%,

while treatment failure in the control Gamma Knife group occurred in only 3%. In the wait-and-

see group 38% experienced more than 10 db of hearing loss and 2 of them became deaf. At 3, 4,

and 5 years, the useful hearing preservation rates were 75%, 52%, and 41% in the wait-and-see

group and 77%, 70%, and 64% in the Gamma Knife group, respectively 247.

The chances of maintaining functional hearing and avoiding further intervention were 60% at 5

years in cases treated by Gamma Knife and 14% at 5 years in cases managed by the wait-and-see

strategy 247. The consequence is a policy change with a general recommendation of earlier Gamma

Knife treatment even without relevant mass effect due to an improved chance of functional

preservation.

Hearing Preservation

Many patients with acoustic neuromas have hearing function at diagnosis the current priority is to

maintain it.

In 2009 Yang, Parsa and colleagues had performed a systematic analysis of the literature in English

reviewing a total of 254 published studies reported assessable and quantifiable outcome data of

patients undergoing radiosurgery for vestibular schwannomas. A total of 74 articles including 5825

patients met the inclusion criteria of reporting assessable and quantifiable outcome data of patients

with preserved hearing with an average overall follow-up of 41.2 months. It was shown that

practitioners who delivered an average prescription dose of 12.5 Gy reported having a higher

hearing preservation rate (12.5 Gy=59% vs. >12.5 Gy=53%, p=0.0285). Age of the patient was not a

significant prognostic factor for hearing preservation rates (<65 years=58% vs. >65 years=62%;

p=0.4317). The average overall follow-up was 41.2 months. The data suggest that an overall hearing

preservation rate of about 57% can be expected after radiosurgical treatment 248.

In a similar review from the same group published in 2010 it was shown that tumor volume was not

Page 51: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

51

predictive for hearing preservation and that age was not a significant prognostic factor for hearing

preservation rates either. In fact, there was a trend toward improved hearing preservation rates in

older patients (56% at < 65 years vs 71% at > or = 65 years of age; p < 0.1134) 249.

In an attempt to distinguish the impact of radiosurgery from the natural course of hearing

deterioration, Yomo, Regis and colleagues showed mean annual hearing decrease rates before and

after Gamma Knife treatment of 5.39 dB/year and 3.77 dB/year, respectively (p > 0.05). A maximum

cochlear dose of less than 4 Gy was found to be the sole prognostic factor for hearing preservation.

This study demonstrated the absence of an increase in annual hearing decrease rate after

radiosurgery as compared with the preoperative annual hearing decrease rate. There was even a

trend indicating a reduction in the annual hearing loss after radiosurgery over the long term 250.

This study demonstrated the absence of an increase in annual hearing decrease rate after

radiosurgery as compared with the preoperative annual hearing decrease rate. There was even a

trend indicating a reduction in the annual hearing loss after radiosurgery over the long term 250.

After Gamma Knife radiosurgery in the management of 415 patients with unilateral vestibular

schwannoma 65.97% had preservation of serviceable hearing (Pure tone average < or = 50 db and

Speech discrimination > or = 50%) at minimum one-year audiological follow-up. The tumors were

stable or declining in size in 90.44% of cases 242.

Tamura, Regis and colleagues evaluated the long-term hearing preservation after gamma knife

radiosurgery for vestibular schwannomas in 74 patients with initially normal or subnormal hearing

(Gardner-Robertson Class 1) after a minimum of 3 years of follow-up (median marginal dose was 12

Gy; range, 9-13 Gy). At the time of the last follow-up evaluation, 78.4% of the patients had

preserved functional hearing. Tumor control was achieved in 93% of the cases. The probability of

preserving functional hearing was higher in patients who had an initial symptom other than hearing

decrease (91.1%), in patients younger than 50 years (83.7%), and in those treated with a dose to the

cochlea of less than 4 Gy (90.9%) 251.

In a separarately published analysis, Regis showed that the functional hearing preservation at 3

years is 77.8% when the patient is initially in Gardner-Robertson stage 1, 80% in patients with

Page 52: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

52

tinnitus as a first symptom and 95% when the patient has both. In these patients, the probability

of functional preservation at 5 years is 84% 252.

Following Gamma Knife radiosurgery for acoustic neuroma in 77 patients who had serviceable

hearing (Gardner-Robertson [GR] Class I or II), Kano, Lunsford and colleagues evaluated tumor

control and hearing preservation with respect to tumor volume, imaging characteristics, and nerve

and cochlear radiation dose; (median tumor volume 0.75 cm3 ; range 0.07-7.7 cm3, median

prescription dose 12.5 Gy; range 12-13 Gy). At diagnosis, a greater distance from the lateral tumor

to the end of the internal auditory canal correlated with better hearing function. Serviceable

hearing was preserved in 71% of all patients and in 89% (46 patients) of those with GR Class I

hearing. Significant prognostic factors for maintaining the same GR class included pre-SRS GR Class

I hearing, a speech discrimination score (SDS) >or= 80%, a pure tone average (PTA) < 20 dB, and a

patient age < 60 years. Significant prognostic factors for serviceable hearing preservation were (all

pre-SRS) GR Class I hearing, an SDS >or= 80%, a PTA < 20 dB, a patient age < 60 years, an

intracanalicular tumor location, and a small tumor volume (< 0.75 cm3) 253.

Han, Jung and colleagues proposed a classification system based on PTA score and the IL I-V of

the auditory brainstem response to predict the rate of hearing preservation in each individual.

The mean baseline values for the interlatency (IL) of waves I through V (IL I-V) on auditory brainstem

response was determined and the pure tone average was calculated by averaging audiometric

masked bone conduction responses at 500 Hz, 1000 Hz, and 2000 Hz in 119 patients with vestibular

schwannoma with serviceable hearing who had undergone Gamma Knife treatment. In

multivariate analysis, the PTA score and IL I-V were significant and independent factors for

hearing preservation: By using the PTA score and IL I-V, the patients were classified into 4 groups

with serviceable hearing after SRS being 89.6%, 64.0%, 25.8%, and 6.7%, respectively 254. Others

confirmed in a multivariate analysis that pretreatment ipsilateral pure tone average (p < 0.001) was

statistically significantly associated with time to non-serviceable hearing 255.

Younger GR Class I patients had a significantly higher probability of retaining functional hearing

even at the 10-year follow-up: Boari, Mortini and associates studied 379 patients who underwent

Gamma Knife radiosurgery for vestibular schwannomas (median clinical follow-up 69.5 months;

median tumor volume median 1.2 cm3, range 0.013-14.3 cm3, median prescription dose 13 Gy;

Page 53: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

53

range 11-15 Gy): Control of the tumor with Gamma Knife was achieved in 97.1% of the patients. The

overall rate of preservation of functional hearing at the long-term follow-up was 71% for patients

with hearing classified as Gardner-Robertson (GR) Class I, and 93% among cases of GR Class I hearing

in patients younger than 55 years 256.

In patients with vestibular schwannoma and well preserved hearing (Gardner-Robertson class I) who

had been treated with Gamma Knife radiosurgery, serviceable hearing retention rates (GR grade I

or II) were 100% for patients who had no subjective hearing loss before Gamma Knife (n=25). In

patients who reported subjective hearing loss before Gamma Knife, serviceable hearing retention

rates were 81% at 1 year, 60% at 2 years, and 57% at 3 years after radiosurgery. Patients with a pure

tone average (PTA) <15 dB before GKRS had significantly higher rates of preservation of GR class I

or II hearing 257.

At median cochlear maximum doses of 6.9 Gy (range 1.6-16 Gy) and a median audiological follow-

up of 35 months (range 6-58 months), the 1-, 2-, and 3-year actuarial rates of maintaining

serviceable hearing were 93%, 77%, and 74%, respectively, in 40 patients with vestibular

schwannoma with serviceable hearing who had been treated using Gamma Knife surgery. Patients

who received a mean cochlear dose less than 3 Gy had a 2-year hearing preservation rate of 91%

compared with 59% in those who received a mean cochlear dose of 3 Gy or greater (p = 0.029) 258.

A cohort of 60 patients with unilateral sporadic intracanalicular vestibular schwannomas were

studied: at a median follow-up duration of 62 months, the actuarial rates of serviceable hearing

preservation were 70%, 63%, and 55% at 1, 2, and 5 years after radiosurgery, respectively. In

multivariate analysis, transient volume expansion of ≥20% from initial tumor size was a statistically

significant risk factor for loss of serviceable hearing and hearing deterioration, while the cochlear

radiation dose did not reach statistical significance 259.

Delbrouck, Massager and associates studied 415 patients with unilateral vestibular schwannoma

who had undergone Gamma Knife radiosurgery. Serviceable hearing was preserved in 65.97% of

144 patients who had serviceable hearing at the time of the treatment (GR class I and GR class II).

Of GR class I patients 74.64% preserved serviceable hearing. The tumor was stable or declining in

size in 90.44% of cases 242.

A total of 68 patients with vestibular schwannoma and well preserved hearing (Gardner-Robertson

Page 54: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

54

class I) were treated with Gamma Knife radiosurgery. In patients with no subjective hearing loss,

serviceable hearing retention rates (GR grade I or II) were 100% compared with 81% at 1 year, 60%

at 2 years, and 57% at 3 years after GKRS for patients with subjective hearing loss. At 2 to 3 years

after GKRS, patients without subjective hearing loss or a PTA <15 dB had higher rates of grade I or

II hearing preservation 257 .

Threshold Doses for the Preservation of Hearing

In a review of the published literature in 2009 Yang, Parsa and colleagues concluded that patients

treated with a prescription dose of 12.5 Gy were more likely to have preserved hearing 248.

Tamura, Regis and colleagues found that the probability of preserving functional hearing was higher

in patients who had been treated with a dose to the cochlea of less than 4 Gy 251. In 154 patients

with vestibular schwannoma who had undergone Gamma Knife surgery Yomo, Regis and colleagues

showed that maximum cochlear dose of less than 4 Gy was found to be the sole prognostic factor

for hearing preservation 250. Following Gamma Knife radiosurgery for acoustic neuroma in 77

patients who had serviceable hearing (Gardner-Robertson [GR] Class I or II), Kano, Lunsford and

colleagues showed that all patients < 60 years of age who had received a cochlear dose < 4.2 Gy

retained serviceable hearing at 2 years post-SRS 253.

Baschnagel and colleagues found that patients who received a mean cochlear dose less than 3 Gy

had a 2-year hearing preservation rate of 91% compared with 59% in those who received a mean

cochlear dose of 3 Gy or greater (p = 0.029). No patient who received a mean cochlear dose less

than 2 Gy experienced serviceable hearing loss (p = 0.035) 258.

Van Eck and Horstmann had reduced the maximum dose that was applied to the acoustic neuroma

to 20 Gy showing that functional hearing was preserved in 83.4% of the patients with functional

hearing preoperatively 260.

The relevance of the mentioned cochlear threshold doses is still questioned by others: In a group of

60 patients (mean marginal dose 12.2±0.1 Gy; range, 11.5-13.0 Gy) the actuarial rates of serviceable

Page 55: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

55

hearing preservation were 70%, 63%, and 55% at 1, 2, and 5 years after radiosurgery, but in

multivariate analysis, transient volume expansion of ≥20% from initial tumor size was a statistically

significant risk factor for loss of serviceable hearing and hearing deterioration, while the cochlear

radiation dose did not reach statistical significance 259 .

Malignant Transformation

In 2014 Seferis performed a search using PubMed and found a total of 29 reported cases of

malignant transformation in a vestibular schwannoma after radiation therapy261. The authors also

retrieved papers reporting 30 cases of malignant vestibular schwannoma in patients who had not

undergone radiation treatment. Of the irradiated cases of malignant transformation, 40.7% were

patients with neurofibromatosis. In those cases in which histology showed tumors with previously

benign characteristics, the median delay to malignant expression was 72 months. The present meta-

analysis indicates that the risk of malignancy over 20 years in cases in which no radiation treatment

has occurred is 1.32-2.08 per 100,000, and this risk decreases to 1.09-1.74 per 100,000 if cases of

neurofibromatosis are excluded. After radiation treatment, the overall risk over 20 years is 25.1 per

100,000, and this risk decreases to 15.6 per 100,000 if cases of NF are excluded 261 .

In a previous retrospective cohort study Rowe, Kemeny and colleagues had compared the Sheffield

radiosurgery patient database set comprising approximately 5000 patients and 30,000 patient-years

of follow-up with national mortality and cancer registries. In this material, a single new astrocytoma

was diagnosed, whereas, based on national incidence figures, 2.47 cases would have been predicted.

It was concluded that no increased risk of malignancy was detected in this series, supporting the

safety of radiosurgery. The authors concluded that the risks of radiosurgery-induced malignancy

seems small, particularly if such risks are considered in the context of the other risks faced by

patients with intracranial pathologies requiring radiosurgical treatments 262.

Among 1309 patients with intracranial AVMs treated with Gamma Knife radiosurgery at University

of Virginia, Starke, Sheehan and colleagues identified a total of 3 cases of radiosurgically induced

neoplasia 263. More than 10 years after GKRS, 2 patients were found to have an incidental, uniformly

enhancing, dural-based mass lesion near the site of the AVM with radiologic characteristics of a

meningioma. As the lesions have shown no evidence of mass effect, they are being followed with

Page 56: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

56

serial neuroimaging. A third patient was found to have neurologic decline from a tumor in

immediate proximity to an AVM previously treated with proton beam radiosurgery and GKRS. The

patient underwent resection, demonstrating a high-grade glioma. The 3-, 10-, and 15-year incidence

of a radiation-induced tumor is 0% (0/812), 0.3% (1/358), and 2.6% (2/78), respectively. The

cumulative rate of radiosurgically induced tumors in those with a minimum of 10-year follow-up

is 3 in 4692 person-years or 64 in 100,000 person-years. It was concluded that patients had a

0.64% chance of developing a radiation-induced tumor within ≥10 years following GKRS 263.

Facial Nerve Palsy

Side effects after Gamma Knife radiosurgery of vestibular schwannoma are in fact rare. In 2009 a

review of 23 articles representing reporting quantifiable outcome data obtained in 2204 patients

who underwent Gamma Knife radiosurgery for vestibular schwannoma showed an overall facial

nerve preservation rate of 96.2% (average overall follow-up 54.1 +/- 31.3 months. Patients

receiving a prescription dose of ≤ 13 Gy had a better facial nerve preservation rate than those who

received higher doses (<or=13 Gy = 98.5% vs. >13 Gy = 94.7%, P < 0.0001) 264.

Comparison Gamma Knife vs. Surgery

Myrseth, Lund-Johansen and colleagues conducted a prospective, open, nonrandomized study of

treatment-associated morbidity in 91 patients undergoing microsurgery or gamma knife

radiosurgery for vestibular schwannomas with a maximum tumor diameter of 25 mm in the

cerebellopontine angle. Evidence of reduced facial nerve function (House-Brackmann grade 2 or

poorer) at 2 years was found in 46% (13/28) after open microsurgery and in 1.6% (1/60) after

Gamma Knife. No patient who underwent surgery had serviceable hearing postoperatively, while

68% of Gamma Knife patients who had serviceable hearing before treatment, preserved

serviceable hearing 2 years after treatment. The tinnitus and vertigo visual analog scale score, as

well as balance platform tests, did not change significantly after treatment, and working status did

not differ between the groups at 2 years. Quality of life (according to the Glasgow Benefit

Inventory questionnaire) was significantly better in the GKRS group at 2 years. This prospective

Page 57: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

57

study demonstrated significantly better facial nerve and hearing outcomes after Gamma Knife

radiosurgery when compared to open surgery for small- and medium-sized vestibular

schwannomas 235.

Tamura, Regis and colleagues showed that the risk of dry eye and burning eye was considerably

lower in patients after Gamma Knife treatment compared to patients after microsurgery. They

used a functional questionnaire to evaluate patient complaints related to the eye and taste in a

series of 200 patients 3 years after the Gamma Knife radiosurgery of a unilateral vestibular

schwannoma not previously resected. Their answers were compared with those of a group of 200

patients operated on microsurgically. In the microsurgery group 57.6% of (57/99) presented with

permanent postoperative facial nerve palsy while no patient in the Gamma Knife group (0/80)

experienced facial nerve palsy. The answers about taste showed that After Gamma Knife 8.1% of

patients complained of a sensation of altered taste while this symptom occurred in 45.5% of

patients after microsurgery 265.

Regis, Peragut and colleagues used a patient questionnaire to evaluate the functional outcome and

quality of life in a series of 97 consecutive patients with acoustic neuroma treated with Gamma

Knife compared with 110 patients in the microsurgery group. Questionnaire answers indicated that

100% of patients who underwent GKS compared with 63% of patients who underwent

microsurgery, had no new facial motor disturbance and 91% of patients treated with GKS had no

functional deterioration after treatment compared to 61% in the microsurgery study. The mean

hospitalization stay was 3 days after GKS and 23 days after microsurgery. All the patients who

underwent GKS who had been employed, except one, had kept the same professional activity

(56% in the microsurgery study). The mean time away from work was 7 days for GKS (130 days in

the microsurgery study). Among patients whose preoperative hearing level was Class 1 according

to the Gardner and Robertson scale, 70% preserved functional hearing after GKS (Class 1 or 2)

compared with only 37.5% in the microsurgery group 266.

Combination with Subtotal Resection

In larger vestibular schwannomas, incomplete resection with planned Gamma Knife radiosurgery

Page 58: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

58

offers the option of reduced morbidity during surgery while maintaining a high degree of local tumor

control. In larger acoustic neuromas a combination of surgery and radiosurgery is increasingly

applied in order to provide tumor volume reduction, tumor control and functional preservation.

Such a combined therapy was shown to be beneficial regarding both tumor control and adverse side

effects.

Iwai had published an initial series of 14 patients with large acoustic neuromas using a

combination of surgery and radiosurgery. Excellent facial nerve function (House & Brackmann

Grade I or II) was preserved in 12 patients (85.7%) in the final follow-up 267. Iwai, Yamanaka and

colleagues recently (2015) published and update after treatment of 40 patients with large unilateral

vestibular schwannomas with planned partial tumor removal followed by Gamma Knife surgery for

functional preservation (median maximum tumor diameter 32.5 mm; range 25-52 mm). Gamma

Knife was performed 1-12 months after surgical resection (median interval 3 months; median tumor

volume at GKS 3.3 cm3; range 0.4-10.4 cm3; median prescribed dose 12 Gy; range 10-12 Gy, median

follow-up period 65 months; 18-156 months). At the final follow-up, facial nerve preservation

(House-Brackmann grade I-II) was achieved in 95%; House-Brackmann grade I: 92.5%, II: 2.5%).

Among the 14 patients with preoperative pure tone average (PTA) less than 50 dB, 42.9%

maintained PTA less than 50 dB at the last follow-up. Five- and 10-year tumor growth control

occurred in 86% of patients 268.

Very similar results had been achieved in a small series from Marseille: After subtotal resection of 8

large acoustic neuroma with an initial average maximum diameter of 40 mm (range 35-45), the

remaining tumors with a remaining diameter of 18 mm (range 9-20mm) were treated with Gamma

Knife radiosurgery at a mean prescription dose of 11.8 Gy (range 11-13). At a mean follow-up period

of 46 months after radiosurgery, excellent facial nerve function (House-Brackmann grade 1 or 2)

was preserved in 7/8 patients (87.5%) 269.

In 2014 the currently available data were reviewed by Brokinkel, Stummer and colleagues who

identified 6 studies comprising 159 patients on the effects of Gamma Knife radiosurgery following

subtotal resection of vestibular schwannoma. Gamma Knife surgery was performed on average 6

months postoperatively with a mean marginal dose of 11.88 Gy (mean target volume 4.42 cm3,

mean diameter 18.45 mm). After tumor resection facial nerve function was serviceable (House and

Page 59: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

59

Brackmann Grades I+II) in 82.8% (125/151 patients). Postoperatively hearing was serviceable in

20.2%. Within a mean follow-up time of 50 months, facial nerve function and hearing after

incomplete resection remained serviceable in 94.0%, and 11.6%, respectively 270.

Subtotal surgery can be performed using different techniques and when surgery for a large

vestibular schwannoma consists of intracapsular resection and decompression, better preservation

of facial and hearing function can be obtained. It was clearly shown by Pan, Yang and colleagues

that intracapsular decompression followed by Gamma Knife afforded a better neurological

outcome and quality of life than radical extracapsular resection followed by Gamma Knife. In a

study comprising 35 patients, the outcomes of intracapsular decompression followed by Gamma

Knife surgery (Group I) were compared with with outcomes of standard microsurgery followed by

radiosurgery (Group II). After the operation, 89% in Group I retained excellent facial function,

whereas only 35% in Group II had excellent facial function (p < 0.01). In Group I, 100% retained

hearing function after the operation. In Group II, none retained hearing function postoperatively (p

< 0.001). No patients experienced adverse effects after GKS. The mean return-to-work times were

2.4 ± 0.16 and 33.4 ± 4.3 weeks in Groups I and II, respectively (p < 0.001). According to the results

obtained using the 36-Item Short Form Health Survey (SF-36), patients in Group I enjoyed more

significant improvements in quality of life than patients in Group II (p < 0.001) 271.

Pituitary adenomas

In pituitary adenomas medication and microsurgery are typically used as first-line treatments. When

hormone-producing tumors cannot be removed completely or when the tumor recurs after surgery,

further treatment is imperative in order to stop excess hormone secretion. Previously, fractionated

radiotherapy has been applied as alternative treatment, which is currently thoroughly questioned

due to a long latency to achieve endocrine normalization, high risk of delayed radiation induced

pituitary dysfunction and even secondary tumor development. When hormone-producing tumors

cannot be removed completely or when the tumor recurs after surgery, further treatment is

imperative in order to stop excess hormone secretion.

Gamma Knife radiosurgery is ideally suited in this situation since the required high radiation doses

can be applied locally with an extreme precision while complex treatment planning helps to protect

the surrounding structures. In these cases Gamma Knife radiosurgery has been shown to normalize

Page 60: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

60

the excessive hormone production and stop further tumor growth.

Typically, high prescription doses of 25 Gy are applied in hormonally active pituitary adenoma. After

radiosurgery, the elevated hormone levels generally decrease slowly within several months. Due to

this known latency period, however, stereotactic radiosurgery is generally not preferred over a

surgical resection, which ideally provides immediate endocrine normalization. Several studies

indicate that withdrawal of anti-secretory medication prior to radiosurgery improves the chance of

endocrine remission.

Non-Secreting Pituitary Adenomas

In cases of smaller non-secreting pituitary adenoma Gamma Knife radiosurgery appears to be a safe

option providing consistent and reproducible tumor control with very low risk for immediate side

effects. Surgery and possibly medication are the main factors in the treatment of pituitary

adenomas, but not all symptoms can be abolished and not all tumors stopped with these

approaches and not always an endocrinological normalisation is achieved through medication.

Recent developments of stereotactic radiosurgery have demonstrated a high local effect allowing

reproducible tumor control and even endocrinological normalisation in treatment refractory

pituitary adenoma, thus offering a new therapeutical alternative.

In 2013 a systematic review and meta-analysis revealed a total of 17 studies to determine the

efficacy and safety of gamma knife surgery for the treatment of nonfunctioning pituitary

adenomas. There were significant differences in the rate of tumor control and in the radiosurgery-

induced endocrinological deficits depending on the tumor volume (P<0.001) 272:

In patients with tumor volumes <2 ml the rate of tumor control was 99%, the rate of

radiosurgery-induced optic neuropathy was 1%, and the rate of radiosurgery-induced

endocrinological deficits was 1% 272.

In patients with tumor volumes from 2 to 4 ml rate of tumor control was 96%, the rate of

radiosurgery-induced optic neuropathy was 0%, and the rate of radiosurgery-induced

endocrinological deficits was 7% 272.

In patients with tumor volumes larger than 4 ml the rate of tumor control was 91%, the rate

of radiosurgery-induced optic neuropathy 2%, and the rate of radiosurgery-induced

endocrinological deficits was 22% 272.

Page 61: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

61

Under the auspices of the North American Gamma Knife Consortium, 9 Gamma Knife surgery (GKS)

centers retrospectively combined their outcome data obtained in 512 patients with nonfunctional

pituitary adenomas. Overall tumor control was achieved in 93.4% of patients at last follow-up;

actuarial tumor control was 98%, 95%, 91%, and 85% at 3, 5, 8, and 10 years postradiosurgery,

respectively 273. Smaller adenoma volume (p = 0.006) and absence of suprasellar extension (p =

0.064) were associated with progression-free tumor survival. New or worsened hypopituitarism

after radiosurgery was noted in 21% of patients, with thyroid and cortisol deficiencies reported as

the most common postradiosurgery endocrinopathies. History of prior radiation therapy and

greater tumor margin doses were predictive of new or worsening endocrinopathy after GKS. New

or progressive cranial nerve deficits were noted in 9% of patients; 6.6% had worsening or new onset

optic nerve dysfunction. No patient died as a result of tumor progression 273.

Several retrospective studies after stereotactic radiosurgery for non-secreting adenoma have

shown a successful long-term control: After Gamma Knife treatment with a median minimum

treatment dose of 16 Gy a group of 62 patients was followed a median of 64 months. Tumor growth

control was 95% at 7 years after radiosurgery with a 5-year risk of developing new anterior pituitary

deficits of 18% for patients with a tumor volume of < = 4.0 cm3 compared with 58% for patients with

a tumor volume >4.0 cm3. No patient had a decline in visual function after a median maximum point

dose of 9.5 Gy to the optic apparatus (range, 5.0-12.6 Gy) 274. A previous series of 140 patients with

non-secreting pituitary adenoma treated with Gamma Knife radiosurgery (median prescription dose

20Gy) showed no adenoma growth after a median follow-up of 60 months; 89% of treated

adenomas decreased in size, with a median volume reduction of 61% 275. Almost identical results

were reported by Mingione, Steiner and colleagues who had treated 100 consecutive patients with

non-secretory pituitary macroadenoma (mean prescription dose 18.5 Gy; mean imaging follow-up :

44.9 months).Tumor volume decreased in 65.6% and remained unchanged in 26.7%, and increased

in 7.8%. The minimal effective peripheral dose was 12 Gy; peripheral doses greater than 20 Gy did

not seem to provide additional benefit. Of 61 patients with a partially or fully functioning pituitary

gland and follow-up data, 12 (19.7%) suffered new hormone deficits following Gamma Knife

treatment. No neurological morbidity or death was related to treatment 276.

Page 62: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

62

Park, Kondziolka and colleagues evaluated the outcomes of Gamma Knife radiosurgery in 125

patients with non-functional pituitary adenomas over an interval of 22 years (median target volume

3.5 cm3; range, 0.4-28.1 cm3; median tumor margin dose 13.0 Gy; range, 10-25 Gy).Tumor volume

decreased in 53%, remained stable in 37% and increased in 10.4% during a median of 62 months

of imaging follow-up. The actuarial tumor control rates at 1, 5, and 10 years were 99%, 94%, and

76%, respectively. Factors associated with a reduced progression-free survival included larger

tumor volume (≥ 4.5 cm3) 277.

Another recent study with 48 patients with a nonfunctioning pituitary adenoma performed a

median follow-up of 80.5 months after Gamma Knife and a median endocrinological follow-up 95

months, respectively. New hormone deficiency after GKRS occurred in 39%. Overall, control of

tumor volume was achieved in 83%. Tumor volume decreased in 75%, was unchanged in 8% and

increased in 17%. Tumor volumes greater than 5 cm3 at the time of GKRS were associated with a

significantly greater rate of growth (P = .003) compared with an adenoma with a volume of 5 cm3

or less 278.

It is notable that tumor volumes greater than 5 cm3 at the time of GKRS were associated with a

significantly greater rate of recurrence 278, since virtually identical results were reported from

Pittsburgh, where a larger tumor volume of ≥ 4.5 cm3 resulted in reduced progression-free survival.

The actuarial tumor control rates were 94% and 76% after 5 and 10 years, respectively in 125

patients after a median follow-up of 62 months 279. Similarly Pollock showed a threshold of 4.0 cm3

for the 5-year risk of developing new anterior pituitary deficits 274.

In a retrospective review of 62 patients with nonfunctioning pituitary adenomas undergoing Gamma

Knife radiosurgery (median tumor volume 4.0 cm3; range, 0.8-12.9; median treatment dose 16 Gy;

range, 11-20; median follow-up 64 months; range, 23-161), tumor growth control was 95% at 3 and

7 years after radiosurgery. The 5-year risk of developing new anterior pituitary deficits was 18% for

patients with a tumor volume of < or = 4.0 cm3 compared with 58% for patients with a tumor volume

>4.0 cm3. No patient had a decline in visual function 274.

Over a period of 10 years 140 patients with nonsecreting pituitary adenoma were treated by Gamma

Knife at Na Homolce Hospital, Prague with 79 patients being followed up for longer than 3 years

Page 63: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

63

(median prescription dose 20Gy; range 12-35 Gy, median follow-up 60 months (range 36 -122

months). No adenoma growth was detected; 89% of treated adenomas decreased in size, with a

median volume reduction of 61%. There was no vision impairment after radiosurgery, while 4 out

of 52 patients with abnormal perimeter vision reported improvement 275.

In a long-term follow-up 23 patients with non-functioning pituitary adenomas (median volume 1.1

cm3; median prescription dose 20 Gy) were followed at a median of 78 months with MR imaging

and 97 months clinically. Tumor growth control was 100%, One recurrence was discovered outside

radiation field and no new hypopituitarism was developed 280.

Side effects: Visual Dysfunction

An analysis of 217 patients with recurrent secretory (n = 131) and non-secretory (n = 86) pituitary

adenomas treated with Gamma Knife surgery was performed to determine the incidence of and risk

factors for subsequent development of visual dysfunction. Nine patients (4%) developed new visual

dysfunctions, and these occurred within 6 hours to 34 months following radiosurgery. Three of these

patients had permanent deficits whereas in 6 the deficits resolved. Prior GKS or radiotherapy (in

5/9) resulted in a significant increase in the incidence of cranial nerve dysfunction 281.

Threshold Doses

Specific structures in the brain have specific tolerances for the applied radiation. This is particularly

important in the treatment of tumors close to the optic system.

A retrospective review of 222 patients having Gamma Knife radiosurgery for benign tumors adjacent

to the anterior visual pathway concluded that the chance of radiation-induced optic neuropathy

according to the maximum radiation dose received by the anterior visual pathway was 0% below

12Gy and 10% (95% CI 0-43.0%) for patients receiving >12 Gy, respectively. One patient (0.5%) who

received an unusually high maximum radiation dose of 12.8 Gy to the anterior visual pathways had

developed unilateral blindness 18 months after SRS 282.

Another retrospective review from the same team around Pollock compared visual function before

and after SRS in 133 patients (266 sides) with pituitary adenomas having SRS showed risks of

developing a radiation-induced optic neuropathy at the 8-Gy, 10-Gy, and 12-Gy volumes (95%

Page 64: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

64

confidence interval) of 0% to 2.6%, 0% to 4.7%, and 0% to 13.9%, respectively. The conclusion was

that patients without prior radiation treatments can safely receive radiation doses up to 12 Gy with

a low risk of radiation-induced optic neuropathy 283.

Marek, Liscak and colleagues reported that hypopituitarism after Gamma Knife developed only in

2.2% of patients irradiated with a mean dose to pituitary <15 Gy in contrast to 72.5% patients

irradiated with a mean dose to pituitary >15Gy 284.

In another analysis a significant increase of new endocrine insufficiencies was seen at doses when

6.5 Gy were applied to the pituitary stalk (vs. 4.1Gy) (p = 0.004) and when more than 12.4 Gy was

applied to the pituitary gland (p = 0.05). That has been found after analysis of 108 Gamma Knife-

treated patients with pituitary adenoma where spot dosimetry was used to determine doses

delivered to structures of the hypothalamopituitary axis. The overall tumor control rate was 97%

and the endocrinological cure rate was 61.2%. The results of this study show that patients in whom

the pituitary stalk and pituitary gland receive a high mean point dose are more likely to develop

pituitary insufficiencies after GKS than those who receive a lower dose 285.

Tab 1. Gamma Knife radiosurgery for non- secreting pituitary adenoma

Years of

treatment

Number of

patients

Tumor

volume

(cm3)

Prescription

doses

Months of

FU complications Tumor control

Gopalan R, et al.

Neurosurgery. 2011

Aug;69(2):284-93

1990-2004 48 5.1

(0.9-27)

18.4 Gy

(8-25)

80.5

(median)

31.4 %

risk of pituitary deficit

at median 5y

83%

Park KJ, et al.

Neurosurgery. 2011

Dec;69(6):1188-99.

1987-2009 125 3.5

(0.4-28.1)

13 Gy

(10-25)

62

(median)

24 %

risk of pituitary deficit

at median 2y

89.6%

Pollock BE, et al.

Int J Radiat Oncol Biol

Phys. 2008 Apr 1;70(5):1325-9.

1992-2004 62 4

(0.8-12.9)

16 Gy

(11-20)

64

(median)

32 %

risk of pituitary deficit

at 5y

no visual deficit

95 %

at 7 years

Page 65: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

65

Liscák R, et al.

Acta Neurochir (Wien).

2007 Oct;149(10):999-

1006

1993-2003 79 3.45

(0.1-31.3)

20 Gy

(12-35)

60

(median)

no visual deficit

100 %

control

89% decreased

Mingione V, et al.

J Neurosurg. 2006

Jun;104(6):876-83.

1989-2004 100 18.5 Gy

(5-25)

44.9

(mean)

25 %

risk of pituitary deficit

after 2y

no visual deficit

92 %

control

65.5%

decreased

Gamma Knife Radiosurgery as Primary Management of Non-Secreting Pituitary

Adenomas

In general, the initial treatment in pituitary adenomas is surgical removal. With regard to the

favorable results after stereotactic radiosurgery however, it has been discussed if Gamma Knife

radiosurgery can be applied as first line treatment in uncomplicated non-secretory pituitary

adenomas.

Among a total of 569 patients who were treated using Gamma Knife radiosurgery for nonfunctioning

pituitary adenomas, 41 patients underwent Gamma Knife treatment as primary management

because of an advanced age, multiple comorbidities, or patient preference. Patients received a

median tumor margin dose of 12 Gy (range 6.2-25.0 Gy) at a median isodose of 50%. The overall

tumor control rate was 92.7%, and the actuarial tumor control rate was 94% and 85% at 5 and 10

years post-radiosurgery, respectively. New or worsened hypopituitarism developed in 24% at a

median interval of 37 months after GKRS. Delayed hypopituitarism was observed more often in

patients who had received a tumor margin dose > 18 Gy (p = 0.038) and a maximum dose > 36 Gy

(p = 0.025). One patient suffered new-onset cranial nerve palsy. No other radiosurgical

complications were noted 286.

Acromegaly

Acromegaly affects 86–240 individuals per million 287. Recently the results of the German Acromegaly

register comprising 1344 patients followed in 42 centers were published: 89% of the patients had at

least one surgical intervention, but after surgery only 38.8% of the patients were controlled without

further therapy and among the medically treated patients 65.2% were controlled 288 . In general three

Page 66: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

66

types of drugs are used for patients with acromegaly and postoperative persistent hormonal activity

dopamine agonists (DAs) like bromocriptine and cabergoline, somatostatin receptor ligands (SSAs) such

as octreotide LAR and lanreotide and GH receptor antagonists (GHRH) such as pegvisomant. However,

of 557 medically treated patients with acromegaly, 65.2% had a normal IGF1 and 86% had at least one

operation, 28% were operated and had received adjunctive radiotherapy 289.

Since neither anti-secretory drugs nor surgical adenoma resection will guarantee endocrine

normalization in all cases of pituitary adenoma, there is need for an additional treatment option such as

radiosurgery.

After conventional fractionated radiotherapy for acromegaly endocrinological normalization could be

achieved in some patients, but there is a very long latency between radiotherapy and effect: Jenkins and

colleagues reported in a very large multicenter study of 884 patients from 14 centers throughout the

United Kingdom a normalization of serum GH (< 2.5 mg/L) in 22% at 2 years and 60% at 10 years 290.

Barrande demonstrated hormonal normalization in 7% of his patients at 2 years, in 35% at 5 years and

in 53% 10 years after treatment 291. A long-term study of 72 acromegalic patients treated with

fractionated radiotherapy showed that normal age-matched IGF-I levels were reached in 16% after 10

years 292.

The side effects induced by fractionated radiotherapy were significant: The risk for secondary

radiation induced hypopituitarism increased progressively after radiotherapy and 10 years after

radiotherapy, 80% of the patients had gonadotropic deficiency, 78% showed thyrotropic

insufficiency, and 82% had developed a corticotropic deficiency 291 .

Epaminonda demonstrated secondary radiation induced tumors in 4.5% of patients with

acromegaly 9-22 years after conventional fractionated radiotherapy 293 , which was confirmed by

others 294,295 .

Stereotactic radiosurgery of acromegaly

In 2010 Yang and colleagues performed a comprehensive search of the English language literature

revealing 970 patients published between June 1998 and September 2009 with new, recurrent, or

persistent acromegaly who had been treated using radiosurgery with assessable and quantifiable

outcome data. The review showed an overall disease control rate without medication in 48%-53%,

while the overall disease control rate with or without medication was 73% at a mean reported

follow-up of 48.5 ± 25.8 months (mean overall tumor volume 2.11 ± 1.16 cc) 296 . Others showed a

Page 67: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

67

rate of remission of 58.3% at 5 years in 103 acromegalic patients treated with Gamma Knife. Further

14.6% were in remission after Gamma Knife while on treatment with somatostatin analogues. No

serious side effects occurred after GK 297.

A similar remission rate of 54.5% was achieved after 60 months of follow-up in 22 patients with

acromegaly treated at the Endocrinology-Metabolism Clinic of Cerrahpasa Medical School in

Istanbul 298. Tumor growth was well controlled in 95.2%.

Castinetti, Brue and colleagues followed 82 patients with acromegaly prospectively studied over a

decade, with a mean follow-up of 49.5 months showing that 17% percent of the patients were in

remission without any treatment and 23% previously uncontrolled on somatostatin agonists fulfilled

the same criteria after Gamma Knife while maintained on medical treatment 299. J Clin Endocrinol

Metab. 2005 Aug;90(8):4483-8. Epub 2005 May 17. Outcome of gamma knife radiosurgery in 82

patients with acromegaly: correlation with initial hypersecretion. Castinetti F, Taieb D, Kuhn JM,

Chanson P, Tamura M, Jaquet P, Conte-Devolx B, Régis J, Dufour H, Brue T.

Endocrine remission was achieved in 47.5 % and endocrine control in additional 10.0 % after

Gamma Knife radiosurgery at the University of Pittsburgh in a series of patients with acromegaly

(n= 40; median follow-up 72 months; range 24-145). Patients with lower IGF-1 level and with tumors

that were less invasive of the cavernous sinus before GKRS were associated with better GH

remission rates. Imaging-defined local tumor control was achieved in 97.5 %, while 40.0 %

eventually developed a new pituitary axis deficiency at a median onset of 36 months after

radiosurgery. No patient developed new visual dysfunction. Almost one half of the patients no

longer required long term medication suppression 300.

Adenoma Subtypes

While patients who have a sparsely granulated somatotroph-cell adenoma may be less responsive

to medical therapy, they exhibited similar responses to Gamma Knife radiosurgery as patients with

a densely granulated somatotroph-cell adenoma adenoma. After Gamma Knife radiosurgery the

actuarial remission rates in the densely granulated somatotroph-cell adenoma group at 2, 4, and

6 years post-radiosurgery were 35.1, 71.4, and 79.3 %, respectively, while those in sparsely

Page 68: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

68

granulated somatotroph-cell adenoma group were 35.4, 73.1, and 82.1 %, respectively 301.

Latency of Endocrinological Normalisation

In a retrospective long-term analysis with a median follow-up of 10 years after Gamma Knife

treatment of acromegalic patients it was demonstrated that cure rates improved over time to 46%

at 10 years. Normal IGF-I values were observed in 82% of patients at their last visit. No visual

impairment, disease recurrence, tumor growth or secondary cerebral tumor occurred 302.

Liu, Lunsford and colleagues found gradually increasing endocrine remission rates of 18.6% , 44.5%

and 67.6 % at 3, 5 and 7 years after radiosurgery, respectively 300. The team from Bergen showed

that normal IGF-I levels were achieved in 45% 3 years after Gamma Knife radiosurgery, 58% after 5

years , and in 86% after 10 years 303. Virtually identical results were published by Jezková and

colleagues from the Gamma Knife group in Prague with a normalization of serum GH (< 2.5 μg/l) in

23.9% 1 year after radiosurgery , in 47.1% at 2 years, in 67.4% at 5 years and 85.7% 8 years after

treatment 304. In general Gamma Knife radiosurgery resulted in endocrine normalization (defined as

GH values <2.5 ng/ml, and normalization of the age and gender appropriate IGF 1) in 40-60%

between 2 and 5 years after radiosurgery. The results demonstrate a faster endocrine normalization

after Gamma Knife when compared to fractionated conventional radiotherapy, which previously

had been demonstrated by Landolt, who showed that the mean time to endocrine normalization

was 1.4 years Gamma Knife cohort and 7.1 years in the group treated with fractionated radiotherapy

with significant difference 305.

Sheehan and colleagues reviewed a total of 418 patients who underwent Gamma Knife radiosurgery

at the University of Virginia with a median follow-up of 31 months (median 31 months) showing

that in patients with a secretory pituitary adenoma, the median time to endocrine remission was

48.9 months. In 90.3% of patients there was tumor control 306.

Radiosurgical Salvage Treatment after Failed Conventional Radiotherapy

A cohort of 25 patients with pituitary adenomas who had not responded to a previous treatment of

Page 69: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

69

conventional external beam radiotherapy was treated with Gamma Knife radiosurgery. Following

GK, mean GH fell by 49% at 1 year in patients with somatotroph tumors. Serum IGF1 fell by 32%

at 1 year and by 38% at 2 years. At final investigation, 80% of the patients with acromegaly have

achieved normalisation of IGF1, and 30% have also achieved a mean GH level of <1.8 ng/ml

correlating with normalised mortality. Prior to GK, 72% of the patients were panhypopituitary, and

42% of the remainder developed new anterior pituitary hormone deficiencies. No other adverse

events have been detected at a mean follow-up of 36.4 months 307.

Radiation Doses

Typically, rather high prescription doses of >25 Gy directed to the tumor margin are applied in

hormonally active pituitary adenoma. However, two series could not establish a relation between

outcome in acromegaly and doses after median doses of 21 Gy 300 and 30 Gy 308. Relative low doses

(median 18.9Gy) were given in a series by Kobayashi and colleagues with slightly less effective

outcome 309. Further studies will have to elucidate if prescription doses can be reduced without

increasing the risk of therapeutic failures.

Kobayashi, Fujitani and colleagues concluded that normalization of GH and IGF-1 secretion was

difficult to achieve in cases with large tumors and low-dose radiation. They had treated 67 patients

for residual or recurrent growth hormone (GH)-producing pituitary adenomas using Gamma Knife

radiosurgery (mean tumor volume 5.4 cm3) and applied a relatively low mean margin dose of 18.9

Gy. After a mean follow-up duration of 63.3 months (range 13-142 months) the tumor response

rate was 68.3%, and the control rate 100%. Growth hormone was normalized (GH < 1.0 ng/ml) in

4.8%, was nearly normal in 11.9% (< 2.0 ng/ml) and significantly decreased (< 5.0 ng/ml) in 23.8%,

decreased in 21.4%, unchanged in 21.4%, and increased in 16.7%. Serum insulin-like growth factor

(IGF)-1 was significantly decreased (IGF-1 < 400 ng/ml) in 40.7% 309.

Relevance of Pre-operative Hormonal Levels

Similarly to a previously cited study after conventional fractionated radiotherapy 291, Jeskova found

that the less active adenomas were normalized earlier and the effect of Gamma Knife treatment

depended on the baseline hormonal activity of the adenoma with impact of the levels of GH and

more significantly of IGF-1 304. Similarly Pollock and colleagues demonstrated a lower pre-Gamma

Page 70: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

70

Knife IGF-1 level (less than 2.25 times the upper limit) was associated with a higher likelihood of

endocrine remission 310.

Radioprotective Effect of Anti-Secretory Therapy

Landolt had found that withdrawal of somatostatin suppression prior to Gamma Knife improves the

chance of endocrine remission 311, which has been corrobated by others 310,312 . As a practical policy,

it is currently recommended to suspend all anti-secretory medication before radiosurgery.

Side Effects in Radiosurgery of Pituitary Adenoma

Treatment induced secondary hypopituitarism can appear very late, and has been described

between 6–74 months after Gamma Knife treatment 308 . The Pittsburgh Gamma Knife group

reported pituitary deficit in 34 % after a median of at 36 months 300 and Pollock described a 33 %

risk of pituitary deficit after 5 years 313. This highlightens the need for long-term clinical control of

patients with radiotherapeutical or radiosurgically treated pituitary adenoma.

Hence, secondary hypopituitarism with hormonal deficiency is a phenomenon and problem

affecting all forms of radiation of the hypophysis at various degrees. The reason for radiation

induced functional defects is still a matter of intense discussion. Feigl and colleagues had initially

demonstrated that the pituitary stalks in patients with deterioration of pituitary function received

a statistically higher dose of radiation, 7.7 +/- 3.7 Gy compared with 5.5 +/- 3 Gy (p = 0.03) after spot

dosimetry was performed in the hypothalamic region, the pituitary gland, and pituitary stalk in 59

patients following gamma knife radiosurgery for pituitary adenomas 314 .

The Gamma Knife group in Prague concluded that rather the radiation dose applied to the adjacent

pituitary gland is responsible for late deficits since in the group of patients developing

hypopituitarism, the mean dose on the adjacent pituitary gland was significantly increased 35.4 Gy

(17.4–63.1Gy). No T4 or sex hormone deficiency developed if the medium pituitary dose did not

exceed 15 Gy, and no cortisol deficiency developed if the medium dose was below 18 Gy 308. The

cumulative risk of adrenocortical deficiency development was calculated at 85% after 90 months

when the mean dose on the healthy pituitary exceeded 20 Gy 308. The same group applied these

dose thresholds prospectively in patients with pituitary adenoma, where the dose to the pituitary

gland was reduced to <15 Gy which resulted in a reduction of hypopituitarism after Gamma Knife

Page 71: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

71

treatment to only 2.2% (1/45), in contrast to 72.5% of patients who had been irradiated with a mean

dose >15 Gy to the pituitary gland 284.

Tab 5. Gamma Knife radiosurgery for Acromegaly

Years of

treatment

Number of

patients

Tumor

volume

Prescription

doses

Months of

FU complications

Endocrine

Normalization

Franzin A, et al.

Int J Endocrinol.

2012;2012:342034.

1994-2009 103 71

7.8 %

pituitary deficit

58.3%

at 5 years

Liu X, et al.

J Neurooncol. 2012

Aug;109(1):71-9.

1988-2009 40 2 21 Gy

(12-30)

72

median

34 %

pituitary deficit

median

at 36 months

47.5%

Jagannathan J, et al.

Neurosurgery. 2008

Jun;62(6):1262-96-9

1988-2006 95

3.1

in 77%

0.68

in 23%

22 Gy

(mean)

57

(mean

endocrine)

34 %

pituitary deficit

4.2%

visual deficit

53 %

at median

23.5 months

Vik-Mo EO, et al.

Eur J Endocrinol. 2007

Sep;157(3):255-63

1993-2003 53 1.23

(0.01-6.6)

25 Gy

in 81 %

66

(mean)

15.1 %

risk of pituitary deficit

3.7 %

visual deficit

normal IGF-I

and GH <5 mIU/l

58 %

at 5 years

40% w/o

medication

Pollock BE, et al.

J Neurosurg. 2007

May;106(5):833-8.

1991-2004 46 3.3

(0.5-18)

20 Gy

(14.4-30)

63

(median)

33 %

risk of pituitary deficit

after 5y

60 %

at 5 years

w/o medication

Jezková J, et al.

Clin Endocrinol (Oxf). 2006

May;64(5):588-95.

after 1993 96 1.35

(0.09-12.7)

30 Gy

(median)

53.7

(mean)

31% thyroidal deficit

14 % adrenal deficit

41% gonadal deficit

50 %

at 54 mo

Castinetti F, et al.

J Clin Endocrinol Metab.

2005 Aug;90(8):4483-8.

1993-2003 84 not given 12-40Gy 49.5 17 %

risk of pituitary deficit

40 %

at mean 36 mo

23% normalized

with medication

Kobayashi T. et al.

J Neurosurg. 2005 Jan;102

Suppl:119-23.

after 1991 67 5.4 18.9 Gy 63.3

(mean)

14.6 %

risk of pituitary deficit

40.7 %

Page 72: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

72

11.1%

deteriorated vision

Prolactinoma

About 40% and thereby the most common of all pituitary adenomas are prolactinoma with a

prevalence ranging betwee 6–50 per 100,000 315. Hyperprolactinemia interrupts the pulsatile

secretion of gonadotrophin-releasing hormone, inhibits the release of luteinizing hormone and

follicle-stimulating hormone, and directly impairs gonadal steroidogenesis 316 . Sometimes mixed

growth hormone and prolactin-secreting tumors can occur, presenting as acromegaly in together

with hyperprolactinemia 316.

Prolactinoma can be large, but medical therapy using dopaminergic agonists such as bromocriptine

and cabergoline are first-line treatment even for prolactin macroadenoma. In many cases the

medication alone reduces the tumor volume and restores the pituitary function 316. In

microprolactinoma the primary treatmet goal is to restore gonadal function by normalizing prolactin

levels 316.

A fraction of prolactinomas are resistant to dopamine agonist therapy and a trans-sphenoidal tumor

resection should be considered in symptomatic patients, who cannot tolerate high doses of

cabergoline or who are not responsive to dopamine agonist therapy 315.

A recent study evaluated the endocrinological efficacy of transsphenoidal surgery in prolactinoma

patients reporting typical results with postoperative remission in 63% of microprolactinomas and

60% of noninvasive macroprolactinomas, and none of the invasive macroprolactinomas. Better

remission rate was independently predicted by lower diagnostic prolactin levels and by the lack of

abnormal postoperative residual tissue (P<0.05). A recurrence of hyperprolactinemia was observed

in 34% of patients after a median follow-up period of 36 (7-164) months 317. The success rate of

surgery appears to be higher for patients with prolactin levels lower than 200μg/l (4000 mIU/l) 316.

In rare patients who fail both medical treatment and surgical resection, radiation therapy of

prolactinoma can be considered.

Gamma Knife Radiosurgery for Prolactinoma

Prolactinomas are generally treated with dopamine agonists, which induce both tumor volume

regression and endocrine remission. In case of failures of the medical therapy, some tumors are

Page 73: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

73

resected. When both medication and surgery fails, stereotactic radiosurgery may be a remaining

option. Radiosurgery may provide some endocrine improvement in a number of patients, but the

rates for complete endocrinological normalization and the number of patients with complete

remissions after stereotactic radiosurgery are limited. Endocrine normalization has been defined as

normal serum prolactin <20 ng/ml, in the absence of dopamine agonist therapy 318.

Prolactinoma are generally considered an endocrine disease and a direct relationship between

imaging outcomes and endocrine remissions are not always observed. At higher radiation doses

73.1% of macro-prolactinoma decreased in size 319. In a recent series from Charlottesville, Virginia,

46% of patients achieved at least a 20% reduction of their in tumor size and 43% of patients had a

stable tumor volume on follow-up, while endocrine normalization was achieved in 37.1% 318. In

2015 an update was published by the same group, now with a total of 38 patients with prolactinoma

who had been treated with Gamma Knife radiosurgery at the University of Virginia and endocrine

remission was achieved in 50 %. GKRS induced hypopituitarism occurred in 30.3 % 320. In a recent

series from Pittsburgh endocrine normalization was achieved in only 27% with improvement in 54%

after using rather low median marginal (prescription) doses of 15Gy. In univariate analysis, a

prolactin level less than 200 ng/ml before Gamma Knife treatment were associated with higher rate

of endocrine normalization 321. A virtually identical outcome was reported by the group from

Charlottesville with endocrine remission in 26% after slightly higher prescription doses of 18.6 Gy

318, while Tanaka, Pollock and colleagues reported a 4-year actuarial rate of normalization in 17%

and additional 14% of patients with normal serum prolactin while on dopamine agonist medication

322.

The Gamma Knife group in Prague used considerably higher radiation doses with a median

prescription dose of 34 Gy (range 20-49 Gy) and reported a higher response rate with 37.1% of

patients with endocrine normalization and 42.9 % normal serum prolactin during continued

medication 319. Similarly, others had shown that margin tumor dose of 30 Gy or more seemed to be

associated with a higher endocrinological normalization rate 323. Two series demonstrated a better

outcome after radiosurgery for smaller prolactinoma with volumes of less than 3cc 318,324.

The average time to remission was 24.5 months ranging between 8 and 48 months 318 and 11.5

months (range 2–42 months) in another recent series 324 .

In line with the other types of pituitary adenoma, it was demonstrated even for prolactinoma, that

antisecretory medication with dopamine agonists during Gamma Knife treatment was associated

Page 74: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

74

with a lower rate of remission 318, which was not confirmed by Jezkova and colleagues 319 .

In summary, the radiosurgical results after Gamma Knife treatment of prolactinoma show a

normalization only in a minority of patients with better outcome for smaller tumors (<3cc) 318,321,

higher prescription doses 319,323 and in patients where the anti-secretory medication was

suspended during radiosurgery.

Tab 6. Gamma Knife radiosurgery for Prolactinoma

Years of

treatment

Number of

patients

Tumor

volume

(cm3)

Prescription

doses years of FU complications

Endocrine

Normalization

Liu X, et al.

Pituitary. 2013

Mar;16(1):68-75.

1994-2009 22 3

(0.3-11.6)

15 Gy

( 12–25 Gy)

36 months

(12–185).

3/22 delayed tumor

progression

1/22

with new pituitary axis

deficiency

27.3%

normalization

54.5%

endocrine

improvement

Tanaka S. et al.

World Neurosurg. 2010

Jul;74(1):147-52.

1994-2006 22 2.2

(0.4-29)

25 Gy

( 16–30 Gy)

60 mo

(16-129)

42% new anterior

pituitary deficits was

at 4 years

17%

4-year actuarial

rate of

normalization

14%normal

serum PRL on

dopamine

agonist med.

Jezková J, et al.

Clin Endocrinol (Oxf).

2009 May;70(5):732-41.

1993-2005 35 0.9cc 34 Gy

( 20-49 Gy)

66 mo

(median)

37.1%

normalization

42.9 %normal

serum PRL on

dopamine

agonist med.

Pouratian N, et al.

Neurosurgery. 2006

Aug;59(2):255-66

1990-2003 23 18.6 Gy

(0.3–25) Gy 55mo

29% new anterior

pituitary deficits after

mean of 44 months

(33–51 mo).

26% after

average 24.5mo

(8-48)

Pan L, et al.

J Neurosurg. 2000

Dec;93 Suppl 3:10-3.

1993-1997 128

31.2 Gy

mean

(9-35 Gy)

33.2 mo

mean

(6-72)

52%

”clinical cure”

Page 75: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

75

Cushing’s disease

Due to the rarity of the condition, the published series after stereotactic radiosurgery for Cushing’s

disease are rather small and only few studies report more than 20 patients. Three series report the

results of stereotactic radiosurgery for Cushing’s disease after treatment with Gamma Knife with

subsequent endocrine normalization in 35%-63% 325-327. In the largest available study from

Charlottesville, Virginia, Jagannathan described that normal 24-hour urinary free cortisol levels

were achieved in 54% (49 patients), at an average time of 13 months after treatment (2–67 months),

however 20% experienced a relapse of Cushing’s disease after initial remission at a median of 27

months 325 . The mean prescription dose in these patients was 22 Gy.

In 2010 a published review of the literature provided a total of 29 published series that have

reported the results for 445 patients with Cushing’s disease treated with postoperative radiosurgery

or with primary radiosurgery. The mean radiosurgical margin dose (i.e. dose to the margin of the

adenoma) for these series ranged from 14.7 to 32 Gy. Exclusion of series with less than 10 patients

and less than 2-years follow-up resulted in endocrine remission rates from 17–83% and remission

is usually observed within 1–2 years. Published tumor control rates range from 50–100% 328.

The visibility of the adenoma in patients with Cushing’s disease can be an issue and accordingly, the

tumor cannot always be outlined on the planning MR image 325 . Without an accurate target, the

efficacy of stereotactic radiosurgery is significantly compromised. Hormone deficiencies after

Gamma Knife occurred in 22% at a mean of 16 months after radiosurgery (4–36 months). In 5

patients (5.6%) and ophtalmoplegia developed after radiosurgery, in 4/5 patients had previously

undergone radiosurgical procedures 325.

Kobayashi reported a normalization of endocrine levels patients (<50 pg/ml for ACTH level and <

than 10 microg/dl for serum cortisol) in 35% of patients with additional significantly improved levels

in additional 25% 326 after Gamma Knife treatment of Cushing’s disease at mean margin dose of 28.7

Gy (range 15–70 Gy) in relatively large adenomas with mean volume of 3.24 cm3 (0.07–10.5 cm3).

A high control rate of 83.2% was achieved when the maximum dose was higher than 55 Gy and/or

Page 76: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

76

a margin dose exceeded 40 Gy, thus indicating the importance of applied doses326.

Castinetti and colleagues reported similar results after Gamma Knife treatment with normalization

in 42.5% after a mean of 22 months 329. The target volume was significantly smaller in patients where

remission could be achieved (0.44 vs. 0.91 cm3). Interestingly, patients who were off anticortisolic

drugs at the time of Gamma Knife had a better chance of remission (48% vs 20%) 329. The

phenomenon of potential radioprotection by anti-secretory drugs has also been described in the

treatment of acromegaly.

The additive effect of radiation has to be considered when patients are treated with either repeated

radiosurgery of with a combination of fractionated radiotherapy and radiosurgery: so far no

stratified dose escalation studies are available but in the Charlottesville series 5 patients (5.6%)

developed ophtalmoplegia after radiosurgery, four of these five patients had previously undergone

radiosurgical procedures 325 .

In 2013 Sheehan published a retrospective review of 96 patients with Cushing's disease treated with

Gamma Knife surgery the University of Virginia (mean tumor margin dose 22 Gy; median follow-up

48 months; range 12-209.8 months) and showed that remission of Cushing's disease occurred in

70% of patients 330. The median time to remission among all patients was 16.6 months (range 1-

165.7 months). The median time to remission in those who had temporarily stopped taking

ketoconazole at the time of Gamma Knife was 12.6 months, whereas it was 21.8 months in those

who continued to receive ketoconazole (p < 0.012). Tumor control was achieved in 98% of patients.

New loss of pituitary function occurred in 36% of patients. New or worsening cranial neuropathies

developed in 5 patients after GKS, with the most common involving cranial nerves II and III 330.

Similarly, hormonal normalization was reached in 80.7 % of 26 patients with Cushing's disease who

had been followed-up after Gamma Knife radiosurgery in Prague. The median time to normalization

was 30 months (6-54 months) 331.

In summary, stereotactic radiosurgery offers an additional option in the treatment of surgically

therapy refractory patients with Cushing’s disease. It has to be considered that the adenoma has

to be visible on the stereotactic pre-treatment MRI and that relatively high prescription doses

have to be applied. Series with lower prescription doses or impaired diagnostic visibility provide

Page 77: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

77

lower therapeutic effects. Normalization of endocrine levels patients ( <50 pg/ml for ACTH level

and < than 10 microg/dl for serum cortisol) in 35% of patients with additional significantly

improved levels in additional 25% can be expected. Empirically established threshold doses of optic

nerves, chiasm and cranial nerves have to be considered in particular when several radiation

treatments are applied. Potential side effects comprise secondary endocrine deficits that may

appear several years after radiosurgery. The risk for other clinical side effects is low after

stereotactic radiosurgery.

Nelson's syndrome

Nelson's syndrome is a significant and frequent risk for patients with Cushing's disease who undergo

bilateral adrenalectomy. Gamma Knife appears to provide prophylaxis for Nelson's syndrome when

used before bilateral adrenalectomy 332.

Whole-Sellar Stereotactic Radiosurgery

Functioning pituitary adenomas (FPAs) can be difficult to delineate on postoperative magnetic

resonance imaging, making them difficult targets for stereotactic radiosurgery. In such cases,

Gamma Knife treatment with radiation delivery to the entire sella has been utilized as a radiosurgical

equivalent of a total hypophysectomy in 64 patients with acromegaly, Cushing disease and

prolactinoma (median margin dose of 25 Gy, n=64; median endocrine follow-up 41 months) The 2-

, 4-, and 6-year actuarial remission rates were 54%, 78%, and 87%, respectively. New-onset

neurological deficit was found in 6.3%, new-onset hypopituitarism in 43.5% 333.

Second Intervention after Failed Resection in Cushing’s Disease

Fifty-two patients with relapse of Cushing’s disease after transsphenoidal adenomectomy were

enrolled and randomly underwent a second surgery or Gamma knife radiosurgery as the next

therapeutic approach. The statistical analysis showed higher recurrence-free interval in the Gamma

knife group compared with the transsphenoidal adenomectomy group 334.

Page 78: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

78

Tab 3. Gamma Knife radiosurgery for Cushing’s disease

Years of

treatment

Number of

patients

Tumor

volume

(cm3)

Prescription

doses

Months of

FU complications

Endocrine

Normalization

Jagannathan J, et al.

Neurosurg. 2007

Jun;106(6):980-7.

1990-2005 90 not given

23 Gy

(8-30Gy)

45 mo

5.6 %

with new cranial

nerve deficit

54 %

13 months

20% relapse

at 27 mo

Castinetti F. et al

Eur J Endocrinol. 2007

Jan;156(1):91-8

1993-2003 40 0.5

29.5 Gy

(median)

(15-40)

54.7 mo

(mean)

15 %

new endocrine deficit

42.5%

(mean 22 mo)

Kobayashi T, et al.

J Neurosurg. 2002

Dec;97(5 Suppl):422-8.

after 1991 25 3.24

(0.07-10.5)

28.7 Gy

(15-70Gy)

64.1

(mean)

no

new endocrine deficit

no visual deficit

35 %

Sheehan JM, et al.

Neurosurg. 2000

Nov;93(5):738-42.

1990-1998 43 not given not given 44

16 %

new endocrine deficit

2.3 % visual deficit

63 %

12.1 months

11% relapse

(3pat)

at 19-38 mo

Page 79: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

79

Gamma Knife Radiosurgery for Malignant Gliomas and Glioblastomas

Glioblastoma (GBM) is the most common primary CNS tumor in the USA and European countries.

The incidence rate for glioblastoma per 100,000 population/year is 3.32 in males and 2.24 in females

335,336. Population-based studies report that the overall survival of patients with newly diagnosed

GBM is 17 – 30% at 1 year, and only 3 – 5% at 2 years, despite access to state-of-the-art modalities

of therapy. Observed survival rates in 715 patients with glioblastoma (2004) were 42.4% at 6 months,

17.7% at 1 year, and 3.3% at 2 years 335,336. According to the SEER database that collects cancer

incidence and survival data representing 28% of the US population, the median survival time of adult

patients with glioblastoma was 9 months (in 2010) 337.

Standard therapy of malignant glioma and glioblastoma consists of surgical resection, radiotherapy

and chemotherapy. It appears that tumor recurrences are inevitable in glioblastoma.

Initial Retrospective evidence appeared to suggest that the survival of patients who were treated

with a radiosurgery boost was superior to historical series in particular for RTOG classes 3 and 4 338-

344, but the prospective randomized RTOG study 9305, did not confirm any survival benefit for the

patients who had been treated with a radiosurgical boost as part of their upfront treatment for

glioblastoma 345.

Hence, as also stated in an American Society for Therapeutic Radiology and Oncology (ASTRO)

evidence-based review, it is now generally accepted, that radiosurgery has no additive effect when

given as boost or in connection to initial standard treatment with surgical resection and fractionated

radiotherapy 346 . This is in line with other studies that failed to demonstrate a beneficial effect of a

fractionated dose escalation as upfront treatment 347-350.

Surgical debulking is the mainstay of initial treatment of malignant gliomas and glioblastoma, but

surgery alone is inadequate for obtaining reliable tumor control. Conventional fractionated

radiation is generally applied as standard treatment of malignant glioma, but it appears that tumor

recurrences are inevitable in glioblastoma. After standard fractionated radiotherapy 77%-90% pa-

tients relapsed centrally within 2 cm of the original gadolinium-enhanced mass on MRI 351-353. The

virtually inevitable glioblastoma recurrence after standard treatment allows for only very few

Page 80: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

80

remaining therapeutic options, since a second surgical resection is not always feasible, conventional

fractionated re-irradiation is impossible and clinical benefit of chemotherapy appears to be limited.

In this situation stereotactic radiosurgery has been applied. Due to the inherent limited and

extremely focused radiation effect, stereotactic radiosurgery offers a potential option to treat

glioblastoma recurrences in this otherwise highly critical situation.

Despite recent advances in treatment, tumor recurrence after initial therapy is inevitable and

typically leads to death within a matter of months. After recurrence the remaining therapeutic

options are 1. re-operation, 2. chemotherapy or 3. re-irradiation using hypo-fractionated

stereotactic radiotherapy or stereotactic radiosurgery. Reoperation is a theoretical option for the

treatment of recurrent glioblastoma but requires a good preoperative clinical condition. The evalua-

tion of surgical studies is significantly impaired by the clinical bias inherently related to select

patients for surgery and by the difficulties to systematically compare tumor infiltration in various

anatomical tumor locations. Based on a variety of studies with rather consistent outcomes the me-

dian survival time after reoperation of recurrent glioblastoma can be estimated with 3.5 – 9

months354-356, provided that the patients are in good preoperative clinical condition (KPS>60) 355,356.

FDA approved of bevacizumab for recurrent glioblastoma. In the treatment of recurrent

glioblastoma with bevacizumab, the median survival after progressive disease the median overall

survival was 4.5 months 357. Iwamoto FM, Abrey LE, Beal K, et al. Patterns of relapse and prognosis

after bevacizumab failure in recurrent glioblastoma. Neurology 2009;73(15): 1200-6.

In the situation of an almost inevitable glioblastoma recurrence, the treatment is highly complex;

as fractionated radiotherapy cannot be repeated and the surgical options may be limited due to the

patients’ frequently reduced clinical condition. Under these circumstances stereotactic radiosurgery

has been proven to be an effective alternative treatment option and patients treated at the time of

progression,358,359,338,339,360-365. The reported the survival after radiosurgical treatment of the

glioblastoma recurrences ranges between 6.5 and 30 months, which compares favorably to the

outcome after reoperation of recurrent glioblastoma (3.5-9 months) 354-356,366, the treatment with

temozolomide 367 and the median overall survival on bevacizumab after glioblastoma recurrence

(4.5 months) 357.

Page 81: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

81

Clinical Outcome Criteria

It is important to note that survival after radiosurgery for glioma is strongly related to five selection

variables (grading, age, decreased KPS, increased volume, multifocality). Much of variation in

survival reported can be attributed to differences of these variables. The median survival of patients

with primary glioblastoma after Gamma Knife treatment was 86 weeks if ‘brachytherapy criteria’

were satisfied 368. A recent prospective cohort study demonstrated that stereotactic radiosurgery

with Gamma Knife or Linac significantly prolonged survival as a salvage treatment in patients with

recurrent glioblastomas (23 months vs 12 months; P < .0001) when compared to a historical control

group from the same institution with patients who had not been treated with radiosurgery for their

recurrence 365. And a case-control study showed that the combination of salvage Gamma Knife

radiosurgery followed by bevacizumab could add potential survival benefit and could reduce the

risk of adverse radiation effects in patients with recurrent glioblastoma (33.2 months vs.

26.7months) 361.

Favorable outcome was reported after Gamma Knife treatment of glioblastoma recurrences were

reported in a recent study from Norway by Skeie and colleagues: Median survival after Gamma Knife

radiosurgery for recurrences was 12 months in 51 patients compared with 6 months in the cohort

that was treated with reoperation alone (P = 0.001) 360. The overall survival from the time of primary

diagnosis was 19 months in the Gamma Knife cohort versus 16 months for the surgical patients. The

complication rate was 9.8% after Gama Knife and 25.2% after reoperation 360.

It is intuitive that younger age and a smaller tumor volume are predictive of better outcome. In

cases of recurrent glioblastoma, for the cohort of younger patients (<70) in better clinical condition

(KPS >60) the survival from the date of radiosurgery was 14.3 months and 10 months when these

criteria were not fulfilled 368. In addition, it has been demonstrated by Shrieve that patients with

tumor volumes smaller than 10.1 cm3 survived longer after stereotactic radiosurgery 364.

Side Effects after Radiosurgery of Recurrent Glioblastoma

Side effects after the retreatment in the situation of recurrent glioblastoma are an important issue.

When radiosurgery is used as salvage treatment, even this local and highly focal radiation is biologi-

cally additive to the initially applied standard fractionated radiotherapy. The occurrence of adverse

Page 82: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

82

radiation effects reflects that problem. Side effects after radiosurgery are a function of irradiated

volume and radiation dose. Ideally the dose should be as low as possible in a small volume to avoid

adverse effects, but there might be a threshold of efficacy for the radiosurgical treatment.

Radiological adverse radiation effects were reported in 7.7% 369, 14%363, 23%358, 24%365, 33% 370,

37% 371 and even 46% 361, but not all adverse radiation effects that are detected in the MRI have

clinical consequences. Park described adverse radiation effects in 46% in a historical control group,

with 23% being clinically symptomatic 361. Similarly, Kong observed adverse radiation effects in

24.4% of 114 patients including 49 patients with WHO III gliomas 365, which necessitated surgical

resections in only 4 cases. Maranzano and colleagues reported adverse radiation effects in 23% that

were asymptomatic and required no specific treatment 358.

The risk for adverse radiation effects is not necessarily related to the treatment modality but

depends on the specific dose volume parameters, which are not provided in the reports. Using

Gamma Knife treatments with relatively low median prescription doses of 12.2 Gy (8-20Gy), Skeie

reported a series with a low risk for adverse radiation effects of 9.8%, which compared favorably

with the 33.8% complication rate after open surgery 360. In another recent Gamma Knife studies side

effects were comparably low with only 11% grade 3/4 toxicities attributable to treatment 340 and

other observed focal radiation necrosis in 7% 344.

When lower doses and restricted tumor volumes are treated with radiosurgery, a low risk for side

effects can be expected. In many radiosurgical series median prescription doses of 15-18Gy were

applied.

Outcome after Radiosurgical Salvage Treatment of Recurrent Glioblastoma

In 2005 Mahajan et al. reported a case-control study of stereotactic Linac radiosurgery for recurrent

glioblastoma multiforme in 41 patients 362. A control group of patients who did not undergo SRS was

created from an institutional database, and each case was matched for known prognostic factors in

glioblastoma with control patients undergoing at least one more surgical intervention for tumor

resection as part of salvage therapy. The median duration of overall survival from diagnosis was 26

months in the radiosurgically treated group and 23 months in the surgical control group. From the

date of radiosurgery, the median duration of survival was 11 months in the case group and 10

Page 83: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

83

months in the surgical control group, a difference that was not statistically significant. This entire

patient population had longer survival duration than that reported in the literature for patients with

similar prognostic factors 362.

In 2005 Combs et al reported 32 patients with recurrent glioblastoma multiforme who were treated

with Linac radiosurgery. The median applied dose was 15 Gy (range, 10-20 Gy) prescribed to the

80% isodose line that encompassed the target volume. The median interval between primary

irradiation and re-irradiation was 10 months. No concomitant chemotherapy was applied. The

median overall survival from primary diagnosis of the tumor was 22 months (range, 9-133 months)

and median overall survival after SRS was 10 months 359 .

In 2005 Hsieh reported 51 consecutive patients with glioblastoma who were treated with gamma

knife radiosurgery: 25 as an "upfront" adjuvant therapy after surgery or 26 at the time of tumor

progression. When Gamma Knife radiosurgery was performed after the time of initial tumor resec-

tion, an overall median survival of 10 months was reached 370.

In 2008 Kong and colleagues treated 65 patients with recurrent glioblastoma with either Gamma

Knife or Linac radiosurgery as a salvage treatment and compared these patients with a historical

control group from the same institution that had been treated without radiosurgery. It was

demonstrated that salvage radiosurgery significantly prolonged survival (23 months vs12 months; P

<.0001). Adverse radiation effects induced by radiosurgery were observed in 24.4% (of the total

population of 114 patients including 49 patients with WHO III glioma) 365 .

In 2009 Patel et al reported 26 patients with pathologically proven recurrent glioblastoma who were

treated with salvage re-irradiation by Linac radiosurgery with a median dose of 18 Gy (range: 12-20

Gy). All patients had previously received radiotherapy with a dose of 50-60 Gy. The time from pri-

mary radiation to re-irradiation was 12.5 months (0.77–119) and median survival time after

radiosurgery for recurrence was 8.5 months 369.

In 2009 Pouratian and colleagues retrospectively reviewed 26 patients with histopathological

diagnoses of glioblastoma at the time of progression who had been treated with Gamma Knife

radiosurgery. Patients survived a median of 9.4 months after treatment for progression. Applied

radiosurgical doses were relatively low and margin dose was a significant prognostic factor on

multivariate analysis. Patients treated at the time of progression had significantly longer overall sur-

vival than those treated on initial presentation (17.4 vs. 15.1 months, P = 0.003) 338.

In 2011 Maranzano and colleagues reported 22 patients with recurrent glioblastoma who were

Page 84: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

84

treated with radiosurgery or fractionated stereotactic radiotherapy, the treatment option being

chosen according to lesion size and location. Median survival from re-irradiation was 11 months,

23% of patients submitted to radiosurgery developed asymptomatic brain radionecrosis 358 .

In 2011 Elliot, Golfinos and colleagues from the New York University Langone Medical Center,

retrospectively analyzed 26 consecutive adults who underwent Gamma Knife radiosurgery for

recurrent high grade gliomas ( Median tumor volume 1.22 cc, median treatment dose 15 Gy) and

showed a median overall survival of 13.5 months for the entire cohort from the time of Gamma

Knife treatment. Values for 12-month actuarial survival from time of Gamma Knife radiosurgery

for glioblastoma multiforme (GBM; n=16), anaplastic mixed oligoastrocytoma (n=5) and

anaplastic astrocytoma (n=5) were 37%, 20% and 80%. Local failure occurred in 9 patients (37.5%)

at a median time of 5.8 months, and 18 patients (75%) experienced distant progression at a median

of 4.8 months. Complications included radiation necrosis in two patients and transient worsening

of hemiparesis in one patient 372.

In a case-control study Park and colleagues evaluated the efficacy and safety of gamma knife

stereotactic radiosurgery followed by bevacizumab in 11 patients with recurrent glioblastoma in

comparison to 44 case-matched controls who underwent Gamma Knife treatment without

additional bevacizumab. The median tumor volume was 13.6 cm3 and the median prescription dose

was 16 Gy (13-18 Gy). The patients who received bevacizumab had significantly prolonged overall

survival (33.2 months vs. 26.7months, P = 0.048), and were less likely to develop an adverse

radiation effect (9 vs. 46%, P = 0.037) 361.

In 2012 Skeie and colleagues published a comparative retrospective study: Gamma Knife was used

for recurrent glioblastoma (in 77 patients) and compared with the outcome after re-operation only:

median survival after retreatment was 12 months for the 51 patients receiving GKS compared with

6 months for reoperation only (P = 0.001), and median survival was 19 months versus 16 months

from the time of primary diagnosis (P = 0.021). The study showed significantly longer survival for

patients treated with Gamma Knife, both from retreatment (P = 0.013) and from primary diagnosis

(P = 0.002). The adjusted results were still significant after separate analysis according to tumor

volumes <5 mL, 5 to 20 mL, and >20 mL. The complications rate was 9.8% after GKS and 25.2% after

re-operation 360.

In 2014 Dodoo, Lippitz and colleagues assessed gamma knife radiosurgery in the management of

high-grade glioma recurrences in 55 consecutive patients with WHO III and WHO IV glioma. All

patients had undergone previous microsurgery and radiochemotherapy (median follow-up 17.2

Page 85: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

85

months; 2.5-114.2 months; median tumor volume 5.2 cm3, median prescription dose 20 Gy; 14-22

Gy), The patients with WHO III tumors showed a median survival of 49.6 months with and a 2-year

survival of 90%. After Gamma Knife radiosurgery of the recurrent tumors, these patients showed a

median survival of 24.2 months and a 2-year survival of 50%. The patients with WHO IV tumors had

a median survival of 24.5 months with a 2-year survival of 51.4%. After the recurrence was treated

with Gamma Knife radiosurgery, the median survival was 11.3 months and a 2-year survival: 22.9%

for the WHO IV patients 373.

In 2015 Niranjan, Lunsford and colleagues performed Gamma Knife SRS in 297 patients with

histologically proven glioblastomas (median tumor volume 14 cm3; range 0.26-84.2 cm3; median

prescription dose 15 Gy; range 9-25 Gy). All patients had received prior fractionated radiation

therapy, and 66% had undergone one or more chemotherapy regimens. Radiosurgery at the time

of tumor progression was associated with a median survival of 21.8 months 374 .

In conclusion, salvage stereotactic radiosurgery can be applied in the treatment of glioblastoma

recurrences. In the vast majority of studies the overall median survival ranged between 18 and 33.2

months, which is more favorable than the outcome in the best prognostic group of glioblastoma

patients (RPA class 3) after conventional treatment. In the salvage situation the median survival was

10-30 months after radiosurgery of the recurrence, which compares favorably to the outcome after

reoperation of recurrent glioblastoma (3.5-9 months), the treatment with temozolomide and the

median overall survival on bevacizumab after glioblastoma recurrence (4.5 months). For patients

with recurrent glioblastoma the outcome after use of stereotactic radiosurgery appeared to be at

least equivalent to repeated surgical resection and more effective than chemotherapy alone. Hence

the retrospective evidence shows that stereotactic salvage radiosurgery in general and Gamma

Knife radiosurgery in particular offers a safe and effective minimal invasive treatment strategy for

glioblastoma recurrences in a situation when only very few alternative options exist.

Table 2. : Radiosurgery as salvage treatment of recurrent glioblastoma:

(Series where radiosurgery was used for different cohorts for primary treatment and for recurrences may appear as separate groups in table 1 and 3: ,,, When series contain cohorts of patients treated

with various modalities, the number of radiosurgically treated patients is given in the table ()

Page 86: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

86

reference year type of

study

radiosurgery

method

median

age

(years)

median

prescription dose

number of

patients

Complication

Rate

(grade III or IV

toxicity)

median survival after

retreatment

(months)

overall median

survival

(months)

Skeie BS et al.

World Neurosurg.

2012,Dec;78(6):658-69.

1996-2007 comparative

retrospective

Gamma

Knife

55.3

(27-73)

12.2 Gy

(8 - 20 Gy) 77 ARE in 9.8% 12 mo 19 mo

Park KJ et al.

J Neurooncol. 2012

Apr;107(2):323-33

1987-2010 case-control

study

Gamma

Knife

without

bevacizumab

64

(41–77)

15 Gy

(10–20)

Tu vol: 9.5cc

44

46%

(ARE)

23% symptomatic

12.2 mo 26.7 mo

(21.8–31.6)

Park KJ et al.

J Neurooncol. 2012

Apr;107(2):323-33

(part of previous study)

2007-2010 case-control

study

Gamma

Knife

with

bevacizumab

62

(46–72)

16 Gy

(13–18)

Tu.vol.13.6 cc

11 9% 17.9 mo 33.2 mo

(23.7-42.7)

Pouratian N et al.

J Neurooncol. 2009

Sep;94(3):409-18

1991–

2007 retrospective

Gamma Knife

60.1

(12.9–

76.9)

6 Gy

(3-15) 26

No recorded

radiation necrosis

9.4 mo

(7.9–10.9)

17.4 mo

(14.4–20.4)

Kong DS et al.

Cancer. 2008 May

1;112(9):2046-51.

2000-2006 Prospective

cohort study

Gamma knife and

Linac

49

(5-75)

16 Gy

(12–50 Gy)

tu vol:10.6cc

65 24.4%

radionecrosis

13 mo

(10.6–16.0)

23.0 mo

(16.2–29.3)

Hsieh PC et al.

Neurosurgery. 2005

Oct;57(4):684-9

1998-2003 retrospective

Gamma Knife

59

(17-81)

12Gy

median vol:

21.6 cm3

26 33% radionecrosis

(17/51) 16.7 mo

Kondziolka D et al.

Neurosurgery. 1997

Oct;41(4):776-83

1987-1995 retrospective Gamma Knife 51

(3-72)

15.5 Gy (12-25

Gy)

median vol: 6.5 ml

(0.88-31.2)

19

GBM

recurrences

30 mo

(2-74)

MST Not

specified

For recurrent

GBM

Larson DA et al.

Int J Radiat Oncol Biol Phys.

1996 Dec 1;36(5):1045-53.

1987-1994 retrospective (?) Gamma Knife

Not

specified

for GBM

16 Gy

(5-37.5)

66

GBM

recurrences

not mentioned

14.3 mo when

brachytherapy criteria

were satisfied

10 mo when

brachytherapy criteria

were not satisfied

median

overall

survival not

specified for

recurrent

GBM

Arterio-venous malformations (AVM)

An arterio-venous malformation (AVM) in the brain is a vascular anomaly resulting in a direct shunt

Page 87: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

87

between arteries and veins. The absence of capillaries creates a lack of resistance and hence a short-

cut for blood to pass directly from arteries to veins. The result is a risk for a bleeding within the brain

potentially resulting in neurological deficits or even death. Other patients with AVMs of the brain

may experience epileptic seizures or other neurological symptoms even without a bleeding.

The overall detection rate of symptomatic AVM appears to be 0.94 per 100,000 person-years (95%

confidence interval, 0.57-1.30/100,000 person-years). The prevalence of detected, active (at risk)

AVM disease is unknown, but it can be inferred from incidence data to be lower than 10.3 per

100,000 population 375.

In general, AVMs are associated with a 2 to 4% annual hemorrhage risk. A deep location for AVMs

is a risk factor for neurological deficit for any possible treatment and is known to be a higher risk for

neurological deficit in case of a hemorrhage. Often deeply seated AVMs are considered inoperable.

According to the natural history of AVMs in the basal ganglia or thalamus and showed that 72% of

the patients presented with an intracranial hemorrhage and 86% of patients who had experienced

a hemorrhage, suffered either from hemiparesis or hemiplegia 376,377.

Patients with aneurysms within the nidus of the AVM, intranidal aneurysms, presented more

frequently with hemorrhage (72% compared with 40%, p < 0.001) and had a significantly increased

risk of bleeding, with a rate of 9.8% per year 378. Hence, patients with aneurysms and an AVM

warrant more aggressive surgical or endovascular treatment to reduce the risk of a hemorrhage in

the latency period after SRS 379. In addition, a venous stenosis increases the risk for hemorrhage 380.

When possible, AVMs are removed to prevent a cerebra hemorrhage. The removal can be achieved

through a classical neurosurgical operation under the microscope. As an alternative it is sometimes

possible to reduce the volume of the AVM or its blood flow using an intravascular approach, through

embolization, when the pathological arterio-venous shunts are occluded using highly selective

catheters. Unfortunately only in exceptions the endovascular approach achieves a complete

occlusion of the AVM.

An increasingly important alternative is the AVM treatment with Gamma Knife radiosurgery. Until

2014 more than 103,000 AVM treatments have been carried out using Gamma Knife radiosurgery

Page 88: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

88

and the results are well documented. The AVM is localized with both angiography and MRI and then

treated with focal Gamma Knife radiation. The result is an irritation of the vessel wall that slowly

occludes the pathological vessels of the AVM 381

The latency between Gamma Knife Radiosurgery and obliteration is dose dependent. In general it is

assumed that the obliteration process takes 2-3 years. This occlusion generally prevents potential

future bleedings and the patient is considered cured 379.

In a typical large of series 400 patients with AVM treated with Gamma Knife radiosurgery it was

shown that at 3 years after treatment 72 % of treated AVMs were obliterated and 87.3% at 5 years 382. The

obliteration rate increases with increasing prescription dose applied to the nidus periphery 383,384

with statistically significant correlation between outcome and dose prescribed to the margin 385-

387 .

Factors leading to a better obliteration rate were: previous hemorrhage (p = 0.0084), smaller nidus (p =

0.0023), higher radiation dose to the lesion's margin (p = 0.0495) and factors leading to an earlier obliteration

of the nidus were : male sex (p = 0.0001), previous hemorrhage (p = 0.0039), smaller nidus diameter (p =

0.0006) 382.

The incidence of adverse radiation effects is related to the volume of the 12 Gy isodose volume and hence

related to the size of the AVM, but the risks of developing permanent symptomatic sequelae from AVM

radiosurgery vary dramatically with location and, to a lesser extent, volume 388.

In general, reasons for treatment failures can be: 1.insufficient definition of Nidus due to suboptimal imaging

technique resulting in incomplete coverage of the AVM, 2.reexpansion after hematoma reabsorption,

3.failure to fill part of the nidus as a result of hemodynamic factors and 4. recanalization after

embolization. Sometimes suboptimal radiation dose (<20 Gy) in larger AVM volumes or in eloquent area

can result in lower obliteration rates. In some patients the AVM has been shown to fail to obliterate

despite correct target definition and adequate dose 389.

Regrowth of an AVM with subsequent hemorrhage has been documented in children and is

attributed to forces acting on the immature vasculature 390,391.

Page 89: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

89

Treatment Doses (AVM)

In an initial attempt to define effective radiosurgical doses in the treatment of AVM, Flickinger,

Lunsford and associates analyzed the results of radiosurgery in 197 AVM patients with 3-year

angiographic follow-up revealing a complete AVM obliteration in 72%. Multivariate logistic

regression analysis of in-field obliteration revealed a significant independent correlation with the

minimum target dose (p = 0.04), but not with volume or maximum dose. A sigmoid dose-response

curve for in-field obliteration was constructed 385. In a recent review (2013) Flickinger confirmed

the previous results by stating that the dose response of stereotactic radiosurgery of AVM is

relatively steep between 12 and 17 Gy, while leveling out at around 20 Gy 386. Even at low doses of

14Gy the chance of obliteration is in the range of 50% .

While prescription doses > 20 Gy are generally considered effective for an obliteration 389 , Inoue

published a series of 114 patients with AVMs who had been treated with Gamma Knife at lower

prescription doses of 20-Gy or less (mean margin dose 19.5 Gy) and demonstrated a total

angiographically documented obliteration in 85.5% 392. Symptomatic edema was seen in only 1.7%.

In two pediatric cohorts, AVM obliteration was achieved in 52% of patients who received a dose of

18-20 Gy and in 16% who received less than 18 Gy 393.

Surprisingly, in 363 pediatric patients Dinca, Kemeny and colleagues found no correlation between

outcome and radiosurgical dose when that dose was between 20 and 25 Gy, thus suggesting that

even the lower of these 2 doses may be effective 394.

.

Page 90: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

90

Dose response curves for AVM obliteration according to prescription dose and history of prior

embolization from 515 patients with >5 years of follow-up. Logistic model, α/β = -45.9; maximum

obliteration at 23 Gy; 95.7% with no embolization, 91.8% with prior embolization. From: Flickinger

et al. 386

Clinical Relevance of Technical Development

The outcomes for 492 patients (564 treatments) with large AVMs >10 cm3 treated by single-stage

Gamma Knife radiosurgery were retrospectively analyzed by the team from the Royal Hallamshire

Hospital, Sheffield, UK. Twenty-eight percent of the patients presented with haemorrhage at a

median age of 29 years (range: 2-75). From 1986 to 1993 (157 patients) plans were simplistic, based

on angiography using a median of 2 isocenters and a marginal dose of 23 Gy covering 45-70% of the

AVM (median volume 15.7 cm3). From 1994 to 2000 (225 patients) plans became more

sophisticated, a median of 5 isocenters was used, covering 64-95% of the AVM (14.6 cm3), with a

marginal dose of 21 Gy. Since 2000, MRI has been used with angiography to plan for 182 patients.

Median isocenters increased to 7 with similar coverage (62-94%) of the AVM (14.3 cm3) and

marginal dose of 21 Gy. Excluding the embolized patients, improvement in planning increased

obliteration rates from 28% to 36% and finally 63%. Improving treatment plans did not

significantly decrease the rate of persisting radiation-induced side effects (12-16.5%).

Complication rate rose with increasing treatment volume. One hundred and twenty-three patients

underwent a second radiosurgical treatment, with a 64% obliteration rate, and mild and rare

complications in 6% 395.

Page 91: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

91

General Obliteration Rate in AVM

In 2007 Liscak and associates summarized the results after gamma knife radiosurgery in 330 patients

with AVMs (median volume 3.9 cm3; range 0.15 to 28.6 cm3). When complete obliteration was not

achieved within 3 years, repeat radiosurgery was performed on 76 patients. AVM obliteration was

achieved in 74% patients after the first round of radiosurgery and in 47 (69%) after the second.

The overall chance of cure was 92% (269 patients). Final angiography verified complete obliteration

by 12 to 96 months (median, 25 mo) after initial radiosurgery. The risk of rebleeding after

radiosurgery was 2.1% annually until full obliteration, and the overall mortality from rebleeding was

1%. The risk of permanent morbidity after the first and second radiosurgery treatments were 2.7

and 2.9%, respectively 396.

Very similar results with obliteration rates of 72% at 3 years and 87.3% at 5 years had been shown

in 400 patients with AVM treated with Gamma Knife at the University of Tokyo as published by Shin,

Kirino and associates 382.

Previously Pollock and colleagues had reported excellent outcomes (obliteration without deficit)

in 66.7% and good outcomes (obliteration with minor deficit) in 6.25% of 144 patients after one

or more radiosurgical procedures 397. In this series 10.4% of patients sustained major deficits and

3.5% died.

In 2008 Douglas and Goodkin summarized the outcome of 95 patients with 99 treatable AVMs after

Gamma Knife treatment at the University of Washington (median AVM volume 3.8 cm3; range 0.12-

32 cm3, median prescription dose 20 Gy; median follow-up 38 months; range 3-91 months). The

Kaplan-Meier estimated 6-year AVM obliteration rate for the entire cohort was 71.4%. The

median time to AVM obliteration was 47 months, 7.4% experienced late toxicities. There were 2

fatal bleeds and 13.8% nonfatal bleeds after Gamma Knife surgery 398.

In 2014 Paúl, Martinez and associates published a retrospective, longitudinal study of 662

consecutive patients with brain AVMs treated with Gamma Knife brain AVMs at the Ruber

International Hospital, Madrid (median AVM volume 3.6 cm3 ; mean follow-up 11 years). The

obliteration rate after a single RS was 69.3%; after multiple RS, it was 75%. Positive predictors of

Page 92: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

92

obliteration included compact nidus, undilated feeders, smaller AVM volume, and higher marginal

dose 399

In a large retrospective study comprising 774 patients, it was demonstrated that non-smokers were

more likely to have obliterated AVMs through radiosurgery. Smoking was not predictive of

treatment complications or post-treatment hemorrhage. In a multivariate analysis a negative

smoking history (OR 1.9, p = 0.006) was a strong independent predictor of AVM obliteration. Of the

patients with obliterated AVMs, 31.9% were smokers, whereas 45% were not (p = 0.05) 400.

In 2014 Paúl, Martinez and associates published a retrospective, longitudinal study of 662

consecutive patients with brain AVMs treated with Gamma Knife brain AVMs at the Ruber

International Hospital, Madrid (median AVM volume 3.6 cm3 ; mean follow-up 11 years). The

obliteration rate after a single RS was 69.3%; after multiple RS, it was 75%. Positive predictors of

obliteration included compact nidus, undilated feeders, smaller AVM volume, and higher marginal

dose 399.

In a series of 102 patients patients treated for an AVM with single-dose or staged-dose Gamma Knife

stereotactic radiosurgery, overall nidus obliteration was achieved in 75% of patients. Morbidity

(19%) correlated with lesion size and Spetzler-Martin grade. For Spetzler-Martin Grade I-III AVMs,

treatment yielded obliteration rates of 100%, 89%, and 86%, respectively and 1% mortality. For

Spetzler-Martin Grade IV and V AVMs, outcomes were less favorable, with obliteration rates of 54%

and 0%, respectively 401.

Starke, Sheehan and associates proposed a grading scale to predict the long-term outcome of

radiosurgically treated AVM patients based on 1012 patients who had been followed-up for more

than 2 years. Obliteration had occurred in 69% of patients, post-radiosurgery hemorrhage

occurred with a yearly incidence of 1.14%, radiation-induced changes 38.2%, symptoms in 9.9%,

and permanent deficits in 2.1% 402.

The Virginia Radiosurgery AVM Scale assigned 1 point each for having an AVM volume of 2-4 cm3,

eloquent AVM location, or a history of hemorrhage, and 2 points for having an AVM volume greater

than 4 cm3. Eighty percent of patients who had a score of 0-1 points had AVM obliteration and no

Page 93: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

93

post-treatment hemorrhage, as did 70% who had a score of 2 points and 45% who had a score of 3-

4 points 402.

Side Effects (AVM)

An early essential study by Flickinger, Lunsford and colleagues had established that the 12 Gy

volume was the only independent variable that correlated significantly with post-radiosurgical

imaging changes (p < 0.0001), while symptomatic post-radiosurgical imaging changes were

correlated with both 12 Gy volume (p = 0.0013) and AVM location (p = 0.0066). In this series of 307

AVM patients who received gamma knife radiosurgery at the University of Pittsburgh between 1987

and 1993 the 7-year actuarial rate for developing persistent symptomatic post-radiosurgical imaging

changes was 5.05% 403. Recently Hayhurst, Zadeh and colleagues confirmed these results and found

in 125 patients with radiosurgically treated AVM that target volume and 12 Gy volume were the

most significant predictors of adverse radiation effects on univariate analysis (p < 0.001) with a

significant target volume threshold of 4 cm3, above which the rate of adverse radiation effects

increased significantly 404.

Radiation-induced imaging changes are newly developed increased T2 signal surrounding the

treated AVM nidi are seen considerably more frequently than clinical side effects: in a large series

of 1426 Gamma Knife procedures for AVMs from the University of Virginia, Charlottesville,

radiation-induced imaging changes were seen in 33.8% of treated AVM at a median latency of 13

months (range 2-124 months). After a median of 22 months the imaging changes disappeared

completely (range 2-128 months). The overall incidence of symptomatic imaging changes was 8.6%

and 1.8% had permanent deficits 405. Similarly, in a subgroup of 140 patients treated with repeat

Gamma Knife at the University of Virginia, Charlottesville, radiation-induced changes were

visualized on magnetic resonance imaging in 39%, while only 3.6% of patients developed

permanent neurological deficits 389.

Hypertension, patient sex, tobacco use, number of draining veins, superficial or deep location of the

lesion, and AVM embolization prior to radiosurgery had no association with the presence of

radiation-induced changes. According to a multivariate analysis, larger AVM volume, worse

Spetzler-Martin grade, and no AVM surgery prior to radiosurgery predicted the occurrence of

Page 94: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

94

radiation-induced changes. Diabetes mellitus had borderline significance 406.

In general, there is a low risk of side effects after Gamma Knife radiosurgery of AVM. Clinical side

effects have been reported in 1.5% 407, 2.3% 379 , 3.2% -3.6% (in children) 394,408, 3.6% 389 and 4.5%

in AVMs of the basal ganglia and thalamus 409 and 6% in Spetzler-Martin Grade III AVM 410.

During a mean long-term follow-up period of 10.2 years (range, 5.4-30.6) in 181 consecutive

patients who underwent Gamma Knife radiosurgery for AVM, 8.3% experienced stereotactic

radiosurgery-related, symptomatic complications. Among these 15 patients, 12 manifested

complications 5 years or more after Gamma Knife 411 .

A higher rate of long-term side-effects in 9.3%, occurring 2 or more years after gamma knife surgery,

was observed by Izawa, Takakura and colleagues in 2005. The complications included hemorrhage

(n=8), delayed cyst formation (n=8), increase of seizure frequency (n=4), and middle cerebral artery

stenosis (n=1) and increased white matter signal intensity on T2-weighted magnetic resonance

imaging (n=1). The long-term complications were associated with larger nidus volume (p < 0.001)

and a lobar location of the AVM (p < 0.01). Delayed cyst formation was associated with a higher

maximal GKS dose (p < 0.001), larger nidus volume (p < 0.001), complete nidus obliteration (p <

0.01), and a lobar location of the AVM (p < 0.05). Out of 237 cases in this series, complete AVM

obliteration was attained in 54.9% 412.

In highly eloquent anatomical regions such as the brainstem permanent neurological deficits due to

adverse radiation effects developed in 10% 413.

Also in larger AVM the risk for symptomatic adverse radiation is known to be higher, which is a clear

limitation of stereotactic radiosurgery. Volume staged treatments are carried out in order to reduce

these potential side effects. Symptomatic adverse radiation were detected after volume-staged

Gamma Knife radiosurgery of larger AVM in 13% of patients 414 and after repeat radiosurgery in

9.5% 415.

Page 95: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

95

Gamma Knife treatment of AVMs Spetzler-Martin Grade III

Based on the functional eloquence of the affected surrounding brain, the size of the AVM and the

type of venous drainage, cerebral AVMs are categorized according to Spetzler-Martin Grades.

Spetzler-Martin Grading allows the prediction of potential side effects associated with a surgical

resection of the AVM. The impact of this grading system cannot be directly translated into

radiosurgical indications, but AVMs that are classified according to Spetzler-Martin Grades Grade III

can be very difficult to remove surgically due to a high risk of potential side effects.

Over the 20-year span from 1989 to 2009 a total of 398 patients with Spetzler-Martin Grade III AVMs

were treated with stereotactic Gamma Knife radiosurgery at the University of Virginia (median AVM

volume 2.8 cm3, median prescription dose 20 Gy; median radiological follow-up intervals 54 months).

Complete AVM obliteration was observed in 69% of Grade III AVM cases at a median time of 46

months after radiosurgery with actuarial obliteration rates at 3 and 5 years were 38% and 60%,

respectively. The obliteration rate was higher in ruptured AVMs than in unruptured ones (p < 0.001)

and in cases with a single draining vein (p = 0.018). The annual risk of postradiosurgery hemorrhage

during the latency period was 1.7%. The rates of symptomatic and permanent radiation-induced

imaging changes were 12% and 4%, respectively 416.

The corresponding series from Pittsburgh comprised 474 patients with Spetzler-Martin Grade III

AVM (median target volume 3.8 ml; range 0.1-26.3 ml; prescription dose 20 Gy; range 13-25 Gy)

showed virtually identical results: At a mean follow-up of 89 months, the total obliteration rates

documented by angiography or MRI increased from 48% at 3 years to 69% at 4 years, 72% at 5 years,

and 77% at 10 years. The cumulative rate of hemorrhage was 2.3% at 1 year, 4.4% at 2 years, 5.5%

at 3 years, 6.4% at 5 years, and 9% at 10 years 410. Symptomatic adverse radiation effects were

detected in 6%.

The consequence is that Gamma Knife radiosurgery can be considered as a safe treatment

alternative in these otherwise very complex and often inoperable AVMs.

Page 96: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

96

Gamma Knife treatment for AVM Spetzler-Martin Grade I or II

Patients with AVMs classified according to Spetzler-Martin Grade I or II are generally considered as

operable. In these cases surgery is generally considered as treatment of choice, since the bleeding

risk disappears immediately with successful surgery and a resection can generally be carried out at

a limited risk for surgical side effects.

However, under specific circumstances surgery is not always possible and Gamma Knife

radiosurgery is an effective and relatively safe option for patients with smaller volume Spetzler-

Martin Grade I or II AVMs who decline initial resection 379.

Kano, Kondziolka and the team from the University of Pittsburgh analyzed the outcome for Gamma

Knife radiosurgery of AVM classified according to Spetzler-Martin Grade I and II in 217 patients

(median target volumes 2.3 cm3; range 0.1-14.1 cm3; median margin dose 22 Gy; range 15-27 Gy).

The actuarial rates of total obliteration determined by angiography or MR imaging after 1 Gamma

Knife procedure were 58%, 87%, 90%, and 93% at 3, 4, 5, and 10 years, respectively 379. The median

time to complete MR imaging-determined obliteration was 30 months. Factors associated with

higher AVM obliteration rates were smaller AVM target volume, smaller maximum diameter, and

greater marginal dose. The annual bleeding risk was 3.7%, 0.3%, and 0.2% for Years 0-1, 1-5, and 5-

10, respectively, after radiosurgery. The presence of a coexisting aneurysm proximal to the AVM

correlated with a significantly higher hemorrhage risk. Temporary symptomatic adverse radiation

effects developed in 2.3% after SRS, and 1% developed delayed cysts 379.

Dural Arteriovenous Fistulas

So far treatment options and management strategies for dural arterio-venous fistulas are ranging

from observation to surgical excision to endovascular embolization and radiosurgery. Niranjan and

Lunsford made an attempt to formulate guidelines about the use of radiosurgery in symptomatic

patients with imaging-identified dural arterio-venous fistulas of the brain and concluded that

Gamma Knife treatment before embolization facilitates the better recognition of the entire target.

For selected dural arterio-venous fistulas patients who are not eligible for embolization or surgery,

radiosurgery alone is an effective treatment option. The dose range for dural arterio-venous fistulas

is similar to that of arterio-venous malformations 417. A total of 40 patients with dural arterio-

Page 97: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

97

venous fistulas underwent Gamma Knife treatment at the University of Pittsburgh (median target

volume 2.0 cm3, median marginal dose 21.0 Gy). The obliteration rate was 83% for patients who

had upfront SRS and embolization. The obliteration rate was lower (67%) for patients managed

with SRS alone 418.

Of 22 patients who underwent Gamma Knife surgery for the management of dural arteriovenous

fistulas at the University of Tokyo Hospital, 55% had an obliteration. The obliteration rate for lesions

without cortical venous drainage was 86% and thus significantly higher than in fistulas with

cortical venous drainage (47%) (p = 0.007). Hemorrhage at presentation (p = 0.03), a target volume

less than 1.5 cm(3) (p = 0.009), and Cognard Type III or IV dAVF (p = 0.005) were factors associated

with a higher obliteration rate419.

During the period between 1972 and 2008, 65 consecutive patients patients with dural arterio-

venous fistulae were treated and followed-up at the Karolinska Hospital Stockholm. Prescription

doses varied considerably, but most commonly it was 20-25 Gy to the 40-60% isodose. There were

59% obliterations, 2 post-treatment hemorrhages and 5 minor adverse radiation effects 420.

In 2013 Pan, Guo and associates presented an update on 321 patients with dural arteriovenous

fistulas (DAVF) treated at the Taipei Veterans General Hospital using Gamma Knife radiosurgery.

The prescribed mean margin dose was 17.2 Gy. For the cavernous sinus DAVFs, 70% showed

complete improvement. For the non-cavernous- sinus DAVFs, 59% showed complete improvement.

The results show that radiosurgery was effective in treating Borden type I lesions with a 72%

complete obliteration rate. However, for Borden type II and III lesions, a lower cure rate was

observed with complete obliteration in 44% 421 . In this series, 98% of patients had a stable or

improved clinical condition after radiosurgery.

Effect of Gamma Knife on Seizure Activity in AVM Patients

Yang, Paek and colleagues reviewed a series of 86 patients with a history of seizure who underwent

stereotactic radiosurgery for unruptured cerebral AVM with a mean follow-up of 89.8 months

showing that 76.7% were seizure-free and 58.1% were medication-free at the last follow-up visit.

Of the patients who achieved AVM obliteration, 96.7% (58/60) were seizure-free while 30.8%

Page 98: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

98

(8/26) of those patients who did not achieve AVM obliteration were seizure-free (p = 0.001). Of the

75 patients with no history of seizure before radiosurgery, 10 had provoked seizures due to the

direct and indirect radiosurgical influences after radiosurgery 422.

Similarly Lim, Rhee and colleagues found after Gamma Knife radiosurgery for AVMs in patients who

had presented with seizures (n=45), that 84.6% of patients with complete obliteration were

seizure-free. In total 53.5% became seizure-free, 23.3% had significant improvement, 18.6% were

unchanged and 4.6% were aggravated 423.

Pediatric AVM

In a comparison between pediatric and adult patients with AVM, Pan, Wong and associates from

the Taipei Veterans General Hospital found in 2008 that AVMs between 3 and 10 cm3 responded

less efficaciously in children (p = 0.042), while obliteration rates of small and extremely large (> 20

cm3) AVMs were similar in the pediatric and adult groups 424.

This team had summarized their data after Gamma Knife surgery for cerebral AVM in 105 pediatric

patients (< 18 years of age) and compared the outcome with the results in 458 adult patients with

AVMs: in pediatric patients, the AVM obliteration rate at 48 months after a primary GKS was 65%.

The efficacy of GKS correlated with the size of the AVM: 91% for small, 86% for medium, and 64%

for large AVMs 424. The treatments were associated with an 8% morbidity rate and < 1% mortality

rate. Post-treatment hemorrhage occurred in 4% 424.

At the National Centre for Stereotactic Radiosurgery, Royal Hallamshire Hospital, Sheffield, 363

pediatric patients (ages 1-16 years) underwent Gamma Knife surgery (mean lesion volume 3.75 ±

5.3 cm3; range 0.01-32.8 cm3, mean prescription dose was 22.7 ± 2.3 Gy; range 15-25 Gy). The most

common presenting symptoms had been: hemorrhage (80.2%), epilepsy (8.3%; overall seizure

prevalence 19.9%), and migrainous headaches (6.3%). Only 0.28% of the AVMs were incidental

findings. The overall obliteration rate was 82.7% and was 71.3% in patients who received one

treatment and 62.5% for retreated patients, with a mean obliteration time of 32.4 and 79.6 months,

respectively. The bleeding rate post-radiosurgery was 2.2%, and the cumulative complication rate

was 3.6% 394.

Page 99: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

99

In 2010 Yen, Sheehan and associates reviewed 186 patients with AVMs who were 18 years of age

or younger were treated with Gamma Knife radiosurgery at the University of Virginia (mean nidus

volume 3.2 cm3, mean prescription dose 21.9 Gy). After initial GKS, 49.5% of patients achieved

total angiographic obliteration. The obliteration rate increased to 58.6% after a second or multiple

GKS. The hemorrhage rate was 5.4% within 2 years after GKS and 0.8% between 2 and 5 years. Six

patients developed neurological deficits along with the radiation-induced changes. Two patients

developed asymptomatic meningiomas 10 and 12 years after GKS. After a mean clinical follow-up

of 98 months, less than 4% of patients had difficulty attending school or developing a career 408.

In another large retrospective analysis Kano, Lunsford and associates from the University of

Pittsburgh reviewed the outcome of 135 pediatric patients with cerebral AVM after Gamma Knife

surgery (median maximum diameter 2.0 cm; range 0.6-5.2 cm; median target volumes 2.5 cm3;

range 0.1-17.5 cm3, median margin dose 20 Gy; range 15-25 Gy). The actuarial rates of total

obliteration documented by angiography or MR imaging were 45%, 64%, 67%, and 72% at 3, 4, 5,

and 10 years, respectively 407. The median time to complete angiographically documented

obliteration was 48.9 months. Factors associated with a higher rate of documented AVM

obliteration were smaller AVM target volume, smaller maximum diameter, and larger margin dose.

In 6% a hemorrhage occurred during the latency interval, and 1 patient died. The rates of AVM

hemorrhage after SRS were 0%, 1.6%, 2.4%, 5.5%, and 10.0% at 1, 2, 3, 5, and 10 years, respectively.

The overall annual hemorrhage rate was 1.8%. Permanent neurological deficits due to adverse

radiation effects developed in 1.5% after SRS, and in 0.7% delayed cyst formation occurred 407.

In 2014 Potts, Gupta and associates published a retrospective review of 2 cohorts of 80 children

with AVMs treated from 1991 to 1998 and from 2000 to 2010 (median target volume 3.1 cm3 ; range

0.09-62.3 cm3, median prescription dose 17.5 Gy; range 12-20 Gy). AVM obliteration was achieved

in 52% of patients who received a dose of 18-20 Gy and in 16% who received less than 18 Gy. At 5

years after SRS, the cumulative incidence of hemorrhage was 25%. No permanent neurological

deficits occurred in patients who did not experience post-treatment hemorrhage 393.

In another series of children with arteriovenous malformations who had been treated with Gamma

Knife radiosurgery at the Gazi University, Ankara (median AVM volume 3.5 cm3), complete AVM

Page 100: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

100

obliteration was achieved in 68.9%, while 15.51% of the patients experienced new deficits and 5.1%

experienced intracranial hemorrhage 425. In a series from Tokyo Women's Medical University, 22

children underwent GKS for AVMs (prescribed isodose volume < 4.0 cm3, median prescription dose

22 Gy; range 20-25 Gy). If the volume of the nidus was larger than 4.0 cm3, a second radiosurgical

session was planned for 3-4 years after the first one. Complete obliteration of the AVM was noted

in 77% within a median period of 47 months after the last radiosurgical session. Complete

obliteration of the lesion occurred in 89% of patients after unstaged treatment and in 62.5% after

staged GKS426. Complications included 3 bleeding episodes.

Unruptured cerebral arterio-venous malformations in pediatric patients (age <18 years) had been

excluded from ’A Randomized Trial of Unruptured AVMs’ (ARUBA). Ding, Sheehan and colleagues

analyzed the outcome in 51 unruptured pediatric AVM patients who had been treated with Gamma

Knife radiosurgery (median nidus volume 3.2 cm3 ; radiosurgical margin dose 21.5 Gy; median

radiologic follow-up 45 months). The actuarial AVM obliteration rates at 3, 5, and 10 years were

29 %, 54 %, and 72 %, respectively. Obliteration rates were significantly higher with a margin dose

of at least 22 Gy and for nidi with 2 or fewer draining veins. The incidences of radiologically evident,

symptomatic, and permanent radiation-induced changes were 55 %, 16 %, and 2 %, respectively.

The annual post-radiosurgery hemorrhage rate was 1.3 %, and the incidence of post-radiosurgery

cyst formation was 2 % 427.

Thalamic/Basal Ganglia and Brainstem AVM

Nagy, Kemeny and associates conducted a retrospective analysis of 356 thalamic/basal ganglia and

160 brainstem AVMs treated with gamma knife radiosurgery. Median volume was 2cm3; (range

0.02-50) for supra-tentorial and 0.5cm3 (0.01-40) for brainstem AVMs; the marginal treatment doses

were 17.5-25Gy. After single treatment, obliteration was achieved in 65% of the brainstem, in 69%

of the supra-tentorial, and 40% of the peri-tectal AVMs. Complications were rare (6-15%) and mild

(≤MRS2). Rebleed rate increased with size, but was not higher than before treatment. AVMs >4cm

in the brainstem were treated with unacceptable morbidity and low cure rate 428.

In 2012 Kano, Lunsford and colleagues from the University of Pittsburgh reviewed the outcome of

133 patients in centrally located AVM: 56 patients who had been treated with Gamma Knife surgery

Page 101: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

101

had AVMs of the basal ganglia and 77 had AVMs of the thalamus (median target volume 2.7 cm3;

range 0.1-20.7 cm3; median margin dose 20 Gy, range 15-25 Gy). The actuarial rates of total

obliteration after radiosurgery were 57%, 70%, 72%, and 72% at 3, 4, 5, and 10 years, respectively

409. Factors associated with a higher rate of AVM obliteration included AVMs located in the basal

ganglia, a smaller target volume, a smaller maximum diameter, and a higher margin dose. Eleven

percent (of 133 patients) suffered a hemorrhage during the latency period and 7 patients died. The

rate of post-SRS AVM hemorrhage was 4.5%, 6.2%, 9.0%, 11.2%, and 15.4% at 1, 2, 3, 5, and 10

years, respectively. The overall annual hemorrhage rate was 4.7%. Permanent neurological deficits

due to adverse radiation effects developed in 4.5%, and in 1 patient a delayed cyst developed 56

months after SRS 409.

In 67 patients had brainstem AVMs treated by the same group (median target volume 1.4 cm3; range

0.1-13.4 cm3; median margin dose 20 Gy; range 14-25.6 Gy) showed actuarial rates of total

obliteration were 41%, 70%, 70%, and 76% at 3, 4, 5, and 10 years, respectively with AVM

hemorrhage after SRS in 3.0%, 3.0%, and 5.8% at 1, 5, and 10 years, respectively. The overall annual

hemorrhage rate was 1.9%. Permanent neurological deficits due to adverse radiation effects

developed in 10% after SRS, and a delayed cyst developed in 3%. Higher 12-Gy volumes and higher

Spetzler-Martin grades were associated with a higher risk of symptomatic AREs 413.

In 2009 Kiran, Mahapatra et al. had studied the outcome of 53 patients with central AVMs in basal

ganglia, thalamus and brainstem (central/deep) treated with Gamma knife radiosurgery at the

Neuro Sciences Centre, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India (mean

marginal dose 23.3 Gy; range 16-25 Gy; mean follow-up was 28 months (range 12-96 months) and

published a complete obliteration of the AVM was documented in 14 of the 19 patients with

complete angiographic follow-up 429.

In AVMs located in the basal ganglia, thalamus, or brainstem (n=56; median volume 3.8 cm3; median

prescription dose 18 Gy) Pollock and associates found an obliteration in 43% after one radiosurgical

procedure and in 57% after one or more interventions 377.

Volume-staged Gamma Knife Radiosurgery in Large AVM

Page 102: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

102

Large AVM have a reduced chance of obliteration and increased risk for side effects after single

session radiosurgery. As a result, volume-staged Gamma Knife radiosurgery has been proposed in

order to reduce the risk for side effects after radiosurgery.

Volume-staged SRS for large AVMs unsuitable for surgery has potential benefit but often requires

more than 2 procedures to complete the obliteration process. To have a reasonable chance of

benefit, the minimum margin dose should be 17 Gy or greater 414.

Patients with large AVMs underwent volume-staged Gamma Knife radiosurgery at the University of

Pittsburgh School of Medicine (n=47). The median interval between the two SRS procedures was

4.9 months (range, 3-14 months). The median nidus volume was 11.5 cm3 (range, 4.0-26 cm3) in

the first stage of SRS and 9.5 cm3 in the second. The median margin dose was 16 Gy (range, 13-18

Gy) for both SRS stages. The actuarial rates of total obliteration after 2-staged SRS were 7, 20, 28

and 36% at 3, 4, 5 and 10 years, respectively. Symptomatic adverse radiation effects were

detected in 13% of patients 430.

In 2012 Yamamoto and colleagues described 31 patients in whom 2-stage GKS was intentionally

planned at the time of initial treatment because the volume of the AVM nidus was larger than 10

cm3 (mean nidus volume 16.2 cm3, maximum 55.8 cm3; low prescription doses: 12-16 Gy) The

second GKS was scheduled for at least 36 months after the first. The cumulative complete

obliteration rate in this series was 76.2% (16 of 21 eligible patients), while 22.6% experienced

bleeding. The bleeding rates were 9.7%, 16.1%, 16.1%, and 26.1%, respectively, at 1, 2, 5, and 10

years post-GKS. There were 2 deaths and 3 cases of morbidity (persistent coma, mild hemimotor

weakness, and hemianopsia in 1 patient each). Mild symptomatic GKS-related complications

occurred in 6.5%.The rate of complications in the pediatric cases was 33.3%, whereas that in the

adolescent and adult cases was 0% (p = 0.0323) 431.

The rates for complete angiographic occlusion in very large AVMs are considerably lower than in

series with smaller AVMs as demonstrated in a small cohort of 18 patients with very large AVMs (>

15 cm3) who underwent prospective staged-volume radiosurgery at the Langone Medical Center,

New York (median AVM volume 22.9 cm3; range, 15.7-50 cm3). Separate anatomic volumes were

Page 103: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

103

irradiated at 3- to 9-month intervals (median volume, 10.9 cm3; range, 5.3-13.4 cm3; median

marginal dose, 15 Gy; range, 15-17 Gy). Actuarial rates of complete angiographic occlusion were

29% and 89% at 5 and 10 years, while 27.8% had a hemorrhage after radiosurgery 432.

Retreatment after Failed Obliteration

In general, AVMs can be retreated when the first radiosurgical session failed to provide a

complete obliteration.

With regard to the additional radiation In 105 patients who had incompletely obliterated AVMs,

repeat Gamma Knife radiosurgery was performed at a median of 40.9 months after initial SRS (range

27.5-139 months). Median AVM target volume was 6.4 cm3 (range 0.2-26.3 cm3) at initial SRS and

was reduced to 2.3 cm3 (range 0.1-18.2 cm3) at the time of the second procedure. The median

margin dose at both initial SRS and repeat SRS was 18 Gy. The actuarial rate of total obliteration by

angiography or MR imaging after repeat SRS was 35%, 68%, 77%, and 80% at 3, 4, 5, and 10 years,

respectively. The median time to complete angiographic or MR imaging obliteration after repeat

SRS was 39 months. Factors associated with a higher rate of AVM obliteration were smaller residual

AVM target volume (p = 0.038) and a volume reduction of 50% or more after the initial procedure

(p = 0.014). Factors associated with a higher risk of hemorrhage after repeat SRS were a greater

number of prior hemorrhages (p = 0.008), larger AVM target volume at initial SRS (p = 0.010).

Symptomatic adverse radiation effects developed in 4.8% after initial SRS and in 9.5% after repeat

SRS 415. A smaller group of patients who had undergone prospectively staged volume radiosurgery

had been published earlier by the same group 433.

Repeat Gamma Knife surgery yielded a total angiographic obliteration in 55% and subtotal

obliteration in 6.4% in a series of 140 patients with cerebral AVMs from the University of Virginia,

Charlottesville. Although radiation-induced changes as visualized on magnetic resonance imaging

occurred in 39%, only 3.6% developed permanent neurological deficits 389.

Gamma Knife Thalamotomy for Tremor

Page 104: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

104

Medication is the standard treatment for the management of patients with movement disorders.

However, in a fraction of patients with insufficient effect or bothersome side-effects of

pharmaceuticals, surgical interventions are considered, among those deep brain stimulation (DBS)

and surgical lesioning of the thalamus and basal ganglia as established invasive neurosurgical

procedures. Lesioning in functional neurosurgery has maintained an important role in the treatment

of movement disorders and a variety of lesioning technologies are still available. In a recent

worldwide survey of a total of 353 neurosurgeons from 51 countries who had operated on 13,200

patients showed that ablative surgery is still used by about 65% of neurosurgeons, regardless of

their country's economic status 434 In addition to these surgical techniques, alternatives are needed

since in some patients invasive options are contraindicated. In addition, DBS is a very costly

treatment with high expenses for the implanted electrodes and with necessity for changing of the

implanted batteries on a regular basis.

Functional lesions can also be safely and accurately applied using radiosurgical Gamma Knife

techniques. Stereotactic Gamma Knife radiosurgery provides a non-invasive alternative option in

movement disorders and even in selected cases of psychosurgery. Radiosurgery can be necessary

for medication-intolerable patients. The use of functional Gamma Knife lesioning to treat movement

disorders is particularly attractive when surgery is contra-indicated, as sometimes in older patients

and in patients with relevant co-morbidities or coagulopathies.

With a recent emphasis on health economy, the use of radiosurgery for functional lesions has

regained new interest. A recent review showed that Gamma Knife thalamotomy provided favorable

outcomes in the treatment of tremor, with published success rates ranging from 80-100% 435 .

In 2013 Kooshkabadi, Kondziolka and colleagues evaluated outcomes in 86 patients who had

undergone unilateral Gamma Knife thalamotomy for tremor during a 15-year period that spanned

the era of MRI-based target selection (1996-2011). A single 4-mm isocenter was used to deliver a

maximum dose of 140 Gy to the posterior-inferior region of the nucleus ventralis intermedius. After

Gamma Knife, 66% of treated patients showed improvement in both tremor, handwriting and

drinking score, 13% in 2 scores, and 2% in just 1 score 436. In 19% there was a failure to improve in

any score. Two patients developed a temporary contralateral hemiparesis, 1 patient noted

dysphagia, and 1 sustained facial sensory loss 436 .

Page 105: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

105

From June 2012 to August 2013, 7 patients with an intractable tremor underwent Gamma Knife

thalamotomy with a maximal dose of 130 Gy to the left ventralis intermedius (VIM) nucleus of the

thalamus at the Samsung Medical Center, Seoul, Korea. Six out of seven patients showed objective

improvement in the Fahn-Tolosa-Marin tremor rating scale (TRS) score. Excluding one patient who

demonstrated tremor progression, there was 28.9% improvement in the TRS score 437.

In 2012 a multicenter collaborative trial at 6 Japanese institutions was conducted comprising a total

of 72 patients with Parkinson disease and essential tremor who had been treated with Gamma knife

thalamotomy in the lateral part of the ventralis intermedius nucleus with a single 130-Gy shot using

a 4-mm collimator. Excellent or good results were seen in 81.1% out of 53 patients who completed

24 months of follow-up 438. No permanent clinical complications were observed.

In 2010 Lim and colleagues published the outcome after unilateral Gamma Knife radiosurgery for

18 patients with disabling tremor refractory to medications who were treated at the University of

Malaya, Kuala Lumpur, Malaysia. After radiosurgery the Fahn-Tolosa-Marin Tremor Rating Scale

activities of daily living scores improved significantly but in this series radiosurgery provided only

modest antitremor efficacy 439.

In 2010 Young and associates published the outcome of VIM thalamotomy procedures with the

Gamma Knife in 161 patients who underwent a total of 203 thalamotomies (119 unilateral and 42

bilateral) with a mean postoperative follow-up duration of 44 +/- 33 months. Overall, 81% of

patients showed improvements in drawing scores, for writing, 77% of patients showed

improvement 440. There were side effects in 8.7%, a total of 5% were permanent. All complications

were related to lesions that grew larger than expected 440.

Kondziolka analyzed the outcome of 26 patients after Gamma Knife treatment for disabling essential

tremor using a single 4-mm isocenter at a maximum dose of 130 or 140 Gy to the nucleus ventralis

intermedius with a median follow-up of 36 months. After radiosurgery, 69% of treated patients

showed improvement in both action tremor and writing scores, 23% only in action tremor scores,

and 12% in neither tremor nor writing 441. Permanent mild right hemiparesis and speech

impairment developed in 1 patient 6 months after radiosurgery. Another patient had transient mild

Page 106: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

106

right hemiparesis and dysphagia 441.

Between 1993 and 2005 Ohye and colleagues at the Hidaka Hospital, Gunma, Japan had performed

Gamma Knife radiosurgery in 70 patients with Parkinson’s disease. The thalamic reaction observed

on MR or CT imaging varied with one lesion type being represented by a circumscribed round high

signal area of approximately 7- to 8-mm diameter surrounding a smaller low signal area. The other

is characterized by an irregular-shaped high signal zone extending into the medial thalamic area

and/or the internal capsule 442.

Similarly in 1998 Duma published a series of 38 patients using gamma knife radiosurgery for

thalamotomies to treat parkinsonian tremor (median maximum radiation dose130 Gy; range 100-

165 Gy) showing excellent relief of tremor in 26% and complete abolition of tremor in 24 %. Good

improvement was seen in 29%. mild improvement was seen in 10.5% and 10.5% of thalamotomies

failed 443. The median time to onset of improvement was 2 months.

In 1999 Niranjan published a series of 15 stereotactic Gamma Knife procedures for patients with

intractable tremor performed at the University of Pittsburgh Medical Center. Radiosurgery usually

was performed in elderly patients with concurrent medical problems. For radiosurgery, a median

dose of 140 Gy (range 130-150 Gy) was delivered using a single 4-mm collimator. Of 12 evaluable

radiosurgery patients, 10 noted excellent relief and 2 had partial relief 444.

The average delay of clinical improvement is about 6 months after radiosurgery 445.

In a small series with patients suffering from multiple sclerosis tremor all 6 patients experienced

improvement in tremor after a median latency period of 2.5 months after Gamma Knife. More

improvement was noted in tremor amplitude than in writing and drawing ability. In four patients,

the tremor reduction led to functional improvement. One patient suffered from transient

contralateral hemiparesis, which resolved after brief corticosteroid administration 446.

Target Definition (VIM)

Page 107: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

107

Sato, Ohye and associates performed depth recording around the region of the estimated optimum

target and determined optimal targets as points 6-8 mm anterior to the posterior commissure, 4-6

mm dorsal to the level of the intercommissural line, and 15-17 mm lateral from the midline.

Rhythmic discharge time-locked to tremor and/or kinesthetic neurons were found within the

expected target area in all patients 447.

For target definition (VIM) Ohye proposed to initially refer to the posterior commissure for an

approximate determination of the lateral part of the ventral intermediate nucleus. Then the point

is further corrected by anatomical landmark (45% of the thalamic length) to compensate the

individual difference 445. Ohye uses then Gamma Knife lesion using a 4-mm collimator, with a

maximum dose of 130 Gy in 1 shot 445.

GK thalamotomy for tremor treatment in published series. From Elaimy et al. 435

1st Author (Year) # Patients

(Age Range)

Radiation

Dose (Gy)

Follow-up

(Range)

Improvement Rate Complications Observed Complication

Rate

Young [9] (2010)

Young RF, Li F, Vermeulen S,

Meier R: Gamma Knife

thalamotomy for treatment of

essential tremor: long-term

161 (18-93yrs) 141-152 Mean: 44 ± 33

months

Drawing: 81%

Writing: 77%

sensory loss, motor

impairments, dysarthria

8.4%

Page 108: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

108

results.

J Neurosurg 2010, 112:1311-

1317.

Lim (2010)

Lim SY, Hodaie M, Fallis M, Poon

YY, Mazzella F, Moro E: Gamma

knife thalamotomy for disabling

tremor: a blinded evaluation.

Arch Neurol 2010, 67:584-588.

18 (64-83 yrs) 130-140 Mean: 19.2

months (7-30

months)

NR edema, hemorrhage,

dysarthria, hemiparesis, lip

and finger numbness

16.7%

Kondziolka [5] (2008)

Gamma Knife thalamotomy for

essential tremor.

J Neurosurg 2008, 108:111-117.

31 (52-92 yrs) 130-140 Median: 36

months (4-96

months)

92% hemiparesis, dysphagia,

dysarthria

7.7%

Duma (1999)

Duma CM, Jacques D, Kopyov

OV: The treatment of

movement disorders using

Gamma Knife stereotactic

radiosurgery.

Neurosurg Clin N Am 1999,

10:379-389.

38 (60-84 yrs) 120-160 Median: 30

months (6-72

months)

90% dysarthria 2.6%

Young (1998)

Young RF, Shumway-Cook A,

Vermeulen SS, Grimm P, Blasko J,

Posewitz A, Burkhart WA, Goiney

RC: Gamma knife radiosurgery

as a lesioning technique in

movement disorder surgery.

J Neurosurg 1998, 89:183-193.

27 (73.3 ± 7.2

yrs**)

120-160 Mean: 22.3*

months (12-44*

months)

89% none 0%

Young (2000)

Young RF, Jacques S, Mark R,

Kopyov O, Copcutt B, Posewitz A,

Li F: Gamma knife thalamotomy

for treatment of tremor: long-

term results.

J Neurosurg 2000, 93(Suppl

3):128-135.

PD: 102 (71.3 ± 8

yrs) ET: 52 (73.8 ±

9.4 yrs) Other: 4

(64.3 ± 7 yrs)

120-160 <12-96 months PD: 88.3% ET: 92.1%

Other: 50%

balance disturbance,

paresthesias, weakness,

dysphasia

1.3%

Niranjan (1999)

Niranjan A, Jawahar A,

Kondziolka D, Lunsford LD: A

comparison of surgical

approaches for the

management of tremor:

radiofrequency thalamotomy,

gamma knife thalamotomy and

thalamic stimulation.

Stereotact Funct Neurosurg

1999, 72:178-184.

12 (38-78 yrs) 130-150 Median: 24

months (4-40

months)

100% dysarthria, weakness 8.3%

Niranjan (2000)

Niranjan A, Kondziolka D, Baser

S, Heyman R, Lunsford LD:

Functional outcomes after

gamma knife thalamotomy for

11 (38-92 yrs) 130-150 Median: 6 months

(2-11 months)

100% dysarthria, weakness 9.1%

Page 109: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

109

essential tremor and MS-

related tremor.

Neurology 2000, 55:443-446.

Mathieu (2007)

Mathieu D, Kondziolka D,

Niranjan A, Flickinger J, Lunsford

LD: Gamma knife thalamotomy

for multiple sclerosis tremor.

Surg Neurol 2007, 68:394-399.

6 (31-57 yrs) 130-150 Median: 27.5

months (5-46

months)

100% hemiparesis 16.7%

Friedman (1999)

Friedman DP, Goldman HW,

Flanders AE, Gollomp SM, Curran

WJ Jr: Stereotactic radiosurgical

pallidotomy and thalamotomy

with the gamma knife: MR

imaging findings with clinical

correlation--preliminary

experience.

Radiology 1999, 212:143-150.

15 (37-84 yrs) 120-140 3 months 93.3% edema, incoordination,

action tremor

46.7%

Trigeminal neuralgia

Trigeminal neuralgia is a disorder of the fifth cranial (trigeminal) nerve that causes sudden bursts of

intense, stabbing, electric shock-like facial pain. The prevalence is four to five per 100,000 448.

The recurrent pain is located in the face in the areas supplied by the trigeminal nerve. These bursts

are often triggered by a light touch around the mouth or face or by talking, eating, or brushing one's

teeth. Usually pain occurs unilaterally. Often medication is effective in the treatment of trigeminal

neuralgia. Most frequently carbamazepine is prescribed, but carbamazepine can cause side effects

Page 110: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

110

including sleepiness, forgetfulness, confusion, drowsiness, dizziness and nausea. There are other

medications that can be used either alone or in combination to control trigeminal neuralgia pain.

They include Lioresal (baclofen), Dilantin (phenytoin), Klonopin (clonazepam), Neurontin

(gabapentin), or Lamictal (lamotrigine).

Intermittent local pressure from vascular loops on the trigeminal nerve can result in painful afferent

discharge from the trigeminal nerve. For patients who continue to experience severe pain despite

medication or who experience side effects from medication, an invasive surgical procedure is

recommended. The most frequently applied surgical procedures are: radiofrequency

electrocoagulation (RFE), glycerol injection (GLY), balloon microcompression (BMC), microvascular

decompression (MVD) and Gamma Knife radiosurgery (GKRS). Some of the procedures treat the

trigeminal nerve at the skull base (GLY, RFE, BMC), microvascular decompression and Gamma Knife

radiosurgery treat the Trigeminal neuralgia at the origin of pain, close to the brain stem where the

nerve is irritated by closely associated vessels. All of the invasive methods can be effective for

periods of time, but frequently a treatment has to repeated or combined with an alternative

approach.

The neurosurgical microvascular decompression (MVD) is often effective and is generally

recommended when patients are in a clinical condition that allows an invasive neurosurgical

intervention. Microvascular decompression separates the trigeminal nerve from the adjacent

pulsating vessel thereby removing the intermittent mechanical irritation of the nerve close to the

brain stem.

Gamma Knife radiosurgery is the least invasive neurosurgical treatment for trigeminal neuralgia and

achieves similar clinical results. Through highly focal irradiation of the trigeminal nerve close to the

brain stem the facial pain can be treated without impairment of other neural functions. Most

published series show a similar outcome with significant pain relief between 73 and 86 % of patients

after the Gamma Knife intervention (see references below).

The statistics from the Leksell Gamma Knife Society demonstrate that in North America 33,194 cases

with trigeminal neuralgia have been treated with Gamma Knife radiosurgery between 1991 and

2014.

Results after Gamma Knife treatment of Trigeminal Neuralgia in larger series (1968 patients)

Page 111: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

111

Study year target number of

patients

maximum

radiation dose

pain free

at 1 year

significantly

improved at 1

year

significantly

improved at 5

year

recurrence of

pain

New

sensory

deficit

Maesawa S, Salame C,

Flickinger JC, Pirris S, Kondziolka

D, Lunsford D. J Neurosurg

2001;94: 14–20.

2001 DREZ 220 60-90 Gy 70,3% 85,8% 55,8% 13,6% 10,2%

Pollock BE, Phuong LK, Foote

RL, Stafford SL, Gorman

DA.Neurosurgery. 2001

Jul;49(1):58-62; discussion 62-4

2001 DREZ? 27 70 Gy 41%

(at last fu) 24,1%

8%

(bothersome)

Pollock BE, Phuong LK, Foote

RL, Stafford SL, Gorman

DA.Neurosurgery. 2001

Jul;49(1):58-62; discussion 62-4

2001 DREZ? 61 90 Gy 61%

(at last fu)

24.1%

(in entire series)

32%

(bothersome)

Massager N, Lorenzoni J,

Devriendt D, Desmedt F,

Brotchi J, Levivier M.. J

Neurosurg 2004;100:597–605.

2004 retro-

gasserian 47 90Gy

83%

(good and excellent

initially)

71%

(good or excellent at

42 mo)

8%

34%

(mild

numbness)

Sheehan J, Pan HC, Stroila M,

Steiner L.. J Neurosurg

2005;102:434–41.

2005 DREZ 151 50-90 Gy 47% 90%

(some improvement)

70%

(Cave 3-y FU) 27% 9%

Urgosik D, Liscak R, Novotny J,

Vymazal J, Vladyka V. J

Neurosurg 2005;102:29–33.

2005 DREZ 107 70-80Gy 25% 20%

Balamucki CJ, Stieber VW,

CBranch et al. J Neurosurg

2006;105:730–35.

2006 DREZ

239

(out of 326

ttreated )

80-90 Gy 56% 80%

Longhi M, Rizzo P, Nicolato A,

Foroni R, Reggio M, Gerosa M.

Neurosurgery 2007;61:1254–

61.

2007 DREZ 160 75-95 Gy 61% 90% 9.5%

Little A, Shetter AG, Shetter ME,

Bay C, Rogers CL. Neurosurgery

2008;63:915–24.

2008 DREZ 185 70-90 Gy

(check)

67%

(at 2 years)

63%

( 7-y FU)

5%

(very

bothersome)

Regis J, Arkha Y, Yomo S,

Murata N, Roussel P, Donnet A,

Peragut JC. Neurochirurgie.

2009 Apr;55(2):213-22.

2009 retro-

gasserian 334 80-90 Gy

89%

(initial pain relief)

58%

(painfree) 17%

Clinical Outcome of Gamma Knife Radiosurgery for Trigeminal Neuralgia

Kondziolka had shown in an early study of 220 patients with idiopathic Trigeminal Neuralgia who

were treated with Gamma Knife radiosurgery (median maximum dose 80 Gy; range 60 -90 Gy) that

Page 112: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

112

complete pain relief was achieved in 64.9 % at 6 months, in 70.3% at 1 year, and in 75.4% at 33

months while new or increased facial paresthesia or facial numbness occurred in 10%. A total of

61.4% had prior surgery. After 5 years 55.8% of patients had complete or partial pain relief 449.

Flickinger had shown in a prospective double-blind, randomized study of of patients with trigeminal

neuralgia who were treated buy Gamma Knife radiosurgery, that increasing the treatment volume

by irradiating a longer nerve length does not significantly improve pain relief but may increase

complications 450 . Arai, Kondziolka and colleagues studied whether radiation dose rate affect clinical

outcomes in patients who undergo stereotactic Gamma Knife surgery for typical trigeminal

neuralgia (165 patients; maximum dose 80Gy; 4-mm collimator). The authors divided patients into

a low-dose-rate group (1.21 - 2.05 Gy/minute), and a high-dose-rate group (2.06 to 3.74 Gy/minute).

Initial pain relief was obtained in 71% of patients in the low-dose-rate group group and 78% in the

high-dose-rate group (p = 0.547). Hence the authors concluded that the GKS dose rate did not

affect duration of pain control or morbidity (facial sensory dysfunction) within the range of 1.21-

3.74 Gy/minute 451. The use of carbamazepine or gabapentin at the time of radiosurgery does not

decrease the rates of obtaining partial or complete pain relief after radiosurgery, but gabapentin

may reduce the risks of developing post-radiosurgery trigeminal neuropathy 452.

Between July 1992 and November 2010, 497 patients presenting with trigeminal neuralgia

underwent a single Gamma Knife treatment and prospective evaluation with a follow-up longer

than 1 year at Timone University Hospital in Marseille, France. Megadolichobasilar artery- or

multiple sclerosis-related trigeminal neuralgia were excluded. A single 4-mm isocenter was

positioned in the cisternal portion of the trigeminal nerve at a median distance of 7.8 mm (range

4.5-14 mm) anterior to the emergence of the nerve. A median maximum dose of 85 Gy (range 70-

90 Gy) was delivered. Median follow-up period was 43.75 months (range 12-174.41 months). A total

of 91.34% were initially pain free within a median time of 10 days (range 1-459 days) after GKS. A

substantial number of patients (37.2%) became pain free within the first 48 hours and 42.8%

between posttreatment Day 3 and Day 30, and 20% after 30 days post-GKS. Out of the patients

who initially became free from pain, 34.4% experienced a recurrence of pain with a median delay

of 24 months (range 0.62-150.06 months) 453.

In 2014 the actuarial rate of freedom from pain recurrence (BNI scores I-III) of 149 cases with

Page 113: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

113

therapy refractory trigeminal neuralgia treated with Gamma Knife stereotactic radiosurgery at the

William Beaumont Hospital, Royal Oak were 76%, 69% and 60%, at 1, 2 and 3-years respectively.

Toxicity after first GK SRS was minimal with 25% of cases experiencing only new or worsening post-

treatment numbness 454 .

Between 1999 and 2008, 446 of 777 patients with trigeminal neuralgia underwent Gamma Knife

radiosurgery at the Wake Forest Baptist Medical Center, Winston-Salem, North Carolina and had

evaluable follow-up (median follow-up 21.2 months). Rates of freedom from BNI 4-5 failure at 1,

3, and 5 years were 84.5%, 70.4%, and 46.9%, respectively. While 42% of patients developed

trigeminal dysfunction, multivariate analysis revealed that post-SRS numbness and improved post-

SRS BNI score at 6 months were predictive of a durable pain response 455.

In 2013 Young and colleagues followed up on 250 consecutive patients treated with the Leksell

Gamma Knife unit using a 4-mm isocenter. The isocenter had been placed on the trigeminal nerve

with the 20% isodose line tangential to the pontine surface (18 Gy). A total of 71.4% were pain free

(Classes I and II) and 89.5% had at least 50% pain relief: 43.7% were pain free without medication;

27.7% were pain free with medication, there was more than 90% decrease in pain intensity in 9.7%,

and decrease in pain intensity 8.4%, and 50%-90% decrease in pain intensity in 8.4%, less than 50%

decrease in pain intensity in 4.6%, and pain became worse in 5.9%.

Increase in numbness intensity after GKS was associated with a decrease in pain intensity and pain

length (p < 0.05) 456.

In 2013 Elaimy, Lee and colleagues presented the outcome of 108 patients treated at Gamma Knife

of Spokane for typical trigeminal neuralgia in a median clinical follow-up time of 15 months.

Following the first GKRS procedure, 71% of patients were grouped into BNI class I-IIIb (I = 31%; II

= 3%; IIIa = 19%; IIIb = 18%) and the median duration of pain relief for those patients was

determined to be 11.8 months. New facial numbness was reported in 19% of patients and new

facial paresthesias were reported in 7% of patients after the first GKRS procedure. Following a

second GKRS procedure (n=19) , 73% of patients were grouped into BNI class I-IIIb (I = 44%; II = 6%;

IIIa = 17%, IIIb = 6%) and the median duration of pain relief for those patients was determined to be

4.9 months. For repeat procedures, new facial numbness was reported in 22% of patients and new

facial paresthesias were reported in 6% of patients 457.

Page 114: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

114

In the post radiosurgical follow-up, tractography was more sensitive than conventional gadolinium-

enhanced post-treatment MR to detect changes in the trigeminal nerve. Diffusion tensor imaging

(DTI) provides information on white matter microstructure. DTI parameters accurately detect the

effects of focal radiosurgery on the trigeminal nerve, serving as an in vivo imaging tool to study

trigeminal neuralgia. Radial diffusivities but not axial diffusivities changed markedly, suggesting that

radiosurgery primarily affects myelin. 458.

In 2012 Li reported on 129 patients with idiopathic trigeminal neuralgia who had undergone

Gamma-knife radiosurgery at the West China Hospital of Sichuan University (maximum dose 80-90

Gy; mean follow-up 29.6 months) that 84.5% had complete or partial pain relief and a recurrence

after a mean time of 12.7 months in 8.3% of the initially successfully treated patients A mild-to-

moderate facial numbness occurred in 38% 459.

In the same year Loescher, Rowe and associates reported the outcome of 72 patients treated with

Gamma-knife radiosurgery (maximal dose 80 Gy) at the National Centre for Stereotactic

Radiosurgery in Sheffield, UK. The percentage of patients who were pain free without medication

was 39% after 6 months, 36% after 12 months and 64% after 24 months. The percentage of

patients who reported being very satisfied with treatment was 71% after 6 months, 57% after 12

months and 53% after 24 months. A new trigeminal sensory deficit was reported by 31% of patients

after radiosurgical treatment 460 .

Hayashi, Takakura and colleagues from Tokyo Women's Medical University applied a maximum dose

of 90Gy in 130 patients with trigeminal neuralgia, who were treated using Leksell Gamma Knife and

reported a marked initial relief of the typical paroxysmal facial pain in 98% (n=127) within a median

interval of 3 weeks after treatment. Since the pain re-appeared later in 23 patients, at the time of

the last follow-up 86% 86% were pain-free (n=112). In 24% facial hypesthesia and/or paresthesia

occurred 461 .

In 2010 Verheul, Beute and colleagues had reviewed 365 patents with medically refractory

trigeminal neuralgia, who were treated at the Gamma Knife Center Tilburg, Netherlands. In all

patients 80 Gy was prescribed, with a single 4-mm isocenter located at the root entry zone. In the

idiopathic trigeminal neuralgia group, rates of adequate pain relief were 75%, 60%, and 58% at 1,

3, and 5 years, respectively. Somewhat bothersome numbness was reported by 6% of patients after

Page 115: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

115

the first treatment and by 24% after repeated GKS. Very bothersome numbness was reported in

0.5% after the first GKS and in 2% after the second treatment 462.

In the same year Kano, Lunsford and colleagues had retrospectively reviewed outcomes after

Gamma Knife radiosurgery for recurrent trigeminal neuralgia after failed surgery in 193 patients.

After Gamma Knife, 85% of patients achieved pain relief or improvement. Pain recurrence was

observed in 73 of 168 patients 6 to 144 months after GKSR (median, 6 years). Factors associated

with better long-term pain relief included no relief from the surgical procedure preceding GKSR,

pain in a single branch, typical TN, and a single previous failed surgical procedure. Eighteen patients

(9.3%) developed new or increased trigeminal sensory dysfunction, and 1 developed

deafferentation pain. Patients who developed sensory loss after GKSR had better long-term pain

control. The best candidates for Gamma Knife were patients with recurrence after a single failed

previous operation and those with typical TN in a single trigeminal nerve distribution 463.

A series of 53 patients with trigeminal neuralgia who underwent a single Gamma Knife treatment

were followed by a minimum follow-up of 36 months at Tufts Medical Center (maximum dose 80

Gy, a single 4-mm isocenter). Complete pain relief without medications was seen in 32.1% of

patients, ; complete pain relief with either a decrease or no change in medications in 1.9%; ≥ 50%

pain relief without medications in 3.8%; ≥ 50% pain relief with either a decrease or no change in

medications in 20.8% and < 50% pain relief and/or increase in medications in 41.5% 464.

In 2009 Regis, Peragut and colleagues reviewed the outcome of 262 patients with minimum of 1

year of follow-up after Gamma Knife treatment for trigeminal neuralgia targeting the

retrogasserian cisternal portion of the Vth cranial nerve (median maximal dose 85Gy; range: 70-90).

At 5 years, 58% of the patients remain pain-free and 83% had no trigeminal nerve disturbance.

The median delay for pain cessation was 15 days. The initial pain-relief rate was 89% 465.

In a study from Université de Sherbrooke, Quebec, Canada, Knafo, Mathieu et al. showed

ignificant pain relief in 77.6% of patients after Gamma knife (n=67), including 32.6% who became

pain-free with sensory side effects in 14.9% 466.

Page 116: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

116

Little, Rogers and colleagues studied the long-term outcome of patients treated with gamma knife

radiosurgery for typical trigeminal neuralgia in 136 patients at the Barrow Neurological Institute in

Phoenix, Arizona with clinical follow-up data of more than 4 years (maximum dose 80 Gy targeted

at the root entry zone). Actuarial analysis demonstrated that 32% of patients were pain-free off

medication and 63% had at least a good outcome at 7 years. When GKRS was used as the primary

treatment, 45% of the patients were pain-free at 7 years. Treatment failed in 33% of the cohort

within 2 years, but only an additional 1% relapsed after 4 years. Posttreatment facial numbness was

reported as very bothersome in 5% 467.

In 160 patients with trigeminal neuralgia refractory to medical therapy Longhi, Gerosa and

colleagues reported a Grade I outcome in 61% and a Grade II outcome in 29%. In 10% Gamma Knife

had no effect 468.

In 2005 Sheehan, Steiner and associates had reported 151 cases of trigeminal neuralgia who had

been treated with Gamma knife (maximal radiation doses ranged from 50 to 90 Gy; median follow

up 19 months; range 2-96 months). The mean time to relief of pain was 24 days (range 1-180 days).

At the 1-, 2-, and 3-year follow-up, 47%, 45%, and 34% of patients, respectively, were pain free

without medication and 90%, 77%, and 70% of patients experienced some improvement in pain at

the 1-, 2-, and 3-year follow ups, respectively, while 27% of patients with initial improvement

subsequently experienced pain recurrence a median of 12 months (range 2-34 months) post-

Gamma Knife. New facial numbness was seen in 9% post-GKS. At the last follow up, GKS effected

pain relief in 44% of patients. Some degree of pain improvement at 3 years post-GKS was noted in

70% of patients with TN 469.

Trigeminal Neuralgia Related to Multiple Sclerosis

Among patients with multiple sclerosis there is a high incidence of trigeminal neuralgia, and

outcomes after treatment seem inferior to those in patients suffering from idiopathic trigeminal

neuralgia.

Patients with Trigeminal neuralgia related to multiple sclerosis with more than 1 year of follow-up

were operated with Gamma Knife radiosurgery and prospectively evaluated in the Timone

Page 117: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

117

University Hospital, Marseille (n=43). A single 4-mm isocenter was positioned at a median distance

of 8 mm anterior to the emergence of the nerve. A median maximum dose of 85 Gy (range 75-90)

was delivered. The median follow-up period was 53.8 months (12-157.1). While 90.7% were initially

pain free, the actuarial probability of remaining pain free without medication at 6 months, 1, 3

and 5 years was 87.2, 71.8, 43.1 and 38.3%, respectively. The hypoesthesia actuarial rate at 6

months, 1 and 2 years was 11.5, 11.5 and 16%, and remained stable till 12 years 470. In the multiple

sclerosis-related trigeminal neuralgia group from Tilburg, Netherlands, the rates of adequate pain

relief were reported to be 56%, 30%, and 20% at 1, 3, and 5 years after Gamma Knife treatment,

respectively. 462.

In 2012 Mathieu, Séguin and colleagues retrospectively compared the clinical outcomes in patients

with MS-related trigeminal neuralgia after Gamma Knife surgery with those obtained using

percutaneous retrogasserian glycerol rhizotomy in 45 patients with multiple sclerosis -related TN.

Reasonable pain control (BNI Pain Scale Scores I-IIIb) was noted in 81.5% who underwent GKS and

in 100% who underwent PRGR. For patients who underwent GKS, the median time to pain relief was

6 months; for those who underwent PRGR, pain relief was immediate. As of the last follow-up after

a median of 39 months, complete or reasonable pain control was finally achieved in 85.2% in the

GKS group and in 88.9% in the percutaneous retrogasserian glycerol rhizotomy group. The

morbidity rate was 22.2% in the GKS group (all due to sensory loss and paresthesia) and 66.7% in

the percutaneous retrogasserian glycerol rhizotomy group (mostly hypalgesia, with 2 patients

having corneal reflex loss and 1 patient suffering from meningitis). It can be concluded from this

study that Gamma Knife surgery has a lower rate of sensory and overall morbidity than

percutaneous retrogasserian glycerol rhizotomy 471.

Treatment Doses (Trigeminal Neuralgia)

In Gamma Knife radiosurgery of trigeminal neuralgia typically maximum doses of 80-85 Gy are

applied in a single isocenter using a 4-mm collimator.

Initially Kondziolka had shown in 1996 that a maximum radiosurgical dose of > 70 Gy was associated

with a significantly greater chance of complete pain relief (72% vs. 9%, p = 0.0003) 472 . Pollock had

compared a group of patients treated at maximum doses of 90Gy vs 70Gy. At last follow-up

Page 118: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

118

examination, 41% of the low-dose group remained pain-free compared with 61% high-dose cohort

(P = 0.17). High-dose radiosurgery was associated with an increased rate of permanent trigeminal

nerve dysfunction (54% versus 15%, P = 0.003) and among high-dose patients 8% developed corneal

numbness and 32% bothersome dysesthesias, whereas only 1 low-dose patient had this

complication (P = 0.01). The authors concluded that the incidence of significant trigeminal nerve

dysfunction is markedly increased after radiosurgery for patients receiving maximum doses of

90Gy 473 .

Flickinger had shown in a prospective double-blind, randomized study that increasing the treatment

volume to include a longer nerve length for trigeminal neuralgia radiosurgery does not significantly

improve pain relief but may increase complications 450 . The Gamma Knife dose rate does not affect

duration of pain control or morbidity 451. Similarly Balamucki, Branch and colleagues had previously

exmined whether the decrease in dose rate over 4.6 years between Co source replacements

affected the control rates of facial pain in patients undergoing Gamma Knife treatment (239

procedures) showing that dose rate was not significantly associated with either the control rate

or degree of pain relief 474.

Target Location (Trigeminal Neuralgia)

The typical target point the in Gamma Knife radiosurgery of trigeminal neuralgia is the trigeminal

nerve either at its dorsal root entry zone or slightly further anterior in the nerve’s retrogasserian

portion. While initially the target point in the dorsal root entry zone had been introduced, Regis

proposed to use an anterior ’retrogasserian’ target in order to avoid the rather constant occurence

of paraesthesia or facial numbness that was associeted with the more posterior target point.

Regis demonstrated in 83 patients who had been treated with gamma knife surgery to the

retrogasserian cisternal portion of the fifth cranial nerve at a typical maximum dose of 85 Gy (range

70-90 Gy) that 70% (58/83) had stopped taking medication during the study and that all quality-of-

life parameters were improved (p < 0.001). With this modified target point only 7.2% (6/83)

reported facial generally mild paresthesia, and 4.8% reported mild hypesthesia 475.

In 2008 Matsuda, Ono and colleagues compared results using the two Gamma Knife target points,

the dorsal root entry zone and the retrogasserian portion, in a series of 100 patients with intractable

Page 119: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

119

trigeminal neuralgia by applying 80 Gy at the proximal trigeminal nerve (posterior group) or 90 Gy

at the retrogasserian portion (anterior group). The rate of initial complete remission was higher in

the posterior group than in the anterior group (p = 0.003). Against expectations, more

complications were observed in the anterior group than in the posterior group (p = 0.009). The

authors found that the posterior targeting group had better pain control and a lower complication

rate 476.

A subgroup of 65 patients was studied for whom 6-month follow-up MRI showed focal contrast

enhancement of the trigeminal nerve showing that the median deviation of the coordinates

between the intended target and the center of contrast enhancement was 0.91 mm in Euclidean

space. The radiation doses fitting within the borders of the contrast enhancement of the trigeminal

nerve root ranged from 49 to 85 Gy (median value, 77 +/- 8.7 Gy).The median deviation found in

clinical assessment of gamma knife treatment for trigeminal neuralgia is low and compatible with

its high rate of efficiency 477.

Retreatment with Gamma Knife (Trigeminal Neuralgia)

In patients with recurrent pain after radiosurgery, the treatment can be repeated. Repeat Gamma

Knife radiosurgery was performed in 119 patients with recurrent trigeminal neuralgia. The median

interval between procedures was 26 months. The median target dose for repeat Gamma Knife was

70 Gy (range, 50-90 Gy) and the median cumulative dose was 145 Gy (range, 120-170 Gy). The

median follow-up was 48 months (range, 6-187 months) after repeat GKSR. After repeat Gamma

Knife, 87% of patients achieved initial pain relief (Barrow Neurological Institute pain score I-IIIb).

Pain relief was maintained at 1 year in 87.8%, at 3 years in 69.8% and at 5 years in 44.2%,

respectively. Facial sensory dysfunction occurred in 21% of patients within 18 months after GKSR.

A cumulative edge of brainstem dose ≥ 44 Gy was more likely to be associated with the

development of sensory loss. The best responses were observed in patients who had good pain

control after the first procedure and those who developed new sensory dysfunction in the affected

trigeminal distribution 478. The rate of pain relief of 27 patients who underwent repeat GK SRS at

the William Beaumont Hospital, Royal Oak was 70% and 62% at 1 and 2 years, respectively 454.

In patients unsuitable for other microsurgical or percutaneous strategies, especially those receiving

Page 120: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

120

long-term oral anticoagulation or antiplatelet agents, Gamma Knife radiosurgery repeated for a

third time may be a satisfactory, low risk option.

In 2015 a retrospective review of all patients who underwent 3 Gamma Knife radiosurgery

procedures for trigeminal neuralgia at 4 participating centers of the North American Gamma Knife

Consortium from 1995 to 2012 was performed and 17 patients were identified; 47.1% reported

initial complete pain relief following their third Gamma Knife radiosurgery and 47.1% experienced

at least partial relief. The average time to initial response was 2.9 months following the third

Gamma Knife radiosurgery. Although 17.6% developed new facial sensory dysfunction following

primary Gamma Knife radiosurgery and 2 patients 11.8% experienced new or worsening sensory

disturbance following the second Gamma Knife radiosurgery, no patient sustained additional

sensory disturbances after the third procedure. At a mean interval of 19.1 months following the

third Gamma Knife treatment, 23.5% suffered from recurrent trigeminal neuralgia 479.

Comparative effectiveness: Gamma Knife vs. Microvascular Decompression

It is virtually impossible to compare a highly invasive treatment such as microvascular

decompression with a non-invasive option such as Gamma Knife radiosurgery, since in general, the

non-invasive option is chosen in order to avoid the associated side-effects and complications

associated with the open neurosurgical procedure. Frequently Gamma Knife is applied in elderly

patients or in patients with relevant co-morbidities, when a highly invasive microvascular

decompression cannot be carried out at a low risk level.

In elderly patients Oh, Lim and associates compared the outcome after micro-vascular

decompression (MVD) with Gamma knife radiosurgery (GKRS). According to Pain Intensity Scale,

MVD group showed better prognosis with 63% (n=17) cases in grade I-II versus 55.6% (n=10) in GKRS

group after the treatment. The rate for recurrence of pain (11.1 %) was identical for both treatment

options. After the treatment, 2 cases of facial numbness, and 1 case each of herpes zoster,

cerebrospinal fluid (CSF) leakage, hearing disturbance, and subdural hematoma occurred in MVD

Group. In GKRS, there was 1 (5.6%) case of dysesthesia but was not permanent 480.

Page 121: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

121

Linskey compared his experience with microvascular decompression ([MVD], 36 patients) and

Gamma Knife surgery ([GKS], 44 patients) in 80 consecutive patients with trigeminal neuralgia.

Respective initial and latest follow-up raw pain-free rates were 100 and 80.6% for MVD and 77.3

and 45.5% for GKS. The median time to the maximal benefit after GKS was 4 weeks (range 1 week-

6 months). Respective initial, 2-, and 5-year actuarial pain-free rates were 100, 88, and 80% for

MVD and 78, 50, and 33% for GKS (p = 0.0002). The rates of permanent mild and severe sensory

loss were 5.6 and 0% for patients in the MVD group, as opposed to 6.8 and 2.3% for patients in the

GKS group. Anesthesia dolorosa did not occur during the study. Both procedures enjoyed a high

degree of early patient satisfaction (95-100%). Microvascular decompression maintained the same

rate of patient satisfaction, but satisfaction with GKS decreased to 75% as pain control waned. In

the MVD group there were complications of cerebrospinal fluid leakage, hearing loss, and persistent

diplopia (1 case each in the MVD group), which were not seen after Gamma Knife 481.

In cases of failed Gamma Knife radiosurgery for trigeminal neuralgia a trigeminal micro-vascular

decompression still remains a viable alternative if further surgery is required, as in surgery after

Gamma Knife no apparent changes in the trigeminal nerve itself or in any other tissue were found

that would have made surgery technically more difficult. 482.

It can be concluded that, despite the delayed and slightly lower degree of pain relief, for

trigeminal neuralgia patients with advanced age or in patients with co-morbidities precluding an

open invasive neurosurgical procedure, Gamma Knife radiosurgery can be preferable due to the

lower rate of surgical complications that arise owing to the old age 480.

Page 122: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

122

References

1. Seung SK, Larson DA, Galvin JM, et al. American College of Radiology (ACR) and

American Society for Radiation Oncology (ASTRO) Practice Guideline for the Performance of

Stereotactic Radiosurgery (SRS). Am J Clin Oncol 2013; 36(3): 310-5. doi:

10.1097/COC.0b013e31826e053d.

2. Lindquist C, Paddick I. The Leksell Gamma Knife Perfexion and comparisons with its

predecessors. Neurosurgery 2007; 61(3 Suppl): 130-40; discussion 40-1.

3. Verellen D, Linthout N, Bel A, et al. Assessment of the uncertainties in dose delivery

of a commercial system for linac-based stereotactic radiosurgery. Int J Radiat Oncol Biol Phys

1999; 44(2): 421-33.

4. Chang SD, Main W, Martin DP, Gibbs IC, Heilbrun MP. An analysis of the accuracy of

the CyberKnife: a robotic frameless stereotactic radiosurgical system. Neurosurgery 2003; 52(1):

140-6; discussion 6-7.

Page 123: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

123

5. Murphy MJ, Cox RS. The accuracy of dose localization for an image-guided frameless

radiosurgery system. Med Phys 1996; 23(12): 2043-9.

6. Murphy MJ. Intrafraction geometric uncertainties in frameless image-guided

radiosurgery. Int J Radiat Oncol Biol Phys 2009; 73(5): 1364-8.

7. Papagiannis P, Karaiskos P, Kozicki M, et al. Three-dimensional dose verification of

the clinical application of gamma knife stereotactic radiosurgery using polymer gel and MRI. Phys

Med Biol 2005; 50(9): 1979-90.

8. McDonald D, Schuler J, Takacs I, Peng J, Jenrette J, Vanek K. Comparison of radiation

dose spillage from the Gamma Knife Perfexion with that from volumetric modulated arc

radiosurgery during treatment of multiple brain metastases in a single fraction. J Neurosurg 2014;

121(Suppl): 51-9. doi: 10.3171/2014.7.GKS141358.

9. Zytkovicz A, Daftari I, Phillips TL, Chuang CF, Verhey L, Petti PL. Peripheral dose in

ocular treatments with CyberKnife and Gamma Knife radiosurgery compared to proton

radiotherapy. Phys Med Biol 2007; 52(19): 5957-71.

10. Regis J, Tamura M, Guillot C, et al. Radiosurgery with the world's first fully robotized

Leksell Gamma Knife PerfeXion in clinical use: a 200-patient prospective, randomized, controlled

comparison with the Gamma Knife 4C. Neurosurgery 2009; 64(2): 346-55; discussion 55-6.

11. Kaul D, Badakhshi H, Gevaert T, et al. Dosimetric comparison of different treatment

modalities for stereotactic radiosurgery of meningioma. Acta Neurochir (Wien) 2015; 157(4): 559-

63; discussion 63-4. doi: 10.1007/s00701-014-2272-9. Epub 2014 Nov 21.

12. Ma L, Nichol A, Hossain S, et al. Variable dose interplay effects across radiosurgical

apparatus in treating multiple brain metastases. Int J Comput Assist Radiol Surg 2014; 20: 20.

13. Abacioglu U, Ozen Z, Yilmaz M, et al. Critical appraisal of RapidArc radiosurgery with

flattening filter free photon beams for benign brain lesions in comparison to GammaKnife: a

treatment planning study. Radiat Oncol 2014; 9:119.(doi): 10.1186/748-717X-9-119.

14. Chuang CF, Larson DA, Zytkovicz A, Smith V, Petti PL. Peripheral dose measurement

for CyberKnife radiosurgery with upgraded linac shielding. Med Phys 2008; 35(4): 1494-6.

15. Suh JH, Stea B, Nabid A, et al. Phase III study of efaproxiral as an adjunct to whole-

brain radiation therapy for brain metastases. J Clin Oncol 2006; 24(1): 106-14.

16. Heisterkamp C, Haatanen T, Schild SE, Rades D. Dose escalation in patients receiving

whole-brain radiotherapy for brain metastases from colorectal cancer. Strahlenther Onkol 2010;

186(2): 70-5.

Page 124: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

124

17. Robinet G, Thomas P, Breton JL, et al. Results of a phase III study of early versus

delayed whole brain radiotherapy with concurrent cisplatin and vinorelbine combination in

inoperable brain metastasis of non-small-cell lung cancer: Groupe Francais de Pneumo-

Cancerologie (GFPC) Protocol 95-1. Ann Oncol 2001; 12(1): 59-67.

18. Rades D, Heisterkamp C, Huttenlocher S, et al. Dose escalation of whole-brain

radiotherapy for brain metastases from melanoma. Int J Radiat Oncol Biol Phys 2010; 77(2): 537-

41.

19. Meyners T, Heisterkamp C, Kueter JD, et al. Prognostic factors for outcomes after

whole-brain irradiation of brain metastases from relatively radioresistant tumors: a retrospective

analysis. BMC Cancer 2010; 10: 582.

20. Ogura M, Mitsumori M, Okumura S, et al. Radiation therapy for brain metastases

from breast cancer. Breast Cancer 2003; 10(4): 349-55.

21. Bartsch R, Fromm S, Rudas M, et al. Intensified local treatment and systemic therapy

significantly increase survival in patients with brain metastases from advanced breast cancer - a

retrospective analysis. Radiother Oncol 2006; 80(3): 313-7.

22. Nieder C, Thamm R, Astner ST, Geinitz H, Molls M. Is whole-brain radiotherapy

effective and safe in elderly patients with brain metastases? Oncology 2007; 72(5-6): 326-9.

23. Nieder C, Andratschke N, Grosu AL, Molls M. Recursive partitioning analysis (RPA)

class does not predict survival in patients with four or more brain metastases. Strahlenther Onkol

2003; 179(1): 16-20.

24. Nieder C, Norum J, Dalhaug A, Aandahl G, Pawinski A. Radiotherapy versus best

supportive care in patients with brain metastases and adverse prognostic factors. Clin Exp

Metastasis 2013.

25. Priestman TJ, Dunn J, Brada M, Rampling R, Baker PG. Final results of the Royal

College of Radiologists' trial comparing two different radiotherapy schedules in the treatment of

cerebral metastases. Clin Oncol (R Coll Radiol) 1996; 8(5): 308-15.

26. Lagerwaard FJ, Levendag PC, Nowak PJ, Eijkenboom WM, Hanssens PE, Schmitz PI.

Identification of prognostic factors in patients with brain metastases: a review of 1292 patients. Int

J Radiat Oncol Biol Phys 1999; 43(4): 795-803.

27. Langley RE, Stephens RJ, Nankivell M, et al. Interim data from the Medical Research

Council QUARTZ Trial: does whole brain radiotherapy affect the survival and quality of life of

Page 125: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

125

patients with brain metastases from non-small cell lung cancer? Clin Oncol (R Coll Radiol) 2013;

25(3): e23-30.

28. Broadbent AM, Hruby G, Tin MM, Jackson M, Firth I. Survival following whole brain

radiation treatment for cerebral metastases: an audit of 474 patients. Radiother Oncol 2004;

71(3): 259-65.

29. Karam I, Nichol A, Woods R, Tyldesley S. Population-based outcomes after whole

brain radiotherapy and re-irradiation in patients with metastatic breast cancer in the trastuzumab

era. Radiat Oncol 2011; 6:181.(doi): 10.1186/748-717X-6-181.

30. Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant whole-brain radiotherapy versus

observation after radiosurgery or surgical resection of one to three cerebral metastases: results of

the EORTC 22952-26001 study. J Clin Oncol 2011; 29(2): 134-41.

31. Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus whole-brain

radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a

randomized controlled trial. JAMA 2006; 295(21): 2483-91.

32. Rades D, Pluemer A, Veninga T, Hanssens P, Dunst J, Schild SE. Whole-brain

radiotherapy versus stereotactic radiosurgery for patients in recursive partitioning analysis classes

1 and 2 with 1 to 3 brain metastases. Cancer 2007; 110(10): 2285-92.

33. Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain metastases

treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled

trial. Lancet Oncol 2009; 10(11): 1037-44.

34. Soffietti R, Kocher M, Abacioglu UM, et al. A European Organisation for Research and

Treatment of Cancer phase III trial of adjuvant whole-brain radiotherapy versus observation in

patients with one to three brain metastases from solid tumors after surgical resection or

radiosurgery: quality-of-life results. J Clin Oncol 2013; 31(1): 65-72.

35. Welzel G, Fleckenstein K, Schaefer J, et al. Memory function before and after whole

brain radiotherapy in patients with and without brain metastases. Int J Radiat Oncol Biol Phys

2008; 72(5): 1311-8.

36. Slotman BJ, Mauer ME, Bottomley A, et al. Prophylactic cranial irradiation in

extensive disease small-cell lung cancer: short-term health-related quality of life and patient

reported symptoms: results of an international Phase III randomized controlled trial by the EORTC

Radiation Oncology and Lung Cancer Groups. J Clin Oncol 2009; 27(1): 78-84.

Page 126: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

126

37. Doyle DM, Einhorn LH. Delayed effects of whole brain radiotherapy in germ cell

tumor patients with central nervous system metastases. Int J Radiat Oncol Biol Phys 2008; 70(5):

1361-4.

38. DiBiase SJ, Chin LS, Ma L. Influence of gamma knife radiosurgery on the quality of life

in patients with brain metastases. Am J Clin Oncol 2002; 25(2): 131-4.

39. Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy with or

without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III

results of the RTOG 9508 randomised trial. Lancet 2004; 363(9422): 1665-72.

40. Sperduto PW, Kased N, Roberge D, et al. Summary report on the graded prognostic

assessment: an accurate and facile diagnosis-specific tool to estimate survival for patients with

brain metastases. J Clin Oncol 2012; 30(4): 419-25.

41. Serizawa T, Yamamoto M, Sato Y, et al. Gamma Knife surgery as sole treatment for

multiple brain metastases: 2-center retrospective review of 1508 cases meeting the inclusion

criteria of the JLGK0901 multi-institutional prospective study. J Neurosurg 2010; 113 Suppl: 48-52.

42. Caballero JA, Sneed PK, Lamborn KR, et al. Prognostic factors for survival in patients

treated with stereotactic radiosurgery for recurrent brain metastases after prior whole brain

radiotherapy. Int J Radiat Oncol Biol Phys 2012; 83(1): 303-9.

43. Kim HJ, Im SA, Keam B, et al. Clinical outcome of central nervous system metastases

from breast cancer: differences in survival depending on systemic treatment. J Neurooncol 2012;

106(2): 303-13. doi: 10.1007/s11060-011-0664-8. Epub 2011 Sep 22.

44. Muacevic A, Kreth FW, Horstmann GA, et al. Surgery and radiotherapy compared

with gamma knife radiosurgery in the treatment of solitary cerebral metastases of small diameter.

J Neurosurg 1999; 91(1): 35-43.

45. O'Neill BP, Iturria NJ, Link MJ, Pollock BE, Ballman KV, O'Fallon JR. A comparison of

surgical resection and stereotactic radiosurgery in the treatment of solitary brain metastases. Int J

Radiat Oncol Biol Phys 2003; 55(5): 1169-76.

46. Schoggl A, Kitz K, Reddy M, et al. Defining the role of stereotactic radiosurgery versus

microsurgery in the treatment of single brain metastases. Acta Neurochir (Wien) 2000; 142(6):

621-6.

47. Muacevic A, Wowra B, Siefert A, Tonn JC, Steiger HJ, Kreth FW. Microsurgery plus

whole brain irradiation versus Gamma Knife surgery alone for treatment of single metastases to

the brain: a randomized controlled multicentre phase III trial. J Neurooncol 2008; 87(3): 299-307.

Page 127: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

127

48. Linskey ME, Andrews DW, Asher AL, et al. The role of stereotactic radiosurgery in the

management of patients with newly diagnosed brain metastases: a systematic review and

evidence-based clinical practice guideline. J Neurooncol 2010; 96(1): 45-68.

49. Kim SH, Weil RJ, Chao ST, et al. Stereotactic radiosurgical treatment of brain

metastases in older patients. Cancer 2008; 113(4): 834-40. doi: 10.1002/cncr.23625.

50. Watanabe S, Yamamoto M, Sato Y, et al. Stereotactic radiosurgery for brain

metastases: a case-matched study comparing treatment results for patients 80 years of age or

older versus patients 65-79 years of age. J Neurosurg 2014; 121(5): 1148-57. doi:

10.3171/2014.6.JNS132790. Epub 2014 Jul 25.

51. Lee CC, Yen CP, Xu Z, Schlesinger D, Sheehan J. Large intracranial metastatic tumors

treated by Gamma Knife surgery: outcomes and prognostic factors. J Neurosurg 2014; 120(1): 52-

9. doi: 10.3171/2013.9.JNS131163. Epub 2013 Oct 25.

52. Yomo S, Hayashi M. A minimally invasive treatment option for large metastatic brain

tumors: long-term results of two-session Gamma Knife stereotactic radiosurgery. Radiat Oncol

2014; 9:132.(doi): 10.1186/748-717X-9-132.

53. Yomo S, Hayashi M, Nicholson C. A prospective pilot study of two-session Gamma

Knife surgery for large metastatic brain tumors. J Neurooncol 2012; 109(1): 159-65. doi:

10.1007/s11060-012-0882-8. Epub 2012 Apr 29.

54. Eaton BR, Gebhardt B, Prabhu R, Shu HK, Curran WJ, Jr., Crocker I. Hypofractionated

radiosurgery for intact or resected brain metastases: defining the optimal dose and fractionation.

Radiat Oncol 2013; 8:135.(doi): 10.1186/748-717X-8-135.

55. Wegner RE, Leeman JE, Kabolizadeh P, et al. Fractionated stereotactic radiosurgery

for large brain metastases. Am J Clin Oncol 2015; 38(2): 135-9. doi:

10.1097/COC.0b013e31828aadac.

56. Jagannathan J, Yen CP, Ray DK, et al. Gamma Knife radiosurgery to the surgical cavity

following resection of brain metastases. J Neurosurg 2009; 111(3): 431-8. doi:

10.3171/2008.11.JNS08818.

57. Iorio-Morin C, Masson-Cote L, Ezahr Y, Blanchard J, Ebacher A, Mathieu D. Early

Gamma Knife stereotactic radiosurgery to the tumor bed of resected brain metastasis for

improved local control. J Neurosurg 2014; 121(Suppl): 69-74. doi: 10.3171/2014.7.GKS141488.

Page 128: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

128

58. Ojerholm E, Lee JY, Thawani JP, et al. Stereotactic radiosurgery to the resection bed

for intracranial metastases and risk of leptomeningeal carcinomatosis. J Neurosurg 2014;

121(Suppl): 75-83. doi: 10.3171/2014.6.GKS14708.

59. Ogiwara H, Kalakota K, Rakhra SS, et al. Intracranial relapse rates and patterns, and

survival trends following post-resection cavity radiosurgery for patients with single intracranial

metastases. J Neurooncol 2012; 108(1): 141-6. doi: 10.1007/s11060-012-0808-5. Epub 2012 Mar

18.

60. Nakazaki K, Higuchi Y, Nagano O, Serizawa T. Efficacy and limitations of salvage

gamma knife radiosurgery for brain metastases of small-cell lung cancer after whole-brain

radiotherapy. Acta Neurochir (Wien) 2013; 155(1): 107-13; discussion 13-4. doi: 10.1007/s00701-

012-1520-0. Epub 2012 Oct 13.

61. Harris S, Chan MD, Lovato JF, et al. Gamma knife stereotactic radiosurgery as salvage

therapy after failure of whole-brain radiotherapy in patients with small-cell lung cancer. Int J

Radiat Oncol Biol Phys 2012; 83(1): e53-9. doi: 10.1016/j.ijrobp.2011.11.059. Epub 2 Feb 17.

62. Yomo S, Hayashi M. The efficacy and limitations of stereotactic radiosurgery as a

salvage treatment after failed whole brain radiotherapy for brain metastases. J Neurooncol 2013;

113(3): 459-65. doi: 10.1007/s11060-013-1138-y. Epub 2013 May 10.

63. Nakazaki K, Kano H. Evaluation of mini-mental status examination score after gamma

knife radiosurgery as the first radiation treatment for brain metastases. J Neurooncol 2013; 112(3):

421-5. doi: 10.1007/s11060-013-1071-0. Epub 2013 Feb 14.

64. Chao ST, Thakkar VV, Barnett GH, et al. Prospective study of the short-term adverse

effects of gamma knife radiosurgery. Technol Cancer Res Treat 2012; 11(2): 117-22.

65. Lee CC, Wintermark M, Xu Z, Yen CP, Schlesinger D, Sheehan JP. Application of

diffusion-weighted magnetic resonance imaging to predict the intracranial metastatic tumor

response to gamma knife radiosurgery. J Neurooncol 2014; 118(2): 351-61. doi: 10.1007/s11060-

014-1439-9. Epub 2014 Apr 24.

66. Yamamoto M, Kawabe T, Higuchi Y, et al. Delayed complications in patients surviving

at least 3 years after stereotactic radiosurgery for brain metastases. Int J Radiat Oncol Biol Phys

2013; 85(1): 53-60. doi: 10.1016/j.ijrobp.2012.04.018. Epub May 30.

67. Ebinu JO, Lwu S, Monsalves E, et al. Gamma knife radiosurgery for the treatment of

cystic cerebral metastases. Int J Radiat Oncol Biol Phys 2013; 85(3): 667-71. doi:

10.1016/j.ijrobp.2012.06.043. Epub Aug 9.

Page 129: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

129

68. Higuchi F, Kawamoto S, Abe Y, Kim P, Ueki K. Effectiveness of a 1-day aspiration plus

Gamma Knife surgery procedure for metastatic brain tumor with a cystic component. J Neurosurg

2012; 117(Suppl): 17-22. doi: 10.3171/2012.7.GKS121001.

69. Hayashi M, Chernov M, Tamura N, et al. "Donut's shape" radiosurgical treatment

planning for large cystic metastatic brain tumors. Minim Invasive Neurosurg 2011; 54(5-6): 286-9.

doi: 10.1055/s-0031-1297987. Epub 2012 Jan 25.

70. Lee SR, Oh JY, Kim SH. Gamma Knife radiosurgery for cystic brain metastases. Br J

Neurosurg 2015; 11: 1-6.

71. Deibert CP, Ahluwalia MS, Sheehan JP, et al. Bevacizumab for refractory adverse

radiation effects after stereotactic radiosurgery. J Neurooncol 2013; 115(2): 217-23. doi:

10.1007/s11060-013-1214-3. Epub 2013 Aug 9.

72. Boothe D, Young R, Yamada Y, Prager A, Chan T, Beal K. Bevacizumab as a treatment

for radiation necrosis of brain metastases post stereotactic radiosurgery. Neuro Oncol 2013; 15(9):

1257-63. doi: 10.093/neuonc/not085. Epub 2013 Jun 27.

73. Jo KI, Lim DH, Kim ST, et al. Leptomeningeal seeding in patients with brain

metastases treated by gamma knife radiosurgery. J Neurooncol 2012; 109(2): 293-9. doi:

10.1007/s11060-012-0892-6. Epub 2012 May 20.

74. Zairi F, Ouammou Y, Le Rhun E, et al. Relevance of gamma knife radiosurgery alone

for the treatment of non-small cell lung cancer brain metastases. Clin Neurol Neurosurg 2014;

125:87-93.(doi): 10.1016/j.clineuro.2014.07.030. Epub Jul 27.

75. Skeie BS, Skeie GO, Enger PO, et al. Gamma knife surgery in brain melanomas:

absence of extracranial metastases and tumor volume strongest indicators of prolonged survival.

World Neurosurg 2011; 75(5-6): 684-91.

76. Chen JC, Petrovich Z, O'Day S, et al. Stereotactic radiosurgery in the treatment of

metastatic disease to the brain. Neurosurgery 2000; 47(2): 268-79; discussion 79-81.

77. Jawahar A, Shaya M, Campbell P, et al. Role of stereotactic radiosurgery as a primary

treatment option in the management of newly diagnosed multiple (3-6) intracranial metastases.

Surg Neurol 2005; 64(3): 207-12.

78. Kim CH, Im YS, Nam DH, Park K, Kim JH, Lee JI. Gamma knife radiosurgery for ten or

more brain metastases. J Korean Neurosurg Soc 2008; 44(6): 358-63. doi:

10.3340/jkns.2008.44.6.358. Epub Dec 31.

Page 130: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

130

79. Rava P, Leonard K, Sioshansi S, et al. Survival among patients with 10 or more brain

metastases treated with stereotactic radiosurgery. J Neurosurg 2013; 119(2): 457-62. doi:

10.3171/2013.4.JNS121751. Epub 2013 May 10.

80. Salvetti DJ, Nagaraja TG, McNeill IT, Xu Z, Sheehan J. Gamma Knife surgery for the

treatment of 5 to 15 metastases to the brain: clinical article. J Neurosurg 2013; 118(6): 1250-7.

doi: 10.3171/2013.2.JNS121213. Epub 2013 Mar 29.

81. Grandhi R, Kondziolka D, Panczykowski D, et al. Stereotactic radiosurgery using the

Leksell Gamma Knife Perfexion unit in the management of patients with 10 or more brain

metastases. J Neurosurg 2012; 117(2): 237-45.

82. Bhatnagar AK, Kondziolka D, Lunsford LD, Flickinger JC. Recursive partitioning

analysis of prognostic factors for patients with four or more intracranial metastases treated with

radiosurgery. Technol Cancer Res Treat 2007; 6(3): 153-60.

83. Sperduto PW, Kased N, Roberge D, et al. The effect of tumor subtype on the time

from primary diagnosis to development of brain metastases and survival in patients with breast

cancer. J Neurooncol 2013; 112(3): 467-72.

84. Xu Z, Schlesinger D, Toulmin S, Rich T, Sheehan J. Impact of triple-negative

phenotype on prognosis of patients with breast cancer brain metastases. Int J Radiat Oncol Biol

Phys 2012; 84(3): 612-8.

85. Sperduto PW, Chao ST, Sneed PK, et al. Diagnosis-specific prognostic factors, indexes,

and treatment outcomes for patients with newly diagnosed brain metastases: a multi-institutional

analysis of 4,259 patients. Int J Radiat Oncol Biol Phys 2010; 77(3): 655-61.

86. Eichler AF, Kahle KT, Wang DL, et al. EGFR mutation status and survival after

diagnosis of brain metastasis in nonsmall cell lung cancer. Neuro Oncol 2010; 12(11): 1193-9.

87. Lippitz BE. Cytokine patterns in patients with cancer: a systematic review. Lancet

Oncol 2013; 14(6): e218-28.

88. Yamamoto M ST, Shuto T, Akabane A, Higuchi Y, Kawagishi J, Yamanaka K, Sato Y,

Jokura H, Yomo S, Nagano O, Kenai H, Moriki A, Suzuki S, Kida Y, Iwai Y, Hayashi M,, Onishi H GM,

Sato M, Akimitsu T, Kubo K, Kikuchi Y, Shibasaki T, Goto T, Takanashi M, Mori Y, Takakura K, Saeki

N, Kunieda E, Aoyama H, Momoshima S, Tsuchiya K. Stereotactic radiosurgery for patients with

multiple brain metastases (JLGK0901): a multi-institutional prospective observational study.

Lancet Oncol 2014 2014.

Page 131: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

131

89. Yamamoto M, Kawabe T, Sato Y, et al. A case-matched study of stereotactic

radiosurgery for patients with multiple brain metastases: comparing treatment results for 1-4 vs

>/= 5 tumors. J Neurosurg 2013.

90. Yamamoto M, Kawabe T, Sato Y, et al. Stereotactic radiosurgery for patients with

multiple brain metastases: a case-matched study comparing treatment results for patients with 2-

9 versus 10 or more tumors. J Neurosurg 2014; 121(Suppl): 16-25. doi:

10.3171/2014.8.GKS141421.

91. Yamamoto M, Kawabe T, Barfod BE. How many metastases can be treated with

radiosurgery? Prog Neurol Surg 2012; 25:261-72.(doi): 10.1159/000331199. Epub 2012 Jan 6.

92. Serizawa T, Saeki N, Higuchi Y, et al. Gamma knife surgery for brain metastases:

indications for and limitations of a local treatment protocol. Acta Neurochir (Wien) 2005; 147(7):

721-6; discussion 6.

93. Serizawa T, Hirai T, Nagano O, et al. Gamma knife surgery for 1-10 brain metastases

without prophylactic whole-brain radiation therapy: analysis of cases meeting the Japanese

prospective multi-institute study (JLGK0901) inclusion criteria. J Neurooncol 2010; 98(2): 163-7.

doi: 10.1007/s11060-010-0169-x. Epub 2010 Apr 22.

94. Yan ES, Sneed PK, McDermott MW, Kunwar S, Wara WM, Larson DA. Number of

Brain Metastases Is Not an Important Prognostic Factor for Survival Following Radiosurgery for

Newly-Diagnosed Nonmelanoma Brain Metastases. International Journal of Radiation, Oncology,

Biology,Physics 2003; 57(supplement 2): 131-2.

95. Shu HK, Sneed PK, Shiau CY, et al. Factors influencing survival after gamma knife

radiosurgery for patients with single and multiple brain metastases. Cancer J Sci Am 1996; 2(6):

335-42.

96. Petrovich Z, Yu C, Giannotta SL, O'Day S, Apuzzo ML. Survival and pattern of failure in

brain metastasis treated with stereotactic gamma knife radiosurgery. J Neurosurg 2002; 97(5

Suppl): 499-506.

97. Chidel MA, Suh JH, Reddy CA, Chao ST, Lundbeck MF, Barnett GH. Application of

recursive partitioning analysis and evaluation of the use of whole brain radiation among patients

treated with stereotactic radiosurgery for newly diagnosed brain metastases. Int J Radiat Oncol

Biol Phys 2000; 47(4): 993-9.

Page 132: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

132

98. Amendola BE, Wolf AL, Coy SR, Amendola M, Bloch L. Gamma knife radiosurgery in

the treatment of patients with single and multiple brain metastases from carcinoma of the breast.

Cancer J 2000; 6(2): 88-92.

99. Goyal LK, Suh JH, Reddy CA, Barnett GH. The role of whole brain radiotherapy and

stereotactic radiosurgery on brain metastases from renal cell carcinoma. Int J Radiat Oncol Biol

Phys 2000; 47(4): 1007-12.

100. Likhacheva A, Pinnix CC, Parikh NR, et al. Predictors of survival in contemporary

practice after initial radiosurgery for brain metastases. Int J Radiat Oncol Biol Phys 2013; 85(3):

656-61.

101. Bashir A, Hodge CJ, Jr., Dababneh H, Hussain M, Hahn S, Canute GW. Impact of the

number of metastatic brain lesions on survival after Gamma Knife radiosurgery. J Clin Neurosci

2014; 21(11): 1928-33. doi: 10.016/j.jocn.2014.03.026. Epub Jul 15.

102. Chang WS, Kim HY, Chang JW, Park YG, Chang JH. Analysis of radiosurgical results in

patients with brain metastases according to the number of brain lesions: is stereotactic

radiosurgery effective for multiple brain metastases? J Neurosurg 2010; 113 Suppl: 73-8.

103. Yamamoto M, Kawabe T, Sato Y, et al. A case-matched study of stereotactic

radiosurgery for patients with multiple brain metastases: comparing treatment results for 1-4 vs

>/= 5 tumors: clinical article. J Neurosurg 2013; 118(6): 1258-68. doi: 10.3171/2013.3.JNS121900.

Epub 2013 Apr 19.

104. Kondziolka D, Patel A, Lunsford LD, Kassam A, Flickinger JC. Stereotactic radiosurgery

plus whole brain radiotherapy versus radiotherapy alone for patients with multiple brain

metastases. Int J Radiat Oncol Biol Phys 1999; 45(2): 427-34.

105. Chougule PB, Burton-Williams M, Saris S, et al. Randomized treatment of brain

metastasis with gamma knife radiosurgery, whole brain radiotherapy or both. International

Journal of Radiation Oncology Biology and Physics 2000; 48(3, supplement): 114.

106. Baschnagel AM, Meyer KD, Chen PY, et al. Tumor volume as a predictor of survival

and local control in patients with brain metastases treated with Gamma Knife surgery. J Neurosurg

2013; 119(5): 1139-44. doi: 10.3171/2013.7.JNS13431. Epub 2013 Aug 23.

107. Choi SW, Kwon do H, Kim CJ. Clinical outcome in gamma knife radiosurgery for

metastatic brain tumors from the primary breast cancer : prognostic factors in local treatment

failure and survival. J Korean Neurosurg Soc 2013; 54(4): 329-35. doi: 10.3340/jkns.2013.54.4.329.

Epub Oct 31.

Page 133: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

133

108. Muacevic A, Kreth FW, Tonn JC, Wowra B. Stereotactic radiosurgery for multiple

brain metastases from breast carcinoma. Cancer 2004; 100(8): 1705-11.

109. Jo KI, Im YH, Kong DS, Seol HJ, Nam DH, Lee JI. Gamma knife radiosurgery for brain

metastases from breast cancer. J Korean Neurosurg Soc 2013; 54(5): 399-404.

110. Jaboin JJ, Ferraro DJ, DeWees TA, et al. Survival following gamma knife radiosurgery

for brain metastasis from breast cancer. Radiat Oncol 2013; 8:131.(doi): 10.1186/748-717X-8-131.

111. Matsunaga S, Shuto T, Kawahara N, Suenaga J, Inomori S, Fujino H. Gamma Knife

surgery for metastatic brain tumors from primary breast cancer: treatment indication based on

number of tumors and breast cancer phenotype. J Neurosurg 2010; 113 Suppl: 65-72.

112. Kondziolka D, Kano H, Harrison GL, et al. Stereotactic radiosurgery as primary and

salvage treatment for brain metastases from breast cancer. Clinical article. J Neurosurg 2011;

114(3): 792-800.

113. Kased N, Binder DK, McDermott MW, et al. Gamma Knife radiosurgery for brain

metastases from primary breast cancer. Int J Radiat Oncol Biol Phys 2009; 75(4): 1132-40.

114. Goyal S, Prasad D, Harrell F, Jr., Matsumoto J, Rich T, Steiner L. Gamma knife surgery

for the treatment of intracranial metastases from breast cancer. J Neurosurg 2005; 103(2): 218-23.

115. Sperduto PW, Kased N, Roberge D, et al. The effect of tumor subtype on the time

from primary diagnosis to development of brain metastases and survival in patients with breast

cancer. J Neurooncol 2013; 112(3): 467-72. doi: 10.1007/s11060-013-1083-9. Epub 2013 Mar 6.

116. Padovani L, Muracciole X, Regis J. gamma knife radiosurgery of brain metastasis from

breast cancer. Prog Neurol Surg 2012; 25:156-62.(doi): 10.1159/000331189. Epub 2012 Jan 6.

117. Sperduto PW, Kased N, Roberge D, et al. Effect of tumor subtype on survival and the

graded prognostic assessment for patients with breast cancer and brain metastases. Int J Radiat

Oncol Biol Phys 2012; 82(5): 2111-7. doi: 10.1016/j.ijrobp.2011.02.027. Epub Apr 15.

118. Vern-Gross TZ, Lawrence JA, Case LD, et al. Breast cancer subtype affects patterns of

failure of brain metastases after treatment with stereotactic radiosurgery. J Neurooncol 2012;

110(3): 381-8. doi: 10.1007/s11060-012-0976-3. Epub 2012 Sep 24.

119. Xu Z, Schlesinger D, Toulmin S, Rich T, Sheehan J. Impact of triple-negative

phenotype on prognosis of patients with breast cancer brain metastases. Int J Radiat Oncol Biol

Phys 2012; 84(3): 612-8. doi: 10.1016/j.ijrobp.2011.12.054. Epub 2 Mar 19.

Page 134: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

134

120. Yomo S, Hayashi M, Cho N. Impacts of HER2-overexpression and molecular targeting

therapy on the efficacy of stereotactic radiosurgery for brain metastases from breast cancer. J

Neurooncol 2013; 112(2): 199-207. doi: 10.1007/s11060-013-1046-1. Epub 2013 Jan 8.

121. Tam M, Narayana A, Raza S, et al. Role of HER2 status in the treatment for brain

metastases arising from breast cancer with stereotactic radiosurgery. Med Oncol 2014; 31(2): 832.

doi: 10.1007/s12032-013-0832-0. Epub 2014 Jan 5.

122. Chen AM, Jahan TM, Jablons DM, Garcia J, Larson DA. Risk of cerebral metastases

and neurological death after pathological complete response to neoadjuvant therapy for locally

advanced nonsmall-cell lung cancer: clinical implications for the subsequent management of the

brain. Cancer 2007; 109(8): 1668-75.

123. Garsa AA, Badiyan SN, DeWees T, et al. Predictors of individual tumor local control

after stereotactic radiosurgery for non-small cell lung cancer brain metastases. Int J Radiat Oncol

Biol Phys 2014; 90(2): 407-13. doi: 10.1016/j.ijrobp.2014.05.047. Epub Jul 30.

124. Gerosa M, Nicolato A, Foroni R, Tomazzoli L, Bricolo A. Analysis of long-term

outcomes and prognostic factors in patients with non-small cell lung cancer brain metastases

treated by gamma knife radiosurgery. J Neurosurg 2005; 102 Suppl: 75-80.

125. Kim YS, Kondziolka D, Flickinger JC, Lunsford LD. Stereotactic radiosurgery for

patients with nonsmall cell lung carcinoma metastatic to the brain. Cancer 1997; 80(11): 2075-83.

126. Pan HC, Sheehan J, Stroila M, Steiner M, Steiner L. Gamma knife surgery for brain

metastases from lung cancer. J Neurosurg 2005; 102 Suppl: 128-33.

127. Kuremsky JG, Urbanic JJ, Petty WJ, et al. Tumor histology predicts patterns of failure

and survival in patients with brain metastases from lung cancer treated with gamma knife

radiosurgery. Neurosurgery 2013; 73(4): 641-7; discussion 7.

128. Motta M, Del Vecchio A, Attuati L, et al. Gamma Knife Radiosurgery for Treatment of

Cerebral Metastases from Non-Small-Cell Lung Cancer. Int J Radiat Oncol Biol Phys 2011.

129. Li XP, Xiao JP, Chen XJ, Jiang XS, Zhang Y, Xu YJ. Fractionated stereotactic

radiotherapy for small-cell lung cancer patients with brain metastases. J Cancer Res Ther 2014;

10(3): 597-602. doi: 10.4103/0973-1482.137964.

130. Serizawa T, Ono J, Iichi T, et al. Gamma knife radiosurgery for metastatic brain

tumors from lung cancer: a comparison between small cell and non-small cell carcinoma. J

Neurosurg 2002; 97(5 Suppl): 484-8.

Page 135: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

135

131. Kong DS, Lee JI, Nam DH, et al. Prognosis of non-small cell lung cancer with

synchronous brain metastases treated with gamma knife radiosurgery. J Korean Med Sci 2006;

21(3): 527-32.

132. Hoffman R, Sneed PK, McDermott MW, et al. Radiosurgery for brain metastases from

primary lung carcinoma. Cancer J 2001; 7(2): 121-31.

133. Sheehan JP, Sun MH, Kondziolka D, Flickinger J, Lunsford LD. Radiosurgery for non-

small cell lung carcinoma metastatic to the brain: long-term outcomes and prognostic factors

influencing patient survival time and local tumor control. J Neurosurg 2002; 97(6): 1276-81.

134. Wegner RE, Olson AC, Kondziolka D, Niranjan A, Lundsford LD, Flickinger JC.

Stereotactic Radiosurgery for Patients With Brain Metastases From Small Cell Lung Cancer. Int J

Radiat Oncol Biol Phys 2011.

135. Marko NF, Suh JH, Chao ST, et al. Gamma knife stereotactic radiosurgery for the

management of incidentally-identified brain metastasis from non-small cell lung cancer. J

Neurooncol 2011.

136. Abacioglu U, Caglar H, Atasoy BM, Abdulloev T, Akgun Z, Kilic T. Gamma knife

radiosurgery in non small cell lung cancer patients with brain metastases: treatment results and

prognostic factors. J BUON 2010; 15(2): 274-80.

137. Kuremsky JG, Urbanic JJ, Petty WJ, et al. Tumor histology predicts patterns of failure

and survival in patients with brain metastases from lung cancer treated with gamma knife

radiosurgery. Neurosurgery 2013; 73(4): 641-7; discussion 7. doi:

10.1227/NEU.0000000000000072.

138. Wang TJ, Saad S, Qureshi YH, et al. Does lung cancer mutation status and targeted

therapy predict for outcomes and local control in the setting of brain metastases treated with

radiation? Neuro Oncol 2015; 24.

139. Johung KL, Yao X, Li F, et al. A clinical model for identifying radiosensitive tumor

genotypes in non-small cell lung cancer. Clin Cancer Res 2013; 19(19): 5523-32. doi: 10.1158/078-

0432.CCR-13-836. Epub 2013 Jul 29.

140. Bowden G, Kano H, Caparosa E, et al. Gamma Knife radiosurgery for the

management of cerebral metastases from non-small cell lung cancer. J Neurosurg 2015; 122(4):

766-72. doi: 10.3171/2014.12.JNS141111. Epub 2015 Feb 6.

Page 136: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

136

141. Flannery TW, Suntharalingam M, Regine WF, et al. Long-term survival in patients

with synchronous, solitary brain metastasis from non-small-cell lung cancer treated with

radiosurgery. Int J Radiat Oncol Biol Phys 2008; 72(1): 19-23.

142. Park SH, Hwang SK, Kang DH, et al. Gamma knife radiosurgery for multiple brain

metastases from lung cancer. J Clin Neurosci 2009; 16(5): 626-9. doi: 10.1016/j.jocn.2008.08.003.

Epub 9 Mar 5.

143. Sperduto PW, Wang M, Robins HI, et al. A phase 3 trial of whole brain radiation

therapy and stereotactic radiosurgery alone versus WBRT and SRS with temozolomide or erlotinib

for non-small cell lung cancer and 1 to 3 brain metastases: Radiation Therapy Oncology Group

0320. Int J Radiat Oncol Biol Phys 2013; 85(5): 1312-8. doi: 10.016/j.ijrobp.2012.11.042. Epub 3

Feb 4.

144. Lim SH, Lee JY, Lee MY, et al. A randomized phase III trial of stereotactic radiosurgery

(SRS) versus observation for patients with asymptomatic cerebral oligo-metastases in non-small-

cell lung cancer. Ann Oncol 2015; 26(4): 762-8. doi: 10.1093/annonc/mdu584. Epub 2014 Dec 23.

145. Wronski M, Maor MH, Davis BJ, Sawaya R, Levin VA. External radiation of brain

metastases from renal carcinoma: a retrospective study of 119 patients from the M. D. Anderson

Cancer Center. Int J Radiat Oncol Biol Phys 1997; 37(4): 753-9.

146. Mori Y, Kondziolka D, Flickinger JC, Logan T, Lunsford LD. Stereotactic radiosurgery

for brain metastasis from renal cell carcinoma. Cancer 1998; 83(2): 344-53.

147. Powell JW, Chung CT, Shah HR, et al. Gamma Knife surgery in the management of

radioresistant brain metastases in high-risk patients with melanoma, renal cell carcinoma, and

sarcoma. J Neurosurg 2008; 109 Suppl: 122-8.

148. Sheehan JP, Sun MH, Kondziolka D, Flickinger J, Lunsford LD. Radiosurgery in patients

with renal cell carcinoma metastasis to the brain: long-term outcomes and prognostic factors

influencing survival and local tumor control. J Neurosurg 2003; 98(2): 342-9.

149. Muacevic A, Kreth FW, Mack A, Tonn JC, Wowra B. Stereotactic radiosurgery without

radiation therapy providing high local tumor control of multiple brain metastases from renal cell

carcinoma. Minim Invasive Neurosurg 2004; 47(4): 203-8.

150. Shuto T, Inomori S, Fujino H, Nagano H. Gamma knife surgery for metastatic brain

tumors from renal cell carcinoma. J Neurosurg 2006; 105(4): 555-60.

151. Lwu S, Goetz P, Monsalves E, et al. Stereotactic radiosurgery for the treatment of

melanoma and renal cell carcinoma brain metastases. Oncol Rep 2013; 29(2): 407-12.

Page 137: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

137

152. Cochran DC, Chan MD, Aklilu M, et al. The effect of targeted agents on outcomes in

patients with brain metastases from renal cell carcinoma treated with Gamma Knife surgery. J

Neurosurg 2012; 116(5): 978-83. doi: 10.3171/2012.2.JNS111353. Epub 2012 Mar 2.

153. Hoshi S, Jokura H, Nakamura H, et al. Gamma-knife radiosurgery for brain metastasis

of renal cell carcinoma: results in 42 patients. Int J Urol 2002; 9(11): 618-25; discussion 26; author

reply 27.

154. Kano H, Iyer A, Kondziolka D, Niranjan A, Flickinger JC, Lunsford LD. Outcome

predictors of gamma knife radiosurgery for renal cell carcinoma metastases. Neurosurgery 2011;

69(6): 1232-9.

155. Hernandez L, Zamorano L, Sloan A, et al. Gamma knife radiosurgery for renal cell

carcinoma brain metastases. J Neurosurg 2002; 97(5 Suppl): 489-93.

156. Liew DN, Kano H, Kondziolka D, et al. Outcome predictors of Gamma Knife surgery

for melanoma brain metastases. Clinical article. J Neurosurg 2011; 114(3): 769-79.

157. Brown PD, Brown CA, Pollock BE, Gorman DA, Foote RL. Stereotactic radiosurgery for

patients with "radioresistant" brain metastases. Neurosurgery 2008; 62 Suppl 2: 790-801.

158. Gaudy-Marqueste C, Regis JM, Muracciole X, et al. Gamma-Knife radiosurgery in the

management of melanoma patients with brain metastases: a series of 106 patients without whole-

brain radiotherapy. Int J Radiat Oncol Biol Phys 2006; 65(3): 809-16.

159. Mathieu D, Kondziolka D, Cooper PB, et al. Gamma knife radiosurgery in the

management of malignant melanoma brain metastases. Neurosurgery 2007; 60(3): 471-81;

discussion 81-2.

160. Radbill AE, Fiveash JF, Falkenberg ET, et al. Initial treatment of melanoma brain

metastases using gamma knife radiosurgery: an evaluation of efficacy and toxicity. Cancer 2004;

101(4): 825-33.

161. Yu C, Chen JC, Apuzzo ML, et al. Metastatic melanoma to the brain: prognostic

factors after gamma knife radiosurgery. Int J Radiat Oncol Biol Phys 2002; 52(5): 1277-87.

162. Mingione V, Oliveira M, Prasad D, Steiner M, Steiner L. Gamma surgery for

melanoma metastases in the brain. J Neurosurg 2002; 96(3): 544-51.

163. Grob JJ, Regis J, Laurans R, et al. Radiosurgery without whole brain radiotherapy in

melanoma brain metastases. Club de Cancerologie Cutanee. Eur J Cancer 1998; 34(8): 1187-92.

Page 138: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

138

164. Mori Y, Kondziolka D, Flickinger JC, Kirkwood JM, Agarwala S, Lunsford LD.

Stereotactic radiosurgery for cerebral metastatic melanoma: factors affecting local disease control

and survival. Int J Radiat Oncol Biol Phys 1998; 42(3): 581-9.

165. Seung SK, Sneed PK, McDermott MW, et al. Gamma knife radiosurgery for malignant

melanoma brain metastases. Cancer J Sci Am 1998; 4(2): 103-9.

166. Neal MT, Chan MD, Lucas JT, Jr., et al. Predictors of survival, neurologic death, local

failure, and distant failure after gamma knife radiosurgery for melanoma brain metastases. World

Neurosurg 2014; 82(6): 1250-5. doi: 10.016/j.wneu.2013.02.025. Epub Feb 10.

167. Knisely JP, Yu JB, Flanigan J, Sznol M, Kluger HM, Chiang VL. Radiosurgery for

melanoma brain metastases in the ipilimumab era and the possibility of longer survival. J

Neurosurg 2012; 117(2): 227-33.

168. Tazi K, Hathaway A, Chiuzan C, Shirai K. Survival of melanoma patients with brain

metastases treated with ipilimumab and stereotactic radiosurgery. Cancer Med 2015; 4(1): 1-6.

doi: 10.1002/cam4.315. Epub 2014 Aug 28.

169. Mathew M, Tam M, Ott PA, et al. Ipilimumab in melanoma with limited brain

metastases treated with stereotactic radiosurgery. Melanoma Res 2013; 23(3): 191-5. doi:

10.1097/CMR.0b013e32835f3d90.

170. Narayana A, Mathew M, Tam M, et al. Vemurafenib and radiation therapy in

melanoma brain metastases. J Neurooncol 2013; 113(3): 411-6. doi: 10.1007/s11060-013-1127-1.

Epub 2013 Apr 12.

171. Gaudy-Marqueste C, Carron R, Delsanti C, et al. On demand Gamma-Knife strategy

can be safely combined with BRAF inhibitors for the treatment of melanoma brain metastases.

Ann Oncol 2014; 25(10): 2086-91. doi: 10.1093/annonc/mdu266. Epub 2014 Jul 23.

172. Da Silva AN, Nagayama K, Schlesinger DJ, Sheehan JP. Gamma Knife surgery for brain

metastases from gastrointestinal cancer. J Neurosurg 2009; 111(3): 423-30.

173. Kim HJ, Huh JW, Jung TY, et al. Clinical outcome with gamma-knife surgery or surgery

for brain metastases from colorectal cancer. J Clin Neurosci 2013; 20(10): 1417-21. doi:

10.016/j.jocn.2012.12.020. Epub 3 Aug 2.

174. Skeie BS, Enger PO, Ganz JC, et al. Gamma knife surgery of colorectal brain

metastases: a high prescription dose of 25 Gy may improve growth control. World Neurosurg

2013; 79(3-4): 525-36.

Page 139: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

139

175. Matsunaga S, Shuto T, Kawahara N, Suenaga J, Inomori S, Fujino H. Gamma Knife

surgery for brain metastases from colorectal cancer. Clinical article. J Neurosurg 2011; 114(3): 782-

9.

176. Hasegawa T, Kondziolka D, Flickinger JC, Lunsford LD. Stereotactic radiosurgery for

brain metastases from gastrointestinal tract cancer. Surg Neurol 2003; 60(6): 506-14; discussion

14-5.

177. Schoeggl A, Kitz K, Reddy M, Zauner C. Stereotactic radiosurgery for brain metastases

from colorectal cancer. Int J Colorectal Dis 2002; 17(3): 150-5.

178. Skeie BS, Enger PO, Ganz JC, et al. Gamma knife surgery of colorectal brain

metastases: a high prescription dose of 25 Gy may improve growth control. World Neurosurg

2013; 79(3-4): 525-36. doi: 10.1016/j.wneu.2011.09.019. Epub 2 Apr 16.

179. Matsunaga S, Shuto T, Kawahara N, Suenaga J, Inomori S, Fujino H. Gamma Knife

surgery for brain metastases from colorectal cancer. Clinical article. J Neurosurg 2011; 114(3): 782-

9. doi: 10.3171/2010.9.JNS10354. Epub 2010 Oct 15.

180. Park YS, Chang JH, Chang JW, Park YG. The efficacy of gamma knife radiosurgery for

advanced gastric cancer with brain metastases. J Neurooncol 2011; 103(3): 513-21. doi:

10.1007/s11060-010-0405-4. Epub 2010 Oct 19.

181. Iwai Y, Yamanaka K, Ikeda H. Gamma Knife radiosurgery for skull base meningioma:

long-term results of low-dose treatment. J Neurosurg 2008; 109(5): 804-10.

182. Sheehan JP, Starke RM, Kano H, et al. Gamma Knife radiosurgery for sellar and

parasellar meningiomas: a multicenter study. J Neurosurg 2014; 120(6): 1268-77. doi:

10.3171/2014.2.JNS13139. Epub 2014 Mar 28.

183. Jaaskelainen J, Haltia M, Laasonen E, Wahlstrom T, Valtonen S. The growth rate of

intracranial meningiomas and its relation to histology. An analysis of 43 patients. Surg Neurol

1985; 24(2): 165-72.

184. Jung HW, Yoo H, Paek SH, Choi KS. Long-term outcome and growth rate of subtotally

resected petroclival meningiomas: experience with 38 cases. Neurosurgery 2000; 46(3): 567-74;

discussion 74-5.

185. Van Havenbergh T, Carvalho G, Tatagiba M, Plets C, Samii M. Natural history of

petroclival meningiomas. Neurosurgery 2003; 52(1): 55-62; discussion -4.

186. Flannery TJ, Kano H, Lunsford LD, et al. Long-term control of petroclival meningiomas

through radiosurgery. J Neurosurg 2010; 112(5): 957-64.

Page 140: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

140

187. Roche PH, Regis J, Dufour H, et al. Gamma knife radiosurgery in the management of

cavernous sinus meningiomas. J Neurosurg 2000; 93 Suppl 3: 68-73.

188. Lee JY, Kondziolka D, Flickinger JC, Lunsford LD. Radiosurgery for intracranial

meningiomas. Prog Neurol Surg 2007; 20: 142-9.

189. Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD. Gamma knife radiosurgery of

imaging-diagnosed intracranial meningioma. Int J Radiat Oncol Biol Phys 2003; 56(3): 801-6.

190. Maruyama K, Shin M, Kurita H, Kawahara N, Morita A, Kirino T. Proposed treatment

strategy for cavernous sinus meningiomas: a prospective study. Neurosurgery 2004; 55(5): 1068-

75.

191. Santacroce A, Walier M, Regis J, et al. Long-term tumor control of benign intracranial

meningiomas after radiosurgery in a series of 4565 patients. Neurosurgery 2012; 70(1): 32-9;

discussion 9.

192. Nicolato A, Foroni R, Alessandrini F, Maluta S, Bricolo A, Gerosa M. The role of

Gamma Knife radiosurgery in the management of cavernous sinus meningiomas. Int J Radiat Oncol

Biol Phys 2002; 53(4): 992-1000.

193. Aichholzer M, Bertalanffy A, Dietrich W, et al. Gamma knife radiosurgery of skull base

meningiomas. Acta Neurochir (Wien) 2000; 142(6): 647-52; discussion 52-3.

194. Kollova A, Liscak R, Novotny J, Jr., Vladyka V, Simonova G, Janouskova L. Gamma

Knife surgery for benign meningioma. J Neurosurg 2007; 107(2): 325-36.

195. Hayashi M, Chernov M, Tamura N, et al. Gamma knife robotic microradiosurgery for

benign skull base meningiomas: tumor shrinkage may depend on the amount of radiation energy

delivered per lesion volume (unit energy). Stereotact Funct Neurosurg 2011; 89(1): 6-16.

196. Zachenhofer I, Wolfsberger S, Aichholzer M, et al. Gamma-knife radiosurgery for

cranial base meningiomas: experience of tumor control, clinical course, and morbidity in a follow-

up of more than 8 years. Neurosurgery 2006; 58(1): 28-36; discussion 28-36.

197. Starke RM, Williams BJ, Hiles C, Nguyen JH, Elsharkawy MY, Sheehan JP. Gamma

knife surgery for skull base meningiomas. J Neurosurg 2012; 116(3): 588-97.

198. Kondziolka D, Mathieu D, Lunsford LD, et al. Radiosurgery as definitive management

of intracranial meningiomas. Neurosurgery 2008; 62(1): 53-8; discussion 8-60. doi:

10.1227/01.NEU.0000311061.72626.0D.

199. Malik I, Rowe JG, Walton L, Radatz MW, Kemeny AA. The use of stereotactic

radiosurgery in the management of meningiomas. Br J Neurosurg 2005; 19(1): 13-20.

Page 141: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

141

200. Kondziolka D, Patel AD, Kano H, Flickinger JC, Lunsford LD. Long-term Outcomes After

Gamma Knife Radiosurgery for Meningiomas. Am J Clin Oncol 2014; 21: 21.

201. Pollock BE, Stafford SL, Link MJ, Garces YI, Foote RL. Single-fraction radiosurgery for

presumed intracranial meningiomas: efficacy and complications from a 22-year experience. Int J

Radiat Oncol Biol Phys 2012; 83(5): 1414-8. doi: 10.016/j.ijrobp.2011.10.033. Epub Dec 29.

202. Starke RM, Przybylowski CJ, Sugoto M, et al. Gamma Knife radiosurgery of large skull

base meningiomas. J Neurosurg 2015; 122(2): 363-72. doi: 10.3171/2014.10.JNS14198. Epub 2014

Dec 5.

203. Pollock BE, Stafford SL, Link MJ, Garces YI, Foote RL. Single-fraction radiosurgery of

benign cavernous sinus meningiomas. J Neurosurg 2013; 119(3): 675-82. doi:

10.3171/2013.5.JNS13206. Epub 2013 Jun 28.

204. Skeie BS, Enger PO, Skeie GO, Thorsen F, Pedersen PH. Gamma knife surgery of

meningiomas involving the cavernous sinus: long-term follow-up of 100 patients. Neurosurgery

2010; 66(4): 661-8; discussion 8-9. doi: 10.1227/01.NEU.0000366112.04015.E2.

205. Igaki H, Maruyama K, Koga T, et al. Stereotactic radiosurgery for skull base

meningioma. Neurol Med Chir (Tokyo) 2009; 49(10): 456-61.

206. Liscak R, Kollova A, Vladyka V, Simonova G, Novotny J, Jr. Gamma knife radiosurgery

of skull base meningiomas. Acta Neurochir Suppl 2004; 91: 65-74.

207. Takanashi M, Fukuoka S, Hojyo A, Sasaki T, Nakagawara J, Nakamura H. Gamma knife

radiosurgery for skull-base meningiomas. Prog Neurol Surg 2009; 22:96-111.(doi):

10.1159/000163385.

208. Han JH, Kim DG, Chung HT, et al. Gamma knife radiosurgery for skull base

meningiomas: long-term radiologic and clinical outcome. Int J Radiat Oncol Biol Phys 2008; 72(5):

1324-32. doi: 10.016/j.ijrobp.2008.03.028. Epub Oct 14.

209. Bir SC, Ambekar S, Ward T, Nanda A. Outcomes and complications of gamma knife

radiosurgery for skull base meningiomas. J Neurol Surg B Skull Base 2014; 75(6): 397-401. doi:

10.1055/s-0034-1376422. Epub 2014 May 27.

210. Williams BJ, Yen CP, Starke RM, et al. Gamma Knife surgery for parasellar

meningiomas: long-term results including complications, predictive factors, and progression-free

survival. J Neurosurg 2011; 114(6): 1571-7. doi: 10.3171/2011.1.JNS091939. Epub 2011 Feb 11.

Page 142: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

142

211. Starke R, Kano H, Ding D, et al. Stereotactic radiosurgery of petroclival meningiomas:

a multicenter study. J Neurooncol 2014; 119(1): 169-76. doi: 10.1007/s11060-014-1470-x. Epub

2014 May 13.

212. Flannery TJ, Kano H, Lunsford LD, et al. Long-term control of petroclival meningiomas

through radiosurgery. J Neurosurg 2010; 112(5): 957-64. doi: 10.3171/2009.8.JNS09695.

213. Kondziolka D, Madhok R, Lunsford LD, et al. Stereotactic radiosurgery for convexity

meningiomas. J Neurosurg 2009; 111(3): 458-63. doi: 10.3171/2008.8.JNS17650.

214. Sheehan JP, Starke RM, Kano H, et al. Gamma Knife radiosurgery for posterior fossa

meningiomas: a multicenter study. J Neurosurg 2015; 10: 1-11.

215. Park SH, Kano H, Niranjan A, Monaco E, 3rd, Flickinger JC, Lunsford LD. Gamma Knife

radiosurgery for meningiomas arising from the tentorium: a 22-year experience. J Neurooncol

2015; 121(1): 129-34. doi: 10.1007/s11060-014-1605-0. Epub 2014 Sep 4.

216. Ding D, Starke RM, Kano H, et al. Gamma knife radiosurgery for cerebellopontine

angle meningiomas: a multicenter study. Neurosurgery 2014; 75(4): 398-408; quiz . doi:

10.1227/NEU.0000000000000480.

217. Gande A, Kano H, Bowden G, et al. Gamma Knife radiosurgery of olfactory groove

meningiomas provides a method to preserve subjective olfactory function. J Neurooncol 2014;

116(3): 577-83. doi: 10.1007/s11060-013-1335-8. Epub 2014 Jan 8.

218. Hasegawa T, Kida Y, Yoshimoto M, Iizuka H, Ishii D, Yoshida K. Gamma Knife surgery

for convexity, parasagittal, and falcine meningiomas. J Neurosurg 2011; 114(5): 1392-8. doi:

10.3171/2010.11.JNS10112. Epub 2010 Dec 3.

219. Ding D, Xu Z, McNeill IT, Yen CP, Sheehan JP. Radiosurgery for parasagittal and

parafalcine meningiomas. J Neurosurg 2013; 119(4): 871-7. doi: 10.3171/2013.6.JNS13110. Epub

2013 Aug 9.

220. Zada G, Pagnini PG, Yu C, et al. Long-term outcomes and patterns of tumor

progression after gamma knife radiosurgery for benign meningiomas. Neurosurgery 2010; 67(2):

322-8; discussion 8-9. doi: 10.1227/01.NEU.0000371974.88873.15.

221. Mathiesen T, Pettersson-Segerlind J, Kihlstrom L, Ulfarsson E. Meningiomas engaging

major venous sinuses. World Neurosurg 2014; 81(1): 116-24. doi: 10.1016/j.wneu.2013.01.095.

Epub Jan 30.

Page 143: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

143

222. Hardesty DA, Wolf AB, Brachman DG, et al. The impact of adjuvant stereotactic

radiosurgery on atypical meningioma recurrence following aggressive microsurgical resection. J

Neurosurg 2013; 119(2): 475-81. doi: 10.3171/2012.12.JNS12414. Epub 2013 Feb 8.

223. Ferraro DJ, Funk RK, Blackett JW, et al. A retrospective analysis of survival and

prognostic factors after stereotactic radiosurgery for aggressive meningiomas. Radiat Oncol 2014;

9:38.(doi): 10.1186/748-717X-9-38.

224. Tamura M, Kubo K, Okita R, et al. Management of non-benign meningiomas with

Gamma Knife radiosurgery. Acta Neurochir Suppl 2013; 116:91-7.(doi): 10.1007/978-3-7091-1376-

9_14.

225. Mori Y, Tsugawa T, Hashizume C, Kobayashi T, Shibamoto Y. Gamma knife

stereotactic radiosurgery for atypical and malignant meningiomas. Acta Neurochir Suppl 2013;

116:85-9.(doi): 10.1007/978-3-7091-1376-9_13.

226. Hanakita S, Koga T, Igaki H, et al. Role of gamma knife surgery for intracranial atypical

(WHO grade II) meningiomas. J Neurosurg 2013; 119(6): 1410-4. doi: 10.3171/2013.8.JNS13343.

Epub 2013 Sep 27.

227. Kim JW, Kim DG, Paek SH, et al. Radiosurgery for atypical and anaplastic

meningiomas: histopathological predictors of local tumor control. Stereotact Funct Neurosurg

2012; 90(5): 316-24. doi: 10.1159/000338253. Epub 2012 Jul 12.

228. Lunsford LD, Niranjan A, Flickinger JC, Maitz A, Kondziolka D. Radiosurgery of

vestibular schwannomas: summary of experience in 829 cases. J Neurosurg 2005; 102(Suppl): 195-

9.

229. Martin HC, Sethi J, Lang D, Neil-Dwyer G, Lutman ME, Yardley L. Patient-assessed

outcomes after excision of acoustic neuroma: postoperative symptoms and quality of life. J

Neurosurg 2001; 94(2): 211-6.

230. Flickinger JC, Kondziolka D, Niranjan A, Maitz A, Voynov G, Lunsford LD. Acoustic

neuroma radiosurgery with marginal tumor doses of 12 to 13 Gy. Int J Radiat Oncol Biol Phys 2004;

60(1): 225-30.

231. Chung WY, Liu KD, Shiau CY, et al. Gamma knife surgery for vestibular schwannoma:

10-year experience of 195 cases. J Neurosurg 2005; 102(Suppl): 87-96.

232. Hasegawa T, Kida Y, Kobayashi T, Yoshimoto M, Mori Y, Yoshida J. Long-term

outcomes in patients with vestibular schwannomas treated using gamma knife surgery: 10-year

follow up. J Neurosurg 2005; 102(1): 10-6.

Page 144: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

144

233. Regis J, Delsanti C, Roche PH, Thomassin JM, Pellet W. [Functional outcomes of

radiosurgical treatment of vestibular schwannomas: 1000 successive cases and review of the

literature]. Neurochirurgie 2004; 50(2-3 Pt 2): 301-11.

234. Unger F, Walch C, Schrottner O, Eustacchio S, Sutter B, Pendl G. Cranial nerve

preservation after radiosurgery of vestibular schwannomas. Acta Neurochir Suppl 2002; 84: 77-83.

235. Myrseth E, Moller P, Pedersen PH, Lund-Johansen M. Vestibular schwannoma:

surgery or gamma knife radiosurgery? A prospective, nonrandomized study. Neurosurgery 2009;

64(4): 654-61; discussion 61-3. doi: 10.1227/01.NEU.0000340684.60443.55.

236. Raut VV, Walsh RM, Bath AP, et al. Conservative management of vestibular

schwannomas - second review of a prospective longitudinal study. Clin Otolaryngol Allied Sci 2004;

29(5): 505-14.

237. Yamakami I, Uchino Y, Kobayashi E, Yamaura A. Conservative management, gamma-

knife radiosurgery, and microsurgery for acoustic neurinomas: a systematic review of outcome

and risk of three therapeutic options. Neurol Res 2003; 25(7): 682-90.

238. Regis J, Carron R, Park MC, et al. Wait-and-see strategy compared with proactive

Gamma Knife surgery in patients with intracanalicular vestibular schwannomas. J Neurosurg 2010;

113(Suppl): 105-11.

239. Breivik CN, Nilsen RM, Myrseth E, et al. Conservative management or gamma knife

radiosurgery for vestibular schwannoma: tumor growth, symptoms, and quality of life.

Neurosurgery 2013; 73(1): 48-56; discussion -7. doi: 10.1227/01.neu.0000429862.50018.b9.

240. Tveiten OV, Carlson ML, Goplen F, Vassbotn F, Link MJ, Lund-Johansen M. Long-term

Auditory Symptoms in Patients With Sporadic Vestibular Schwannoma: An International Cross-

Sectional Study. Neurosurgery 2015; 4: 4.

241. Murphy ES, Barnett GH, Vogelbaum MA, et al. Long-term outcomes of Gamma Knife

radiosurgery in patients with vestibular schwannomas. J Neurosurg 2011; 114(2): 432-40. doi:

10.3171/2009.12.JNS091339. Epub 2010 Jan 22.

242. Delbrouck C, Hassid S, Choufani G, De Witte O, Devriendt D, Massager N. Hearing

outcome after gamma knife radiosurgery for vestibular schwannoma: a prospective Belgian clinical

study. B-ENT 2011; 7(Suppl 17): 77-84.

243. Sun S, Liu A. Long-term follow-up studies of Gamma Knife surgery with a low margin

dose for vestibular schwannoma. J Neurosurg 2012; 117(Suppl): 57-62. doi:

10.3171/2012.7.GKS12783.

Page 145: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

145

244. Wangerid T, Bartek J, Jr., Svensson M, Forander P. Long-term quality of life and

tumour control following gamma knife radiosurgery for vestibular schwannoma. Acta Neurochir

(Wien) 2014; 156(2): 389-96. doi: 10.1007/s00701-013-1924-5. Epub 2013 Nov 6.

245. Timmer FC, van Haren AE, Mulder JJ, et al. Quality of life after gamma knife

radiosurgery treatment in patients with a vestibular schwannoma: the patient's perspective. Eur

Arch Otorhinolaryngol 2010; 267(6): 867-73. doi: 10.1007/s00405-009-1140-3. Epub 2009 Nov 6.

246. Park SS, Grills IS, Bojrab D, et al. Longitudinal assessment of quality of life and

audiometric test outcomes in vestibular schwannoma patients treated with gamma knife surgery.

Otol Neurotol 2011; 32(4): 676-9. doi: 10.1097/MAO.0b013e3182138fc5.

247. Regis J, Carron R, Park MC, et al. Wait-and-see strategy compared with proactive

Gamma Knife surgery in patients with intracanalicular vestibular schwannomas: clinical article. J

Neurosurg 2013; 119(Suppl): 105-11.

248. Yang I, Aranda D, Han SJ, et al. Hearing preservation after stereotactic radiosurgery

for vestibular schwannoma: a systematic review. J Clin Neurosci 2009; 16(6): 742-7. doi:

10.1016/j.jocn.2008.09.023. Epub 9 Mar 20.

249. Yang I, Sughrue ME, Han SJ, et al. A comprehensive analysis of hearing preservation

after radiosurgery for vestibular schwannoma. J Neurosurg 2010; 112(4): 851-9. doi:

10.3171/2009.8.JNS0985.

250. Yomo S, Carron R, Thomassin JM, Roche PH, Regis J. Longitudinal analysis of hearing

before and after radiosurgery for vestibular schwannoma. J Neurosurg 2012; 117(5): 877-85. doi:

10.3171/2012.7.JNS10672. Epub 2012 Aug 31.

251. Tamura M, Carron R, Yomo S, et al. Hearing preservation after gamma knife

radiosurgery for vestibular schwannomas presenting with high-level hearing. Neurosurgery 2009;

64(2): 289-96; discussion 96. doi: 10.1227/01.NEU.0000338256.87936.7C.

252. Regis J, Tamura M, Delsanti C, Roche PH, Pellet W, Thomassin JM. Hearing

preservation in patients with unilateral vestibular schwannoma after gamma knife surgery. Prog

Neurol Surg 2008; 21:142-51.(doi): 10.1159/000156901.

253. Kano H, Kondziolka D, Khan A, Flickinger JC, Lunsford LD. Predictors of hearing

preservation after stereotactic radiosurgery for acoustic neuroma. J Neurosurg 2009; 111(4): 863-

73. doi: 10.3171/2008.12.JNS08611.

Page 146: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

146

254. Han JH, Kim DG, Chung HT, et al. Hearing preservation in patients with unilateral

vestibular schwannoma who undergo stereotactic radiosurgery: Reinterpretation of the auditory

brainstem response. Cancer 2012; 118(21): 5441-7. doi: 10.1002/cncr.27501. Epub 2012 May 8.

255. Carlson ML, Jacob JT, Pollock BE, et al. Long-term hearing outcomes following

stereotactic radiosurgery for vestibular schwannoma: patterns of hearing loss and variables

influencing audiometric decline. J Neurosurg 2013; 118(3): 579-87. doi: 10.3171/2012.9.JNS12919.

Epub 2012 Oct 26.

256. Boari N, Bailo M, Gagliardi F, et al. Gamma Knife radiosurgery for vestibular

schwannoma: clinical results at long-term follow-up in a series of 379 patients. J Neurosurg 2014;

121(Suppl): 123-42. doi: 10.3171/2014.8.GKS141506.

257. Mousavi SH, Kano H, Faraji AH, et al. Hearing Preservation up to 3 Years After

Gamma Knife Radiosurgery for Gardner-Robertson Class I Patients With Vestibular Schwannomas.

Neurosurgery 2015; 76(5): 584-91. doi: 10.1227/NEU.0000000000000674.

258. Baschnagel AM, Chen PY, Bojrab D, et al. Hearing preservation in patients with

vestibular schwannoma treated with Gamma Knife surgery. J Neurosurg 2013; 118(3): 571-8. doi:

10.3171/2012.10.JNS12880. Epub 2012 Dec 7.

259. Kim YH, Kim DG, Han JH, et al. Hearing outcomes after stereotactic radiosurgery for

unilateral intracanalicular vestibular schwannomas: implication of transient volume expansion. Int

J Radiat Oncol Biol Phys 2013; 85(1): 61-7. doi: 10.1016/j.ijrobp.2012.03.036. Epub May 12.

260. van Eck AT, Horstmann GA. Increased preservation of functional hearing after

gamma knife surgery for vestibular schwannoma. J Neurosurg 2005; 102(Suppl): 204-6.

261. Seferis C, Torrens M, Paraskevopoulou C, Psichidis G. Malignant transformation in

vestibular schwannoma: report of a single case, literature search, and debate. J Neurosurg 2014;

121(Suppl): 160-6. doi: 10.3171/2014.7.GKS141311.

262. Rowe J, Grainger A, Walton L, Silcocks P, Radatz M, Kemeny A. Risk of malignancy

after gamma knife stereotactic radiosurgery. Neurosurgery 2007; 60(1): 60-5; discussion 5-6.

263. Starke RM, Yen CP, Chen CJ, et al. An updated assessment of the risk of radiation-

induced neoplasia after radiosurgery of arteriovenous malformations. World Neurosurg 2014;

82(3-4): 395-401. doi: 10.1016/j.wneu.2013.02.008. Epub Feb 9.

264. Yang I, Sughrue ME, Han SJ, et al. Facial nerve preservation after vestibular

schwannoma Gamma Knife radiosurgery. J Neurooncol 2009; 93(1): 41-8. doi: 10.1007/s11060-

009-9842-3. Epub 2009 May 9.

Page 147: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

147

265. Tamura M, Murata N, Hayashi M, Roche PH, Regis J. Facial nerve function

insufficiency after radiosurgery versus microsurgery. Prog Neurol Surg 2008; 21:108-18.(doi):

10.1159/000156718.

266. Regis J, Pellet W, Delsanti C, et al. Functional outcome after gamma knife surgery or

microsurgery for vestibular schwannomas. J Neurosurg 2002; 97(5): 1091-100.

267. Iwai Y, Yamanaka K, Ishiguro T. Surgery combined with radiosurgery of large acoustic

neuromas. Surg Neurol 2003; 59(4): 283-9; discussion 9-91.

268. Iwai Y, Ishibashi K, Watanabe Y, Uemura G, Yamanaka K. Functional Preservation

After Planned Partial Resection Followed by Gamma Knife Radiosurgery for Large Vestibular

Schwannomas. World Neurosurg 2015; 16(15): 00247-8.

269. Fuentes S, Arkha Y, Pech-Gourg G, Grisoli F, Dufour H, Regis J. Management of large

vestibular schwannomas by combined surgical resection and gamma knife radiosurgery. Prog

Neurol Surg 2008; 21:79-82.(doi): 10.1159/000156709.

270. Brokinkel B, Sauerland C, Holling M, et al. Gamma Knife radiosurgery following

subtotal resection of vestibular schwannoma. J Clin Neurosci 2014; 21(12): 2077-82. doi:

10.1016/j.jocn.2014.03.037. Epub Jul 22.

271. Pan HC, Sheehan J, Sheu ML, Chiu WT, Yang DY. Intracapsular decompression or

radical resection followed by Gamma Knife surgery for patients harboring a large vestibular

schwannoma. J Neurosurg 2012; 117(Suppl): 69-77. doi: 10.3171/2012.6.GKS12697.

272. Chen Y, Li ZF, Zhang FX, et al. Gamma knife surgery for patients with volumetric

classification of nonfunctioning pituitary adenomas: a systematic review and meta-analysis. Eur J

Endocrinol 2013; 169(4): 487-95. doi: 10.1530/EJE-13-0400. Print 2013 Oct.

273. Sheehan JP, Starke RM, Mathieu D, et al. Gamma Knife radiosurgery for the

management of nonfunctioning pituitary adenomas: a multicenter study. J Neurosurg 2013;

119(2): 446-56. doi: 10.3171/2013.3.JNS12766. Epub 2013 Apr 26.

274. Pollock BE, Cochran J, Natt N, et al. Gamma knife radiosurgery for patients with

nonfunctioning pituitary adenomas: results from a 15-year experience. Int J Radiat Oncol Biol Phys

2008; 70(5): 1325-9.

275. Liscak R, Vladyka V, Marek J, Simonova G, Vymazal J. Gamma knife radiosurgery for

endocrine-inactive pituitary adenomas. Acta Neurochir (Wien) 2007; 149(10): 999-1006; discussion

276. Mingione V, Yen CP, Vance ML, et al. Gamma surgery in the treatment of

nonsecretory pituitary macroadenoma. J Neurosurg 2006; 104(6): 876-83.

Page 148: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

148

277. Park KJ, Kano H, Parry PV, et al. Long-term outcomes after gamma knife stereotactic

radiosurgery for nonfunctional pituitary adenomas. Neurosurgery 2011; 69(6): 1188-99. doi:

10.227/NEU.0b013e318222afed.

278. Gopalan R, Schlesinger D, Vance ML, Laws E, Sheehan J. Long-term outcomes after

Gamma Knife radiosurgery for patients with a nonfunctioning pituitary adenoma. Neurosurgery

2011; 69(2): 284-93.

279. Park KJ, Kano H, Parry PV, et al. Long-term outcomes after gamma knife stereotactic

radiosurgery for nonfunctional pituitary adenomas. Neurosurgery 2011; 69(6): 1188-99.

280. Hoybye C, Rahn T. Adjuvant Gamma Knife radiosurgery in non-functioning pituitary

adenomas; low risk of long-term complications in selected patients. Pituitary 2009; 12(3): 211-6.

doi: 10.1007/s11102-008-0163-x.

281. Cifarelli CP, Schlesinger DJ, Sheehan JP. Cranial nerve dysfunction following Gamma

Knife surgery for pituitary adenomas: long-term incidence and risk factors. J Neurosurg 2012;

116(6): 1304-10. doi: 10.3171/2012.2.JNS111630. Epub 2012 Mar 16.

282. Leavitt JA, Stafford SL, Link MJ, Pollock BE. Long-term evaluation of radiation-induced

optic neuropathy after single-fraction stereotactic radiosurgery. Int J Radiat Oncol Biol Phys 2013;

87(3): 524-7. doi: 10.1016/j.ijrobp.2013.06.47.

283. Pollock BE, Link MJ, Leavitt JA, Stafford SL. Dose-volume analysis of radiation-induced

optic neuropathy after single-fraction stereotactic radiosurgery. Neurosurgery 2014; 75(4): 456-

60; discussion 60. doi: 10.1227/NEU.0000000000000457.

284. Marek J, Jezkova J, Hana V, et al. Is it possible to avoid hypopituitarism after

irradiation of pituitary adenomas by the Leksell gamma knife? Eur J Endocrinol 2011; 164(2): 169-

78.

285. Feigl GC, Pistracher K, Berghold A, Mokry M. Pituitary insufficiency as a side effect

after radiosurgery for pituitary adenomas: the role of the hypothalamus. J Neurosurg 2010;

113(Suppl): 153-59.

286. Lee CC, Kano H, Yang HC, et al. Initial Gamma Knife radiosurgery for nonfunctioning

pituitary adenomas. J Neurosurg 2014; 120(3): 647-54. doi: 10.3171/2013.11.JNS131757. Epub

2014 Jan 3.

287. Ribeiro-Oliveira A, Jr., Barkan A. The changing face of acromegaly--advances in

diagnosis and treatment. Nat Rev Endocrinol 2012; 8(10): 605-11.

Page 149: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

149

288. Schofl C, Franz H, Grussendorf M, et al. Long-term outcome in patients with

acromegaly: analysis of 1344 patients from the German Acromegaly Register

The changing face of acromegaly--advances in diagnosis and treatment

Update on the natural history of cavernous malformations and factors predicting aggressive

clinical presentation. Eur J Endocrinol 2013; 168(1): 39-47.

289. Schofl C, Franz H, Grussendorf M, et al. Long-term outcome in patients with

acromegaly: analysis of 1344 patients from the German Acromegaly Register. Eur J Endocrinol

2013; 168(1): 39-47. doi: 10.1530/EJE-12-0602. Print 2013 Jan.

290. Jenkins PJ, Bates P, Carson MN, Stewart PM, Wass JA. Conventional pituitary

irradiation is effective in lowering serum growth hormone and insulin-like growth factor-I in

patients with acromegaly. J Clin Endocrinol Metab 2006; 91(4): 1239-45.

291. Barrande G, Pittino-Lungo M, Coste J, et al. Hormonal and metabolic effects of

radiotherapy in acromegaly: long-term results in 128 patients followed in a single center. J Clin

Endocrinol Metab 2000; 85(10): 3779-85.

292. Cozzi R, Barausse M, Asnaghi D, Dallabonzana D, Lodrini S, Attanasio R. Failure of

radiotherapy in acromegaly. Eur J Endocrinol 2001; 145(6): 717-26.

293. Epaminonda P, Porretti S, Cappiello V, Beck-Peccoz P, Faglia G, Arosio M. Efficacy of

radiotherapy in normalizing serum IGF-I, acid-labile subunit (ALS) and IGFBP-3 levels in

acromegaly. Clin Endocrinol (Oxf) 2001; 55(2): 183-9.

294. Tsang RW, Laperriere NJ, Simpson WJ, Brierley J, Panzarella T, Smyth HS. Glioma

arising after radiation therapy for pituitary adenoma. A report of four patients and estimation of

risk. Cancer 1993; 72(7): 2227-33.

295. Minniti G, Traish D, Ashley S, Gonsalves A, Brada M. Risk of second brain tumor after

conservative surgery and radiotherapy for pituitary adenoma: update after an additional 10 years.

J Clin Endocrinol Metab 2005; 90(2): 800-4.

296. Yang I, Kim W, De Salles A, Bergsneider M. A systematic analysis of disease control in

acromegaly treated with radiosurgery. Neurosurg Focus 2010; 29(4): E13. doi:

0.3171/2010.7.FOCUS10170.

297. Franzin A, Spatola G, Losa M, Picozzi P, Mortini P. Results of gamma knife

radiosurgery in acromegaly. Int J Endocrinol 2012; 2012: 342034.

Page 150: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

150

298. Erdur FM, Kilic T, Peker S, Celik O, Kadioglu P. Gammaknife radiosurgery in patients

with acromegaly. J Clin Neurosci 2011; 18(12): 1616-20. doi: 10.016/j.jocn.2011.03.023. Epub Oct

15.

299. Castinetti F, Taieb D, Kuhn JM, et al. Outcome of gamma knife radiosurgery in 82

patients with acromegaly: correlation with initial hypersecretion. J Clin Endocrinol Metab 2005;

90(8): 4483-8. Epub 2005 May 17.

300. Liu X, Kano H, Kondziolka D, et al. Gamma knife radiosurgery for clinically persistent

acromegaly. J Neurooncol 2012; 109(1): 71-9. doi: 10.1007/s11060-012-0862-z. Epub 2012 Apr 26.

301. Lee CC, Vance ML, Lopes MB, Xu Z, Chen CJ, Sheehan J. Stereotactic radiosurgery for

acromegaly: outcomes by adenoma subtype. Pituitary 2015; 18(3): 326-34. doi: 10.1007/s11102-

014-0578-5.

302. Ronchi CL, Attanasio R, Verrua E, et al. Efficacy and tolerability of gamma knife

radiosurgery in acromegaly: a 10-year follow-up study. Clin Endocrinol (Oxf) 2009; 71(6): 846-52.

doi: 10.1111/j.365-2265.009.03589.x. Epub 2009 Mar 28.

303. Vik-Mo EO, Oksnes M, Pedersen PH, et al. Gamma knife stereotactic radiosurgery for

acromegaly. Eur J Endocrinol 2007; 157(3): 255-63.

304. Jezkova J, Marek J, Hana V, et al. Gamma knife radiosurgery for acromegaly--long-

term experience. Clin Endocrinol (Oxf) 2006; 64(5): 588-95.

305. Landolt AM, Haller D, Lomax N, et al. Stereotactic radiosurgery for recurrent

surgically treated acromegaly: comparison with fractionated radiotherapy. J Neurosurg 1998;

88(6): 1002-8.

306. Sheehan JP, Pouratian N, Steiner L, Laws ER, Vance ML. Gamma Knife surgery for

pituitary adenomas: factors related to radiological and endocrine outcomes. J Neurosurg 2011;

114(2): 303-9. doi: 10.3171/2010.5.JNS091635. Epub 2010 Jun 11.

307. Swords FM, Monson JP, Besser GM, et al. Gamma knife radiosurgery: a safe and

effective salvage treatment for pituitary tumours not controlled despite conventional

radiotherapy. Eur J Endocrinol 2009; 161(6): 819-28. doi: 10.1530/EJE-09-0493. Epub 2009 Sep 22.

308. Jezkova J, Marek J, Hana V, et al. Gamma knife radiosurgery for acromegaly--long-

term experience. Clin Endocrinol (Oxf) 2006; 64(5): 588-95.

309. Kobayashi T, Mori Y, Uchiyama Y, Kida Y, Fujitani S. Long-term results of gamma knife

surgery for growth hormone-producing pituitary adenoma: is the disease difficult to cure? J

Neurosurg 2005; 102(Suppl): 119-23.

Page 151: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

151

310. Pollock BE, Jacob JT, Brown PD, Nippoldt TB. Radiosurgery of growth hormone-

producing pituitary adenomas: factors associated with biochemical remission. J Neurosurg 2007;

106(5): 833-8.

311. Landolt AM, Haller D, Lomax N, et al. Octreotide may act as a radioprotective agent

in acromegaly. J Clin Endocrinol Metab 2000; 85(3): 1287-9.

312. Jagannathan J, Sheehan JP, Pouratian N, Laws ER, Jr., Steiner L, Vance ML. Gamma

knife radiosurgery for acromegaly: outcomes after failed transsphenoidal surgery. Neurosurgery

2008; 62(6): 1262-9; discussion 9-70. doi: 10.27/01.neu.0000333297.41813.3d.

313. Pollock BE, Jacob JT, Brown PD, Nippoldt TB. Radiosurgery of growth hormone-

producing pituitary adenomas: factors associated with biochemical remission. J Neurosurg 2007;

106(5): 833-8.

314. Feigl GC, Bonelli CM, Berghold A, Mokry M. Effects of gamma knife radiosurgery of

pituitary adenomas on pituitary function. J Neurosurg 2002; 97(5 Suppl): 415-21.

315. Melmed S, Casanueva FF, Hoffman AR, et al. Diagnosis and treatment of

hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011;

96(2): 273-88.

316. Casanueva FF, Molitch ME, Schlechte JA, et al. Guidelines of the Pituitary Society for

the diagnosis and management of prolactinomas. Clin Endocrinol (Oxf) 2006; 65(2): 265-73.

317. Primeau V, Raftopoulos C, Maiter D. Outcomes of transsphenoidal surgery in

prolactinomas: improvement of hormonal control in dopamine agonist-resistant patients. Eur J

Endocrinol 2012; 166(5): 779-86.

318. Pouratian N, Sheehan J, Jagannathan J, Laws ER, Jr., Steiner L, Vance ML. Gamma

knife radiosurgery for medically and surgically refractory prolactinomas. Neurosurgery 2006; 59(2):

255-66; discussion -66.

319. Jezkova J, Hana V, Krsek M, et al. Use of the Leksell gamma knife in the treatment of

prolactinoma patients. Clin Endocrinol (Oxf) 2009; 70(5): 732-41.

320. Cohen-Inbar O, Xu Z, Schlesinger D, Vance ML, Sheehan JP. Gamma Knife

radiosurgery for medically and surgically refractory prolactinomas: long-term results. Pituitary

2015; 12: 12.

321. Liu X, Kano H, Kondziolka D, et al. Gamma knife stereotactic radiosurgery for drug

resistant or intolerant invasive prolactinomas. Pituitary 2013; 16(1): 68-75.

Page 152: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

152

322. Tanaka S, Link MJ, Brown PD, Stafford SL, Young WF, Jr., Pollock BE. Gamma knife

radiosurgery for patients with prolactin-secreting pituitary adenomas. World Neurosurg 2010;

74(1): 147-52.

323. Pan L, Zhang N, Wang EM, Wang BJ, Dai JZ, Cai PW. Gamma knife radiosurgery as a

primary treatment for prolactinomas. J Neurosurg 2000; 93 Suppl 3: 10-3.

324. Liu X, Kano H, Kondziolka D, et al. Gamma knife stereotactic radiosurgery for drug

resistant or intolerant invasive prolactinomas. Pituitary 2013; 16(1): 68-75.

325. Jagannathan J, Sheehan JP, Pouratian N, Laws ER, Steiner L, Vance ML. Gamma Knife

surgery for Cushing's disease. J Neurosurg 2007; 106(6): 980-7.

326. Kobayashi T, Kida Y, Mori Y. Gamma knife radiosurgery in the treatment of Cushing

disease: long-term results. J Neurosurg 2002; 97(5 Suppl): 422-8.

327. Sheehan JM, Vance ML, Sheehan JP, Ellegala DB, Laws ER, Jr. Radiosurgery for

Cushing's disease after failed transsphenoidal surgery. J Neurosurg 2000; 93(5): 738-42.

328. Starke RM, Williams BJ, Vance ML, Sheehan JP. Radiation therapy and stereotactic

radiosurgery for the treatment of Cushing's disease: an evidence-based review. Curr Opin

Endocrinol Diabetes Obes 2010; 17(4): 356-64.

329. Castinetti F, Nagai M, Dufour H, et al. Gamma knife radiosurgery is a successful

adjunctive treatment in Cushing's disease. Eur J Endocrinol 2007; 156(1): 91-8.

330. Sheehan JP, Xu Z, Salvetti DJ, Schmitt PJ, Vance ML. Results of gamma knife surgery

for Cushing's disease. J Neurosurg 2013; 119(6): 1486-92. doi: 10.3171/2013.7.JNS13217. Epub

2013 Aug 9.

331. Marek J, Jezkova J, Hana V, et al. Gamma knife radiosurgery for Cushing's disease and

Nelson's syndrome. Pituitary 2015; 18(3): 376-84. doi: 10.1007/s11102-014-0584-7.

332. Mehta GU, Sheehan JP, Vance ML. Effect of stereotactic radiosurgery before bilateral

adrenalectomy for Cushing's disease on the incidence of Nelson's syndrome. J Neurosurg 2013;

119(6): 1493-7. doi: 10.3171/2013.7.JNS13389. Epub 2013 Aug 16.

333. Lee CC, Chen CJ, Yen CP, et al. Whole-sellar stereotactic radiosurgery for functioning

pituitary adenomas. Neurosurgery 2014; 75(3): 227-37; discussion 37. doi:

10.1227/NEU.0000000000000425.

334. Bodaghabadi M, Riazi H, Aran S, et al. Repeated transsphenoidal surgery or gamma

knife radiosurgery in recurrent cushing disease after transsphenoidal surgery. J Neurol Surg A Cent

Eur Neurosurg 2014; 75(2): 91-7. doi: 10.1055/s-0033-1345688. Epub 2013 Aug 21.

Page 153: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

153

335. Ohgaki H, Dessen P, Jourde B, et al. Genetic pathways to glioblastoma: a population-

based study. Cancer Res 2004; 64(19): 6892-9.

336. Adamson C, Kanu OO, Mehta AI, et al. Glioblastoma multiforme: a review of where

we have been and where we are going. Expert Opin Investig Drugs 2009; 18(8): 1061-83.

337. Johnson DR, Leeper HE, Uhm JH. Glioblastoma survival in the United States improved

after Food and Drug Administration approval of bevacizumab: A population-based analysis. Cancer

2013.

338. Pouratian N, Crowley RW, Sherman JH, Jagannathan J, Sheehan JP. Gamma Knife

radiosurgery after radiation therapy as an adjunctive treatment for glioblastoma. J Neurooncol

2009; 94(3): 409-18.

339. Kondziolka D, Flickinger JC, Bissonette DJ, Bozik M, Lunsford LD. Survival benefit of

stereotactic radiosurgery for patients with malignant glial neoplasms. Neurosurgery 1997; 41(4):

776-83; discussion 83-5.

340. Einstein DB, Wessels B, Bangert B, et al. Phase II trial of radiosurgery to magnetic

resonance spectroscopy-defined high-risk tumor volumes in patients with glioblastoma

multiforme. Int J Radiat Oncol Biol Phys 2012; 84(3): 668-74.

341. Shrieve DC, Alexander E, 3rd, Black PM, et al. Treatment of patients with primary

glioblastoma multiforme with standard postoperative radiotherapy and radiosurgical boost:

prognostic factors and long-term outcome. J Neurosurg 1999; 90(1): 72-7.

342. Sarkaria JN, Mehta MP, Loeffler JS, et al. Radiosurgery in the initial management of

malignant gliomas: survival comparison with the RTOG recursive partitioning analysis. Radiation

Therapy Oncology Group. Int J Radiat Oncol Biol Phys 1995; 32(4): 931-41.

343. Lipani JD, Jackson PS, Soltys SG, Sato K, Adler JR. Survival following CyberKnife

radiosurgery and hypofractionated radiotherapy for newly diagnosed glioblastoma multiforme.

Technol Cancer Res Treat 2008; 7(3): 249-55.

344. Nwokedi EC, DiBiase SJ, Jabbour S, Herman J, Amin P, Chin LS. Gamma knife

stereotactic radiosurgery for patients with glioblastoma multiforme. Neurosurgery 2002; 50(1): 41-

6; discussion 6-7.

345. Souhami L, Seiferheld W, Brachman D, et al. Randomized comparison of stereotactic

radiosurgery followed by conventional radiotherapy with carmustine to conventional radiotherapy

with carmustine for patients with glioblastoma multiforme: report of Radiation Therapy Oncology

Group 93-05 protocol. Int J Radiat Oncol Biol Phys 2004; 60(3): 853-60.

Page 154: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

154

346. Tsao MN, Mehta MP, Whelan TJ, et al. The American Society for Therapeutic

Radiology and Oncology (ASTRO) evidence-based review of the role of radiosurgery for malignant

glioma. Int J Radiat Oncol Biol Phys 2005; 63(1): 47-55.

347. Chan JL, Lee SW, Fraass BA, et al. Survival and failure patterns of high-grade gliomas

after three-dimensional conformal radiotherapy. J Clin Oncol 2002; 20(6): 1635-42.

348. Massaccesi M, Ferro M, Cilla S, et al. Accelerated intensity-modulated radiotherapy

plus temozolomide in patients with glioblastoma: a phase I dose-escalation study (ISIDE-BT-1). Int J

Clin Oncol 2012.

349. Tsien C, Moughan J, Michalski JM, et al. Phase I three-dimensional conformal

radiation dose escalation study in newly diagnosed glioblastoma: Radiation Therapy Oncology

Group Trial 98-03. Int J Radiat Oncol Biol Phys 2009; 73(3): 699-708.

350. Coughlin C, Scott C, Langer C, Coia L, Curran W, Rubin P. Phase II, two-arm RTOG trial

(94-11) of bischloroethyl-nitrosourea plus accelerated hyperfractionated radiotherapy (64.0 or

70.4 Gy) based on tumor volume (> 20 or < or = 20 cm(2), respectively) in the treatment of newly-

diagnosed radiosurgery-ineligible glioblastoma multiforme patients. Int J Radiat Oncol Biol Phys

2000; 48(5): 1351-8.

351. Sherriff J, Tamangani J, Senthil L, et al. Patterns of relapse in glioblastoma multiforme

following concomitant chemoradiotherapy with temozolomide. Br J Radiol 2013; 86(1022):

20120414.

352. Dobelbower MC, Burnett Iii OL, Nordal RA, et al. Patterns of failure for glioblastoma

multiforme following concurrent radiation and temozolomide. J Med Imaging Radiat Oncol 2011;

55(1): 77-81.

353. Chamberlain MC. Radiographic patterns of relapse in glioblastoma. J Neurooncol

2011; 101(2): 319-23.

354. Barker FG, 2nd, Chang SM, Gutin PH, et al. Survival and functional status after

resection of recurrent glioblastoma multiforme. Neurosurgery 1998; 42(4): 709-20; discussion 20-

3.

355. Young B, Oldfield EH, Markesbery WR, et al. Reoperation for glioblastoma. J

Neurosurg 1981; 55(6): 917-21.

356. Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy

of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent

gliomas. The Polymer-brain Tumor Treatment Group. Lancet 1995; 345(8956): 1008-12.

Page 155: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

155

357. Iwamoto FM, Abrey LE, Beal K, et al. Patterns of relapse and prognosis after

bevacizumab failure in recurrent glioblastoma. Neurology 2009; 73(15): 1200-6.

358. Maranzano E, Anselmo P, Casale M, et al. Treatment of recurrent glioblastoma with

stereotactic radiotherapy: long-term results of a mono-institutional trial. Tumori 2011; 97(1): 56-

61.

359. Combs SE, Widmer V, Thilmann C, Hof H, Debus J, Schulz-Ertner D. Stereotactic

radiosurgery (SRS): treatment option for recurrent glioblastoma multiforme (GBM). Cancer 2005;

104(10): 2168-73.

360. Skeie BS, Enger PO, Brogger J, et al. gamma knife surgery versus reoperation for

recurrent glioblastoma multiforme. World Neurosurg 2012; 78(6): 658-69.

361. Park KJ, Kano H, Iyer A, et al. Salvage gamma knife stereotactic radiosurgery followed

by bevacizumab for recurrent glioblastoma multiforme: a case-control study. J Neurooncol 2012;

107(2): 323-33.

362. Mahajan A, McCutcheon IE, Suki D, et al. Case-control study of stereotactic

radiosurgery for recurrent glioblastoma multiforme. J Neurosurg 2005; 103(2): 210-7.

363. Hall WA, Djalilian HR, Sperduto PW, et al. Stereotactic radiosurgery for recurrent

malignant gliomas. J Clin Oncol 1995; 13(7): 1642-8.

364. Shrieve DC, Alexander E, 3rd, Wen PY, et al. Comparison of stereotactic radiosurgery

and brachytherapy in the treatment of recurrent glioblastoma multiforme. Neurosurgery 1995;

36(2): 275-82; discussion 82-4.

365. Kong DS, Lee JI, Park K, Kim JH, Lim DH, Nam DH. Efficacy of stereotactic radiosurgery

as a salvage treatment for recurrent malignant gliomas. Cancer 2008; 112(9): 2046-51.

366. Ammirati M, Galicich JH, Arbit E, Liao Y. Reoperation in the treatment of recurrent

intracranial malignant gliomas. Neurosurgery 1987; 21(5): 607-14.

367. Hart MG, Garside R, Rogers G, Stein K, Grant R. Temozolomide for high grade glioma.

Cochrane Database Syst Rev 2013; 4: CD007415.

368. Larson DA, Gutin PH, McDermott M, et al. Gamma knife for glioma: selection factors

and survival. Int J Radiat Oncol Biol Phys 1996; 36(5): 1045-53.

369. Patel M, Siddiqui F, Jin JY, et al. Salvage reirradiation for recurrent glioblastoma with

radiosurgery: radiographic response and improved survival. J Neurooncol 2009; 92(2): 185-91.

Page 156: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

156

370. Hsieh PC, Chandler JP, Bhangoo S, et al. Adjuvant gamma knife stereotactic

radiosurgery at the time of tumor progression potentially improves survival for patients with

glioblastoma multiforme. Neurosurgery 2005; 57(4): 684-92; discussion -92.

371. Cho KH, Hall WA, Gerbi BJ, Higgins PD, McGuire WA, Clark HB. Single dose versus

fractionated stereotactic radiotherapy for recurrent high-grade gliomas. Int J Radiat Oncol Biol

Phys 1999; 45(5): 1133-41.

372. Elliott RE, Parker EC, Rush SC, et al. Efficacy of gamma knife radiosurgery for small-

volume recurrent malignant gliomas after initial radical resection. World Neurosurg 2011; 76(1-2):

128-40; discussion 61-2. doi: 10.1016/j.wneu.2010.12.053.

373. Dodoo E, Huffmann B, Peredo I, et al. Increased survival using delayed gamma knife

radiosurgery for recurrent high-grade glioma: a feasibility study. World Neurosurg 2014; 82(5):

e623-32. doi: 10.1016/j.wneu.2014.06.011. Epub Jun 13.

374. Niranjan A, Kano H, Iyer A, Kondziolka D, Flickinger JC, Lunsford LD. Role of adjuvant

or salvage radiosurgery in the management of unresected residual or progressive glioblastoma

multiforme in the pre-bevacizumab era. J Neurosurg 2015; 122(4): 757-65. doi:

10.3171/2014.11.JNS13295. Epub 2015 Jan 16.

375. Berman MF, Sciacca RR, Pile-Spellman J, et al. The epidemiology of brain

arteriovenous malformations. Neurosurgery 2000; 47(2): 389-96; discussion 97.

376. Fleetwood IG, Marcellus ML, Levy RP, Marks MP, Steinberg GK. Deep arteriovenous

malformations of the basal ganglia and thalamus: natural history. J Neurosurg 2003; 98(4): 747-50.

377. Pollock BE, Gorman DA, Brown PD. Radiosurgery for arteriovenous malformations of

the basal ganglia, thalamus, and brainstem. J Neurosurg 2004; 100(2): 210-4.

378. Redekop G, TerBrugge K, Montanera W, Willinsky R. Arterial aneurysms associated

with cerebral arteriovenous malformations: classification, incidence, and risk of hemorrhage. J

Neurosurg 1998; 89(4): 539-46.

379. Kano H, Lunsford LD, Flickinger JC, et al. Stereotactic radiosurgery for arteriovenous

malformations, Part 1: management of Spetzler-Martin Grade I and II arteriovenous

malformations. J Neurosurg 2012; 116(1): 11-20. doi: 10.3171/2011.9.JNS101740. Epub 2011 Nov

11.

380. Mansmann U, Meisel J, Brock M, Rodesch G, Alvarez H, Lasjaunias P. Factors

associated with intracranial hemorrhage in cases of cerebral arteriovenous malformation.

Neurosurgery 2000; 46(2): 272-9; discussion 9-81.

Page 157: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

157

381. Schneider BF, Eberhard DA, Steiner LE. Histopathology of arteriovenous

malformations after gamma knife radiosurgery. J Neurosurg 1997; 87(3): 352-7.

382. Shin M, Maruyama K, Kurita H, et al. Analysis of nidus obliteration rates after gamma

knife surgery for arteriovenous malformations based on long-term follow-up data: the University

of Tokyo experience. J Neurosurg 2004; 101(1): 18-24.

383. Flickinger JC, Kondziolka D, Maitz AH, Lunsford LD. An analysis of the dose-response

for arteriovenous malformation radiosurgery and other factors affecting obliteration. Radiother

Oncol 2002; 63(3): 347-54.

384. Karlsson B, Lindquist C, Steiner L. Prediction of obliteration after gamma knife

surgery for cerebral arteriovenous malformations. Neurosurgery 1997; 40(3): 425-30; discussion

30-1.

385. Flickinger JC, Pollock BE, Kondziolka D, Lunsford LD. A dose-response analysis of

arteriovenous malformation obliteration after radiosurgery. Int J Radiat Oncol Biol Phys 1996;

36(4): 873-9.

386. Flickinger JC, Kano H, Niranjan A, Kondziolka D, Lunsford LD. Dose selection in

stereotactic radiosurgery. Prog Neurol Surg 2013; 27:49-57.(doi): 10.1159/000341623. Epub 2012

Dec 11.

387. Wegner RE, Oysul K, Pollock BE, et al. A modified radiosurgery-based arteriovenous

malformation grading scale and its correlation with outcomes. Int J Radiat Oncol Biol Phys 2011;

79(4): 1147-50. doi: 10.016/j.ijrobp.2009.12.056. Epub 10 Jun 3.

388. Flickinger JC, Kondziolka D, Lunsford LD, et al. Development of a model to predict

permanent symptomatic postradiosurgery injury for arteriovenous malformation patients.

Arteriovenous Malformation Radiosurgery Study Group. Int J Radiat Oncol Biol Phys 2000; 46(5):

1143-8.

389. Yen CP, Jain S, Haq IU, et al. Repeat gamma knife surgery for incompletely

obliterated cerebral arteriovenous malformations. Neurosurgery 2010; 67(1): 55-64; discussion .

390. Gabriel EM, Sampson JH, Wilkins RH. Recurrence of a cerebral arteriovenous

malformation after surgical excision. Case report. J Neurosurg 1996; 84(5): 879-82.

391. Freudenstein D, Duffner F, Ernemann U, Rachinger J, Grote E. Recurrence of a

cerebral arteriovenous malformation after surgical excision. Cerebrovasc Dis 2001; 11(1): 59-64.

Page 158: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

158

392. Inoue HK. Long-term results of Gamma Knife surgery for arteriovenous

malformations: 10- to 15-year follow up in patients treated with lower doses. J Neurosurg 2006;

105(Suppl): 64-8.

393. Potts MB, Sheth SA, Louie J, et al. Stereotactic radiosurgery at a low marginal dose

for the treatment of pediatric arteriovenous malformations: obliteration, complications, and

functional outcomes. J Neurosurg Pediatr 2014; 14(1): 1-11. doi: 0.3171/2014.3.PEDS13381. Epub

2014 Apr 25.

394. Dinca EB, de Lacy P, Yianni J, et al. Gamma knife surgery for pediatric arteriovenous

malformations: a 25-year retrospective study. J Neurosurg Pediatr 2012; 10(5): 445-50. doi:

10.3171/2012.8.PEDS1241. Epub 2012 Aug 31.

395. Nagy G, Rowe JG, Radatz MW, Hodgson TJ, Coley SC, Kemeny AA. A historical analysis

of single-stage gamma knife radiosurgical treatment for large arteriovenous malformations:

evolution and outcomes. Acta Neurochir (Wien) 2012; 154(3): 383-94. doi: 10.1007/s00701-011-

1245-5. Epub 2011 Dec 16.

396. Liscak R, Vladyka V, Simonova G, et al. Arteriovenous malformations after Leksell

gamma knife radiosurgery: rate of obliteration and complications. Neurosurgery 2007; 60(6):

1005-14; discussion 15-6.

397. Pollock BE, Gorman DA, Coffey RJ. Patient outcomes after arteriovenous

malformation radiosurgical management: results based on a 5- to 14-year follow-up study.

Neurosurgery 2003; 52(6): 1291-6; discussion 6-7.

398. Douglas JG, Goodkin R. Treatment of arteriovenous malformations using Gamma

Knife surgery: the experience at the University of Washington from 2000 to 2005. J Neurosurg

2008; 109(Suppl): 51-6. doi: 10.3171/JNS/2008/109/12/S9.

399. Paul L, Casasco A, Kusak ME, Martinez N, Rey G, Martinez R. Results for a series of

697 arteriovenous malformations treated by gamma knife: influence of angiographic features on

the obliteration rate. Neurosurgery 2014; 75(5): 568-83; dicussion 82-3; quiz 83. doi:

10.1227/NEU.0000000000000506.

400. Dalyai R, Starke RM, Chalouhi N, et al. Smoking is a negative predictor of

arteriovenous malformation posttreatment obliteration: analysis of vascular risk factors in 774

patients. Neurosurg Focus 2014; 37(3): E3. doi: 10.3171/20145.FOCUS14121.

Page 159: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

159

401. Koltz MT, Polifka AJ, Saltos A, et al. Long-term outcome of Gamma Knife stereotactic

radiosurgery for arteriovenous malformations graded by the Spetzler-Martin classification. J

Neurosurg 2013; 118(1): 74-83. doi: 10.3171/2012.9.JNS112329. Epub 2012 Oct 19.

402. Starke RM, Yen CP, Ding D, Sheehan JP. A practical grading scale for predicting

outcome after radiosurgery for arteriovenous malformations: analysis of 1012 treated patients. J

Neurosurg 2013; 119(4): 981-7. doi: 10.3171/2013.5.JNS1311. Epub 2013 Jul 5.

403. Flickinger JC, Kondziolka D, Pollock BE, Maitz AH, Lunsford LD. Complications from

arteriovenous malformation radiosurgery: multivariate analysis and risk modeling. Int J Radiat

Oncol Biol Phys 1997; 38(3): 485-90.

404. Hayhurst C, Monsalves E, van Prooijen M, et al. Pretreatment predictors of adverse

radiation effects after radiosurgery for arteriovenous malformation. Int J Radiat Oncol Biol Phys

2012; 82(2): 803-8. doi: 10.1016/j.ijrobp.2010.12.014. Epub 1 Feb 23.

405. Yen CP, Matsumoto JA, Wintermark M, et al. Radiation-induced imaging changes

following Gamma Knife surgery for cerebral arteriovenous malformations. J Neurosurg 2013;

118(1): 63-73. doi: 10.3171/2012.10.JNS12402. Epub 2012 Nov 9.

406. Quigg M, Yen CP, Chatman M, et al. Risks of history of diabetes mellitus,

hypertension, and other factors related to radiation-induced changes following Gamma Knife

surgery for cerebral arteriovenous malformations. J Neurosurg 2012; 117(Suppl): 144-9. doi:

10.3171/2012.6.GKS1245.

407. Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arteriovenous

malformations, part 2: management of pediatric patients. J Neurosurg Pediatr 2012; 9(1): 1-10.

doi: .3171/2011.9.PEDS10458.

408. Yen CP, Monteith SJ, Nguyen JH, Rainey J, Schlesinger DJ, Sheehan JP. Gamma Knife

surgery for arteriovenous malformations in children. J Neurosurg Pediatr 2010; 6(5): 426-34. doi:

10.3171/2010.8.PEDS10138.

409. Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arteriovenous

malformations, Part 4: management of basal ganglia and thalamus arteriovenous malformations. J

Neurosurg 2012; 116(1): 33-43. doi: 10.3171/2011.9.JNS11175. Epub 2011 Nov 11.

410. Kano H, Flickinger JC, Yang HC, et al. Stereotactic radiosurgery for Spetzler-Martin

Grade III arteriovenous malformations. J Neurosurg 2014; 120(4): 973-81. doi:

10.3171/2013.12.JNS131600. Epub 2014 Jan 31.

Page 160: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

160

411. Yamamoto M, Kawabe T, Barfod BE. Long-term side effects of radiosurgery for

arteriovenous malformations. Prog Neurol Surg 2013; 27:97-106.(doi): 10.1159/000341771. Epub

2012 Dec 11.

412. Izawa M, Hayashi M, Chernov M, et al. Long-term complications after gamma knife

surgery for arteriovenous malformations. J Neurosurg 2005; 102(Suppl): 34-7.

413. Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arteriovenous

malformations, Part 5: management of brainstem arteriovenous malformations. J Neurosurg 2012;

116(1): 44-53. doi: 10.3171/2011.9.JNS11176. Epub 2011 Nov 11.

414. Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arteriovenous

malformations, Part 6: multistaged volumetric management of large arteriovenous malformations.

J Neurosurg 2012; 116(1): 54-65. doi: 10.3171/2011.9.JNS11177. Epub 2011 Nov 11.

415. Kano H, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for arteriovenous

malformations, Part 3: outcome predictors and risks after repeat radiosurgery. J Neurosurg 2012;

116(1): 21-32. doi: 10.3171/2011.9.JNS101741. Epub 2011 Nov 11.

416. Ding D, Yen CP, Starke RM, Xu Z, Sun X, Sheehan JP. Radiosurgery for Spetzler-Martin

Grade III arteriovenous malformations. J Neurosurg 2014; 120(4): 959-69. doi:

10.3171/2013.12.JNS131041. Epub 2014 Jan 24.

417. Niranjan A, Lunsford LD. Stereotactic radiosurgery guidelines for the management of

patients with intracranial dural arteriovenous fistulas. Prog Neurol Surg 2013; 27:218-26.(doi):

10.1159/000341799. Epub 2012 Dec 11.

418. Yang H, Kano H, Kondziolka D, et al. Stereotactic radiosurgery with or without

embolization for intracranial dural arteriovenous fistulas. Prog Neurol Surg 2013; 27:195-204.(doi):

10.1159/000341796. Epub 2012 Dec 11.

419. Hanakita S, Koga T, Shin M, Shojima M, Igaki H, Saito N. Role of Gamma Knife surgery

in the treatment of intracranial dural arteriovenous fistulas. J Neurosurg 2012; 117(Suppl): 158-63.

doi: 10.3171/2012.7.GKS12967.

420. Soderman M, Dodoo E, Karlsson B. Dural arteriovenous fistulas and the role of

gamma knife stereotactic radiosurgery: the Stockholm experience. Prog Neurol Surg 2013; 27:205-

17.(doi): 10.1159/000341798. Epub 2012 Dec 11.

421. Pan DH, Wu HM, Kuo YH, Chung WY, Lee CC, Guo WY. Intracranial dural

arteriovenous fistulas: natural history and rationale for treatment with stereotactic radiosurgery.

Prog Neurol Surg 2013; 27:176-94.(doi): 10.1159/000341793. Epub 2012 Dec 11.

Page 161: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

161

422. Yang SY, Kim DG, Chung HT, Paek SH. Radiosurgery for unruptured cerebral

arteriovenous malformations: long-term seizure outcome. Neurology 2012; 78(17): 1292-8. doi:

10.12/WNL.0b013e31825182c5. Epub 2012 Mar 28.

423. Lim YJ, Lee CY, Koh JS, Kim TS, Kim GK, Rhee BA. Seizure control of Gamma Knife

radiosurgery for non-hemorrhagic arteriovenous malformations. Acta Neurochir Suppl 2006; 99:

97-101.

424. Pan DH, Kuo YH, Guo WY, et al. Gamma Knife surgery for cerebral arteriovenous

malformations in children: a 13-year experience. J Neurosurg Pediatr 2008; 1(4): 296-304. doi:

10.3171/PED/2008/1/4/296.

425. Borcek AO, Emmez H, Akkan KM, et al. Gamma Knife radiosurgery for arteriovenous

malformations in pediatric patients. Childs Nerv Syst 2014; 30(9): 1485-92. doi: 10.007/s00381-

014-2469-5. Epub 2014 Jun 28.

426. Tamura N, Hayashi M, Chernov M, et al. Outcome after Gamma Knife surgery for

intracranial arteriovenous malformations in children. J Neurosurg 2012; 117(Suppl): 150-7. doi:

10.3171/2012.7.GKS12998.

427. Ding D, Xu Z, Yen CP, Starke RM, Sheehan JP. Radiosurgery for unruptured cerebral

arteriovenous malformations in pediatric patients. Acta Neurochir (Wien) 2015; 157(2): 281-91.

doi: 10.1007/s00701-014-2305-4. Epub 2014 Dec 17.

428. Nagy G, Major O, Rowe JG, et al. Stereotactic radiosurgery for arteriovenous

malformations located in deep critical regions. Neurosurgery 2012; 70(6): 1458-69; discussion 69-

71. doi: 10.227/NEU.0b013e318246a4d0.

429. Kiran NA, Kale SS, Kasliwal MK, et al. Gamma knife radiosurgery for arteriovenous

malformations of basal ganglia, thalamus and brainstem--a retrospective study comparing the

results with that for AVMs at other intracranial locations. Acta Neurochir (Wien) 2009; 151(12):

1575-82. doi: 10.007/s00701-009-0335-0. Epub 2009 May 5.

430. Kano H, Kondziolka D, Flickinger JC, et al. Multistaged volumetric management of

large arteriovenous malformations. Prog Neurol Surg 2013; 27:73-80.(doi): 10.1159/000341629.

Epub 2012 Dec 11.

431. Yamamoto M, Akabane A, Matsumaru Y, Higuchi Y, Kasuya H, Urakawa Y. Long-term

follow-up results of intentional 2-stage Gamma Knife surgery with an interval of at least 3 years for

arteriovenous malformations larger than 10 cm(3). J Neurosurg 2012; 117(Suppl): 126-34. doi:

10.3171/2012.6.GKS12757.

Page 162: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

162

432. Huang PP, Rush SC, Donahue B, et al. Long-term outcomes after staged-volume

stereotactic radiosurgery for large arteriovenous malformations. Neurosurgery 2012; 71(3): 632-

43; discussion 43-4. doi: 10.1227/NEU.0b013e31825fd247.

433. Sirin S, Kondziolka D, Niranjan A, Flickinger JC, Maitz AH, Lunsford LD. Prospective

staged volume radiosurgery for large arteriovenous malformations: indications and outcomes in

otherwise untreatable patients. Neurosurgery 2008; 62(Suppl 2): 744-54. doi:

10.1227/01.neu.0000316278.14748.87.

434. Jourdain VA, Schechtmann G. Health economics and surgical treatment for

Parkinson's disease in a world perspective: results from an international survey. Stereotact Funct

Neurosurg 2014; 92(2): 71-9. doi: 10.1159/000355215. Epub 2014 Jan 30.

435. Elaimy AL, Arthurs BJ, Lamoreaux WT, et al. Gamma knife radiosurgery for movement

disorders: a concise review of the literature. World J Surg Oncol 2010; 8:61.(doi): 10.1186/477-

7819-8-61.

436. Kooshkabadi A, Lunsford LD, Tonetti D, Flickinger JC, Kondziolka D. Gamma Knife

thalamotomy for tremor in the magnetic resonance imaging era. J Neurosurg 2013; 118(4): 713-8.

doi: 10.3171/2013.1.JNS121111. Epub 2013 Feb 1.

437. Cho KR, Kim HR, Im YS, Youn J, Cho JW, Lee JI. Outcome of gamma knife

thalamotomy in patients with an intractable tremor. J Korean Neurosurg Soc 2015; 57(3): 192-6.

doi: 10.3340/jkns.2015.57.3.192. Epub Mar 20.

438. Ohye C, Higuchi Y, Shibazaki T, et al. Gamma knife thalamotomy for Parkinson

disease and essential tremor: a prospective multicenter study. Neurosurgery 2012; 70(3): 526-35;

discussion 35-6. doi: 10.1227/NEU.0b013e3182350893.

439. Lim SY, Hodaie M, Fallis M, Poon YY, Mazzella F, Moro E. Gamma knife thalamotomy

for disabling tremor: a blinded evaluation. Arch Neurol 2010; 67(5): 584-8. doi:

10.1001/archneurol.2010.69.

440. Young RF, Li F, Vermeulen S, Meier R. Gamma Knife thalamotomy for treatment of

essential tremor: long-term results. J Neurosurg 2010; 112(6): 1311-7. doi:

10.3171/2009.10.JNS09332.

441. Kondziolka D, Ong JG, Lee JY, Moore RY, Flickinger JC, Lunsford LD. Gamma Knife

thalamotomy for essential tremor. J Neurosurg 2008; 108(1): 111-7. doi:

10.3171/JNS/2008/108/01/0111.

Page 163: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

163

442. Ohye C, Shibazaki T, Sato S. Gamma knife thalamotomy for movement disorders:

evaluation of the thalamic lesion and clinical results. J Neurosurg 2005; 102(Suppl): 234-40.

443. Duma CM, Jacques DB, Kopyov OV, Mark RJ, Copcutt B, Farokhi HK. Gamma knife

radiosurgery for thalamotomy in parkinsonian tremor: a five-year experience. J Neurosurg 1998;

88(6): 1044-9.

444. Niranjan A, Jawahar A, Kondziolka D, Lunsford LD. A comparison of surgical

approaches for the management of tremor: radiofrequency thalamotomy, gamma knife

thalamotomy and thalamic stimulation. Stereotact Funct Neurosurg 1999; 72(2-4): 178-84.

445. Ohye C. From selective thalamotomy with microrecording to gamma thalamotomy

for movement disorders. Stereotact Funct Neurosurg 2006; 84(4): 155-61. Epub 2006 Aug 10.

446. Mathieu D, Kondziolka D, Niranjan A, Flickinger J, Lunsford LD. Gamma knife

thalamotomy for multiple sclerosis tremor. Surg Neurol 2007; 68(4): 394-9.

447. Sato S, Ohye C, Shibazaki T, Zama A, Cai X. Neurophysiological evaluation of the

optimum target in gamma thalamotomy: indirect evidence. Stereotact Funct Neurosurg 2005;

83(2-3): 108-12; discussion 13-4. Epub 2005 Aug 3.

448. Yen CP, Schlesinger D, Sheehan JP. Gamma Knife(R) radiosurgery for trigeminal

neuralgia. Expert Rev Med Devices 2011; 8(6): 709-21. doi: 10.1586/erd.11.46.

449. Kondziolka D, Lunsford LD, Flickinger JC. Stereotactic radiosurgery for the treatment

of trigeminal neuralgia. Clin J Pain 2002; 18(1): 42-7.

450. Flickinger JC, Pollock BE, Kondziolka D, et al. Does increased nerve length within the

treatment volume improve trigeminal neuralgia radiosurgery? A prospective double-blind,

randomized study. Int J Radiat Oncol Biol Phys 2001; 51(2): 449-54.

451. Arai Y, Kano H, Lunsford LD, et al. Does the Gamma Knife dose rate affect outcomes

in radiosurgery for trigeminal neuralgia? J Neurosurg 2010; 113(Suppl): 168-71.

452. Flickinger JC, Jr., Kim H, Kano H, et al. Do carbamazepine, gabapentin, or other

anticonvulsants exert sufficient radioprotective effects to alter responses from trigeminal

neuralgia radiosurgery? Int J Radiat Oncol Biol Phys 2012; 83(4): e501-6. doi:

10.1016/j.ijrobp.2012.01.016. Epub Mar 13.

453. Tuleasca C, Carron R, Resseguier N, et al. Patterns of pain-free response in 497 cases

of classic trigeminal neuralgia treated with Gamma Knife surgery and followed up for least 1 year.

J Neurosurg 2012; 117(Suppl): 181-8. doi: 10.3171/2012.8.GKS121015.

Page 164: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

164

454. Baschnagel AM, Cartier JL, Dreyer J, et al. Trigeminal neuralgia pain relief after

gamma knife stereotactic radiosurgery. Clin Neurol Neurosurg 2014; 117:107-11.(doi):

10.1016/j.clineuro.2013.12.003. Epub Dec 15.

455. Lucas JT, Jr., Nida AM, Isom S, et al. Predictive nomogram for the durability of pain

relief from gamma knife radiation surgery in the treatment of trigeminal neuralgia. Int J Radiat

Oncol Biol Phys 2014; 89(1): 120-6. doi: 10.1016/j.ijrobp.2014.01.023. Epub Mar 7.

456. Young B, Shivazad A, Kryscio RJ, St Clair W, Bush HM. Long-term outcome of high-

dose gamma knife surgery in treatment of trigeminal neuralgia. J Neurosurg 2013; 119(5): 1166-

75. doi: 10.3171/2013.1.JNS12875. Epub 2013 Apr 19.

457. Elaimy AL, Lamm AF, Demakas JJ, et al. Gamma knife radiosurgery for typical

trigeminal neuralgia: An institutional review of 108 patients. Surg Neurol Int 2013; 4:92.(doi):

10.4103/2152-7806.115163. eCollection 2013.

458. Hodaie M, Chen DQ, Quan J, Laperriere N. Tractography delineates microstructural

changes in the trigeminal nerve after focal radiosurgery for trigeminal neuralgia. PLoS One 2012;

7(3): e32745. doi: 10.1371/journal.pone.0032745. Epub 2012 Mar 6.

459. Li P, Wang W, Liu Y, Zhong Q, Mao B. Clinical outcomes of 114 patients who

underwent gamma-knife radiosurgery for medically refractory idiopathic trigeminal neuralgia. J

Clin Neurosci 2012; 19(1): 71-4. doi: 10.1016/j.jocn.2011.03.020. Epub Dec 9.

460. Loescher AR, Radatz M, Kemeny A, Rowe J. Stereotactic radiosurgery for trigeminal

neuralgia: outcomes and complications. Br J Neurosurg 2012; 26(1): 45-52. doi:

10.3109/02688697.2011.591849. Epub 2011 Aug 4.

461. Hayashi M, Chernov M, Tamura N, et al. Stereotactic radiosurgery of essential

trigeminal neuralgia using Leksell Gamma Knife model C with automatic positioning system:

technical nuances and evaluation of outcome in 130 patients with at least 2 years follow-up after

treatment. Neurosurg Rev 2011; 34(4): 497-508. doi: 10.1007/s10143-011-0330-9. Epub 2011 Jun

24.

462. Verheul JB, Hanssens PE, Lie ST, Leenstra S, Piersma H, Beute GN. Gamma Knife

surgery for trigeminal neuralgia: a review of 450 consecutive cases. J Neurosurg 2010; 113(Suppl):

160-7.

463. Kano H, Kondziolka D, Yang HC, et al. Outcome predictors after gamma knife

radiosurgery for recurrent trigeminal neuralgia. Neurosurgery 2010; 67(6): 1637-44; discussion 44-

5. doi: 10.227/NEU.0b013e3181fa098a.

Page 165: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

165

464. Riesenburger RI, Hwang SW, Schirmer CM, et al. Outcomes following single-

treatment Gamma Knife surgery for trigeminal neuralgia with a minimum 3-year follow-up. J

Neurosurg 2010; 112(4): 766-71. doi: 10.3171/2009.8.JNS081706.

465. Regis J, Arkha Y, Yomo S, et al. [Radiosurgery in trigeminal neuralgia: long-term

results and influence of operative nuances]. Neurochirurgie 2009; 55(2): 213-22. doi:

10.1016/j.neuchi.2009.01.020. Epub Apr 1.

466. Knafo H, Kenny B, Mathieu D. Trigeminal neuralgia: outcomes after gamma knife

radiosurgery. Can J Neurol Sci 2009; 36(1): 78-82.

467. Little AS, Shetter AG, Shetter ME, Bay C, Rogers CL. Long-term pain response and

quality of life in patients with typical trigeminal neuralgia treated with gamma knife stereotactic

radiosurgery. Neurosurgery 2008; 63(5): 915-23; discussion 23-4. doi:

10.1227/01.NEU.0000327689.05823.28.

468. Longhi M, Rizzo P, Nicolato A, Foroni R, Reggio M, Gerosa M. Gamma knife

radiosurgery for trigeminal neuralgia: results and potentially predictive parameters--part I:

Idiopathic trigeminal neuralgia. Neurosurgery 2007; 61(6): 1254-60; discussion 60-1.

469. Sheehan J, Pan HC, Stroila M, Steiner L. Gamma knife surgery for trigeminal

neuralgia: outcomes and prognostic factors. J Neurosurg 2005; 102(3): 434-41.

470. Tuleasca C, Carron R, Resseguier N, et al. Multiple sclerosis-related trigeminal

neuralgia: a prospective series of 43 patients treated with gamma knife surgery with more than

one year of follow-up. Stereotact Funct Neurosurg 2014; 92(4): 203-10. doi: 10.1159/000362173.

Epub 2014 Jul 8.

471. Mathieu D, Effendi K, Blanchard J, Seguin M. Comparative study of Gamma Knife

surgery and percutaneous retrogasserian glycerol rhizotomy for trigeminal neuralgia in patients

with multiple sclerosis. J Neurosurg 2012; 117(Suppl): 175-80. doi: 10.3171/2012.6.GKS12987.

472. Kondziolka D, Lunsford LD, Flickinger JC, et al. Stereotactic radiosurgery for

trigeminal neuralgia: a multiinstitutional study using the gamma unit. J Neurosurg 1996; 84(6):

940-5.

473. Pollock BE, Phuong LK, Foote RL, Stafford SL, Gorman DA. High-dose trigeminal

neuralgia radiosurgery associated with increased risk of trigeminal nerve dysfunction.

Neurosurgery 2001; 49(1): 58-62; discussion -4.

Page 166: Clinical Indications for Gamma Knife Radiosurgery A Review ...beating-gbm.org/wp-content/uploads/2016/01/Lippitz... · indication Gamma Knife radiosurgery is generally used as a complement

166

474. Balamucki CJ, Stieber VW, Ellis TL, et al. Does dose rate affect efficacy? The outcomes

of 256 gamma knife surgery procedures for trigeminal neuralgia and other types of facial pain as

they relate to the half-life of cobalt. J Neurosurg 2006; 105(5): 730-5.

475. Regis J, Metellus P, Hayashi M, Roussel P, Donnet A, Bille-Turc F. Prospective

controlled trial of gamma knife surgery for essential trigeminal neuralgia. J Neurosurg 2006;

104(6): 913-24.

476. Matsuda S, Serizawa T, Nagano O, Ono J. Comparison of the results of 2 targeting

methods in Gamma Knife surgery for trigeminal neuralgia. J Neurosurg 2008; 109(Suppl): 185-9.

doi: 10.3171/JNS/2008/109/12/S28.

477. Massager N, Abeloos L, Devriendt D, Op de Beeck M, Levivier M. Clinical evaluation

of targeting accuracy of gamma knife radiosurgery in trigeminal neuralgia. Int J Radiat Oncol Biol

Phys 2007; 69(5): 1514-20. Epub 2007 Aug 8.

478. Park KJ, Kondziolka D, Berkowitz O, et al. Repeat gamma knife radiosurgery for

trigeminal neuralgia. Neurosurgery 2012; 70(2): 295-305; discussion . doi:

10.1227/NEU.0b013e318230218e.

479. Tempel ZJ, Chivukula S, Monaco EA, 3rd, et al. The results of a third Gamma Knife

procedure for recurrent trigeminal neuralgia. J Neurosurg 2015; 122(1): 169-79. doi:

10.3171/2014.9.JNS132779.

480. Oh IH, Choi SK, Park BJ, Kim TS, Rhee BA, Lim YJ. The Treatment Outcome of Elderly

Patients with Idiopathic Trigeminal Neuralgia : Micro-Vascular Decompression versus Gamma

Knife Radiosurgery. J Korean Neurosurg Soc 2008; 44(4): 199-204. doi:

10.3340/jkns.2008.44.4.199. Epub Oct 30.

481. Linskey ME, Ratanatharathorn V, Penagaricano J. A prospective cohort study of

microvascular decompression and Gamma Knife surgery in patients with trigeminal neuralgia. J

Neurosurg 2008; 109(Suppl): 160-72. doi: 10.3171/JNS/2008/109/12/S25.

482. Shetter AG, Zabramski JM, Speiser BL. Microvascular decompression after gamma

knife surgery for trigeminal neuralgia: intraoperative findings and treatment outcomes. J

Neurosurg 2005; 102(Suppl): 259-61.