Delayed radiation-induced vasculitic leukoencephalopathy related to stereotactic radiosurgery (SRS) of brain metastases has been reported to manifest clinically 9 to 18 months after treatment. Immune-modulating therapies have been introduced to treatment regimens for malignancies with metastatic predilection to the brain. The interaction of these systemic therapies with other modalities of treatment for brain metastases, namely, SRS, has not been fully characterized. We report two patients with metastatic malignancies to the brain who received SRS followed by immunotherapy with monoclonal antibodies (mAb) to programmed death 1 (PD-1). Both patients appeared to have early clinical and radiologic progression of their treated lesions, which was highly suspicious for tumor progression. Both patients underwent surgical resection of their lesions and the material was submitted for histopathologic examination. Pathologic examination in both cases showed predominantly radiation-induced changes characterized by reactive astrocytosis and vascular wall infiltration by T lymphocytes. The accelerated response to SRS in these two patients was temporally related to the initiation of immunotherapy. We propose a possible biologic interaction between SRS and the PD-1 mAbs. Additionally, awareness of this potential occurrence is critical for accurate interpretation and proper management of clinical and radiologic findings in these patients. Cancer Immunol Res; 4(6); 481–7. ©2016 AACR.

Radiographic changes—unrelated to tumor after stereotactic radiosurgery (SRS) for brain metastasis are well documented (1–5). Classically, these changes manifest clinically months to years after SRS (5, 6). The addition of immune-modulating therapies to systemic treatment paradigms is becoming standard of care in some malignancies that tend to metastasize to the brain, in particular melanoma and non–small cell lung cancer (NSCLC; refs. 7–9). A number of studies have shown activity of immune checkpoint inhibitors in patients with brain metastases, and combinations of these drugs with SRS may be used more frequently in the future (10–12). However, the effects of immunotherapy after SRS on brain tumors and brain tissue adjacent to SRS-treated tumors have not been fully characterized. Programmed death 1 (PD-1) is a human cell-surface coinhibitory receptor expressed on activated T cells that suppresses T-cell effector function when engaged by PD ligand (PD-L1 or PD-L2; ref. 13). PD-1 antagonist antibodies, such as nivolumab and pembrolizumab, can relieve inhibition of antitumor CD8+ T cells, resulting in durable clinical and radiographic responses (14). In this report, we describe the clinical, radiologic, and pathologic findings in two patients—one with melanoma and one with NSCLC metastatic to the brain—who were treated with SRS to brain metastases prior to systemic treatment with PD-1 monoclonal antibodies (mAb). In both cases, the time frames for clinical progression and the development of radiologic findings were highly suspicious for tumor progression. However, pathologic examination showed findings consistent with an accelerated response to SRS treatment.

Case 1

History and clinical course.

A 45-year-old woman was initially diagnosed with a 2.3-mm ulcerated melanoma on the dorsum of the right foot in 2008. Her sentinel lymph nodes were uninvolved. In 2010 she presented with an inguinal mass comprising four lymph nodes positive for melanoma that harbored a BRAFV600E mutation. She received adjuvant interferon-α for 12 months and was then found to have lung and brain metastases. She was subsequently treated with a number of systemic therapies between 2010 and 2013, including ipilimumab (anti–CTLA-4), vemurafenib (BRAFV600E inhibitor), carboplatin and paclitaxel (chemotherapy), and dabrafenib and trametinib (MEK inhibition). During that period of time, five brain metastatic lesions ranging in size between 0.3 cm and 0.6 cm were treated with SRS. Three of these lesions were treated before the patient started vemurafenib. The other two were treated while the patient was on vemurafenib. After an initial slight increase in size, these lesions regressed and remained stable at a size approximately similar to their original size. She also remained asymptomatic throughout the entire follow-up period. In 2014, MRI of the brain showed a new lesion in the right anterior temporal lobe measuring 1.3 cm in its greatest dimension. This lesion was also treated with SRS in the form of 22 Gy to the 50% isodose surface. Six weeks after SRS, the patient developed new-onset seizures that were controlled by levetiracetam. Five months after this last radiosurgical treatment, she started therapy with pembrolizumab at 2 mg/kg/3 weeks. One month later, she reported progressively worsening headaches associated with an increase in seizure frequency not controlled with medical therapy. A repeat MRI showed increase in the size of the enhancing temporal lesion with statistically significant increase in surrounding edema. Given the early timing of the MRI changes, there was concern about progressive tumor growth, and the patient underwent craniotomy for local lesion management. The lesion was resected en bloc. Postoperatively, her headaches and seizures resolved.

Radiologic examination.

The patient has had multiple brain metastases during the course of her disease. Of interest is the 1.3 × 1.1 × 1.0 cm gadolinium enhancing lesion identified in the right anterior temporal lobe found on surveillance MRI (Fig. 1A and B). Two months after SRS, this lesion had decreased in size to 1.0 cm in its greatest dimension (Fig. 1C and D). One month after the initiation of treatment with pembrolizumab (6 months after SRS), gadolinium-enhanced MRI showed that the temporal lesion had increased in size to 2.0 cm. This change was associated with significant increase in surrounding T2 and FLAIR signal abnormality consistent with perilesional edema (Fig. 1E and F). Postoperative MRI 3 months after surgical resection confirmed resection of the temporal lesion and resolution of right temporal lobe edema (Fig. 1G and H).

Figure 1.

A and B, T1-weighted and FLAIR images showing contrast-enhancing lesion in the right anterior temporal lobe with associated perilesional edema. C and D, 2 months after SRS, the lesion and the associated FLAIR signal have decreased in size. E and F, 1 month after pembrolizumab (6 months after SRS), the lesion and the associated perilesional edema have significantly increased in size. G and H, postresection images showing mild residual FLAIR signal.

Figure 1.

A and B, T1-weighted and FLAIR images showing contrast-enhancing lesion in the right anterior temporal lobe with associated perilesional edema. C and D, 2 months after SRS, the lesion and the associated FLAIR signal have decreased in size. E and F, 1 month after pembrolizumab (6 months after SRS), the lesion and the associated perilesional edema have significantly increased in size. G and H, postresection images showing mild residual FLAIR signal.

Close modal

Pathologic examination.

A biopsy of a left occipital brain lesion previously performed to confirm metastatic disease showed a pleomorphic malignant neoplasm staining positive with Melan A and HMB-45, consistent with metastatic melanoma. Histologic examination of the treated anterior temporal lesion, however, showed a central zone of predominantly extracellular melanin pigment and shadows of dying cells with intracellular melanin pigment (Fig. 2A and B), surrounded by a rim of necrosis with no viable tumor cells (Fig. 2C). At the periphery of the necrosis were areas of prominent eosinophilic hyalinization of blood vessel wall and intense vasculitis characterized by infiltration by CD3+ T lymphocytes and CD68+ microglial cells of the vessel walls (Fig. 2D–F). CD20 highlighted rare B lymphocytes, and PD-L1 (E1L3N antibody) showed no staining in lesional tissue. These features were consistent with delayed treatment-related inflammatory changes that have been previously reported post-SRS.

Figure 2.

A and B, representative hematoxylin and eosin (H&E) sections showing necrotic tumoral tissue with abundant melanin pigment and admixed hemorrhage (×4 and ×20). C, an H&E section of the tissue surrounding the melanin nodule revealing only necrotic material (×20). D and E, the brain tissue further out from the melanin nodule showing marked radiation-induced changes characterized by vascular wall thickening, hyalinization, and lymphocytic infiltration (×10 and ×40). F, CD3 immunostaining highlighting the predominantly lymphocytic nature of the inflammatory infiltrate in the vasculitic lesions (×40).

Figure 2.

A and B, representative hematoxylin and eosin (H&E) sections showing necrotic tumoral tissue with abundant melanin pigment and admixed hemorrhage (×4 and ×20). C, an H&E section of the tissue surrounding the melanin nodule revealing only necrotic material (×20). D and E, the brain tissue further out from the melanin nodule showing marked radiation-induced changes characterized by vascular wall thickening, hyalinization, and lymphocytic infiltration (×10 and ×40). F, CD3 immunostaining highlighting the predominantly lymphocytic nature of the inflammatory infiltrate in the vasculitic lesions (×40).

Close modal

Follow-up.

The patient restarted pembrolizumab within 2 weeks of surgery and remains headache and seizure free 4 months after surgery. Her last MRI scan 3 months after surgery showed markedly reduced edema in the right temporal lobe and stable appearance of other lesions. She continues surveillance MRI for follow-up.

Case 2

Clinical course.

A 59-year-old woman with smoking history presented with confusion and speech difficulty. MRI of the brain showed a left frontotemporal enhancing lesion. Further workup revealed a right upper lobe lung mass and several enlarged hilar lymph nodes. Mediastinoscopy was performed, and pathology showed poorly differentiated adenocarcinoma of the lung. The brain lesion was therefore presumed to be a brain metastasis and treated with SRS comprising 20 Gy to the 50% isodose surface. One month after SRS, MRI showed that the size of the lesion and perilesional edema had decreased. Systemic therapy was started with nivolumab and ipilimumab on a clinical trial (NCT01454102). Within 2 months of initiation of immunotherapy, extracerebral imaging demonstrated remarkable response to therapy. However, brain MRI showed significant increase in the size of the treated lesion and perilesional edema causing mild midline shift. The patient developed symptoms of intermittent confusion, requiring treatment with steroids, and craniotomy was performed for local lesional control.

Radiologic examination.

Initial MRI showed a 2.3 × 2.0 cm enhancing mass in the left frontotemporal region. FLAIR sequence showed marked perilesional edema (Fig. 3A and B). MRI performed 1 month after SRS showed a marked decrease in the size of the lesion (1.4 × 1.2 cm) and mild improvement in the perilesional edema (Fig. 3C and D). MRI 2 months after the initiation of immunotherapy and 3 months after SRS showed that the lesion had regrown (2.7 × 2.3 cm), and the increased perilesional edema was now associated with mild midline shift (Fig. 3E and F). The right upper lobe lung lesion, conversely, showed marked response to immunotherapy, decreasing in size from 5.2 × 4.0 cm before treatment to 3.6 × 2.3 cm after 2 months of treatment. Similarly, several enlarged mediastinal lymph nodes showed reduction in size following therapy. Six-week postoperative brain MRI showed complete resection of the lesion and marked decrease in the amount of perilesional edema with resolution of midline shift (Fig. 3G and H).

Figure 3.

A and B, T1-weighted and FLAIR images showing contrast-enhancing lesion in the left frontotemporal region with associated perilesional edema. C and D, 1 month after SRS, the lesion has decreased in size with minimal improvement in the associated edema. E and F, 2 months after treatment with nivolumab and ipilimumab (3 months after SRS), the lesion and the associated perilesional edema have significantly increased in size with mild midline shift. G and H, postresection images showing near-total resection with markedly reduced FLAIR signal.

Figure 3.

A and B, T1-weighted and FLAIR images showing contrast-enhancing lesion in the left frontotemporal region with associated perilesional edema. C and D, 1 month after SRS, the lesion has decreased in size with minimal improvement in the associated edema. E and F, 2 months after treatment with nivolumab and ipilimumab (3 months after SRS), the lesion and the associated perilesional edema have significantly increased in size with mild midline shift. G and H, postresection images showing near-total resection with markedly reduced FLAIR signal.

Close modal

Pathologic examination.

Tissue from the patient's mediastinal lymph node biopsy showed metastatic adenocarcinoma staining positively for CK7 and TTF-1, confirming its lung origin. Molecular profiling showed no genetic alterations in EGFR, ALK, KRAS, or ROS. Pathologic examination of the patient's irradiated left frontotemporal lesion showed a large central area of necrosis with minute foci of residual tumor cells encased within the necrosis (Fig. 4A–C). Surrounding the necrosis was evidence of marked vasculitis, reactive astrocytosis, and hyalinization of blood vessels (Fig. 4D). The blood vessel walls were infiltrated by CD3+ T cells and CD68-positive microglial cells (Fig. 4E and F). Similar to the first case, rare B cells were present, and no PD-L1 staining of the lesional tissue was identified.

Figure 4.

A and B, representative H&E sections of the resected lesion following SRS showing predominant necrosis and minute collections of tumor cells (upper right; ×4 and ×10). C, CK7 immunostain highlights the small collection of tumoral cells (×10). D, a representative H&E section of the surrounding brain tissue depicting an intense vasculitic process characterized by hyalinization of vessel walls and infiltration by inflammatory cells (×20). E, CD3 immunostain highlights the T lymphocytes infiltrating the vessel wall (×20). F, CD68 immunostain highlights the macrophages and microglial cells infiltrating vessel walls and the surrounding brain tissue (×20).

Figure 4.

A and B, representative H&E sections of the resected lesion following SRS showing predominant necrosis and minute collections of tumor cells (upper right; ×4 and ×10). C, CK7 immunostain highlights the small collection of tumoral cells (×10). D, a representative H&E section of the surrounding brain tissue depicting an intense vasculitic process characterized by hyalinization of vessel walls and infiltration by inflammatory cells (×20). E, CD3 immunostain highlights the T lymphocytes infiltrating the vessel wall (×20). F, CD68 immunostain highlights the macrophages and microglial cells infiltrating vessel walls and the surrounding brain tissue (×20).

Close modal

Follow-up.

The patient was able to resume systemic therapy on the clinical trial within 2 weeks of surgery, and she remains asymptomatic neurologically 6 weeks after surgery.

Immune checkpoint inhibitors are revolutionizing the ability to treat metastatic cancer (15–18). Many clinical trials are under way using these agents to treat a variety of cancers, and inhibitors of CTLA-4 and PD-1 have been approved for melanoma (19). Pembrolizumab and nivolumab, inhibitors of PD-1, were the first to be approved for melanoma and are being studied in other diseases and in combination with other drugs (nivolumab was recently approved for squamous cells carcinoma of the lung). The interaction between immune checkpoint inhibitors and standard treatments for brain metastases, such as SRS, however, remains underinvestigated (20, 21).

SRS is well established as a treatment modality for brain metastases (22, 23). Although initially intended to minimize the effect of radiation on the whole brain, several reports have now documented that, in a subset of patients, the high focal dose of radiation to the small rim of normal brain surrounding the metastasis can result in a delayed appearance of progressively enlarging gadolinium-enhancing areas surrounding the isocenter of radiation—typically 9 to 18 months after SRS (2–4). Histologic examination of these areas shows a delayed radiation-induced vasculitic leukoencephalopathy (DRIVL), characterized by myelin loss, infiltrating macrophages, marked reactive astrocytosis, and hyalinization and sclerosis of blood vessels. Additionally, these cases consistently showed marked inflammatory changes, including abundant infiltration of CD3+ T lymphocytes seen diffusely scattered within the parenchyma and infiltrating vascular walls giving a vasculitis-like appearance (2).

In the two cases presented here, routine MRI follow-up scans showed an initial decrease in the size of the irradiated lesions after SRS, as expected. However, only 1 to 2 months after initiation of immune checkpoint inhibitor treatment, follow-up MRI showed regrowth in the size of the treated lesions with statistically significant increase in perilesional edema-causing symptoms. In the first patient, symptoms developed almost 6 months after the administration of SRS. Although uncommon, it is not impossible to have symptomatic regrowth of lesions—unrelated to tumor recurrence—as early as 6 months after treatment, especially in larger lesions (5). However, given the initial size of the metastatic lesion in this patient, the temporal relationship with pembrolizumab administration, and the more typical delayed development of radiographic and clinical changes following SRS, it is thought that immunotherapy likely played an important role in the pathogenesis of this patient's post-SRS lesions.

We postulate that immunotherapy may accentuate or accelerate radiation-induced changes in the perilesional brain tissue, especially changes that are immunologically mediated (20). By blocking the interaction between PD-1 and its receptor ligands, PD-L1 and PD-L2, treatment with drugs such as pembrolizumab and nivolumab results in a robust antitumor immune response (14). Similarly, CTLA-4 inhibits the immune-stimulatory interaction between CD28 and B7-1 or B7-2 (24). Ipilimumab and tremilumab, mAbs to CTLA-4, thus prevent this immune-inhibitory process. The combination of ipilimumab and nivolumab could further enhance immune activation (18). However, in addition to the antitumoral immune activation, several undesired immune-mediated adverse effects have been reported, including immune-mediated pneumonitis, colitis, hepatitis, and hypophysitis (25).

In addition to direct damaging effects on tumoral cells, conclusive evidence exists that radiotherapy induces an immunogenic cell death (26, 27). These effects are mediated by release of tumor antigens and expression of damage-associated molecular patterns, such as upregulation of Fas and ICAM-1, and exposure of calretinin on the cell surface (28, 29). Accordingly, it has been postulated that radiotherapy may work as an inducer for the immune system, driving cytotoxic T cell–mediated destruction of cancer cells, thus augmenting the effect of immune therapy (30, 31). Likewise, evidence supports a disruptive effect of radiation on the blood–brain barrier and the vascular endothelial cells, leading to exposure of otherwise protected nontumoral antigens resulting in immunologically mediated damage of surrounding normal brain tissue (2, 32, 33). Accordingly, one can postulate that, in the setting of an activated immune system, this radiation-induced immunologically mediated process in the tumor and surrounding brain tissue might be exacerbated (20). This observation may also explain the temporal relationship between the changes seen in the treated lesions in these two cases and initiation of immune therapy. Another important consideration is the characteristics of the histologic findings. Although the current report did not show significant histologic differences from our previously reported cases of post-SRS leukoencephalopathy (2, 4), it is possible that as more cases are identified in the future, potential subtle differences in histopathologic appearance after SRS with immunotherapy versus SRS alone will be identified.

Surgical management provided confirmation of diagnosis and rapid symptomatic relief and allowed both patients to resume systemic immunotherapy within several weeks. Awareness of the potential interaction between the effect of SRS and PD-1 mAbs is imperative to avoid the misinterpretation of radiologic changes as tumor progression, which could otherwise lead to inappropriate re-irradiation or cessation of an effective therapy and potentially harmful consequences (6). If this significant interaction between SRS and immunotherapy is confirmed at other centers, changes in management options such as first-line surgical management of brain metastases or lower-dose SRS may need to be considered in patients with brain metastases being offered immunotherapy.

A. Chiang reports serving as a consultant/advisory board member for Genentech/Roche. S. Gettinger reports serving as a consultant/advisory board member for Bristol Myers-Squibb. S.B. Goldberg reports receiving commercial research support from AstraZeneca and Merck. H.M. Kluger reports receiving speakers bureau honoraria from Merck and Regeneron. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.K. Alomari, J. Cohen, S. Gettinger, H.M. Kluger, V.L. Chiang

Development of methodology: A.K. Alomari, A.O. Vortmeyer, V.L. Chiang

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.K. Alomari, A.O. Vortmeyer, A. Chiang, S. Gettinger, H.M. Kluger, V.L. Chiang

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.K. Alomari, A.O. Vortmeyer, S. Gettinger, S. Goldberg, V.L. Chiang

Writing, review, and/or revision of the manuscript: A.K. Alomari, J. Cohen, A.O. Vortmeyer, A. Chiang, S. Gettinger, S. Goldberg, H.M. Kluger, V.L. Chiang

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.K. Alomari

Study supervision: S. Gettinger, V.L. Chiang

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Huber
PE
,
Hawighorst
H
,
Fuss
M
,
van Kaick
G
,
Wannenmacher
MF
,
Debus
J
. 
Transient enlargement of contrast uptake on MRI after linear accelerator (linac) stereotactic radiosurgery for brain metastases
.
Int J Rad O B Phys
2001
;
49
:
1339
49
.
2.
Rauch
PJ
,
Park
HS
,
Knisely
JP
,
Chiang
VL
,
Vortmeyer
AO
. 
Delayed radiation-induced vasculitic leukoencephalopathy
.
Int J Radiat Oncol Biol Phys
2012
;
83
:
369
75
.
3.
Patel
TR
,
McHugh
BJ
,
Bi
WL
,
Minja
FJ
,
Knisely
JP
,
Chiang
VL
. 
A comprehensive review of MR imaging changes following radiosurgery to 500 brain metastases
.
AJNR Am J Neuroradiol
2011
;
32
:
1885
92
.
4.
Alomari
A
,
Rauch
PJ
,
Orsaria
M
,
Minja
FJ
,
Chiang
VL
,
Vortmeyer
AO
. 
Radiologic and histologic consequences of radiosurgery for brain tumors
.
J Neuro-Oncol
2014
;
117
:
33
42
.
5.
Kohutek
ZA
,
Yamada
Y
,
Chan
TA
,
Brennan
CW
,
Tabar
V
,
Gutin
PH
, et al
Long-term risk of radionecrosis and imaging changes after stereotactic radiosurgery for brain metastases
.
J Neurooncol
2015
;
125
:
149
56
.
6.
Ruzevick
J
,
Kleinberg
L
,
Rigamonti
D
. 
Imaging changes following stereotactic radiosurgery for metastatic intracranial tumors: differentiating pseudoprogression from tumor progression and its effect on clinical practice
.
Neurosurg Rev
2014
;
37
:
193
201
;
discussion 201
.
7.
Kenchappa
RS
,
Tran
N
,
Rao
NG
,
Smalley
KS
,
Gibney
GT
,
Sondak
VK
, et al
Novel treatments for melanoma brain metastases
.
Cancer Control J Moffitt Cancer Center
2013
;
20
:
298
306
.
8.
Hurst
R
,
White
DE
,
Heiss
J
,
Lee
DS
,
Rosenberg
SA
,
Schwartzentruber
DJ
. 
Brain metastasis after immunotherapy in patients with metastatic melanoma or renal cell cancer: is craniotomy indicated?
J Immunother
1999
;
22
:
356
62
.
9.
Declerck
S
,
Vansteenkiste
J
. 
Immunotherapy for lung cancer: ongoing clinical trials
.
Future Oncol
2014
;
10
:
91
105
.
10.
Silk
AW
,
Bassetti
MF
,
West
BT
,
Tsien
CI
,
Lao
CD
. 
Ipilimumab and radiation therapy for melanoma brain metastases
.
Cancer Med
2013
;
2
:
899
906
.
11.
Knisely
JP
,
Yu
JB
,
Flanigan
J
,
Sznol
M
,
Kluger
HM
,
Chiang
VL
. 
Radiosurgery for melanoma brain metastases in the ipilimumab era and the possibility of longer survival
.
J Neurosurg
2012
;
117
:
227
33
.
12.
Margolin
K
. 
Ipilimumab in a Phase II trial of melanoma patients with brain metastases
.
Oncoimmunology
2012
;
1
:
1197
9
.
13.
Keir
ME
,
Butte
MJ
,
Freeman
GJ
,
Sharpe
AH
. 
PD-1 and its ligands in tolerance and immunity
.
Ann Rev Immunol
2008
;
26
:
677
704
.
14.
Brahmer
JR
,
Drake
CG
,
Wollner
I
,
Powderly
JD
,
Picus
J
,
Sharfman
WH
, et al
Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates
.
J Clin Oncol
2010
;
28
:
3167
75
.
15.
Turcotte
S
,
Rosenberg
SA
. 
Immunotherapy for metastatic solid cancers
.
Adv Surg
2011
;
45
:
341
60
.
16.
Menzies
AM
,
Long
GV
. 
New combinations and immunotherapies for melanoma: latest evidence and clinical utility
.
Thera Adv Med Oncol
2013
;
5
:
278
85
.
17.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
18.
Wolchok
JD
,
Kluger
H
,
Callahan
MK
,
Postow
MA
,
Rizvi
NA
,
Lesokhin
AM
, et al
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med
2013
;
369
:
122
33
.
19.
Domingues
D
,
Turner
A
,
Silva
MD
,
Marques
DS
,
Mellidez
JC
,
Wannesson
L
, et al
Immunotherapy and lung cancer: current developments and novel targeted therapies
.
Immunotherapy
2014
;
6
:
1221
35
.
20.
Kiess
AP
,
Wolchok
JD
,
Barker
CA
,
Postow
MA
,
Tabar
V
,
Huse
JT
, et al
Stereotactic radiosurgery for melanoma brain metastases in patients receiving ipilimumab: safety profile and efficacy of combined treatment
.
Int J Radiat Oncol Biol Phys
2015
;
92
:
368
75
.
21.
Mathew
M
,
Tam
M
,
Ott
PA
,
Pavlick
AC
,
Rush
SC
,
Donahue
BR
, et al
Ipilimumab in melanoma with limited brain metastases treated with stereotactic radiosurgery
.
Melanoma Res
2013
;
23
:
191
5
.
22.
Aoyama
H
,
Shirato
H
,
Tago
M
,
Nakagawa
K
,
Toyoda
T
,
Hatano
K
, et al
Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a randomized controlled trial
.
JAMA
2006
;
295
:
2483
91
.
23.
Mintz
A
,
Perry
J
,
Spithoff
K
,
Chambers
A
,
Laperriere
N
. 
Management of single brain metastasis: a practice guideline
.
Curr Oncol
2007
;
14
:
131
43
.
24.
O'Day
SJ
,
Hamid
O
,
Urba
WJ
. 
Targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4): a novel strategy for the treatment of melanoma and other malignancies
.
Cancer
2007
;
110
:
2614
27
.
25.
Teply
BA
,
Lipson
EJ
. 
Identification and management of toxicities from immune checkpoint-blocking drugs
.
Oncology
2014
;
28 Suppl 3
:
30
8
.
26.
Demaria
S
,
Bhardwaj
N
,
McBride
WH
,
Formenti
SC
. 
Combining radiotherapy and immunotherapy: a revived partnership
.
Int J Rad Oncol Biol Phys
2005
;
63
:
655
66
.
27.
Formenti
SC
,
Demaria
S
. 
Radiation therapy to convert the tumor into an in situ vaccine
.
Int J Rad Oncol Biol Phys
2012
;
84
:
879
80
.
28.
Chakraborty
M
,
Abrams
SI
,
Camphausen
K
,
Liu
K
,
Scott
T
,
Coleman
CN
, et al
Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy
.
J Immunol
2003
;
170
:
6338
47
.
29.
Obeid
M
,
Tesniere
A
,
Ghiringhelli
F
,
Fimia
GM
,
Apetoh
L
,
Perfettini
JL
, et al
Calreticulin exposure dictates the immunogenicity of cancer cell death
.
Nat Med
2007
;
13
:
54
61
.
30.
Okwan-Duodu
D
,
Pollack
BP
,
Lawson
D
,
Khan
MK
. 
Role of radiation therapy as immune activator in the era of modern immunotherapy for metastatic malignant melanoma
.
Am J Clin Oncol
2015
;
38
:
119
25
.
31.
Stamell
EF
,
Wolchok
JD
,
Gnjatic
S
,
Lee
NY
,
Brownell
I
. 
The abscopal effect associated with a systemic anti-melanoma immune response
.
Int J Rad Oncol Biol Phys
2013
;
85
:
293
5
.
32.
Kureshi
SA
,
Hofman
FM
,
Schneider
JH
,
Chin
LS
,
Apuzzo
ML
,
Hinton
DR
. 
Cytokine expression in radiation-induced delayed cerebral injury
.
Neurosurgery
1994
;
35
:
822
9
;
discussion 9–30
.
33.
Moore
AH
,
Olschowka
JA
,
Williams
JP
,
Paige
SL
,
O'Banion
MK
. 
Radiation-induced edema is dependent on cyclooxygenase 2 activity in mouse brain
.
Rad Res
2004
;
161
:
153
60
.