Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

www.redjournal.

org

HyTEC: Organ-Specific Paper

Single- and Multifraction Stereotactic


Radiosurgery Dose/Volume Tolerances of the
Brain
Michael T. Milano, MD, PhD,* Jimm Grimm, PhD,y
Andrzej Niemierko, PhD,z Scott G. Soltys, MD,x Vitali Moiseenko, PhD,k
Kristin J. Redmond, MD,{ Ellen Yorke, PhD,# Arjun Sahgal, MD,**
Jinyu Xue, PhD,yy Anand Mahadevan, MD,y Alexander Muacevic, MD,zz
Lawrence B. Marks, MD,xx and Lawrence R. Kleinberg, MD{
*Department of Radiation Oncology, University of Rochester, Rochester, New York; yDepartment of
Radiation Oncology, Geisinger Cancer Institute, Danville, Pennsylvania; zDepartment of Radiation
Oncology, Massachusetts General Hospital, Boston, Massachusetts; xDepartment of Radiation
Oncology, Stanford University Medical Center, Stanford, California; kDepartment of Radiation
Medicine and Applied Sciences, University of California San Diego, La Jolla, California; {Department
of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of
Medicine, Baltimore, Maryland; #Department of Medical Physics, Memorial SloaneKettering Cancer
Center, New York City, New York; **Department of Radiation Oncology, Odette Cancer Centre,
Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada; yyDepartment of
Radiation Oncology, NYU Langone Medical Center, New York City, NY; zzEuropean Cyberknife Center
Munich-Großhadern, Munich, Germany; and xxDepartment of Radiation Oncology and Lineberger
Cancer Center, University of North Carolina, Chapel Hill, North Carolina

Received Aug 3, 2020. Accepted for publication Aug 3, 2020.

Summary Purpose: As part of the American Association of Physicists in Medicine Working


Pooled published data reveal Group on Stereotactic Body Radiotherapy investigating normal tissue complication
that radiation-induced brain probability (NTCP) after hypofractionated radiation therapy, data from published

Corresponding author: Michael T. Milano, MD, PhD; E-mail: michael_ faculty), Brainlab, and Medtronic Kyphon; researched a research grant
milano@urmc.rochester.edu with Elekta AB; received travel accommodations/expenses from Elekta,
Disclosures: M.T.M. reports royalties from UpToDate and Galera Varian, and Brainlab; and belongs to the Elekta MR Linac Research
Therapeutics, outside the submitted work. J.G. reports grants from Accu- Consortium, Elekta Spine, Oligometastases, and LINAC Based SRS
ray and NovoCure, outside the submitted work, and has a patent DVH Consortia. A.M. reports personal fees from Varian Inc and nonfinancial
Evaluator issued. S.G.S. reports personal fees from Inovio Pharmaceuticals support from Accuray Inc, outside the submitted work. A.M. receives
Inc and Zap Surgical Inc and grants from Novocure, outside the submitted speaker honoraria from Accuray Inc and is former president and chairman
work. K.J.R. reports grants and other from Elekta AB, grants and other of the Radiosurgery Society, therss.org. L.R.K. reports grants, personal
from Accuray, personal fees from BioMimetix, and other from Brainlab, fees, and other from Novocure; grants and personal fees from Accuray; and
outside the submitted work. E.Y. was partly supported by NCI Cancer grants from Arbor, outside the submitted work.
Center Support Grant P30CA008748. A.S. has been an advisor/consultant Data sharing: Research data are stored in an institutional repository and
with Abbvie, Merck, Roche, Varian (Medical Advisory Group), and Elekta will be shared upon request to the corresponding author.
(Gamma Knife Icon); been an ex officio board member to International Supplementary material for this article can be found at https://doi.org/
Stereotactic Radiosurgery Society (ISRS); received honorarium for past 10.1016/j.ijrobp.2020.08.013.
educational seminars with Elekta AB, Accuray Inc, Varian (CNS teaching

Int J Radiation Oncol Biol Phys, Vol. -, No. -, pp. 1e19, 2020
0360-3016/$ - see front matter Ó 2020 Elsevier Inc. All rights reserved.
https://doi.org/10.1016/j.ijrobp.2020.08.013
2 Milano et al. International Journal of Radiation Oncology  Biology  Physics

toxicity after single-fraction reports (PubMed indexed 1995-2018) were pooled to identify dosimetric and clinical
stereotactic radiosurgery for predictors of radiation-induced brain toxicity after single-fraction stereotactic radio-
brain metastases and arterio- surgery (SRS) or fractionated stereotactic radiosurgery (fSRS).
venous malformations is low Methods and Materials: Eligible studies provided NTCPs for the endpoints of radio-
when the volume of tissue necrosis, edema, or symptoms after cranial SRS/fSRS and quantitative dose-volume
receiving 12 Gy is less than 5 metrics. Studies of patients with only glioma, meningioma, vestibular schwannoma,
cm3 but increases with larger or brainstem targets were excluded. The data summary and analyses focused on arte-
treatment volumes. Frac- riovenous malformations (AVM) and brain metastases.
tionated stereotactic radio- Results: Data from 51 reports are summarized. There was wide variability in reported
surgery can reduce toxicity rates of radionecrosis. Available data for SRS/fSRS for brain metastases were more
risk for larger targets. Stan- amenable to NTCP modeling than AVM data. In the setting of brain metastases,
dardized dosimetric and SRS/fSRS-associated radionecrosis can be difficult to differentiate from tumor pro-
toxicity reporting would gression. For single-fraction SRS to brain metastases, tissue volumes (including target
facilitate future pooled ana- volumes) receiving 12 Gy (V1) of 5 cm3, 10 cm3, or >15 cm3 were associated with
lyses and predict brain risks of symptomatic radionecrosis of approximately 10%, 15%, and 20%, respec-
toxicity risks more precisely. tively. SRS for AVM was associated with modestly lower rates of symptomatic radio-
necrosis for equivalent V12. For 3-fraction fSRS for brain metastases, normal brain
tissue V18 <30 cm3 and V23 <7 cm3 were associated with <10% risk of radionecro-
sis.
Conclusions: The risk of radionecrosis after SRS and fSRS can be modeled as a func-
tion of dose and volume treated. The use of fSRS appears to reduce risks of radione-
crosis for larger treatment volumes relative to SRS. More standardized dosimetric and
toxicity reporting is needed to facilitate future pooled analyses that can refine predic-
tive models of brain toxicity risks. Ó 2020 Elsevier Inc. All rights reserved.

1. Clinical Significance Asymptomatic necrosis can be managed with clinical


and radiographic surveillance. For symptomatic necrosis,
Single-fraction radiosurgery (SRS) or hypofractionated (2- corticosteroids are considered first-line therapy,4,5 with the
dose, schedule, and duration of corticosteroids dependent
5 fraction) cranial radiosurgery (fSRS) is commonly used
on the response of symptoms and possible complications of
for various primary and metastatic brain tumors. Radiation-
their treatment (eg, myopathy with motor weakness,
induced necrosis (radionecrosis), edema, and other neuro-
osteopenia, hypertension, extremity edema, obesity with
logic complications are relatively common. Radiation
accumulation of subcutaneous fat, and glucose intoler-
necrosis represents inflammation from tissue death and
ance).4 Emerging data have shown favorable radiographic
breakdown and is generally associated with surrounding
and clinical responses of brain radionecrosis to bev-
edema, which can be either symptomatic or asymptomatic.
Terminology in the literature varies and includes necrosis, acizumab6,7; in addition, bevacizumab may reduce the need
for corticosteroids. A small (n Z 14) randomized double-
radionecrosis, radiation treatment effect, treatment-related
blind, placebo-controlled study, allowing for crossover to
imaging changes, and adverse radiation effect (ARE;
the treatment arm, provided compelling evidence of the
including reactive inflammation, vasculitis, and necrosis)1;
efficacy of bevacizumab for brain radionecrosis,8 although
herein, the terms necrosis, radionecrosis, and radiation
bevacizumab was associated with relatively high risks (in 3
necrosis are used (unless specifying the outcome used in a
of 11 patients) of severe toxicity (aspiration pneumonia,
specific report). In most studies, the onset of necrosis
pulmonary embolus, and superior sagittal sinus throm-
ranges from <6 months to several years after SRS/fSRS,
with median time to onset of roughly a year. bosis), warranting caution before considering this therapy.
An Alliance study planned to randomize 130 patients
Symptomatic necrosis can result from direct brain injury
(currently closed after accrual of 18) with symptomatic
or from necrosis within the target that elicits an inflam-
post-SRS brain radionecrosis to corticosteroids with or
matory cascade,1 which can cause mass effect and/or sur-
without bevacizumab; the primary endpoint was patient-
rounding edema with associated compression (and, at the
reported outcome (symptoms) of radionecrosis, and the
extreme, herniation) of normal brain. Necrosis is thought to
study allowed crossover to the treatment arm
be mediated via vascular endothelial injury.1-3 Edema in the
(NCT02490878). Hyperbaric-oxygen therapy may have
absence of necrosis can also occur. Symptoms of edema
and/or necrosis include headache, nausea, vomiting, ataxia, some efficacy against brain radionecrosis, although this
treatment remains investigational.4,5 Radionecrosis that is
seizure, and focal siteedependent functional deficits.
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 3

resistant to medical management can be considered for Table 1 Scoring systems for brain toxicity (not specific to
surgical resection or laser-interstitial thermal therapy with necrosis)
biopsy, which are often considered when persistent/recur-
RTOG/EORTC
rent tumor is in the differential diagnosis. acute morbidity score
(from 1995)
Endpoints Grade 0 None
Grade 1 Fully functional status (ie,
able to work) with minor
The common brain toxicity endpoints reported in the
neurologic findings; no
literature include necrosis, edema, and neurologic medication needed
compromise. Radionecrosis is often evident as a ring- Grade 2 Neurologic findings present
enhancing lesion, commonly with associated edema. sufficient to require home
For tumors, SRS/fSRS-associated radionecrosis can be care; nursing assistance
difficult to differentiate from tumor progression or pseu- may be required;
doprogression. Necrosis can be symptomatic or asymp- medications including
tomatic, and the diagnosis can be made based on imaging steroids/antiseizure agents
findings or pathologic confirmation (which may reveal a may be required
mixture of necrosis and viable tumor). Pathologic correlates Grade 3 Neurologic findings requiring
with imaging findings are not commonly reported in studies hospitalization for initial
management
of radionecrosis incidence. Most often the diagnosis is
Grade 4 Serious neurologic
made based solely on imaging findings. Techniques such as impairment that includes
positron emission tomography, magnetic resonance (MR) paralysis, coma, or seizures
cerebral perfusion, delayed-contrast MR imaging, and MR >3 per week despite
spectroscopy may help strengthen the diagnostic cer- medication; hospitalization
tainty.2,3,5,9 The sensitivity and specificity of various im- required
aging modalities used in differentiating between CTCAE version 4
radionecrosis and tumor recurrence were recently summa- (from 2009)
rized.4 Overall, the sensitivity is on the order of 60% to Grade 0 None
80% for most of these modalities in differentiating necrosis Grade 1 Asymptomatic; clinical or
from tumor progression; thus, no single test can reliably diagnostic observations
only; intervention not
distinguish radionecrosis from recurrent tumor, potentially
indicated
requiring a multimodal approach for some patients. Grade 2 Moderate symptoms;
Radiomic strategies, such as changes in MR imaging corticosteroids indicated
features, are also being investigated as a means to distin- Grade 3 Severe symptoms; medical
guish radionecrosis from tumor progression.10-12 The lack intervention indicated
of a consistent means to diagnose necrosis13 complicates Grade 4 Life-threatening
the interpretation of the data. Recognizing this challenge, consequences; urgent
the 2015 Response Assessment in Neuro-Oncology Brain intervention indicated
Metastases (RANO-BM) working group report suggested Abbreviations: CTCAE Z Common Terminology Criteria for
that if lesion growth meets criteria for progression (“at least Adverse Events; EORTC Z European Organization for Research and
a 20% increase in the sum longest diameter of central Treatment of Cancer; RTOG Z Radiation Therapy Oncology Group.
nervous system target lesions” relative to the smaller of
baseline or nadir, sustained for 4 weeks) but clinical ev-
idence suggests treatment effect, additional evidence (not Several examples16-18 are shown in Table 1. A more
specified in the report) is needed to distinguish progression detailed toxicity grading system specifically considered
from treatment effect.14 Thus, it is sometimes challenging symptomatic, objective, management, and analytic
to diagnose necrosis with confidence. The imaging mani- criteria,2,19 and although this approach has clear benefits for
festations in patients on immune therapies appear to be brain toxicity, it has not been widely adopted.
particularly challenging with respect to edema and necrosis The most accurate means of diagnosing radionecrosis is
versus immune response; as a consequence, newer imaging pathologic confirmation. If surgery is required for necrosis,
criteria such as immunotherapy response assessment in historically the necrosis was considered at least grade 3;
neuro-oncology are being implemented.15 however, it is conceivable that some patients with no
Necrosis may cause a variety of symptoms, and toxicity symptoms, or grade 1 to 2 symptoms, could undergo a
grading traditionally has been based on the severity of these planned resection to establish a diagnosis of recurrence
symptoms (rather than the extent/severity of the necrosis versus necrosis, and the pathologic findings could be most
itself, for which there is no widely accepted grading scale). consistent with necrosis. Therefore, an endpoint defined
4 Milano et al. International Journal of Radiation Oncology  Biology  Physics

simply as “pathologic confirmation of necrosis” may 1


Flickinger 1998
include lower grade events, potentially overestimating the .9 Voges 1996
incidence of clinically significant necrosis. Nevertheless, .8

Probability of any necrosis


grade 1 to 2 symptoms typically do not warrant a biopsy or
.7
surgery, and thus the determination of necrosis is less ac-

after SRS for AVM


.6
curate in these patients. The more serious complications,
which more often lead to biopsy or surgery, are based on .5

the more reliable pathologic (vs clinical) assessment of .4


necrosis. .3
.2
3. Challenges Defining Volumes .1
0
Defining the major structures of the central nervous system 0 5 10 15 20 25 30
V12 (cc)
is straightforward, although there is no consensus on how to
delineate subregions of the brain.20 Autocontouring soft- Fig. 1. Risk of any necrosis (symptomatic or asymp-
ware may erroneously include portions of the cavernous tomatic) after single-fraction stereotactic radiosurgery
sinus, venous sinuses, or surgical deficits in the skull (ie, (SRS) for arteriovenous malformations (AVM) versus tis-
postcraniotomy) as normal brain; we recommend excluding sue volume receiving 12 Gy in 1 fraction (V12). Data
these regions, as well as cranial nerves and brainstem, from were extracted from studies that subgrouped (binned)
the brain organ at risk. ranges of V10 to V12 and reported the risks of necrosis for
In published reports on necrosis risks after SRS/fSRS, patients/lesions within the individual bins. The figure rep-
there is variability in what volumes are used to correlate resents a rough approximation of data, as (1) the median
dose-volume exposure with toxicity risks. Specific volumes value of V12 for a given bin was estimated as the midpoint
described (Supplemental Tables 1e5) include (1) all tissue in the range of values in that bin, except for the bin with the
(ie, all of the volume encompassed by isodose line, which greatest V12; for the largest-value bins, it was assumed that
includes target volume), (2) “normal” or “nontarget” tissue, the highest values within that bin represented a relatively
and (3) “surrounding brain” or brain. For “brain” or “sur- small proportion of patients (ie, the extremely large V12
rounding brain” tissue, there is some variability in what values were outliers); (2) for data from the Voges et al22
target volume (ie, gross target, clinical target, or planning study that reported V10, the equivalent to V12 was esti-
target volumes) is subtracted from brain. Such variability mated as described in the text (Eq. 2). The error bars
complicates pooling of data. The differences in calculated represent 95% binomial confidence intervals and the solid
volume exposures between these approaches would depend lines represent a fitted logistic model to the data. Studies
on the size and location of the target, as well as the plan- included Voges 199622 and Flickinger 199823; both were
ning approaches used. As an example, assume a simple single-fraction SRS studies.
scenario of 2 spherical target volumes, surrounded by brain
parenchyma, in which the 12-Gy isodose line is 3 mm from studies in print in 2017 that were published and indexed
the target surface (not accounting for sharper dose-falloffs online in 2016); search criteria are detailed in Appendix E1.
with smaller targets21). For a 0.4-cm target (0.034 cm3), One additional study from 2018 was also included.41 All of
the brain V12 (0.49 cm3) is similar to tissue V12 (0.52 the searches included the word “volume” or “volumes.”
cm3). For a 2.5-cm target (8.2 cm3), brain V12 (7.4 cm3) is Studies included in this review (listed in Table E1) reported
less than half of tissue V12 (15.6 cm3). With a 2.5-cm endpoints of symptomatic and/or asymptomatic necrosis
spherical target (tissue V12 of 14.1 cm3) abutting the and/or edema after SRS/fSRS and correlated toxicity risks
brain surface, with 30% of the 12-Gy volume outside of with target, tissue and/or brain volume or dose-volume
brain, the brain V12 (5.2 cm3) would be about one-third of metrics. We also included studies that used more general-
tissue V12. From the models in Figures 1 to 6, it can be ized endpoints of radiation-induced neurologic (noncranial
seen that in the flat regions a change of 1 cm3 in brain nerve) complications (which generally comprise necrosis
volume has less than a 1% effect on risk, but in the steepest and/or edema). Studies that analyzed only potential effects
dose/volume response regions, a 5% change in risk could of prescribed dose on toxicity risks (ie, no analyses of
result from about 1 cm3 of volume uncertainty. target volume or normal tissue volume exposure) were not
included in the Supplemental Tables or analyses (although
4. Review of Outcomes Data some will be discussed) because the manner in which the
dose is prescribed is highly variable and does not neces-
Published studies sarily reflect normal tissue dose-volume exposure. Target
volumes also do not sufficiently describe normal tissue
PubMed searches were performed for reports published dose-volume exposure, but studies that analyzed target
from January 1995 through December 2016 (including volume without analyzing normal tissue exposure were
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 5

included here because target volume is a reproducible studies included symptomatic brain necrosis (with some
measure. Studies of central nervous system radiotoxicity studies specifying toxicity grade), any necrosis, edema,
after conventionally fractionated or >5-fraction hypo- neurologic complications (without specifying from ne-
fractionated stereotactic radiation therapy were omitted crosis or edema), and others (Table E2). Such variability
(unless a subset of 1-5 fraction SRS/fSRS patients was complicates data pooling.
analyzed separately). A few studies reported only P values for the correlation
We excluded studies reporting toxicity outcomes in a of risks with dosimetric parameters, not the risks within
cohort composed solely of (1) brainstem targets, because subgroups; therefore, data from these studies did not allow
the brainstem appears to have a different dose tolerance binning subsets into groups for modeling. Table E2 also
than normal brain (see section 7); (2) patients with glioma, summarizes the reported timing of the onset of necrosis,
because many gliomas have tumor-related necrosis before which in most studies ranges from <6 months to several
SRS/fSRS; (3) patients with vestibular schwannoma/ years after SRS/fSRS, with median time to onset of roughly
acoustic neuromadbecause these tumors are not within the a year. Actuarial risks are only reported in some studies;
brain parenchyma, intratumoral necrosis after SRS/fSRS is therefore, our graphs that summarize published data (Figs.
fairly common for vestibular schwannoma/acoustic neu- 1-4) and NTCP models (section 6 and Figs. 5 and 6) use
roma, and neurologic radiotoxicity from these tumors is crude toxicity risks.
mostly related to cranial nerve, cochlear, and/or brainstem For the data sets presented in Figures 1 to 4 (in this
injury; and (4) patients with edema after SRS/fSRS for section), with the data binned into subgroups of Vx (ie, by
meningioma, because the etiology of postradiation edema quartiles or median splits), the logarithmic form of the lo-
may be unique for meningiomas.42 Nevertheless, some of gistic model (described in section 6) was used (Equation 1).
the studies included in this review did include a subset of 1
NTCP Z  4g50 ð1Þ
patients with glioma, meningioma, vestibular schwannoma,
or brainstem targets, but generally these represented a small 1þ Vx50
Vx
subset of patients/tumors in these studies (specified in Table
E1), and these patients were not included in the pooled
analyses specific for brain metastases and arteriovenous
1
malformations (AVM). Flickinger 1998 Flickinger 2000 Herbert 2012
.9 Cetin 2012 Kano 2017
Table E1 summarizes 51 published reports, from which
Probability of symptomatic necrosis

9828 patients and 13,913 targets were studied. Some re- .8


ports23,24,43-45,33,46,22,27 included a number of patients who .7
after SRS for AVM

were analyzed in 1 or more earlier reports. With estimation .6


(owing to partial overlap of some patients in multiple .5
studies, as described in Table E1) these reports considered
.4
approximately 7814 unique patients. Most patients were
.3
treated for AVM or brain metastases: An estimated 3848
patients had 3858 AVM, and 3966 patients had 7903 brain .2

metastases. Most of these studies/patients were not .1


amenable to modeling analyses but are included in the 0
discussion and Supplemental Tables because they offered 0 5 10 15 20 25
V12 (cc)
30 35 40 45
insight into NTCP risks.
Some studies included only patients with Fig. 2. Risk of symptomatic necrosis after single fraction
AVM,23,24,22,27,47-52,25,26 others included only patients stereotactic radiosurgery (SRS) for arteriovenous malfor-
with benign and/or malignant tumors,43-45,33,46,53,54,29 mations (AVM) versus tissue volume receiving 12 Gy in 1
,34,55,30,31,32,35,56,57,38,39,36,58-67,28,68,69,68,71,70 fraction (V12). Data were extracted from studies that re-
and 4 others
included patients with tumors or AVM72,73,40,74 (of which 3 ported necrosis risks and median V12 among all patients or
analyzed the different disease groups separately). In 10 subgrouped (binned) ranges of V12 and reported the risks of
studies, some46,56 or all45,35,57,59,66,69,71,70 of the patients necrosis for patients/lesions within the individual bins. The
underwent SRS/fSRS to the surgical cavity of a resected figure represents a rough approximation of data, as the me-
brain metastasis. dian value of V12 for a given bin was estimated as described
Various endpoints (Table E2) and dosimetric measures in the caption from Figure 1. The error bars represent 95%
(Tables E3-5) were used to characterize toxicity risks. For binomial confidence intervals and the solid lines represent a
example, where Vx reflects the volume receiving greater fitted logistic model to the data. Studies included Flickinger
than or equal to a dose of x Gy, several studies analyzed 1998,23 Flickinger 2000,24 Cetin 2012,25 Herbert 2012,26 and
tissue Vx (ie, volume of x Gy isodose line), others Kano 201727; all were single-fraction SRS studies. The data
analyzed normal tissue Vx (ie, excluding target volume), from Herbert et al were obtained from the authors. Three of
and others analyzed brain Vx (ie, excluding target volume the 5 studies included patients from 1 institution (University
and nonbrain normal tissue). Endpoints in different of Pittsburgh).
6 Milano et al. International Journal of Radiation Oncology  Biology  Physics

A 1 Minniti 2016 Blonigen 2010 Minniti 2011


B 1
Minniti 2016 Dore 2017 Ernst-Stecken 2006
Korytko 2006 Ohtakara 2012 Minniti 2013 Minniti 2014

Probability of any necrosis after fSRS


.9 Sneed 2015 .9
after SRS for brain metastases

.8 Single fraction .8 Multi-fraction


Probability of any necrosis

for brain metastases


.7 .7

.6 .6

.5 .5

.4 .4

.3 .3

.2 .2

.1 .1

0 0
0 5 10 15 20 25 30 0 5 10 15 20 25 30 35 40 45 50
V12 (cc) V12 (cc)

Fig. 3. Risk of any necrosis (symptomatic or asymptomatic) after single-fraction stereotactic radiosurgery (SRS) for brain
metastases versus tissue (target plus nontarget) or brain (excluding the target volume and nonbrain normal tissue) volume
receiving 12 Gy equivalent in 1 fraction (V12). (A) Data from single-fraction SRS studies; solid data points represent tissue
V12 (target plus nontarget) and hollow data points represent brain V12 (excluding the target volume and nonbrain normal
tissue). (B) shows data from multifraction SRS studies (all reporting brain Vx; that is, again excluding the target volume and
nonbrain normal tissue). Data were extracted from studies that reported necrosis risks and median Vx among all patients/
lesions or subgrouped (binned) ranges of Vx and reported the risks of necrosis for patients/lesions within the individual bins.
One study (Minniti 201628) in (A) calculated Vx per patient, and the others calculated Vx per lesion (see text); all of the
studies in (B) calculated Vx per patient. These figures represent a rough approximation of data, as (1) the median value of
V12 for a given bin was estimated as described in the caption from Figure 1; (2) for multifraction SRS studies, the linear-
quadratic model with alpha-beta ratio of 2 was used to convert dose to single-fraction equivalent dose. The error bars
represent 95% binomial confidence intervals and the solid lines represent a fitted logistic model to the data. When analyzed
separately, there was no significant difference between the tissue V12 and brain V12 logistic models; they are therefore
combined in (A), recognizing that tissue V12 would have a larger value than brain V12, and that the figure is meant to be
descriptive. Studies included in (A) were Korytko 2006,29 Blonigen 2010,30 Minniti 2011,31 Ohtakara 2012,32 Sneed 2015,33
and Minniti 2016.28 Studies included in (B) were Ernst-Stecken 200634 and Minniti 201335 (with 1 resected brain metastasis
per patient); Minniti 2014,36 Minniti 2016,28 and Dore 201737 (with w1 resected brain metastasis per patient).

Arteriovenous malformations Figures 1 and 2 show the reported risks of any or symp-
tomatic necrosis after single-fraction SRS for AVM, relative
For AVM, the concept of high-dose volume exposure with to tissue V12, in those studies that grouped/binned patients/
SRS correlating with toxicity risks was recognized in the lesions by V10-V12. The combined factors of converting
1970s to 1980s.75,76 Flickinger et al published several V10 in the Voges et al study22 to V12 (using Equation 2 in
studies in the 1980s and 1990s analyzing the relationship section 6), and estimating the median values of V12 (see
between necrosis/toxicity risks and dose-volume exposure figure captions), produce uncertainties in the data compila-
in the treatment of tumors and AVM.77-80 The AVM Study tion. Logistic models are shown merely as a descriptive way
Group described the effects of AVM location and tissue to summarize the data (and not intended to be predictive). For
volume (including target) receiving 12 Gy (V12) in 1 these logistic models, the logarithm of Vx (as opposed to Vx)
fraction in predicting permanent sequelae.24 Studies from was used for the independent variable (Equation 1) to avoid
several institutions have also demonstrated the importance predictions of a nonzero probability of necrosis for Vx Z 0;
of V10-12 in predicting toxicity risks of AVM for the plots in Figures 1 to 2, the inverse log of that inde-
patients.23,22,48,51,52,25,26,72 Most studies of AVM analyzed pendent variable was used (to recreate Vx).
tissue V10-12 (ie, included the target volume, as opposed to Target volume is also predictive of toxicity risks in AVM
a V10-12, which excludes the target), which seems logical patients.22,49,50,52,25,26,72 With AVM, the reported rate of
because the AVM interdigitates with brain tissue. In the radionecrosis ranges from w0% to 20% for small lesions
AVM Study Group report, nontarget V12 did not signifi- and w15% to 40% for lesions >10 cm3; similar event rates
cantly improve the risk-prediction model; both target and are seen with low V12 versus V12 >10 cm3, with vari-
nontarget V8-12 were significant predictors of toxicity in ability between reports likely partially attributable to dif-
another study.52 ferences in analyzed endpoints (Tables E3-5). In a multi-
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 7

1 Single fraction Multi-fraction radiation-induced radiographic changes of 24% versus 0%


.9
Blonigen 2010
Sneed 2015
Minniti 2011
Korytko 2006
Dore 2017
Inoue 2013
for V10 >10 cm3 versus 10 cm3 (P < .0001), respec-
Probability of symptomatic necrosis
after SRS/fSRS for brain metastases

.8 Ohktakara 2012 Inoue 2014 tively. This study is a landmark publication, being among
the first to correlate toxicity risks with SRS dose-volume
.7
metrics for tumors.72 Two other early (2001) studies,
.6
which grouped together benign intracranial tumors,
.5
.4
.3
1.0
.2 Observed data
0.9 Logisitic model
.1
Korytko 2006

Probability of Edema or Necrosis


0
0.8
0 5 10 15 20 25 30 0.7
V12 (cc)
0.6
Fig. 4. Risk of symptomatic necrosis after single-fraction
0.5
stereotactic radiosurgery (SRS) for brain metastases versus
volume of tissue (target plus nontarget; solid symbols) or 0.4
brain (excluding the target volume and nonbrain normal 0.3
tissue; open symbols) receiving 12 Gy equivalent in 1
0.2
fraction (V12). All of the studies reported Vx per lesion
(see text). Data were extracted from studies that reported 0.1
necrosis risks and median Vx among all patients/lesions or 0.0
0 10 20 30 40 50 60 70 80
subgrouped (binned) ranges of Vx and reported the risks of
3
necrosis for patients/lesions within the individual bins. All Brain including GTV, 1fx equivalent V12Gy’ cm
of the multifraction fSRS studies used the endpoint of
Fig. 5. Aggregate model of edema or necrosis as a
symptomatic necrosis requiring resection, whereas all of
function of V12Gy, including gross target volume, for pa-
the single-fraction SRS studies used the endpoint of any
tients treated with cranial single fraction stereotactic radi-
symptomatic necrosis. The figure represents a rough
osurgery (SRS)/fractionated stereotactic radiosurgery
approximation of data, as (1) the median value of V12 for a
(fSRS), using individual patient data from Chin 2001,40
given bin was estimated as described in the caption from
Korykto 2006,29 Inoue 2013 and 2014,38,39 and Peng
Figure 1; (2) for multifraction SRS studies, the linear-
2018,41 including 34, 129, 145, 78, and 57 patients (34,
quadratic model with alpha-beta ratio of 2 was used to
198, 159, 85, and 294 lesions), respectively. Eq. (1) (shown
convert dose to single-fraction equivalent dose; (3) in those
in the text) was used to approximate V12Gy for the
studies that reported V10 or V14, the equivalent to V12 was
aggregated data points from studies by Chin et al,40 Inoue
estimated as described in the text (Eq. 2). The error bars
et al,38,39 and Peng et al.41 The vertical error bars are
represent 95% binomial confidence intervals, and the solid
binomial 68% confidence intervals calculated using the
and dashed lines represent fitted logistic models to the
score method and the horizontal error bars are the standard
single-fraction tissue V12 and multifraction brain V12 (in
deviations for V12 for a particular bin. Because there were
single-fraction equivalent) data, respectively. Single-
relatively few targets associated with V12Gy >50 cm3, the
fraction SRS studies included Korytko 2006,29 Blonigen
bins in that range tended to quantize toward 0% or 100%.
2010,30 Minniti 2011,31 Ohtakara 2012,32 and Sneed
The model parameters for the pooled data are Vx,50 Z
2015.33 Multifraction fSRS studies included Inoue 2013
63.2 (95% CI, 49.2-97.0) and g50 Z 0.87 (95% CI, 0.74-
and 201438,39 and Dore 201737 (with w1 resected brain
1.03) with P < .001 (calculated using c2, based on likeli-
metastasis per patient); all 3 used brain Vx.
hood ratio by comparing best fit against a horizontal line
through incidence averaged for all patients). Because the
institutional study of 2236 AVM patients with target vol- reported risks from Korytko et al were substantially greater
umes ranging from 0.1 to 24 cm3 (median, 3), multivariate than in other studies, the unmodified logistic model from
analyses demonstrated that doses >24 Gy to the target their report (dashed line) is shown separately from the
margin, versus lower doses, significantly (P Z .004) pre- others (solid line), and only the other studies are included in
dicted a >1.5-fold risk of radiation-induced changes; no the observed data (dots with error bars). The Korytko et al29
dose-volume metrics were analyzed.81 data points were published in terms of V12Gy. Abbrevia-
tions: GTV Z 1fx Z single-fraction equivalent dose using
Tumors linear quadratic with a/bZ 2 Gy; CI Z confidence inter-
val; gross tumor volume; V12Gy Z the volume of
Among 135 patients with AVM, benign skull base tumors, total brain exceeding an equivalent dose of 12 Gy in 1
and other benign tumors, Voges et al reported risks of fraction.
8 Milano et al. International Journal of Radiation Oncology  Biology  Physics

A B
1.0

Probability of Pathologically Confirmed Necrosis


1.0
0.9 Observed data 0.9 Observed data
Logisitic model Logisitic model
Probability of Edema or Necrosis

0.8 0.8
0.7 0.7

0.6 0.6
0.5 0.5
0.4 0.4
0.3 0.3

0.2 0.2
0.1 0.1
0.0 0.0
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70
Brain including GTV, 1fx equivalent V14Gy’ cm3 Brain including GTV, 1fx equivalent V14Gy’ cm3

Fig. 6. Volume response normal tissue complication probability models for brain necrosis in patients treated with single-
fraction stereotactic radiosurgery (SRS)/fractionated stereotactic radiosurgery (fSRS) for brain metastases: (A) for grade 1 to
3 edema or necrosis and (B) for grade 3 surgically removed and pathologically confirmed necrosis. Data are pooled from 3
studies: Inoue 2013,38 Inoue 2014,39 and Peng 2018,41 including 145, 78, and 57 patients (159, 85, and 294 lesions),
respectively. The vertical error bars are binomial 68% confidence intervals calculated using the score method and the hor-
izontal error bars are the standard deviations for V14 for a particular bin. The model parameters for the pooled data are for
(A), Vx,50 Z 45.8 (95% CI, 33.0-106.2) and g50 Z 0.88 (95% CI, 0.68-1.11) with P Z .003; for (B), Vx,50 Z 42.6 (95%
CI, 33.8-75.5) and g50 Z 1.58 (95% CI, 1.17-2.09) with P < .001. P values were calculated using c2, based on likelihood
ratio by comparing best fit against a horizontal line through incidence averaged for all patients. Abbreviations: 1fx Z single-
fraction equivalent dose using linear quadratic with a/b Z 2 Gy; CI Z confidence interval; GTV Z gross tumor volume;
V14Gy Z the volume of total brain exceeding an equivalent dose of 14 Gy in 1 fraction.

gliomas, brain metastases, and AVM in their analyses, for lesions 2.0 cm treated to peripheral dose of 24 Gy,
evaluated SRS dose-volume metrics associated with ne- 20% for lesions 2.1 to 3.0 cm treated to 18 Gy, and 14% for
crosis and complication risks.73,40 Chin et al compared the lesions 3.1 to 4.0 cm treated to 15 Gy. In multivariate an-
dosimetry in 17 patients who developed symptomatic ne- alyses, tumor diameter was associated with a significantly
crosis versus 17 controls who did not and reported that increased risk of grade 3 neurotoxicity, with 7.3- and 16-
total-volume V10 (median 28.4 vs 7.8 c cm3, P Z .007), fold increased risks for tumors 2.1 to 3.0 and 3.1 to 4.0 cm,
brain V10 (19.8 vs 7.1 cm3, P Z .005) and target volume respectively, versus tumors 2.0 cm. A 2017 study from
(4.4 vs 1.5 cm3, P Z .04) were significantly greater in the the Cleveland Clinic,83 with 896 patients with 3034 brain
necrosis group.40 Nakamura et al reported that among 657 metastases 2 cm, showed that tumor diameter >1 cm (vs
evaluable patients with 1244 targets, prescription volume <1 cm) was associated (on multivariate analysis) with a
(P Z .009), target volume (P Z .007), and nontarget 2.1-fold increased risk of necrosis and a 4.8-fold increased
volume (P Z .03) correlated with grade 3 complications risk of symptomatic necrosis; similar risks (on univariate
risks73; prescription volumes of 0.66 cm3, 0.67 to 3.0 analyses) were seen with tumor volume >0.1 cm3. Another
cm3, 3.1 to 8.6 cm3, and 8.7 cm3 were associated with study reported that maximum dose divided by lesion
grade 3 complication rates of 0%, 3%, 7%, and 9%, diameter was predictive of grade 3 to 4 necrosis risks.84
respectively. For smaller lesions that might be safely treated to rela-
tively high doses, clinicians generally opt against exceeding
Brain metastases a prescribed dose of 24 Gy. A University of Kentucky study
of patients with brain metastases 2.0 cm in size reported
For metastatic malignant brain tumors, earlier studies on nonsignificantly greater grade 3 to 4 toxicity rates (5.9% vs
toxicity risks focused on peripheral dose and tumor size. 1.9%, P Z .078) with peripheral SRS doses >20 Gy (vs
The Radiation Therapy Oncology Group 90-05 phase 1 lower doses) after whole brain radiation therapy (WBRT).85
study (which included recurrent, previously irradiated brain This finding may not be applicable for patients treated with
metastases and gliomas) reported 2-year actuarial neuro- SRS alone (ie, no WBRT). A prescribed dose of >17 Gy
toxicity risks of 11%.82 With lesions grouped by maximum was predictive of any (asymptomatic or symptomatic) ne-
diameter, crude rates of grade 3 neurotoxicity were 10% crosis (hazard ratio [HR] 3.3, P Z .03) in another study.68
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 9

In the aforementioned study from the Cleveland Clinic >41.2 cm3 was associated with 6%, 13%, and 25% radio-
(including lesions 2 cm),83 dose >24 Gy was not asso- necrosis risks, respectively. In studies from Inoue et al,38,39
ciated with increased necrosis risks. nontarget V23.1 (3 fractions) <5 cm3 and <7 cm3 were
Target volume correlates with toxicity risks in many associated with 1.4% and 0% risks of symptomatic edema
studies. The University of Pittsburgh described risks of and necrosis requiring surgery, respectively; and V28.8 (5
neurologic complications of 3% versus 16% at 5 years for fractions) <3 cm3 and <7 cm3 were associated with 0%
target volume 2 cm3 versus >2 cm3 (P Z .0085).53 These risks of symptomatic edema and necrosis requiring surgery,
findings are comparable to other reports9,38,39,73 (Tables respectively. A V23.1 in 3 fractions and V28.8 in 5 fractions
E3-5), 2 of which converted 3 to 5 fraction fSRS to a were considered dose-equivalent to 14 Gy in 1 fraction
single-fraction equivalent dose.38,39 In 2 studies, tumor (using an alpha-beta ratio of 2 Gy). Escalating prescribed
volume did not predict risk of neurologic complica- dose from 9 to 10 Gy  3 for brain metastases >3 cm
tions.54,55 In a BC Cancer Agency study of brain metastases increased the risks of radionecrosis, without apparent
<3 cm in size, the reported necrosis risks were relatively improvement in tumor control, in a recent dose-escalation
high compared with other studies; for lesions greater versus study.88 In a 2017 study of 2-fraction SRS,89 published
less than 0.4 cm3, necrosis rates were 12% versus 6%.64 after our 2016 cut-off, brain V20 to 87.8 values were
Beginning in the early 2000s, several studies described analyzed as factors potentially associated with necrosis
dose-volume measures predictive of radionecrosis, edema, risks (albeit with only 4 events in 39 patients), with sig-
and neurologic complications after SRS/fSRS for brain nificant P values at V44.5 to 87.8.
metastases. The University of California San Francisco The reported risks of any necrosis (Fig. 3) and symp-
(UCSF) published one of the largest studies of patients with tomatic necrosis (Fig. 4) after SRS or fSRS for brain me-
brain metastases, in which the 1-year risks of symptomatic tastases, relative to tissue or brain V12 in a single-fraction
ARE were 1%, 6%, and 13% for tissue V12 of 1.84 cm3, dose equivalent, are shown for those studies that reported
1.86 to 3.30 cm3, and >3.3 cm3, respectively.33 In a Uni- necrosis risks and median Vx among all patients/lesions or
versity of Cincinnati study,30 necrosis (asymptomatic or that grouped patients/lesions by tissue or brain Vx. As with
symptomatic) risks were 4.7% and 11.9% for V12 <1.6 Figures 1 to 2 (see previous discussion) the logarithm of Vx
cm3 and 1.6 to 4.7, respectively, comparable to the UCSF was used for risk calculation in the model to avoid pre-
study. A much higher toxicity rate was reported in a Case dictions of a nonzero probability of necrosis for Vx Z 0.
Western Reserve study, with 23% of brain metastases with A 2020 analysis (published after our literature search) of
V12 <5 cm3 developing symptomatic necrosis and 15% 5-fraction fSRS for intact brain metastases (n Z 132) re-
developing asymptomatic necrosis.29 With V12 in excess of ported that a brain (minus clinical target volume [CTV])
10 cm3, necrosis rates were >50% in the University of V30 of <10.5 cm3 versus 10.5 cm3 was associated with a
Cincinnati and Case Western Reserve studies29,30 and lower significantly greater risk of symptomatic ARE (13% vs
in the USCF study,33 in which a 15% necrosis rate (11% 61%), with a >7 fold relative-risk on multivariable anal-
symptomatic) was reported with V12 of 7 to 35 cm3. A ysis.90 The 61% risk is appreciably higher than what is
study by Minniti et al analyzed brain V12 (as opposed to reported in most fSRS series (Fig. 4), although few patients
tissue V12), showing a similar trend of increased V12 in these other studies had many lesions with brain (minus
associated with increased toxicity risks, and a similarly CTV) V30 >10.5 cm3.
high rate (w50%) of toxicity with V12 >10 cm3.31 In the An issue specific to multitarget fSRS/SRS (a common
Minniti et al 2016 study of 151 patients with brain metas- situation with brain metastases) is whether Vx is reported
tases >2 cm,28 brain V12 remained significant (P Z .02) per lesion or per patient (ie, from the composite plan) and
for radionecrosis risks, with a V12 >18.2 cm3 associated whether risks of necrosis are reported per lesion or per
with a w50% risk of radionecrosis. patient. In a 2020 study of single-fraction SRS in 40 pa-
Several studies (n Z 15, dating back to 2006 in the tients with 10 brain metastases, the V12 of individual
Supplemental Tables) have reported neurotoxicity risks targets predicted risks of posttreatment changes suggestive
after fSRS. Patients selected for fSRS often have bulkier of necrosis, as opposed to the net V12.91 Of the studies
disease and/or tumors in critical locations. Toxicity after shown in Figures 3 to 4, one study34 analyzed composite
fSRS appears to be relatively lower (vs. single-fraction Vx and necrosis risks per patient, 2 studies36,28 analyzed
SRS) (Tables E3-5), with meta-analyses suggesting that necrosis risks per patient as grouped by lesion (generally
tumor control is not compromised, with the current fSRS the largest) Vx (G. Minniti, personal communication), 2
dose-fractionation schedules commonly used.86,87 Minniti studies35,70 included patients with 1 or w1 target (resected
et al examined V15 to 24 with a 3-fraction schedule for brain metastasis) per patient, and all of the others analyzed
patients with intact36 or resected35 brain metastases; <25 Vx and necrosis risks per lesion. All of the studies in
cm3 of normal brain receiving >18 Gy36 and <17 cm3 Figure 3B reported necrosis risks per patient (including the
receiving >24 Gy35 were associated with <5% necrosis 2 studies of SRS/fSRS for resection cavities and 2 studies
risks.35,36 In the Minniti et al 2016 study of 138 patients that analyzed lesion Vx); all of the studies in Figure 4 re-
with brain metastases >2 cm treated to a dose of 9 Gy  ported necrosis risks and Vx per lesion. The logistic models
3,28 a brain V18 of 22.8 to 30.2 cm3, 30.2 to 41.2 cm3, and of the data demonstrated significantly greater risks after
10 Milano et al. International Journal of Radiation Oncology  Biology  Physics

SRS versus fSRS for both any necrosis (Fig. 3, P < .0001) generally being more susceptible to symptomatic
and symptomatic necrosis (Fig. 4, P < .0001), with the toxicity23,24,46,27,47,48,50,51,29,55,31,32,63,64,74dalbeit with
caveat that for symptomatic necrosis (Fig. 4) the endpoint location being defined somewhat inconsistently. Confor-
in all of the fSRS studies was symptomatic necrosis mity index/conformality was a significant factor in some
requiring surgical resection versus the less stringent studies46,31,32,73 and not others54,29,35,36,58; counterintui-
endpoint of any symptomatic necrosis that was used in the tively, lower conformity index (ie, greater conformality)
SRS studies. was associated with an increased risk in 1 study,73 perhaps
For any given value of Vx, the Vx for tissue exposure a spurious finding. Among patients with brain metastases,
should represent less irradiated normal brain than the Vx prior WBRT was generally not a significant factor for
for brain (because the tissue includes both normal brain and toxicity risk,33,53,30,32,58,71,40,83 although it was implicated
other tissues), and thus the risk of toxicity should be less for in some studies.29,63,71
tissue versus brain (at the same Vx). As shown in Figures 3 The potential impact of histology of brain metastases
and 4, this is an expected finding that was not seen in the remains unclear. In 3 studies,54,62,70 cancer type was not a
reported clinical data (perhaps reflecting heterogeneity in significant variable. However, several other studies reported
study populations and reporting methods/standards and different findings (although it remains unclear whether the
challenges in comparing data across institutions). In Fig- associations of some cancer types with greater risks of
ures E1 to E3, risks of necrosis are plotted against tissue necrosis in these studies are from inherent differences be-
V12 (similar to Figs. 3 and 4), with tissue V12 estimated tween cancers or differences in systemic therapy used).
(for those studies that reported brain V12) by adding the Kidney cancer (n Z 47 of 316 lesions) was associated with
brain V12 to the estimated target volume (which considered a 2.4-fold risk of severe neurologic complications in 1
the range and median target volumes reported in the study.55 In another study of 435 patients (2200 lesions),
studies). As anticipated, the risk of necrosis relative to kidney cancer (n Z 26 patients with 86 lesions) was
estimated V12 was lower using this method. associated with an increased risk of adverse events among
The combined factors of variable use of brain versus patients with tumors >1 cm3.33 In a 2016 study from the
tissue Vx, variable use of patient versus lesion Vx, variable University of Maryland,65 renal or melanoma (n Z 24
reporting of toxicity risks per patient versus per lesion, patients) versus lung or breast (n Z 83 patients) histology
conversion of single-fraction V10 or V14 to V12, conver- was associated with a >2-fold greater risk of acute (within
sion of multifraction fSRS Vx to single-fraction equivalent 100 days) edema. In a 2016 study from the Cleveland
V12, and estimating median values of V12 (see Equation 2 Clinic, among 1939 patients with 5747 lesions, renal cancer
in section 6 and figure captions) produce uncertainties in (HR 1.78, P Z .03) was associated with increased risks of
the data compilation. Therefore, the logistic models should radionecrosis after SRS; among NSCLC patients (2276
be considered as a descriptive way to summarize the data lesions) adenocarcinoma histology (HR 1.89, P Z .04) and
(and are not intended to be predictive). ALK rearrangement (HR 6.36, P < .01) were also associ-
ated with radionecrosis.93
In patients having both surgery and SRS/fSRS, symp-
5. Factors Affecting Outcomes tomatic necrosis has been reported to be more common
with post- versus pre-resection SRS/fSRS (2 years: 17% vs
Radionecrosis, edema, and neurotoxicity are multifactorial, 3% at 2 years; P Z .01).46 In a 2017 analysis, restricted to
with risks attributable to dose and volume (and perhaps lesions 2 cm diameter ( z4 cm3 volume), the 1-year
surface area) and also likely related to target type (ie, necrosis rates after postoperative SRS, preoperative SRS,
malignant tumor, benign tumor, AVM), target location, and SRS alone were 23% vs 5% and 12%, respectively (P
prior radiation therapy, prior surgery, systemic therapy, < .001).94
comorbid conditions, and so on. Radiation toxicity likely
represents injury to cells within the target and/or sur-
rounding normal brain tissue.3,92 Table E6 summarizes 6. Mathematical/Biological Models
variables, other than target volume and Vx, that were or
were not significantly correlated with toxicity risks in each At the time of the literature search, 4 articles presented
study. For any given variable, there was inconsistency SRS/fSRS dose (in 1-5 fractions), target volume, and
across various studies. Nevertheless, there are some broad toxicity outcome per treated target; an additional article
patterns. with this level of granularity of data was published in 2018.
In general, sex was not a significant risk factor; specif- Data from these 5 studies, summarized in Table 2, were
ically, female sex was implicated as a risk factor in only 1 amenable to NTCP modeling. These studies included
study.45 Age was not a significant factor in most studies; in mostly patients with brain metastases, although 2 studies
only 2 studies was older age an adverse risk factor.45,50 also included patients with glioma and benign tumors. We
Central nervous system target location appears to be asso- did not similarly model necrosis risks after SRS/fSRS for
ciated with toxicity risk, with more eloquent brain areas AVM because the published data did not lend itself to
(eg, the thalamus, basal ganglia, and occipital lobes) NTCP modeling.
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 11

Table 2 Studies included in the dose-response models for NTCP of brain necrosis after SRS for brain metastases
No.
Author targets/
(publication No. SRS Definition of Tumor patients
year) fractions system Vx analyzed brain types (ratio) Endpoint
Chin 1 GK V10Gy Total 10 Gy Mixed NR/233
Pathologic confirmation on MR imaging of radiation
(2001)40 volume necrosis, or development of a necrotic lesion that
resolved over time
Korytko 1 GK V12Gy Total 12 Gy Mixed 198/127 Symptomatic radiation necrosis
(2006)29 volume (1.56)
Inoue 3 CK V14Gy 14 Gy volume Brain 159/145 Datapoints 1: G1-3 edema and necrosis,
(2013)38 SFED excluding GTV metastases (1.10) Datapoints 2: G3 pathologically confirmed
Inoue 5 CK V14Gy 14 Gy volume Brain 85/79 Datapoints 1: G1-3 edema and necrosis,
(2014)39 SFED excluding GTV metastases (1.09) Datapoints 2: G3 pathologically confirmed
Peng 1, 3, 5* CK V14Gy Brain 14 Gy Brain 294/139* Model 1: G1-3 edema and necrosis,
(2018)41 SFED volume (target not metastases (2.12) Model 2: G3 pathologically confirmed
subtracted)
Mixed Z brain metastases; glioma and benign brain tumors (see Table E1 for specific details).
Abbreviations: CK Z CyberKnife; G Z grade; GK Z GammaKnife; GTV Z gross target volume; NR Z not reported; NTCP Z normal tissue
complication probability; SFED Z single fraction equivalent dose calculated using the linear quadratic model with a/b Z 2 Gy; SRS Z single fraction
stereotactic radiosurgery.
* n Z 96, 23, and 20 treatment with 1, 3, and 5 fractions, respectively; 57 patients with 139 treatment plans.

The 2 earliest data sets that were used in our models Vx50 is the volume corresponding to 50% risk of necrosis;
were from studies of patients who underwent single- and g50 is the slope parameter. The best-fitting values were
fraction SRS with Gamma Knife.29,40 The 3 other studies found using the maximum likelihood method, and the 95%
included patients treated with single- and/or multifraction confidence intervals were calculated using the profile-
SRS; the doses from these data sets were converted to likelihood method.98
single-fraction equivalent doses, using the linear quadratic
It was observed that the risk level reported by Korytko
(LQ) model with a/bZ 2 Gy.41,38,39 To form a composite
et al,29 in which 44% of lesions developed symptomatic ne-
model in terms of V12Gy (a commonly used dose metric
crosis, was substantially higher than what was reported in the
for brain SRS), a simple ratio was used to approximate
other data sets. Potential causes of the vastly
V12Gy from V10Gy and V14Gy95,96:
differing reported rates of necrosis are unknown. Therefore,
V12GyzV14Gy  ð1 þ DeltaÞ
ð2Þ the Korytko et al data set was kept separate from the HyTEC
V12GyzV10Gy=ð1 þ DeltaÞ model that included the other 4 studies, as seen in Figure 5.
The average Delta Z 0.5 was measured from the The studies by Inoue et al38,39 and Peng et al41 included
V10Gy, V12Gy, and V14Gy values in the appendix of Peng only patients with brain metastases and comprise the data
et al.41 The impact of elapsed treatment time is not that are modelled and shown in Figure 6. These 3 studies
considered in these calculations. analyzed 2 separate endpoints: (a) any edema or necrosis,
In the articles by Inoue et al,38,39 gross target volume which we have designated as grade 1 to 3; and (2) necrosis
(GTV) was subtracted from the brain contours to calculate requiring surgical resection, allowing pathologic confir-
brain Vx. Before modeling, we added the median GTV to mation of necrosis, which we have designated grade 3. The
brain Vx to approximate tissue Vx. Combined data from studies from Inoue et al38,39 used V14, which was chosen as
the studies listed in Table 2 were fit to a logistic model97 to the dosimetric measure for Figure 6. The Peng et al study
generate curves describing probability of toxicity as a analyzed Vx by patient, and the studies by Inoue et al
function of V12Gy and V14Gy. analyzed Vx by lesion. Most patients in these studies had a
For the data sets presented in Figures 5 to 6 (this sec- single treated lesion, although specific data on number of
tion), using studies that provided individual patient/lesion lesions per data point in the Peng study are not available.
data, the exponential form of the logistic model was used From individual patient/lesion data from these 3 studies, we
(Equation 3): show the volume response NTCP models (Fig. 6) and dose-
   volume risk data (Table 3) for brain necrosis in patients
exp 4g50 Vx50  1
Vx
treated with SRS/fSRS for brain metastases.
NTCP Z    ð3Þ fSRS was used in some (Peng et al) or all (Inoue et al) of
1 þ exp 4g50 VxVx50  1 the patients in the 3 studies included in the NTCP model for
brain metastases. For 1-, 3- and 5-fraction SRS/fSRS, there
were 96, 182, and 105 data points, respectively, with a
where Vx is the volume of tissue exceeding  Gy, which is
median calculated V14 of 3.9 cm3, 11.7 cm3, and 17.5 cm3,
x Z 12 Gy in Figures 1 to 5 and x Z 14 Gy in Figure 6;
12 Milano et al. International Journal of Radiation Oncology  Biology  Physics

Table 3 Estimated risk, from dose-response models for NTCP, of brain radionecrosis after SRS for brain metastases at various dose/
volume levels
Vx metric (x in Gy) Toxicity Toxicity rate for Vx  Studies in NTCP
1 fraction 3 fractions* 5 fractions* grade 5 cm3 10 cm3 20 cm3 model
V12 V19.6 V24.4 1-3 3.6% 4.8% 8.6% Chin 2001y þ Inoue 2013 þ Inoue 2014 þ Peng 2018
V12 V19.6 V24.4 1-3 19.6% 25.8% 41.5% Korytko 2006y
V14 V23.1 V28.8 1-3 4.1% 6.0% 12.1% Inoue 2013 þ Inoue 2014 þ Peng 2018
V14 V23.1 V28.8 3 0.4% 0.8% 3.4% Inoue 2013 þ Inoue 2014 þ Peng 2018

Abbreviations: NTCP Z normal tissue complication probability; SRS Z single fraction stereotactic radiosurgery.
* Equivalent doses are calculated with the linear quadratic model and a/bZ 2 Gy.
y
These studies included patients with brain metastases as well as other intracranial tumors (see Table E1 for specific details).

respectively. For V14 of 0 to 5 cm3, 5 to 10 cm3, 10 to 20 studies from the University of Pittsburgh23,48 and Gunma
cm3, and >20 cm3, there were 71, 71, 216, and 25 data University38,39 (w17%-25% with brainstem lesions; see
points, respectively, with single-fraction SRS accounting Table E1). The most recent AVM study from the University
for 77%, 37%, 6%, and 8% of data points, respectively. of Pittsburgh27 included 55 (7%) of 755 brainstem AVM.
Thus, the risks for calculated V14 >10 cm3 reflect mostly Other studies of fSRS described including some brain le-
fSRS data. sions abutting the brainstem.44,56,36,68 Because we explic-
Notably, for the modeling shown in Figures 1 to 4 itly omitted studies that focused on brainstem toxicity, we
(section 4), the logarithm of volume V12 was used in the have not reported specific dose-volume metrics predictive
logistic model (Equation 1), forcing a 0% risk at 0 Gy, of brainstem toxicity. Notably, in a 2016 multi-institutional
whereas no logarithm of volume was used for the modeling pooled analysis102 of 547 patients with 596 brainstem
shown in Figures 5 and 6. The different approaches to metastases treated with Gamma Knife SRS, 7.4% of lesions
modeling, as well as using binned data extracted from developed grade 3þ toxicity. Factors associated with
published studies (Figs. 1-4) versus more granular indi- significantly increased risks of grade 3þ toxicity included
vidual patient data (Figs. 5 and 6), account for the different tumor volume >1 cm3 (odds ratio [OR] >10, P < .001),
appearance of these curves. Figure E4 shows how the doses >16 Gy to target margin (OR >3, P Z .05), and
models in Figures 3A-B would be different if the logarithm prior WBRT (OR Z 4.7, P Z .002). No grade 3þ toxicity
of volumes were not used (ie, similar to what was done in occurred with margin doses <12 Gy or with tumors <0.1
Figs. 5-6). cm3. Primary cancer type (P > 0.2), brainstem location (P
In the HyTEC review for optic nerve and chiasm,99 Z .30), and tissue V12 (P Z .06) were not significant
limitations of the NTCP modeling were discussed in factors.
detail. These limitations include statistical uncertainties
with data compilation, uncertainties in the a/b value and
applicability of the LQ model, and the absence of data in Resected brain metastases
the low-dose and high-dose extremes. Additionally, the
inclusion of retrospective studies, with different planning, In a 2020 study of 442 patients treated with SRS for 501
delivery methods, and follow-up protocols, is another lim- resection cavities, the 1-year rates of ARE and symptom-
itation; with retrospective studies, necrosis could be atic ARE were 8.9% and 5.9%, respectively.103 In a pooled
underreported. analysis of 36 studies of postresection cavity SRS/fSRS, the
risk of radionecrosis was 6.9%.104 Among the 51 reports
summarized here, in 10 studies some46,56 or
7. Special Situations all45,35,57,59,66,69,71,70 of the patients underwent SRS (2
studies) or fSRS (10 studies) to the surgical cavity of a
Brainstem targets resected brain metastasis. For resected brain metastases, the
X Gy isodose line volume (ie, “tissue Vx”) may not be as
It is well recognized that the brainstem is more susceptible clinically meaningful as brain Vx, given that the target
to symptomatic radiation toxicity; some studies in this volume likely includes fluid (ie, nontissue) within the
analysis,23,24,27,48,50,32 the American Association of Physi- cavity.
cists in Medicine Task Group101 report,100 and QUANTEC In a 2018 analysis (outside of this report’s search) of 90
paper101 suggest relatively stringent SRS/fSRS constraints resection cavities in 80 patients treated with SRS, there were
for the brainstem. Although several of the studies in our 5 necrosis events; greater necrosis risks were associated with
analyses included brainstem targets, they generally repre- greater brain/parenchymal V12 (P Z .05) and smaller total
sented only a small percent of patients (eg, ~1%33,49,26,73 or (ie, tissue) V12 (P Z .03).105 The authors postulated that
2% to 5%50,25,55,31,32,64 in most studies). Exceptions are larger cavities (and therefore greater V12) may have been
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 13

more likely to border the calvarium or ventricles, accounting SRS/fSRS appears to increase toxicity risks compared with
for the counterintuitive finding of lower necrosis risks with patients with no prior radiation therapy.33,40,109 For
greater tissue V12. Although patients with intact or subto- example, in a study from UCSF, prior SRS increased
tally resected brain metastases may develop symptomatic toxicity risks w5-fold33; in those with versus without prior
tumor or brain necrosis after SRS/fSRS, in those with SRS, crude rates of ARE were 14% versus 3%, respec-
completely resected brain metastases, necrosis would only tively, and 1-year cumulative incidences of ARE were 20%
develop within “normal brain,” although this brain may have versus 4%, respectively. A retrospective analysis from
had tumor- or surgery-related functional impairment before Wake Forest of 32 patients with 46 lesions receiving 2
SRS/fSRS. In a 2019 study of patients treated with post- courses of SRS suggested that tissue composite V40 was
operative, frameless, linear acceleratorebased 5-fraction associated (P Z .003) with the development of radio-
fSRS, 10 of 52 (19%) developed radionecrosis, with risks necrosis. The composite V40 values (sum from 2 single-
correlated with “hot-spot” doses >105% outside the cavity, fraction SRS plans) of 0.28, 0.76, and 1.60 cm3 were
and not with maximum doses within the CTV; brain V25, associated with 10%, 20%, and 50% probabilities of radi-
V30, and V35 were not associated with radionecrosis onecrosis, respectively.110 Other reports of repeated courses
risks.106 In a 2020 report, intact metastases were associated of SRS suggest that retreatment is tolerable for AVM and
with a significant >3-fold higher relative-risk of symptom- brain metastases.111-115 We did not analyze or model the
atic ARE versus resection cavities (after accounting for brain effect of repeat SRS owing to limited data. Because dosi-
V30 and other factors).90 metric predictors of toxicity after repeat SRS are limited,
None of the 10 studies reviewed in this report, which caution is warranted when considering reirradiation.
included patients with resected brain metastases, analyzed
tissue/total Vx, and 3 studies specifically reported brain Concurrent systemic therapy
Vx.35,57,70 The study by Broemme et al57 reported 1 event
in 44 treatments and the “normal brain” V10 to 12 (in 1
Data on concurrent systemic therapy and cranial SRS,
fraction) associated with that event (Table E4). The study
although limited, suggest that concurrent cranial SRS and
by Dore et al70 reported a range and median of brain (minus
cytotoxic chemotherapy may not increase the risk of
PTV) V12 and V21 values (in 3 fractions) among those
neurotoxicity or radionecrosis.116,117 and may decrease
who did versus did not develop radiographic evidence of
risks.118 In a retrospective study of 1650 patients with brain
necrosis; V10, V12, and V21 were significantly greater in
metastases treated at the Cleveland Clinic, the 1-year cu-
the group that developed necrosis (Table E3); binned
mulative incidence of necrosis was 6.6% in the lesions
groups of V10, V12, and V21, with associated necrosis
treated with SRS concurrently with a variety of systemic
risks, were not described.
therapies, versus 5.3% in those treated without concurrent
systemic therapies (P Z .14).116 Among those who also
Staged-SRS for AVM had received WBRT, radionecrosis rates were significantly
elevated with concurrent SRS and systemic therapy,
For bulky AVM, for which single-fraction SRS (at doses although there was no significant effect of systemic therapy
needed for an appreciable likelihood of obliteration) would on necrosis risks among those treated with SRS alone.
be associated with unacceptable risks of necrosis, staged- Notably, on subset analyses, significantly increased risks of
SRS is an alternative. One approach is fSRS-termed dose- post-SRS necrosis were observed with concurrent VEGFR
staged SRS, because the entire target is treated with and EGRF tyrosine kinase inhibitors (necrosis rates 14%-
multiple fractions; another approach is volume-staged SRS, 16%) and HER2 antibodies. Thus, it is prudent to consider
in which segmented portions of the target are treated withholding these specific agents when delivering SRS.
sequentially, generally months apart. A 2018 review sum- Despite ALK rearrangement being associated with radio-
marizes the risks of radiation-induced changes as well as necrosis risks in the aforementioned study from the
death (in part because of hemorrhage risks).107 Deep AVM Cleveland Clinic, the use of ALK inhibitors was not.93 In
location (ie, basal ganglia, thalamus, and brainstem) ap- the absence of more data, systemic therapy immediately
pears to be associated with a 5-fold increased risk of before and after SRS/fSRS should only be considered with
radiation-induced changes versus other locations.108 We caution.
opted against including staged-SRS in our analyses because
nearly all studies published to date have included <50 Concurrent immunotherapy
patients and have not focused on dose-volume effects on
necrosis risks.
Immune checkpoint inhibitors (ICI) have a long duration of
potential efficacy and may act synergistically with SRS,
Reirradiation potentially exacerbating toxicity risks.119 Concurrent SRS
and ICI was not associated with increased risks of adverse
Although prior WBRT may not appreciably increase the events in studies from Johns Hopkins,120 Yale,118 or the
risks of brain toxicity after SRS/fSRS (see section 5), prior University of Virginia,121 although a study from Harvard122
14 Milano et al. International Journal of Radiation Oncology  Biology  Physics

suggested that ICI may increase risks of post-SRS radio- appear to be location dependent,24 which we did not ac-
necrosis. A 2019 meta-analysis reported a 0% to 21% risk count for. Notably, after fSRS for brain metastases, a
of radionecrosis (among 5 studies) in patients with brain single-fraction equivalent V12 of 10 cm3 is associated with
metastases treated with SRS and ICI, with risks possibly a w5% to 10% risk of symptomatic necrosis (Fig. 4 and
related to the specific agents used; data were too limited to Fig. E3).
elucidate a temporal relationship between SRS and ICI.123 The 2018 UK Consensus on Normal Tissue Dose Con-
Investigators from Wake Forest showed, among 289 pa- straints project126 and UK Consortium SABR guidelines
tients with 2004 brain metastases, that although receipt of (version 6.1)127 recommended a dose of <12 Gy in 1
immune checkpoint inhibitors increased risks of adverse fraction to 10 cm3 of brain minus GTV (D10 <12 Gy,
radiation effects (HR Z 2.58, P < .01), the timing of which would result in a brain V12 <10 cm3). The Amer-
receipt of ICI (within 30 days or not) was not a significant ican Association of Physicists in Medicine Task Group 101
factor.124 Many randomized controlled trials evaluating the report did not include specific SRS/fSRS constraints for
safety and efficacy of ICI and SRS for brain metastases are brain parenchyma.100 The QUANTEC authors20 noted
accruing.125 substantial variation between studies and emphasized that
“more stringent constraints are needed for eloquent brain
Pediatric patients regions such as brainstem and corpus callosum.” Never-
theless, decisions for individual patients should weigh their
Our analyses did not specifically address brain toxicity after goals of care as well as NTCP and TCP estimates. Pre-
SRS/fSRS in the pediatric population because of insuffi- scribing more “moderate” fSRS/SRS doses for brain me-
cient data. tastases (relative to commonly prescribed dose schedules)
to reduce radionecrosis risks may be considered for select
patients,128 and more study is needed to determine the
9. Recommended Dose/Volume Objectives optimal dose-fractionation schemes with respect to NTCP
and TCP. There is a separate HYTEC TCP effort for brain
For brain metastases treated with SRS, our analyses suggest metastases that might help guide these often-challenging
that the risks of grade 3 necrosis are approximately 0.4%, decisions regarding balancing NTCP and TCP.
0.8%, and 3.4% for the total irradiated volumes (including
target volume) receiving a single-fraction equivalent dose
of 14 Gy (V14) of 5 cm3, 10 cm3, and 20 cm3, respectively. 9. Future Studies
These data can be used in conjunction with much of the
published data that focused on V10 to 12 (with V12 shown Future retrospective and prospective studies should use
in Figs. 1-4 and Figs. E1-3), as will be described. consistent reporting standards (outlined in section 10).
It is notable that fSRS was used in 3 of the studies More consistent means of establishing the diagnosis of
included in our NTCP model and accounted for >90% of radionecrosis versus tumor progression are needed
the V14 >10 cm3 data. Thus, our modeled w3.4% risk of because different institutions use different imaging mo-
grade 3 toxicity for a V14 of 20 cm3 would be most dalities. Furthermore, the endpoints of ARE (which can be
applicable to multifraction regimens, approximately corre- temporary) versus necrosis (which reflect tissue injury)
sponding to a V23 in 3 fractions and V29 in 5 fractions are challenging to characterize at the onset because it is
(using the LQ model with alpha-beta ratio of 2 Gy). A 2018 often unknown whether the symptoms and/or imaging
meta-analysis of 24 studies demonstrated that fSRS for changes will be reversible.33 Large patient numbers and
large (>4 cm3) brain metastases reduced the risk of radi- data pooling are needed to determine which variables, in
onecrosis (at prescribed doses that did not compromise addition to dose and volume, affect the risk of symp-
tumor control probability) relative to commonly used tomatic necrosis. Of particular interest would be a better
single-fraction SRS doses.87 We similarly note significantly description of susceptibility to necrosis/edema in different
reduced NTCP with fSRS versus SRS. These findings regions of the brain for brain metastases patients, similar
suggest that the LQ model may overestimate the single- to the efforts by Flickinger et al for AVM.24,129 Whether
fraction dose equivalent for NTCP. necrosis risks are technology/platform dependent should
Thus, the QUANTEC recommendation to limit single- be studied further,130-132 although differences in risks are
fraction V12Gy to 5 to 10 cm3 remains prudent,20 potentially subject to institutional variations in planning
particularly because the QUANTEC model was based practices (even with the same technology) as well as new
solely on single-fraction SRS data (without the un- and continually evolving technologies. The American
certainties with dose conversions), and in light of the high Society for Radiation Oncology (ASTRO) and the
risks reported in the Korytko et al series.29 From our American Association of Neurologic Surgeons (AANS)
compiled data from published studies, the risk of symp- SRS registry may facilitate pooled analyses, using clinical
tomatic necrosis with a V12 of 10 cm3 is on the order of and dosimetric parameters. Actuarial analyses are needed
15% after SRS for brain metastases (Fig. 4 and Fig. E3), to better understand the time course of risks as well as the
and <10% after SRS for AVM (Fig. 2); however, risks long-term actuarial toxicity risks. Patients with brain
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 15

metastases are living longer than they were in prior de- Fields/technique
-
cades owing to improved imaging (resulting in diagnosis Dose, fractionation
-
of smaller-volume brain metastases earlier in the course B Prior SRS e Y/N
of disease) and improved therapy. Future studies should - Target(s)
seek to better identify which patients benefit the most  Dates of treatment
from fSRS versus SRS with respect to NTCP and TCP,  Prescribed dose
particularly for targets that are smaller and not abutting  Dose fractionation
dose-limiting tissue (ie, brainstem, optic apparatus), for  Controlled (Y/N)
which scant comparative data exist.
It is important to continue investigating factors that may
cause some studies to have a higher incidence of necrosis
• Details of prior cranial surgery/ies and procedures
B Prior resection (Y/N)
than others, such as grading, histology, and other potential
- Lesion(s) resected
reasons.
 Date of surgery
 Surgical technique
10. Reporting Standards
• Details of SRS tumor(s)/target(s)
Necrosis, edema, and brain toxicity should be scored using B Intact versus resected metastasis
standardized toxicity criteria. The Common Terminology B Tumor/target volumes
Criteria for Adverse Events version 4 scores the extent of B Location(s) of treated targets/lesions
toxicity. The RANO-BM report describes the need to
differentiate progression from postradiation change, but • Treatment planning parameters
consensus criteria for defining necrosis are lacking. Stan- B Planning software (and version)
dardized reporting for SRS is needed and reportedly B Planning technique
forthcoming.133 Ideally, the details should be reported per B Dose calculation algorithm used
treatment course and per lesion for all patients whether they B Dose calculation grid size (more relevant for small
had toxicity, so the denominator of all cases would be targets)
known. With respect to toxicity reporting, we propose
recording and reporting the following information for pa- • Methods of treatment set-up and positional verification
tients with and without toxicity (with each bullet point
being an item potentially captured in a registry or • Target and brain dose metrics and/or dose-volume
database): histogram
B Maximum target dose (for each target if applicable)

• Patient age B Prescription target dose (for each target if applicable)

B Dose coverage of each target

• Patient sex - Percent isodose line covering percent target

volume
• Patient performance status B Brain/tissue dose-volume exposure per lesion or per

plan, or at least per stratified groups of patients (eg,


• Primary tumor type/histology quartiles)
- Tissue Vx across broad range of doses

• Date diagnosed with primary cancer - Brain Vx across broad range of doses (with spec-

ification of definition of brain (ie, brain minus


• Date diagnosed with brain metastases GTV or PTV)
- For multilesion treatment: description of lesion Vx

• Symptoms from brain metastases (Y/N) and patient (ie, composite) Vx. For patients with
lesions in close proximity, it may not be feasible to
• Steroids needed for brain metastases (Y/N) specify a lesion Vx, particularly if 2 or more le-
sions are treated as 1 target, but also if each lesion
• Statusof extracranial disease (controlled, uncontrolled, is planned as a separate target (owing to over-
new diagnosis) lapping isodose lines).
- Physical dose including fractionation for each re-

• Details of prior brain radiation therapy ported Vx is preferable (eg, V12Gy/1 fraction or
B WBRT e Y/N V20Gy/3 fractions); even when elaborate radiobi-
- Dates of treatment ological conversions are feasible, the basic
16 Milano et al. International Journal of Radiation Oncology  Biology  Physics

physical dose and fractionation should also be re- 7. Delishaj D, Ursino S, Pasqualetti F, et al. Bevacizumab for the
ported to enable future data pooling initiatives treatment of radiation-induced cerebral necrosis: A systematic review
of the literature. J Clin Med Res 2017;9:273-280.
8. Levin VA, Bidaut L, Hou P, et al. Randomized double-blind placebo-
• Dates of clinical and radiographic follow-up controlled trial of bevacizumab therapy for radiation necrosis of the
central nervous system. Int J Radiat Oncol Biol Phys 2011;79:1487-
1495.
• Imaging studies used to determine control and toxicity 9. Zach L, Guez D, Last D, et al. Delayed contrast extravasation MRI:
A new paradigm in neuro-oncology. Neuro Oncol 2015;17:457-465.
• Control of tumor or obliteration of AVM (Y/N) 10. Zhang Z, Yang J, Ho A, et al. A predictive model for distinguishing
radiation necrosis from tumour progression after gamma knife radi-
osurgery based on radiomic features from MR images. Eur Radiol
• Toxicity (including adverse imaging changes in the
2018;28:2255-2263.
absence of symptoms) 11. Zhou M, Scott J, Chaudhury B, et al. Radiomics in brain tumor:
B Yes/no for each type of event Image assessment, quantitative feature descriptors, and machine-
B If yes, specify learning approaches. AJNR Am J Neuroradiol 2018;39:208-216.
- Date of onset 12. Peng L, Parekh V, Huang P, et al. Distinguishing true progression
from radionecrosis after stereotactic radiation therapy for brain me-
- If >1 lesion/target, specify (if possible) which
tastases with machine learning and radiomics. Int J Radiat Oncol
target is the cause of the event Biol Phys 2018;102:1236-1243.
- Grade (Common Terminology Criteria for Adverse 13. Stockham AL, Ahluwalia M, Reddy CA, et al. Results of a ques-
Events version 4 preferred) tionnaire regarding practice patterns for the diagnosis and treatment
 No intervention of intracranial radiation necrosis after srs. J Neurooncol 2013;115:
469-475.
 Medical intervention
14. Lin NU, Lee EQ, Aoyama H, et al. Response assessment criteria for
 Surgical intervention brain metastases: Proposal from the RANO group. Lancet Oncol
B Date 2015;16:e270-e278.
15. Okada H, Weller M, Huang R, et al. Immunotherapy response
- Criteria used to define toxicity assessment in neuro-oncology: A report of the RANO working
 Computed tomography scan (Y/N) group. Lancet Oncol 2015;16:e534-e542.
 MR imaging (Y/N) 16. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy
 Other imaging (Y/N) Oncology Group (RTOG) and the European Organization for
Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol
 Symptoms/examination (Y/N)
Phys 1995;31:1341-1346.
B Type of symptoms
17. Trotti A, Colevas AD, Setser A, et al. CTCAE v3.0: Development of
- Intervention (Y/N) a comprehensive grading system for the adverse effects of cancer
treatment. Semin Radiat Oncol 2003;13:176-181.
If yes, specify type 18. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for
- If available: Patient-reported outcomes score systematic reviews and meta-analyses: The PRISMA statement.
- If available: Data from validated questionnaires PLoS Med 2009;6:e1000097.
such as FACT-BR 19. [No authors listed]. LENT SOMA scales for all anatomic sites. Int J
Radiat Oncol Biol Phys. 1995;31:1049-1091.
As recommended by other HYTEC reports,134 these data 20. Lawrence YR, Li XA, el Naqa I, et al. Radiation dose-volume effects
should be provided in electronic supplements to facilitate in the brain. Int J Radiat Oncol Biol Phys 2010;76:S20-S27.
data pooling and NTCP modeling. 21. Reynolds TA, Jensen AR, Bellairs EE, et al. Dose gradient index for
stereotactic radiosurgery/radiation therapy. Int J Radiat Oncol Biol
Phys 2020;106:604-611.
22. Voges J, Treuer H, Lehrke R, et al. Risk analysis of linac radio-
References surgery in patients with arteriovenous malformation (avm). Acta
Neurochir Suppl 1997;68:118-123.
1. Fabiano AJ, Prasad D, Qiu J. Adverse radiation effect in the brain 23. Flickinger JC, Kondziolka D, Maitz AH, et al. Analysis of neuro-
during cancer radiotherapy. J Radiat Cancer Res 2017;8:135-140. logical sequelae from radiosurgery of arteriovenous malformations:
2. Schultheiss TE, Kun LE, Ang KK, et al. Radiation response of the How location affects outcome. Int J Radiat Oncol Biol Phys 1998;40:
central nervous system. Int J Radiat Oncol Biol Phys 1995;31:1093- 273-278.
1112. 24. Flickinger JC, Kondziolka D, Lunsford LD, et al. Development of a
3. Chao ST, Ahluwalia MS, Barnett GH, et al. Challenges with the model to predict permanent symptomatic postradiosurgery injury for
diagnosis and treatment of cerebral radiation necrosis. Int J Radiat arteriovenous malformation patients. Arteriovenous malformation
Oncol Biol Phys 2013;87:449-457. radiosurgery study group. Int J Radiat Oncol Biol Phys 2000;46:
4. Mehta S, Shah A, Jung H. Diagnosis and treatment options for 1143-1148.
sequelae following radiation treatment of brain tumors. Clin Neurol 25. Cetin IA, Ates R, Dhaens J, et al. Retrospective analysis of linac-
Neurosurg 2017;163:1-8. based radiosurgery for arteriovenous malformations and testing of
5. Vellayappan B, Tan CL, Yong C, et al. Diagnosis and management of the flickinger formula in predicting radiation injury. Strahlenther
radiation necrosis in patients with brain metastases. Front Oncol Onkol 2012;188:1133-1138.
2018;8:395. 26. Herbert C, Moiseenko V, McKenzie M, et al. Factors predictive of
6. Tye K, Engelhard HH, Slavin KV, et al. An analysis of radiation symptomatic radiation injury after linear accelerator-based stereo-
necrosis of the central nervous system treated with bevacizumab. J tactic radiosurgery for intracerebral arteriovenous malformations. Int
Neurooncol 2014;117:321-327. J Radiat Oncol Biol Phys 2012;83:872-877.
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 17

27. Kano H, Flickinger JC, Tonetti D, et al. Estimating the risks of 46. Patel KR, Burri SH, Asher AL, et al. Comparing preoperative with
adverse radiation effects after gamma knife radiosurgery for arte- postoperative stereotactic radiosurgery for resectable brain metasta-
riovenous malformations. Stroke 2017;48:84-90. ses: A multi-institutional analysis. Neurosurgery 2016;79:279-285.
28. Minniti G, Scaringi C, Paolini S, et al. Single-fraction versus mul- 47. Lax I, Karlsson B. Prediction of complications in gamma knife radi-
tifraction (3  9 Gy) stereotactic radiosurgery for large (>2 cm) osurgery of arteriovenous malformation. Acta Oncol 1996;35:49-55.
brain metastases: A comparative analysis of local control and risk of 48. Flickinger JC, Kondziolka D, Pollock BE, et al. Complications from
radiation-induced brain necrosis. Int J Radiat Oncol Biol Phys 2016; arteriovenous malformation radiosurgery: Multivariate analysis and
95:1142-1148. risk modeling. Int J Radiat Oncol Biol Phys 1997;38:485-490.
29. Korytko T, Radivoyevitch T, Colussi V, et al. 12 Gy gamma knife 49. Miyawaki L, Dowd C, Wara W, et al. Five year results of linac
radiosurgical volume is a predictor for radiation necrosis in non-avm radiosurgery for arteriovenous malformations: Outcome for large
intracranial tumors. Int J Radiat Oncol Biol Phys 2006;64:419-424. AVMs. Int J Radiat Oncol Biol Phys 1999;44:1089-1106.
30. Blonigen BJ, Steinmetz RD, Levin L, et al. Irradiated volume as a 50. Barker FG 2nd, Butler WE, Lyons S, et al. Dose-volume prediction of
predictor of brain radionecrosis after linear accelerator stereotactic radiation-related complications after proton beam radiosurgery
radiosurgery. Int J Radiat Oncol Biol Phys 2010;77:996-1001. for cerebral arteriovenous malformations. J Neurosurg 2003;99:254-263.
31. Minniti G, Clarke E, Lanzetta G, et al. Stereotactic radiosurgery for 51. Friedman WA, Bova FJ, Bollampally S, et al. Analysis of factors
brain metastases: Analysis of outcome and risk of brain radio- predictive of success or complications in arteriovenous malformation
necrosis. Radiat Oncol 2011;6:48. radiosurgery. Neurosurgery 2003;52:296-307; discussion 307-308.
32. Ohtakara K, Hayashi S, Nakayama N, et al. Significance of target 52. Levegrun S, Hof H, Essig M, et al. Radiation-induced changes of
location relative to the depth from the brain surface and high-dose brain tissue after radiosurgery in patients with arteriovenous mal-
irradiated volume in the development of brain radionecrosis after formations: Correlation with dose distribution parameters. Int J
micromultileaf collimator-based stereotactic radiosurgery for brain Radiat Oncol Biol Phys 2004;59:796-808.
metastases. J Neurooncol 2012;108:201-209. 53. Varlotto JM, Flickinger JC, Niranjan A, et al. Analysis of tumor
33. Sneed PK, Mendez J, Vemer-van den Hoek JG, et al. Adverse radiation control and toxicity in patients who have survived at least one year
effect after stereotactic radiosurgery for brain metastases: Incidence, after radiosurgery for brain metastases. Int J Radiat Oncol Biol Phys
time course, and risk factors. J Neurosurg 2015;123:373-386. 2003;57:452-464.
34. Ernst-Stecken A, Ganslandt O, Lambrecht U, et al. Phase II trial of 54. Valery CA, Cornu P, Noel G, et al. Predictive factors of radiation
hypofractionated stereotactic radiotherapy for brain metastases: Re- necrosis after radiosurgery for cerebral metastases. Stereotact Funct
sults and toxicity. Radiother Oncol 2006;81:18-24. Neurosurg 2003;81:115-119.
35. Minniti G, Esposito V, Clarke E, et al. Multidose stereotactic radi- 55. Williams BJ, Suki D, Fox BD, et al. Stereotactic radiosurgery for
osurgery (9 Gy  3) of the postoperative resection cavity for treat- metastatic brain tumors: A comprehensive review of complications. J
ment of large brain metastases. Int J Radiat Oncol Biol Phys 2013; Neurosurg 2009;111:439-448.
86:623-629. 56. Eaton BR, Gebhardt B, Prabhu R, et al. Hypofractionated radio-
36. Minniti G, D’Angelillo RM, Scaringi C, et al. Fractionated stereo- surgery for intact or resected brain metastases: Defining the optimal
tactic radiosurgery for patients with brain metastases. J Neurooncol dose and fractionation. Radiat Oncol 2013;8:135.
2014;117:295-301. 57. Broemme J, Abu-Isa J, Kottke R, et al. Adjuvant therapy after
37. Dore M, Martin S, Delpon G, et al. Stereotactic radiotherapy resection of brain metastases. Frameless image-guided linac-based
following surgery for brain metastasis: Predictive factors for local radiosurgery and stereotactic hypofractionated radiotherapy. Strah-
control and radionecrosis. Cancer Radiother 2017;21:4-9. lenther Onkol 2013;189:765-770.
38. Inoue HK, Seto K, Nozaki A, et al. Three-fraction cyberknife 58. Schuttrumpf LH, Niyazi M, Nachbichler SB, et al. Prognostic factors
radiotherapy for brain metastases in critical areas: Referring to the for survival and radiation necrosis after stereotactic radiosurgery
risk evaluating radiation necrosis and the surrounding brain volumes alone or in combination with whole brain radiation therapy for 1-3
circumscribed with a single dose equivalence of 14 Gy (v14). J cerebral metastases. Radiat Oncol 2014;9:105.
Radiat Res 2013;54:727-735. 59. Ahmed KA, Freilich JM, Abuodeh Y, et al. Fractionated stereotactic
39. Inoue HK, Sato H, Seto K, et al. Five-fraction cyberknife radio- radiotherapy to the post-operative cavity for radioresistant and
therapy for large brain metastases in critical areas: Impact on the radiosensitive brain metastases. J Neurooncol 2014;118:179-186.
surrounding brain volumes circumscribed with a single dose equiv- 60. Kirkpatrick JP, Wang Z, Sampson JH, et al. Defining the optimal
alent of 14 Gy (v14) to avoid radiation necrosis. J Radiat Res 2014; planning target volume in image-guided stereotactic radiosurgery of
55:334-342. brain metastases: Results of a randomized trial. Int J Radiat Oncol
40. Chin LS, Ma L, DiBiase S. Radiation necrosis following gamma Biol Phys 2015;91:100-108.
knife surgery: A case-controlled comparison of treatment parameters 61. Cho YH, Lee JM, Lee D, et al. Experiences on two different ste-
and long-term clinical follow up. J Neurosurg 2001;94:899-904. reotactic radiosurgery modalities of gamma knife and cyberknife in
41. Peng L, Grimm J, Gui C, et al. Updated risk models demonstrate low treating brain metastases. Acta Neurochirurgica 2015;157:2003-
risk of symptomatic radionecrosis following stereotactic radiosurgery 2009.
for brain metastases in the modern era. Surg Neurol Int 2018;10:32. 62. Jeong WJ, Park JH, Lee EJ, et al. Efficacy and safety of fractionated
42. Milano MT, Sharma M, Soltys SG, et al. Radiation-induced edema stereotactic radiosurgery for large brain metastases. J Korean Neu-
after single-fraction or multifraction stereotactic radiosurgery for rosurg Soc 2015;58:217-224.
meningioma: A critical review. Int J Radiat Oncol Biol Phys 2018; 63. Zhuang H, Zheng Y, Wang J, et al. Analysis of risk and predictors of
101:344-357. brain radiation necrosis after radiosurgery. Oncotarget 2016;7:7773-
43. Fahrig A, Ganslandt O, Lambrecht U, et al. Hypofractionated ste- 7779.
reotactic radiotherapy for brain metastasesdResults from three 64. Nichol A, Ma R, Hsu F, et al. Volumetric radiosurgery for 1 to 10
different dose concepts. Strahlenther Onkol 2007;183:625-630. brain metastases: A multicenter, single-arm, phase 2 study. Int J
44. Inoue HK, Sato H, Suzuki Y, et al. Optimal hypofractionated Radiat Oncol Biol Phys 2016;94:312-321.
conformal radiotherapy for large brain metastases in patients with 65. Hanna A, Boggs DH, Kwok Y, et al. What predicts early volumetric
high risk factors: A single-institutional prospective study. Radiat edema increase following stereotactic radiosurgery for brain metas-
Oncol 2014;9:231. tases? J Neurooncol 2016;127:303-311.
45. Eaton BR, La Riviere MJ, Kim S, et al. Hypofractionated radio- 66. Abuodeh Y, Ahmed KA, Naghavi AO, et al. Postoperative stereo-
surgery has a better safety profile than single fraction radiosurgery tactic radiosurgery using 5-Gy  5 sessions in the management of
for large resected brain metastases. J Neurooncol 2015;123:103-111. brain metastases. World Neurosurg 2016;90:58-65.
18 Milano et al. International Journal of Radiation Oncology  Biology  Physics

67. Ishihara T, Yamada K, Harada A, et al. Hypofractionated stereotactic brain metastases: A dose-escalation study [e-pub ahead of print]. J
radiotherapy for brain metastases from lung cancer: Evaluation of Neurosurg. https://doi.org/10.3171/2019.5.JNS19222. Accessed
indications and predictors of local control. Strahlenther Onkol 2016; September 9, 2020.
192:386-393. 89. Dohm A, McTyre ER, Okoukoni C, et al. Staged stereotactic radio-
68. Navarria P, Pessina F, Cozzi L, et al. Hypo-fractionated stereotactic surgery for large brain metastases: Local control and clinical out-
radiotherapy alone using volumetric modulated ARC therapy for comes of a one-two punch technique. Neurosurgery 2017;83:114-
patients with single, large brain metastases unsuitable for surgical 121.
resection. Radiat Oncol 2016;11:76. 90. Faruqi S, Ruschin M, Soliman H, et al. Adverse radiation effect after
69. Pessina F, Navarria P, Cozzi L, et al. Outcome evaluation of oligo- hypofractionated stereotactic radiosurgery in 5 daily fractions for
metastatic patients treated with surgical resection followed by surgical cavities and intact brain metastases. Int J Radiat Oncol Biol
hypofractionated stereotactic radiosurgery (HSRS) on the tumor bed, Phys 2020;106:772-779.
for single, large brain metastases. PLoS One 2016;11:e0157869. 91. Minniti G, Capone L, Nardiello B, et al. Neurological outcome and
70. Bennion NR, Malouff T, Verma V, et al. A comparison of clinical and memory performance in patients with 10 or more brain metastases
radiologic outcomes between frame-based and frameless stereotactic treated with frameless linear accelerator (linac)-based stereotactic
radiosurgery for brain metastases. Pract Radiat Oncol 2016;6:e283- radiosurgery. J Neurooncol 2020;148:47-55.
e290. 92. Milano MT, Usuki KY, Walter KA, et al. Stereotactic radiosurgery
71. Lima L, Sharim J, Levin-Epstein R, et al. Hypofractionated stereo- and hypofractionated stereotactic radiotherapy: Normal tissue dose
tactic radiosurgery and radiotherapy to large resection cavity of constraints of the central nervous system. Cancer Treat Rev 2011;37:
metastatic brain tumors. World Neurosurg 2017;97:571-579. 567-578.
72. Voges J, Treuer H, Sturm V, et al. Risk analysis of linear accelerator 93. Miller JA, Bennett EE, Xiao R, et al. Association between radiation
radiosurgery. Int J Radiat Oncol Biol Phys 1996;36:1055-1063. necrosis and tumor biology after stereotactic radiosurgery for brain
73. Nakamura JL, Verhey LJ, Smith V, et al. Dose conformity of gamma metastasis. Int J Radiat Oncol Biol Phys 2016;96:1060-1069.
knife radiosurgery and risk factors for complications. Int J Radiat 94. Prabhu RS, Press RH, Patel KR, et al. Single-fraction stereotactic
Oncol Biol Phys 2001;51:1313-1319. radiosurgery (SRS) alone versus surgical resection and SRS for large
74. Ganz JC, Reda WA, Abdelkarim K. Adverse radiation effects after brain metastases: A multi-institutional analysis. Int J Radiat Oncol
gamma knife surgery in relation to dose and volume. Acta Neurochir Biol Phys 2017;99:459-467.
(Wien) 2009;151:9-19. 95. Ma L, Petti PL, Smith V, et al. Functional relationship between the
75. Kjellberg RN, Hanamura T, Davis KR, et al. Bragg-peak proton- volume of a near-target peripheral isodose line and its dose value for
beam therapy for arteriovenous malformations of the brain. N Engl J gamma knife radiosurgery. In: McDermott MW, editor. Radiosurgery.
Med 1983;309:269-274. Basel: Karger; 2010. p. 75-83.
76. Marks LB, Spencer DP. The influence of volume on the tolerance of 96. Ma L, Sahgal A, Descovich M, et al. Equivalence in dose fall-off for
the brain to radiosurgery. J Neurosurg 1991;75:177-180. isocentric and nonisocentric intracranial treatment modalities and its
77. Flickinger JC. An integrated logistic formula for prediction of impact on dose fractionation schemes. Int J Radiat Oncol Biol Phys
complications from radiosurgery. Int J Radiat Oncol Biol Phys 1989; 2010;76:943-948.
17:879-885. 97. Okunieff P, Morgan D, Niemierko A, et al. Radiation dose-response
78. Flickinger JC, Schell MC, Larson DA. Estimation of complications of human tumors. Int J Radiat Oncol Biol Phys 1995;32:1227-1237.
for linear accelerator radiosurgery with the integrated logistic for- 98. Levegrun S, Jackson A, Zelefsky MJ, et al. Fitting tumor control
mula. Int J Radiat Oncol Biol Phys 1990;19:143-148. probability models to biopsy outcome after three-dimensional
79. Flickinger JC, Lunsford LD, Kondziolka D. Dose-volume consider- conformal radiation therapy of prostate cancer: Pitfalls in deducing
ations in radiosurgery. Stereotact Funct Neurosurg 1991;57:99-105. radiobiologic parameters for tumors from clinical data. Int J Radiat
80. Flickinger JC, Lunsford LD, Wu A, et al. Predicted dose-volume Oncol Biol Phys 2001;51:1064-1080.
isoeffect curves for stereotactic radiosurgery with the 60Co gamma 99. Milano MT, Grimm J, Soltys SG, et al. Single- and multi-fraction
unit. Acta Oncol 1991;30:363-367. stereotactic radiosurgery dose tolerances of the optic pathways [e-
81. Starke RM, Kano H, Ding D, et al. Stereotactic radiosurgery for pub ahead of print]. Int J Radiat Oncol Biol Phys. https://doi.org/10.
cerebral arteriovenous malformations: Evaluation of long-term out- 1016/j.ijrobp.2018.01.053. Accessed September 9, 2020.
comes in a multicenter cohort. J Neurosurg 2017;126:36-44. 100. Benedict SH, Yenice KM, Followill D, et al. Stereotactic body ra-
82. Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical treat- diation therapy: The report of AAPM Task Group 101. Med Phys
ment of recurrent previously irradiated primary brain tumors and 2010;37:4078-4101.
brain metastases: Final report of RTOG protocol 90-05. Int J Radiat 101. Mayo C, Yorke E, Merchant TE. Radiation associated brainstem
Oncol Biol Phys 2000;47:291-298. injury. Int J Radiat Oncol Biol Phys 2010;76:S36-S41.
83. Mohammadi AM, Schroeder JL, Angelov L, et al. Impact of the 102. Trifiletti DM, Lee CC, Kano H, et al. Stereotactic radiosurgery for
radiosurgery prescription dose on the local control of small (2 cm or brainstem metastases: An international cooperative study to define
smaller) brain metastases. J Neurosurg 2017;126:735-743. response and toxicity. Int J Radiat Oncol Biol Phys 2016;96:280-288.
84. Amsbaugh M, Pan J, Yusuf MB, et al. Dose-volume response relation- 103. Shi S, Sandhu N, Jin MC, et al. Stereotactic radiosurgery for resected
ship for brain metastases treated with frameless single-fraction linear brain metastases: Single-institutional experience of over 500 cavities.
accelerator-based stereotactic radiosurgery. Cureus 2016;8:e587. Int J Radiat Oncol Biol Phys 2020;106:764-771.
85. Shehata MK, Young B, Reid B, et al. Stereotatic radiosurgery of 468 104. Akanda ZZ, Hong W, Nahavandi S, et al. Post-operative stereotactic
brain metastases < or Z2 cm: Implications for SRS dose and whole radiosurgery following excision of brain metastases: A systematic
brain radiation therapy. Int J Radiat Oncol Biol Phys 2004;59:87-93. review and meta-analysis. Radiother Oncol 2020;142:27-35.
86. Shuryak I, Carlson DJ, Brown JM, et al. High-dose and fractionation 105. Patel RA, Lock D, Helenowski IB, et al. Postoperative stereotactic
effects in stereotactic radiation therapy: Analysis of tumor control radiosurgery for patients with resected brain metastases: A volu-
data from 2965 patients. Radiother Oncol 2015;115:327-334. metric analysis. J Neurooncol 2018;140:395-401.
87. Lehrer EJ, Peterson JL, Zaorsky NG, et al. Single versus multi- 106. Tanenbaum DG, Buchwald ZS, Jhaveri J, et al. Dosimetric
fraction stereotactic radiosurgery for large brain metastases: An in- factors related to radiation necrosis after 5-fraction radiosurgery for
ternational meta-analysis of 24 trials. Int J Radiat Oncol Biol Phys patients with resected brain metastases. Pract Radiat Oncol 2020;10:
2019;103:618-630. 36-43.
88. Kim KH, Kong DS, Cho KR, et al. Outcome evaluation of patients 107. Ilyas A, Chen CJ, Ding D, et al. Radiation-induced changes after
treated with fractionated gamma knife radiosurgery for large (>3 cm) stereotactic radiosurgery for brain arteriovenous malformations: A
Volume -  Number -  2020 HyTEC: Brain radiosurgery dose tolerances 19

systematic review and meta-analysis. Neurosurgery 2018;83:365- 122. Martin AM, Cagney DN, Catalano PJ, et al. Immunotherapy and
376. symptomatic radiation necrosis in patients with brain metastases
108. Zengpanpan Y, Xiaolin A, You C. Letter: Radiation-induced changes treated with stereotactic radiation. JAMA Oncol 2018;4:1123-1124.
after stereotactic radiosurgery for brain arteriovenous malformations. 123. Lehrer EJ, Peterson J, Brown PD, et al. Treatment of brain metastases
Neurosurgery 2018;82:E75-E76. with stereotactic radiosurgery and immune checkpoint inhibitors: An
109. Jimenez RB, Alexander BM, Mahadevan A, et al. The impact of international meta-analysis of individual patient data. Radiother
different stereotactic radiation therapy regimens for brain metastases Oncol 2019;130:104-112.
on local control and toxicity. Adv Radiat Oncol 2017;2:391-397. 124. Helis CA, Hughes RT, Glenn CW, et a l. Predictors of adverse ra-
110. McKay WH, McTyre ER, Okoukoni C, et al. Repeat stereotactic diation effect in brain metastasis patients treated with stereotactic
radiosurgery as salvage therapy for locally recurrent brain metastases radiosurgery and immune checkpoint inhibitor therapy. Int J Radiat
previously treated with radiosurgery. J Neurosurg 2017;127:148-156. Oncol Biol Phys 2020;108:295-303.
111. Awad AJ, Walcott BP, Stapleton CJ, et al. Repeat radiosurgery for 125. Lehrer EJ, McGee HM, Peterson JL, et al. Stereotactic radiosurgery
cerebral arteriovenous malformations. J Clin Neurosci 2015;22:945- and immune checkpoint inhibitors in the management of brain me-
950. tastases. Int J Mol Sci 2018;19:3054.
112. Minniti G, Scaringi C, Paolini S, et al. Repeated stereotactic radio- 126. Hanna GG, Murray L, Patel R, et al. UK consensus on normal tissue
surgery for patients with progressive brain metastases. J Neurooncol dose constraints for stereotactic radiotherapy. Clin Oncol (R Coll
2016;126:91-97. Radiol) 2018;30:5-14.
113. Kwon KY, Kong DS, Lee JI, et al. Outcome of repeated radiosurgery 127. UK Consortium: Stereotactic ablative body radiation therapy
for recurrent metastatic brain tumors. Clin Neurol Neurosurg 2007; (SABR): A resource. Available at: https://www.sabr.org.uk/wp-
109:132-137. content/uploads/2019/04/SABRconsortium-guidelines-2019-v6.1.0.
114. Rana N, Pendyala P, Cleary RK, et al. Long-term outcomes after pdf. Accessed May 1, 2020.
salvage stereotactic radiosurgery (SRS) following in-field failure of 128. Cummings M, Youn P, Bergsma DP, et al. Single-fraction radio-
initial SRS for brain metastases. Front Oncol 2017;7:279. surgery using conservative doses for brain metastases: Durable re-
115. Koffer P, Chan J, Rava P, et al. Repeat stereotactic radiosurgery for sponses in select primaries with limited toxicity. Neurosurgery 2017;
locally recurrent brain metastases. World Neurosurg 2017;104:589- 83:437-444.
593. 129. Flickinger JC, Kondziolka D, Lunsford LD, et al. A multi-institu-
116. Kim JM, Miller JA, Kotecha R, et al. The risk of radiation necrosis tional analysis of complication outcomes after arteriovenous
following stereotactic radiosurgery with concurrent systemic thera- malformation radiosurgery. Int J Radiat Oncol Biol Phys 1999;44:
pies. J Neurooncol 2017;133:357-368. 67-74.
117. Shen CJ, Kummerlowe MN, Redmond KJ, et al. Stereotactic radio- 130. Sebastian NT, Glenn C, Hughes R, et al. Linear accelerator-based
surgery: Treatment of brain metastasis without interruption of sys- radiosurgery is associated with lower incidence of radionecrosis
temic therapy. Int J Radiat Oncol Biol Phys 2016;95:735-742. compared with gamma knife for treatment of multiple brain metas-
118. Colaco RJ, Martin P, Kluger HM, et al. Does immunotherapy in- tases. Radiother Oncol 2020;147:136-143.
crease the rate of radiation necrosis after radiosurgical treatment of 131. Navarria P, Clerici E, Carta G, et al. Randomized phase III trial
brain metastases? J Neurosurg 2016;125:17-23. comparing gamma knife and linac based (EDGE) approaches for
119. Tran TT, Jilaveanu LB, Omuro A, et al. Complications associated brain metastases radiosurgery: Results from the Gadget trial. Int J
with immunotherapy for brain metastases. Curr Opin Neurol 2019; Radiat Oncol Biol Phys 2018;102(suppl):S143-S144.
32:907-916. 132. Clerici E, Navarria P, Franzese C, et al. Randomized phase III trial
120. Chen L, Douglass J, Kleinberg L, et al. Concurrent immune check- comparing gamma knife (GK) and linac based (EDGE) radiosurgery
point inhibitors and stereotactic radiosurgery for brain metastases in for brain metastases from solid tumors: Results from the GADGET
non-small cell lung cancer, melanoma, and renal cell carcinoma. Int J trial. Neuro Oncol 2018;20(suppl 3):iii227.
Radiat Oncol Biol Phys 2018;100:916-925. 133. Chung C, Torrens M, Paddick I, et al. Radiosurgery nomenclature: A
121. Shepard MJ, Xu Z, Donahue J, et al. Stereotactic radiosurgery with confusion of tongues. Int J Radiat Oncol Biol Phys 2015;92:512-513.
and without checkpoint inhibition for patients with metastatic non- 134. Kong FS, Moiseenko V, Zhao J, et al. Organs at risk considerations for
small cell lung cancer to the brain: A matched cohort study [e-pub thoracic stereotactic body radiation therapy: What is safe for lung pa-
ahead of print]. J Neurosurg. https://doi.org/10.3171/2019.4. renchyma [e-pub ahead of print]? Int J Radiat Oncol Biol Phys. https://
JNS19822. Accessed September 9, 2020. doi.org/10.1016/j.ijrobp.2018.11.028. Accessed September 9, 2020.

You might also like