Radiotherapy in combination with systemic therapies for brain metastases: current status and progress ===================================================================================================== * Lei Liu * Wanqi Chen * Ruopeng Zhang * Yuekun Wang * Penghao Liu * Xin Lian * Fuquan Zhang * Yu Wang * Wenbin Ma ## Abstract Brain metastases (BMs) are the most common cause of intracranial neoplasms in adults with poor prognosis. Most BMs originate from lung cancer, breast cancer, or melanoma. Radiotherapy (RT), including whole brain radiotherapy (WBRT) and stereotactic radiation surgery (SRS), has been widely explored and is considered a mainstay anticancer treatment for BMs. Over the past decade, the advent of novel systemic therapies has revolutionized the treatment of BMs. In this context, there is a strong rationale for using a combination of treatments based on RT, with the aim of achieving both local disease control and extracranial disease control. This review focuses on describing the latest progress in RT as well as the synergistic effects of the optimal combinations of RT and systemic treatment modalities for BMs, to provide perspectives on current treatments. * Brain metastases * radiotherapy * targeted therapy * immunotherapy * systemic therapy ## Introduction Brain metastases (BMs), the most common intracranial neoplasms in adults with invasive cancer, cause a decline in neurological function and overall quality of life (QoL), and may result in mortality from recurrent or untreatable lesions. Approximately 20%–45% of patients with tumors are diagnosed with BMs in their lifetime1. Most BMs originate from lung cancer, breast cancer, or melanoma2. The current standard management for BMs, consisting of a multimodal approach including surgery and/or radiotherapy (RT), systemic therapy, and symptomatic therapy, remains controversial and ineffective3. RT, in the form of whole brain radiotherapy (WBRT) and stereotactic radiation surgery (SRS), is considered a mainstay anticancer modality in the treatment of BMs from solid tumors. RT provides better local tumor control with relatively fewer systemic adverse effects than chemotherapy4,5. Over the past decade, the advent of novel systemic therapies, including chemotherapy, targeted therapy (TT), and immunotherapy (IT), has revolutionized systemic therapy for several malignancies. Consequently, the combination of RT and systemic therapies has been investigated with the aim of achieving both local and extracranial disease control, and possibly improvement in overall survival (OS). Given the lack of updated guidelines for the treatment of patients with BMs, particularly reflecting the introduction of new therapies, this review aims to describe the effects of combining RT with other systemic treatment modalities for BMs, and to provide perspectives on current treatments. ## RT for brain metastases WBRT is primarily used as an alternative therapy and a palliative therapy for BM when local treatment cannot be applied6. Intracranial progression can be completely or partially controlled after WBRT with a low risk of local recurrence and new metastases7–10. However, whether WBRT should be routinely used in patients with BMs remains controversial, because the OS or QoL have not been found to improve after WBRT, whereas impaired cognitive function and decreased QoL have been observed9,11–14. Compared with WBRT, SRS results in a similar OS with improvement in cognitive function; previous studies have indicated that SRS can be used as a standard therapy for patients with limited BM after surgery15–18. For extensive BM, usually defined by the presence of 5 to 10 metastatic lesions, WBRT is the standard treatment recommended in most guidelines6. However, a group of Japanese observational studies have indicated that the performance of SRS alone for extensive BMs is not inferior to that of SRS for 2 to 4 metastatic lesions, and does not result in significant differences in OS4,19,20. Because SRS has relatively fewer adverse effects, these studies have recommended SRS as a treatment option for extensive BM. Whether WBRT plus SRS should be used in patients with BMs is currently unclear. The results of 3 phase III randomized controlled trials (RCTs) have shown that WBRT after SRS, compared with SRS alone, decreases the intracranial recurrence rate, but causes more significant cognitive impairment and does not result in a difference in OS9,12,14. Therefore, SRS alone should be strongly recommended in patients with limited BMs (1–4) or those younger than 50 years who are about to receive active systemic therapy13,14. Patients with a high risk of central failure might benefit from WBRT combined with SRS. Early studies have recommended postoperative RT7,9, but extremely few studies have defined the optimal therapeutic time interval (TTI) between surgery and RT, and TTIs vary among trials21. Recent retrospective studies have reported that preoperative SRS significantly decreases the incidence of meningeal metastases and symptomatic radiation necrosis compared with postoperative SRS, and the OS of preoperative SRS is even not inferior22,23. Retrospective studies have reported that hyperfractionation results in better local control compared with that of lower dose regimens and is associated with a lower risk of necrosis24. ## RT combined with chemotherapy Systemic therapy, an emerging modality, has also shown a great therapeutic potential and has demonstrated activity in the brain. Although most chemotherapeutic drugs have poor penetration of the blood-brain barrier (BBB), RT might help disrupt the BBB to allow the drugs to penetrate into the BMs25. Owing to their poor BBB penetration, chemotherapeutic drugs are generally not preferred as the first-line treatment but instead are used in addition to local treatment7. Recently, several studies have evaluated temozolomide (TMZ), which can penetrate the BBB, and have suggested that TMZ has therapeutic effects in recurrent and progressive BMs. Phase II trials of RT combined with TMZ have suggested that this combination, compared with TMZ alone, significantly improves local control but not the OS25–27. For non-small cell lung cancer (NSCLC) BMs, platinum, either alone or in combination with other drugs, has been widely used as a standard treatment regimen before or after RT. Compared with RT alone, the combination of RT and chemotherapy improves the local response rate of BMs but not the OS28. However, there is no standard chemotherapy regimen for patients with BMs from breast cancer or melanoma, for which RT, chemotherapy, or chemoradiotherapy has shown only a modest effect on survival6. ## RT combined with TT TT is a topic of interest in BM treatment research. The response rates of specific molecular subtypes to targeted drugs are higher than those to cytotoxic chemotherapy drugs. However, owing to their poor prognosis, patients with BMs have long been excluded from clinical trials evaluating systemic therapies in primary solid tumors. Thus, the role of systemic therapies, particularly in combination with RT for BMs, is poorly understood. The use of a combination of RT and TT might be effective, but the safety and exact efficacy in patients with various types of BMs remain unclear. Most reported studies have been retrospective or phase I/II clinical trials with small samples and inconsistent conclusions. Hence, high-level evidence, such as from large-scale phase III RCTs, is urgently needed. Additionally, previous studies have lacked stratification of BMs according to factors such as mutational status. The dose fraction of RT, the timing of administration, and the dose of combined targeted drugs must also be further explored. ### Lung cancer Targeted drugs for lung cancer, mostly NSCLC, mainly include EGFR-tyrosine kinase inhibitors (EGFR-TKIs) and ALK-tyrosine kinase inhibitors (ALK-TKIs). These drugs have shown a high response rate and safety for EGFR- or ALK-mutant NSCLC BMs29–33. A phase III clinical trial (PL03.05: BRAIN) has reported that icotinib is well tolerated and results in a significantly longer OS than chemoradiotherapy34. Many pre-clinical and *in vitro* studies have reported the role of EGFR in regulating radiosensitivity. EGFR inhibitors (EGFRi) can decrease the radioresistance of tumor cells, and EGFR-mutant cell lines show higher radiosensitivity than wild-type cells35–37. Moreover, RT may in turn disrupt the BBB and consequently increase the ability of EGFR-TKIs to permeate the BBB38. Hence, the use of a combination of RT and TKIs might be very effective because of their synergistic effects. However, the results of current clinical studies on combination therapy are contradictory; hence, no firm conclusions have been reached39–43. Several phase II trials have suggested that WBRT combined with TKIs is well tolerated and either does not increase or even decreases neurocognitive toxicity44–47. Another study has suggested that WBRT combined with TKIs increases the incidence of toxic responses or results in no improvement in OS. The RTOG 0320 (phase III) trial has concluded that grade 3 to 5 toxicity significantly increases and that the survival is not improved after the addition of erlotinib to WBRT and SRS48. A multicenter retrospective study comparing SRS followed by EGFR-TKI, WBRT followed by EGFR-TKI, and EGFR-TKI followed by SRS or WBRT has suggested that patients with NSCLC BMs with EGFR mutations who receive SRS followed by EGFR-TKI have the longest OS and are able to avoid the potential neurocognitive sequelae of WBRT49. Preclinical studies have also reported that concurrent RT plus ALK-TKIs synergistically affect tumor growth and microvessel density50,51, thus potentially resulting in better local control. A multi-institutional retrospective analysis has shown prolonged OS in patients with ALK-rearranged NSCLC BMs treated with SRS plus ALK inhibitors52. However, neither prospective nor more convincing retrospective clinical data have been reported. Notably, the previous studies have enrolled mostly patients with both wild-type and mutant NSCLC and have lacked stratification based on mutation status, thus potentially explaining the inconsistent results across studies. A single-arm phase II trial has reported that most patients with EGFR mutations show an impressive intracranial response rate when erlotinib is added to WBRT45. Another phase II trial has reported that most patients carry wild-type EGFR and has concluded that there is no improvement in the neurological PFS or OS in patients with multiple BMs53. Patients with EGFR or ALK mutations show significantly better efficacy than those with wild-type EGFR or ALK28,47. Therefore, future studies should be conducted in subgroups according to the mutation status. ### Breast cancer The 2018 ASCO guidelines recommend RT as a standard treatment for breast cancer BMs (BCBMs)54. Lapatinib plus capecitabine can be used as the initial treatment for BMs in breast cancer before RT55. Compared with chemotherapy, HER2-targeted drugs and targeted plus chemical drugs result in higher response rates and improved survival56. Pre-clinical studies have shown that the HER2-targeted antibody-drug conjugate T-DM1 may increase radiosensitivity, whereas RT interferes with the permeability of the BBB and enhances breast HER2/neu expression, thus potentially sensitizing cells to the antiproliferative effects of anti-HER2 therapy57. To date, few clinical trials have been conducted to assess the efficacy of RT plus TT in BCBMs, all of which are phase I/II trials with low response rates and few positive results. Two retrospective studies on HER2+ BCBMs conducted by the Curie Institute support the safety and effectiveness of concurrent WBRT and trastuzumab as a radiosensitizer, but further research is needed58. Yomo et al.59 have retrospectively analyzed 40 HER2+ breast cancer patients receiving SRS with or without lapatinib. The 1-year local tumor control rate in the lapatinib group was significantly higher than that that in the non-lapatinib group, thus suggesting that lapatinib has a synergistic effect with SRS. ### Melanoma Vemurafenib, a BRAF inhibitor (BRAFi), is effective in BMs from BRAF-mutated melanoma60–62. Pre-clinical studies have suggested that the MAPK pathway is often upregulated in cancer cells and is activated by exposure to ionizing radiation; BRAFi treatment can increase the radiosensitivity of tumor cells by blocking this pathway. In patients with a BRAF mutation, the radiotherapeutic response can be increased when treated with ionizing radiation combined with TT63,64. The mechanism underlying this improvement is unclear and remains under investigation, and high-level evidence is required. A prospective study involving 80 patients with melanoma brain metastases (MBMs) has indicated an improvement in OS in BRAF-mutant patients treated with SRS combined with BRAFi65. Other studies have found that the administration of BRAFi±MEK inhibitor (BRAFi±MEKi) after RT is safe and effective in patients with MBMs, thus indicating that the combination of the 2 treatments may have a synergistic and superior clinical effect62,66–69. Although the timing and sequence of the 2 therapies suggest a significant synergistic effect, the optimal timing and sequence remain unknown, particularly in patients with multiple BMs. ### Other targeted therapies Other targeted sites are also under investigation, such as those targeted by anti-angiogenic agents. Antiangiogenic agents, including bevacizumab, inhibit tumor angiogenesis by blocking VEGF and promote tumor ischemic necrosis, thus making them a potential RT sensitizer. RT in turn affects the expression of angiogenic factors and tumor growth factors such as VEGF, Ang-2, and Ang-1, and their receptor Tie-2. In mouse models, low-dose radiation promotes tumor growth and metastasis by activating VEGFR270. A single-arm phase I REBECA trial including 19 patients with unresectable solid tumor BMs and has indicated that bevacizumab plus WBRT treatment is tolerable but requires further evaluation71. Higher-level evidence is currently lacking. ## RT combined with IT IT, particularly monoclonal antibodies targeting immune checkpoint pathways (PD-1/PD-L1 and CTLA-4), improves the OS of patients with advanced tumors, such as melanoma and NSCLC, which frequently develop BMs72. Moreover, a synergistic effect has been observed when RT is administered with IT in patients with BM. Hence, more efforts are focusing on evaluating the effects of RT combined with ITs, including antibody-based immune checkpoint blockers, cancer vaccines, and T-cell therapies. RT can induce both antitumor and pro-tumor immune responses in the tumor microenvironment (TME), and the balance of these responses must be further investigated. For the antitumor immune response induced by RT, growing evidence suggests that RT locally interacts with the immune system by inducing the production of immune factors and tumor antigens in the irradiated TME (ITME), which can individually or synergistically prime the immune system, as well as by activating immune cells73–76. Recent studies have also demonstrated that the recruitment of dendritic cells and several other immune effector cell types is mediated by cytokines, such as CXCL16, TNFα, IL-1β, and IL-6, that are secreted by tumor cells after radiation77. Moreover, RT administered with IT has a synergistic effect not only at the irradiated target but also at distant sites, owing to the immune regulation initiated by the local TME (abscopal effect)78. Regarding pro-tumor immune regulation, RT can also upregulate radiation resistance in a manner mediated by radiation-associated antigenic proteins, such as PD-1/PD-L1 and CD47, through the activation of NF-κB79–81. RT increases IFNγ produced by CD8+ T cells, thus resulting in enhanced PD-L1 expression on tumor cells82,83. CTLA-4, which is predominantly expressed by Treg cells, is an immune checkpoint inhibitor (ICI) together with PD-1/PD-L1. Its high expression in cancer cells inhibits immune effector cell activation by increasing CTLA4-CD80/86 binding84. Finally, targeted IT used to block these immunosuppressive pathways can enhance the response rate when it is combined with RT. ### Lung cancer Several retrospective studies have suggested that the use of a combination of IT and RT for BMs from NSCLC might be a safe strategy with promising activity. However, the safety and efficacy of IT administered with brain RT have not been fully clarified. CIs such as PD-1/PD-L1 inhibitors are the primary treatment for BMs from lung cancer. Hubbeling et al.85 have determined the safety of RT combined with ICIs in patients with advanced NSCLC BMs. They have found that treatment with ICI and cranial RT does not significantly increase RT-related adverse events, thus suggesting that the use of RT and ICIs is well tolerated. Two other retrospective studies including 66 patients with NSCLC have characterized the effects of concurrent RT and ICIs on survival outcomes and safety and have not observed increased adverse events in patients with BMs from different solid tumors86,87. Schapira et al.88 have retrospectively reviewed the outcomes of 37 patients with BMs from NSCLC treated with PD-1 pathway inhibitors (83.8% nivolumab, 10.8% atezolizumab, and 5.4% pembrolizumab) and SRS. The patients treated with concurrent SRS and PD-1 pathway inhibitors had longer OS than those treated with SRS before or after IT. NSCLC comprises a higher percentage of BMs than SCLC, and recent studies have suggested that SCLC has a higher biological propensity for the central nervous system89. However, for SCLC, unlike NSCLC, there are very few indications of SCLC-specific targets90. Trials using antibodies targeting PD-1 (nivolumab) and CTLA4 (ipilimumab) on BMs from SCLC are underway. ### Breast cancer Hu et al.91 have found that the overall response rate of breast cancers to IT is only 19%. Dewan et al.92 have found that fractionated RT combined with anti-CTLA4 treatment not only delays the growth of primary tumors but also induces an abscopal effect with enhanced CD8+ T cells in mouse breast models. However, metastasis to the brain from breast cancer remains a major clinical challenge. Recently, chimeric antigen receptor (CAR)-based immune therapy for the treatment of BCBMs has garnered attention. Priceman et al.93 have optimized HER2-CAR T cells containing either CD28 or 4-1BB intracellular costimulatory signaling domains and have demonstrated robust antitumor efficacy after the regional intraventricular delivery of HER2-CAR T cells for the treatment of HER2+ breast cancer metastasis to the brain in a mouse model. Further studies are warranted to validate this approach. ### Melanoma In the past decade, several new systemic drugs have been introduced, including IT with checkpoint inhibitors such as anti-CTLA4 antibodies [ipilimumab (IPI)]94,95, anti-PD1 antibodies (nivolumab and pembrolizumab)96,97, or a combination of these drugs98, and promising results have been observed in patients with melanoma. However, patients with MBMs have been excluded from clinical trials evaluating the efficacy of IT in melanoma. Hence, the role of IT in MBMs is poorly understood, particularly in combination with RT. Opijnen et al.99 have emphasized the role of a combination of RT and IT for MBMs and have found that the combination of IT and SRS is highly effective, with a weighted median OS of 17.4 months. A phase I study aiming to determine the maximum tolerable dose and safety of ipilimumab with SRS or WBRT in patients with MBMs has found that SRS and IT are more effective100. The study has also demonstrated the safety of concurrent ipilimumab 10 mg/kg with SRS. However, other studies evaluating the efficacy of a combination of RT and IT in MBMs have been retrospective, and no prospective randomized studies have been performed. Among these retrospective studies, Stokes et al.101 have examined the largest sample size from the National Cancer Database, with 185 patients receiving both RT and IT. The study suggests that adding IT to RT for MBMs is associated with prolonged survival, with a median of 10.8 months. The factors associated with this improved survival have been found to include stereotactic RT, chemotherapy, and IT. Fang et al.102 have evaluated 137 patients with MBM treated with SRS and anti-CTLA-4 and/or anti-PD-1 antibodies, and have reported a median OS of 16.9 months. They have also characterized radiation necrosis after IT and SRS and demonstrated that the IT type and timing proximity to SRS are not associated with radiation necrosis risk. In addition to the survival benefit, the relative benefit in local control after a combination of RT and IT has been found to be significant103,104. A previous systematic review has reported the outcomes of patients treated with IT and RT for BMs, including 33 studies, 28 of which were associated with MBMs. The pooled median OS from the start of treatment was 15.9 months, and the 1-year OS rate was 55.2%. Moreover, RT administered before or concurrently with IT may provide better results than inverse sequencing105. However, more RCTs or prospective studies are warranted to generate proper evidence that can be used to change the standard of care for patients with MBMs. All the clinical trials for the combined therapies mentioned above are summarized in **Table 1**. View this table: [Table 1](http://www.cancerbiomed.org/content/17/4/910/T1) Table 1 Characteristics of clinical trials examining treatments for brain metastases (RT+chemo, RT+TT, and RT+IT) ## Conclusions and future perspectives BMs are the most common intracranial tumors in adults, but conventional treatments show limited efficacy. RT has been demonstrated to control the intracranial progression of BMs. WBRT is a commonly recommended RT for multiple BMs that can decrease the risk of recurrence and new metastases; however, it can adversely affect neurocognitive function and QoL. Studies examining drugs and techniques to improve neurocognitive impairment *via* WBRT have yielded positive results. In contrast, as an alternative RT for surgery in patients with small, asymptomatic lesions and those with lesions that are not surgically accessible, SRS alone or after surgery can decrease cognitive impairment and toxic responses, while resulting in a prognosis similar to that with WBRT. Moreover, recent research has shown that the clinical efficacy of SRS is not inferior to that of WBRT for extensive BM. Finally, SRS is preferred as the first-line RT, if allowed after evaluation. At present, most studies recommend RT postoperatively. However, the TTI between surgery and RT, and the dose fractionation of RT remain controversial. Hence, further studies should be conducted. Furthermore, ongoing clinical trials on neoadjuvant RT before surgery have shown promising results, thus providing a new perspective on the delivery of RT. An increasing number of systemic treatments, particularly TT and IT, have demonstrated activity in the brain, and this modality might be reasonable for some patients, such as those with asymptomatic BMs, with initial systemic therapy instead of SRS or WBRT. Most treatment decisions are based on the primary tumor type. Multidisciplinary treatment of BMs, including RT combined with systemic therapy, has shown promising results. Because of the low chemosensitivity of BMs and the poor penetration of chemical drugs into the BBB, chemotherapy alone is used not as a first-line treatment but as an addition to local treatment. Most chemical drugs are not effective against BMs, and only several of them, such as TMZ, improve patient survival when added to RT. Moreover, TT shows better effects. RT and TT have been found to show mutually enhanced effects in pre-clinical studies; TT functions as a radiosensitizer, thus indicating that this combination shows promise in BM treatment. However, the existing retrospective and clinical trials have not reached a consensus. Research has focused primarily on BMs from NSCLC, whereas very few phase III clinical trials have been performed; the existing trials have yielded negative results and have focused on first-generation drugs. With the development of targeted drugs, more research is urgently needed to determine the efficacy of different drugs combined with different RT methods in patients with various mutations. For BMs from other cancers, such as BC and multiple myeloma, limited and low-level studies have suggested a promising outlook that urgently must be explored. RT and IT have synergistic effects not only in the ITME but also at distant sites. At present, RT combined with IT is used primarily as a treatment for BMs from melanoma and lung cancer. Systematic reviews of the current literature have affirmed the safety and role of IT combined with RT, particularly SRS, in patients with BMs. Furthermore, retrospective studies have suggested that IT added to RT as treatment for BMs may be associated with a decreased incidence of new BMs and with favorable survival outcomes, without increased rates of adverse events. However, rigorous and reliable data from clinical trials are currently lacking and are urgently needed to support the efficacy of RT combined with systemic treatment modalities to treat BMs. Moreover, additional studies are needed to explore the mechanisms of central permeability and drug-radiation synergy. In addition, more immune-related approaches such as immune checkpoint blockers, CD47 blockers, cancer vaccines, and T cell therapy, should be investigated as new options. In the future, stratified studies of combination therapy should be based on the number of BMs, the pathological type of the primary tumor, and the genotype of the primary tumor, as well as the metastatic foci. Furthermore, treatment administration, such as the best method for administering RT and systemic drugs, the optimal dose, and the drug order, are needed to achieve individualized, multidisciplinary, and precise treatment. ## Acknowledgements This work was supported by grants from Beijing Municipal Natural Science Foundation (Grant No. 7202150), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (Grant No. 2016-I2M-2-001). ## Footnotes * *These authors contributed equally to this work. * **Conflict of interest statement** No potential conflicts of interest are disclosed. * Received April 20, 2020. * Accepted July 9, 2020. * Copyright: © 2020, Cancer Biology & Medicine [https://creativecommons.org/licenses/by/4.0/](https://creativecommons.org/licenses/by/4.0/) This is an open access article distributed under the terms of the [Creative Commons Attribution License (CC BY) 4.0](https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. ## References 1. 1. Achrol AS, Rennert RC, Anders C, Soffietti R, Ahluwalia MS, Nayak L, et al. Brain metastases. Nat Rev Dis Primers. 2019; 5: 5. 2. 2. Tabouret E, Chinot O, Metellus P, Tallet A, Viens P, Goncalves A. Recent trends in epidemiology of brain metastases: an overview. Anticancer Res. 2012; 32: 4655–62. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImFudGljYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjMyLzExLzQ2NTUiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 3. 3. Soffietti R, Abacioglu U, Baumert B, Combs SE, Kinhult S, Kros JM, et al. Diagnosis and treatment of brain metastases from solid tumors: guidelines from the European Association of Neuro-Oncology (EANO). Neuro Oncol. 2017; 19: 162–74. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/neuonc/now241&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28391295&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 4. 4. Yamamoto M, Serizawa T, Shuto T, Akabane A, Higuchi Y, Kawagishi J, et al. Stereotactic radiosurgery for patients with multiple brain metastases (JLGK0901): a multi-institutional prospective observational study. Lancet Oncol. 2014; 15: 387–95. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1470-2045(14)70061-0&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24621620&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000334301900036&link_type=ISI) 5. 5. Tsao MN, Xu W, Wong RK, Lloyd N, Laperriere N, Sahgal A, et al. Whole brain radiotherapy for the treatment of newly diagnosed multiple brain metastases. Cochrane Database Syst Rev. 2018; 1: CD003869. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 6. 6. Nabors LB, Portnow J, Ammirati M, Baehring J, Brem H, Butowski N, et al. NCCN guidelines insights: central nervous system cancers, version 1.2017. J Natl Compr Canc Netw. 2017; 15: 1331–45. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NToiam5jY24iO3M6NToicmVzaWQiO3M6MTA6IjE1LzExLzEzMzEiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7. Patchell RA, Tibbs PA, Regine WF, Dempsey RJ, Mohiuddin M, Kryscio RJ, et al. Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. J Am Med Assoc. 1998; 280: 1485–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.280.17.1485&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=9809728&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000076757200021&link_type=ISI) 8. 8. 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. J Am Med Assoc. 2006; 295: 2483–91. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.295.21.2483&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=16757720&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000238057300023&link_type=ISI) 9. 9. Kocher M, Soffietti R, Abacioglu U, Villa S, Fauchon F, Baumert BG, et al. Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 study. J Clin Oncol. 2011; 29: 134–41. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjg6IjI5LzIvMTM0IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 10. 10. Mulvenna P, Nankivell M, Barton R, Faivre-Finn C, Wilson P, McColl E, et al. Dexamethasone and supportive care with or without whole brain radiotherapy in treating patients with non-small cell lung cancer with brain metastases unsuitable for resection or stereotactic radiotherapy (QUARTZ): results from a phase 3, non-inferiority, randomised trial. Lancet. 2016; 388: 2004–14. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0140-6736(16)30825-X&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=27604504&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 11. 11. Chang EL, Wefel JS, Hess KR, Allen PK, Lang FF, Kornguth DG, et al. Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled trial. Lancet Oncol. 2009; 10: 1037–44. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1470-2045(09)70263-3&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19801201&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000271852800012&link_type=ISI) 12. 12. Soffietti R, Kocher M, Abacioglu UM, Villa S, Fauchon F, Baumert BG, et al. A European Organisation for Research and Treatment of Cancer phase III trial of adjuvant whole-brain radiotherapy versus observation in patients with one to three brain metastases from solid tumors after surgical resection or radiosurgery: quality-of-life results. J Clin Oncol. 2013; 31: 65–72. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjc6IjMxLzEvNjUiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 13. 13. Aoyama H, Tago M, Shirato H, Japanese Radiation Oncology Study Group I. Stereotactic radiosurgery with or without whole-brain radiotherapy for brain metastases: secondary analysis of the JROSG 99-1 randomized clinical trial. JAMA Oncol. 2015; 1: 457–64. 14. 14. Brown PD, Jaeckle K, Ballman KV, Farace E, Cerhan JH, Anderson SK, et al. Effect of radiosurgery alone vs radiosurgery with whole brain radiation therapy on cognitive function in patients with 1 to 3 brain metastases: a randomized clinical trial. J Am Med Assoc. 2016; 316: 401–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1001/jama.2016.9839&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=27458945&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 15. 15. Brown PD, Ballman KV, Cerhan JH, Anderson SK, Carrero XW, Whitton AC, et al. Postoperative stereotactic radiosurgery compared with whole brain radiotherapy for resected metastatic brain disease (NCCTG N107C/CEC.3): a multicentre, randomised, controlled, phase 3 trial. Lancet Oncol. 2017; 18: 1049–60. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1470-2045(17)30441-2&link_type=DOI) 16. 16. Kayama T, Sato S, Sakurada K, Mizusawa J, Nishikawa R, Narita Y, et al. Effects of surgery with salvage stereotactic radiosurgery versus surgery with whole-brain radiation therapy in patients with one to four brain metastases (JCOG0504): a phase III, noninferiority, randomized controlled trial. J Clin Oncol. 2018; 36: 3282–9. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 17. 17. Akanda ZZ, Hong W, Nahavandi S, Haghighi N, Phillips C, Kok DL. Post-operative stereotactic radiosurgery following excision of brain metastases: a systematic review and meta-analysis. Radiother Oncol. 2020; 142: 27–35. 18. 18. Mahajan A, Ahmed S, McAleer MF, Weinberg JS, Li J, Brown P, et al. Post-operative stereotactic radiosurgery versus observation for completely resected brain metastases: a single-centre, randomised, controlled, phase 3 trial. Lancet Oncol. 2017; 18: 1040–8. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1470-2045(17)30414-X&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28687375&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 19. 19. Yamamoto M, Kawabe T, Sato Y, Higuchi Y, Nariai T, Barfod BE, et al. A case-matched study of stereotactic radiosurgery for patients with multiple brain metastases: comparing treatment results for 1-4 vs >/= 5 tumors: clinical article. J Neurosurg. 2013; 118: 1258–68. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.3171/2013.3.JNS121900&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23600938&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 20. 20. Yamamoto M, Kawabe T, Sato Y, Higuchi Y, Nariai T, Watanabe S, et al. Stereotactic radiosurgery for patients with multiple brain metastases: a case-matched study comparing treatment results for patients with 2-9 versus 10 or more tumors. J Neurosurg. 2014;121 Suppl: 16–25. 21. 21. Yusuf MB, Amsbaugh MJ, Burton E, Nelson M, Williams B, Koutourousiou M, et al. Increasing time to postoperative stereotactic radiation therapy for patients with resected brain metastases: investigating clinical outcomes and identifying predictors associated with time to initiation. J Neurooncol. 2018; 136: 545–3. 22. 22. Patel KR, Burri SH, Asher AL, Crocker IR, Fraser RW, Zhang C, et al. Comparing preoperative with postoperative stereotactic radiosurgery for resectable brain metastases: a multi-institutional analysis. Neurosurgery. 2016; 79: 279–85. 23. 23. Routman DM, Yan E, Vora S, Peterson J, Mahajan A, Chaichana KL, et al. Preoperative stereotactic radiosurgery for brain metastases. Front Neurol. 2018; 9: 959. 24. 24. Musunuru HB, Witt JS, Yadav P, Francis DM, Kuczmarska-Haas A, Labby ZE, et al. Impact of adjuvant fractionated stereotactic radiotherapy dose on local control of brain metastases. J Neurooncol. 2019; 145: 385–90. 25. 25. Antonadou D, Paraskevaidis M, Sarris G, Coliarakis N, Economou I, Karageorgis P, et al. Phase II randomized trial of temozolomide and concurrent radiotherapy in patients with brain metastases. J Clin Oncol. 2002; 20: 3644–50. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyMC8xNy8zNjQ0IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 26. 26. Verger E, Gil M, Yaya R, Vinolas N, Villa S, Pujol T, et al. Temozolomide and concomitant whole brain radiotherapy in patients with brain metastases: a phase II randomized trial. Int J Radiat Oncol Biol Phys. 2005; 61: 185–91. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ijrobp.2004.04.061&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15629610&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 27. 27. Chua D, Krzakowski M, Chouaid C, Pallotta MG, Martinez JI, Gottfried M, et al. Whole-brain radiation therapy plus concomitant temozolomide for the treatment of brain metastases from non-small-cell lung cancer: a randomized, open-label phase II study. Clin Lung Cancer. 2010; 11: 176–81. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.3816/CLC.2010.n.022&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20439193&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 28. 28. Singh R, Lehrer EJ, Ko S, Peterson J, Lou Y, Porter AB, et al. Brain metastases from non-small cell lung cancer with EGFR or ALK mutations: a systematic review and meta-analysis of multidisciplinary approaches. Radiother Oncol. 2019; 144: 165–79. 29. 29. Zimmermann S, Dziadziuszko R, Peters S. Indications and limitations of chemotherapy and targeted agents in non-small cell lung cancer brain metastases. Cancer Treat Rev. 2014; 40: 716–22. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ctrv.2014.03.005&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24759599&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 30. 30. Costa DB, Shaw AT, Ou SH, Solomon BJ, Riely GJ, Ahn MJ, et al. Clinical experience with crizotinib in patients with advanced ALK-rearranged non-small-cell lung cancer and brain metastases. J Clin Oncol. 2015; 33: 1881–8. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMy8xNy8xODgxIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 31. 31. Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, et al. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med. 2017; 377: 829–38. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 32. 32. Gadgeel S, Peters S, Mok T, Shaw AT, Kim DW, Ou SI, et al. Alectinib versus crizotinib in treatment-naive anaplastic lymphoma kinase-positive (ALK+) non-small-cell lung cancer: CNS efficacy results from the ALEX study. Ann Oncol. 2018; 29: 2214–22. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/annonc/mdy405&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=30215676&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 33. 33. Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. 2018; 378: 113–25. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa1713137&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=29151359&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 34. 34. Wu YL, Yang JJ, Zhou CC, Feng JF, Lu S, Song Y, et al. BRAIN: a phase III trial comparing WBI and chemotherapy with icotinib in NSCLC with brain metastases harboring EGFR mutations (CTONG 1201). J Thorac Oncol. 2017; 12(Suppl 1):3–4. 35. 35. Das AK, Sato M, Story MD, Peyton M, Graves R, Redpath S, et al. Non-small-cell lung cancers with kinase domain mutations in the epidermal growth factor receptor are sensitive to ionizing radiation. Cancer Res. 2006; 66: 9601–8. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI2Ni8xOS85NjAxIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 36. 36. Gow CH, Chien CR, Chang YL, Chiu YH, Kuo SH, Shih JY, et al. Radiotherapy in lung adenocarcinoma with brain metastases: effects of activating epidermal growth factor receptor mutations on clinical response. Clin Cancer Res. 2008; 14: 162–8. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6ODoiMTQvMS8xNjIiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 37. 37. Johung KL, Yao X, Li F, Yu JB, Gettinger SN, Goldberg S, et al. A clinical model for identifying radiosensitive tumor genotypes in non-small cell lung cancer. Clin Cancer Res. 2013; 19: 5523–32. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjE5LzE5LzU1MjMiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 38. 38. Zeng YD, Liao H, Qin T, Zhang L, Wei WD, Liang JZ, et al. Blood-brain barrier permeability of gefitinib in patients with brain metastases from non-small-cell lung cancer before and during whole brain radiation therapy. Oncotarget. 2015; 6: 8366–76. 39. 39. Luo S, Chen L, Chen X, Xie X. Evaluation on efficacy and safety of tyrosine kinase inhibitors plus radiotherapy in NSCLC patients with brain metastases. Oncotarget. 2015; 6: 16725–34. 40. 40. Chen Y, Yang J, Li X, Hao D, Wu X, Yang Y, et al. First-line epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor alone or with whole-brain radiotherapy for brain metastases in patients with EGFR-mutated lung adenocarcinoma. Cancer Sci. 2016; 107: 1800–5. 41. 41. Jiang T, Min W, Li Y, Yue Z, Wu C, Zhou C. Radiotherapy plus EGFR TKIs in non-small cell lung cancer patients with brain metastases: an update meta-analysis. Cancer Med. 2016; 5: 1055–65. 42. 42. Jiang T, Su C, Li X, Zhao C, Zhou F, Ren S, et al. EGFR TKIs plus WBRT demonstrated no survival benefit other than that of TKIs alone in patients with NSCLC and EGFR mutation and brain metastases. J Thorac Oncol. 2016; 11: 1718–28. 43. 43. Dong K, Liang W, Zhao S, Guo M, He Q, Li C, et al. EGFR-TKI plus brain radiotherapy versus EGFR-TKI alone in the management of EGFR-mutated NSCLC patients with brain metastases. Transl Lung Cancer Res. 2019; 8: 268–79. 44. 44. Pesce GA, Klingbiel D, Ribi K, Zouhair A, von Moos R, Schlaeppi M, et al. Outcome, quality of life and cognitive function of patients with brain metastases from non-small cell lung cancer treated with whole brain radiotherapy combined with gefitinib or temozolomide. A randomised phase II trial of the Swiss Group for Clinical Cancer Research (SAKK 70/03). Eur J Cancer. 2012; 48: 377–84. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ejca.2011.10.016&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22093943&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000300729800012&link_type=ISI) 45. 45. Welsh JW, Komaki R, Amini A, Munsell MF, Unger W, Allen PK, et al. Phase II trial of erlotinib plus concurrent whole-brain radiation therapy for patients with brain metastases from non-small-cell lung cancer. J Clin Oncol. 2013; 31: 895–902. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjg6IjMxLzcvODk1IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 46. 46. Soon YY, Leong CN, Koh WY, Tham IW. EGFR tyrosine kinase inhibitors versus cranial radiation therapy for EGFR mutant non-small cell lung cancer with brain metastases: a systematic review and meta-analysis. Radiother Oncol. 2015; 114: 167–72. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.radonc.2014.12.011&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25583566&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 47. 47. Fan Y, Huang Z, Fang L, Miao L, Gong L, Yu H, et al. A phase II study of icotinib and whole-brain radiotherapy in Chinese patients with brain metastases from non-small cell lung cancer. Cancer Chemother Pharmacol. 2015; 76: 517–23. 48. 48. Sperduto PW, Wang M, Robins HI, Schell MC, Werner-Wasik M, Komaki R, et al. A phase 3 trial of whole brain radiation therapy and stereotactic radiosurgery alone versus WBRT and SRS with temozolomide or erlotinib for non-small cell lung cancer and 1 to 3 brain metastases: radiation therapy oncology group 0320. Int J Radiat Oncol Biol Phys. 2013; 85: 1312–8. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ijrobp.2012.11.042&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23391814&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 49. 49. Magnuson WJ, Lester-Coll NH, Wu AJ, Yang TJ, Lockney NA, Gerber NK, et al. Management of brain metastases in tyrosine kinase inhibitor-naive epidermal growth factor receptor-mutant non-small-cell lung cancer: a retrospective multi-institutional analysis. J Clin Oncol. 2017; 35: 1070–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1200/JCO.2016.69.7144&link_type=DOI) 50. 50. Sun Y, Nowak KA, Zaorsky NG, Winchester CL, Dalal K, Giacalone NJ, et al. ALK inhibitor PF02341066 (crizotinib) increases sensitivity to radiation in non-small cell lung cancer expressing EML4-ALK. Mol Cancer Ther. 2013; 12: 696–704. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6Im1vbGNhbnRoZXIiO3M6NToicmVzaWQiO3M6ODoiMTIvNS82OTYiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 51. 51. Dai Y, Wei Q, Schwager C, Moustafa M, Zhou C, Lipson KE, et al. Synergistic effects of crizotinib and radiotherapy in experimental EML4-ALK fusion positive lung cancer. Radiother Oncol. 2015; 114: 173–81. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.radonc.2014.12.009&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25592111&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 52. 52. Johung KL, Yeh N, Desai NB, Williams TM, Lautenschlaeger T, Arvold ND, et al. Extended survival and prognostic factors for patients with ALK-rearranged non-small-cell lung cancer and brain metastasis. J Clin Oncol. 2016; 34: 123–9. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjg6IjM0LzIvMTIzIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 53. 53. Lee SM, Lewanski CR, Counsell N, Ottensmeier C, Bates A, Patel N, et al. Randomized trial of erlotinib plus whole-brain radiotherapy for NSCLC patients with multiple brain metastases. J Natl Cancer Inst. 2014; 106: dju151. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/jnci/dju151&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25031274&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 54. 54. Ramakrishna N, Temin S, Chandarlapaty S, Crews JR, Davidson NE, Esteva FJ, et al. Recommendations on disease management for patients with advanced human epidermal growth factor receptor 2-positive breast cancer and brain metastases: ASCO Clinical Practice Guideline update. J Clin Oncol. 2018; 36: 2804–7. 55. 55. Ramakrishna N, Temin S, Lin NU. Recommendations on disease management for patients with advanced human epidermal growth factor receptor 2-positive breast cancer and brain metastases: ASCO Clinical Practice Guideline update summary. J Oncol Pract. 2018; 14: 505–7. 56. 56. Shah N, Mohammad AS, Saralkar P, Sprowls SA, Vickers SD, John D, et al. Investigational chemotherapy and novel pharmacokinetic mechanisms for the treatment of breast cancer brain metastases. Pharmacol Res. 2018; 132: 47–68. 57. 57. Adams SR, Yang HC, Savariar EN, Aguilera J, Crisp JL, Jones KA, et al. Anti-tubulin drugs conjugated to anti-ERBB antibodies selectively radiosensitize. Nat Commun. 2016; 7: 13019. 58. 58. Chargari C, Idrissi HR, Pierga JY, Bollet MA, Dieras V, Campana F, et al. Preliminary results of whole brain radiotherapy with concurrent trastuzumab for treatment of brain metastases in breast cancer patients. Int J Radiat Oncol Biol Phys. 2011; 81: 631–6. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ijrobp.2010.06.057&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20932686&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 59. 59. Yomo S, Hayashi M, Cho N. Impacts of HER2-overexpression and molecular targeting therapy on the efficacy of stereotactic radiosurgery for brain metastases from breast cancer. J Neurooncol. 2013; 112: 199–207. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s11060-013-1046-1&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23296546&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 60. 60. Long GV, Trefzer U, Davies MA, Kefford RF, Ascierto PA, Chapman PB, et al. Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (break-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol. 2012; 13: 1087–95. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1470-2045(12)70431-X&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23051966&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000310570900039&link_type=ISI) 61. 61. Dummer R, Goldinger SM, Turtschi CP, Eggmann NB, Michielin O, Mitchell L, et al. Vemurafenib in patients with BRAF(V600) mutation-positive melanoma with symptomatic brain metastases: final results of an open-label pilot study. Eur J Cancer. 2014; 50: 611–21. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ejca.2013.11.002&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24295639&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 62. 62. Gaudy-Marqueste C, Carron R, Delsanti C, Loundou A, Monestier S, Archier E, et al. On demand gamma-knife strategy can be safely combined with BRAF inhibitors for the treatment of melanoma brain metastases. Ann. Oncol. 2014; 25: 2086–91. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/annonc/mdu266&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25057167&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000343045500032&link_type=ISI) 63. 63. Chung EJ, Brown AP, Asano H, Mandler M, Burgan WE, Carter D, et al. In vitro and in vivo radiosensitization with AZD6244 (ARRY-142886), an inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 kinase. Clin Cancer Res. 2009; 15: 3050–7. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMTUvOS8zMDUwIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 64. 64. Sambade MJ, Peters EC, Thomas NE, Kaufmann WK, Kimple RJ, Shields JM. Melanoma cells show a heterogeneous range of sensitivity to ionizing radiation and are radiosensitized by inhibition of B-RAF with PLX-4032. Radiother Oncol. 2011; 98: 394–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.radonc.2010.12.017&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21295875&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 65. 65. Wolf A, Zia S, Verma R, Pavlick A, Wilson M, Golfinos JG, et al. Impact on overall survival of the combination of BRAF inhibitors and stereotactic radiosurgery in patients with melanoma brain metastases. J Neurooncol. 2016; 127: 607–15. 66. 66. Choong ES, Lo S, Drummond M, Fogarty GB, Menzies AM, Guminski A, et al. Survival of patients with melanoma brain metastasis treated with stereotactic radiosurgery and active systemic drug therapies. Eur J Cancer. 2017; 75: 169–78. 67. 67. Gaudy-Marqueste C, Dussouil AS, Carron R, Troin L, Malissen N, Loundou A, et al. Survival of melanoma patients treated with targeted therapy and immunotherapy after systematic upfront control of brain metastases by radiosurgery. Eur J Cancer. 2017; 84: 44–54. 68. 68. Rauschenberg R, Bruns J, Brutting J, Daubner D, Lohaus F, Zimmer L, et al. Impact of radiation, systemic therapy and treatment sequencing on survival of patients with melanoma brain metastases. Eur J Cancer. 2019; 110: 11–20. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 69. 69. Tetu P, Allayous C, Oriano B, Dalle S, Mortier L, Leccia MT, et al. Impact of radiotherapy administered simultaneously with systemic treatment in patients with melanoma brain metastases within MelBase, a French multicentric prospective cohort. Eur J Cancer. 2019; 112: 38–46. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 70. 70. Sofia Vala I, Martins LR, Imaizumi N, Nunes RJ, Rino J, Kuonen F, et al. Low doses of ionizing radiation promote tumor growth and metastasis by enhancing angiogenesis. PLoS One. 2010; 5: e11222. 71. 71. Levy C, Allouache D, Lacroix J, Dugue AE, Supiot S, Campone M, et al. Rebeca: a phase I study of bevacizumab and whole-brain radiation therapy for the treatment of brain metastasis from solid tumours. Ann Oncol. 2014; 25: 2351–6. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/annonc/mdu465&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25274615&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000345825800007&link_type=ISI) 72. 72. Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015; 520: 373–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature14292&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25754329&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 73. 73. Michelin S, Gallegos CE, Dubner D, Favier B, Carosella ED. Ionizing radiation modulates the surface expression of human leukocyte antigen-G in a human melanoma cell line. Hum Immunol. 2009; 70: 1010–5. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.humimm.2009.07.030&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=19665041&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 74. 74. Ibrahim EC, Aractingi S, Allory Y, Borrini F, Dupuy A, Duvillard P, et al. Analysis of HLA antigen expression in benign and malignant melanocytic lesions reveals that upregulation of HLA-G expression correlates with malignant transformation, high inflammatory infiltration and HLA-A1 genotype. Int J Cancer. 2004; 108: 243–50. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/ijc.11456&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=14639610&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000187224600012&link_type=ISI) 75. 75. Schaue D, McBride WH. Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol. 2015; 12: 527–40. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nrclinonc.2015.120&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26122185&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 76. 76. Assi HI, Kamphorst AO, Moukalled NM, Ramalingam SS. Immune checkpoint inhibitors in advanced non-small cell lung cancer. Cancer. 2018; 124: 248–61. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/cncr.31105&link_type=DOI) 77. 77. Allaoui R, Bergenfelz C, Mohlin S, Hagerling C, Salari K, Werb Z, et al. Cancer-associated fibroblast-secreted CXCL16 attracts monocytes to promote stroma activation in triple-negative breast cancers. Nat Commun. 2016; 7: 13050. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ncomms13050&link_type=DOI) 78. 78. Park SS, Dong H, Liu X, Harrington SM, Krco CJ, Grams MP, et al. Pd-1 restrains radiotherapy-induced abscopal effect. Cancer Immunol Res. 2015; 3: 610–9. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FuaW1tIjtzOjU6InJlc2lkIjtzOjc6IjMvNi82MTAiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 79. 79. Wu ZH, Shi Y, Tibbetts RS, Miyamoto S. Molecular linkage between the kinase ATM and NF-kappaB signaling in response to genotoxic stimuli. Science. 2006; 311: 1141–6. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzMTEvNTc2NC8xMTQxIjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 80. 80. Ahmed KM, Li JJ. NF-kappa B-mediated adaptive resistance to ionizing radiation. Free Radic Biol Med. 2008; 44: 1–13. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.freeradbiomed.2007.09.022&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17967430&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000251931400001&link_type=ISI) 81. 81. Hsuan CF, Hsu HF, Tseng WK, Lee TL, Wei YF, Hsu KL, et al. Glossogyne tenuifolia extract inhibits TNF-alpha-induced expression of adhesion molecules in human umbilical vein endothelial cells via blocking the NF-kB signaling pathway. Molecules. 2015; 20: 16908–23. 82. 82. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, McKenna C, Jones S, Cheadle EJ, et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-l1 blockade. Cancer Res. 2014; 74: 5458–68. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3NC8xOS81NDU4IjtzOjQ6ImF0b20iO3M6MTg6Ii9jYm0vMTcvNC85MTAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 83. 83. Dovedi SJ, Cheadle EJ, Popple AL, Poon E, Morrow M, Stewart R, et al. Fractionated radiation therapy stimulates antitumor immunity mediated by both resident and infiltrating polyclonal T-cell populations when combined with PD-1 blockade. Clin Cancer Res. 2017; 23: 5514–26. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjIzLzE4LzU1MTQiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 84. 84. Walker LS. Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun. 2013; 45: 49–57. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.jaut.2013.06.006&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23849743&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000325448600007&link_type=ISI) 85. 85. Hubbeling HG, Schapira EF, Horick NK, Goodwin KEH, Lin JJ, Oh KS, et al. Safety of combined PD-1 pathway inhibition and intracranial radiation therapy in non-small cell lung cancer. J Thorac Oncol. 2018; 13: 550–8. 86. 86. Pike LRG, Bang A, Ott P, Balboni T, Taylor A, Catalano P, et al. Radiation and PD-1 inhibition: favorable outcomes after brain-directed radiation. Radiother Oncol. 2017; 124: 98–103. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 87. 87. Chen L, Douglass J, Kleinberg L, Ye X, Marciscano AE, Forde PM, et al. Concurrent immune checkpoint inhibitors and stereotactic radiosurgery for brain metastases in non-small cell lung cancer, melanoma, and renal cell carcinoma. Int J Radiat Oncol Biol Phys. 2018; 100: 916–25. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ijrobp.2017.11.041&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=29485071&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 88. 88. Schapira E, Hubbeling H, Yeap BY, Mehan WA Jr.., Shaw AT, Oh K, et al. Improved overall survival and locoregional disease control with concurrent PD-1 pathway inhibitors and stereotactic radiosurgery for lung cancer patients with brain metastases. Int J Radiat Oncol Biol Phys. 2018; 101: 624–9. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 89. 89. Lukas RV, Gondi V, Kamson DO, Kumthekar P, Salgia R. State-of-the-art considerations in small cell lung cancer brain metastases. Oncotarget. 2017; 8: 71223–33. 90. 90. Pezzuto F, Fortarezza F, Lunardi F, Calabrese F. Are there any theranostic biomarkers in small cell lung carcinoma? J Thorac Dis. 2019; 11: S102–12. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.21037/jtd.2018.12.14&link_type=DOI) 91. 91. Hu ZI, Ho AY, McArthur HL. Combined radiation therapy and immune checkpoint blockade therapy for breast cancer. Int J Radiat Oncol Biol Phys. 2017; 99: 153–64. 92. 92. Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS, Formenti SC, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009; 15: 5379–88. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjE1LzE3LzUzNzkiO3M6NDoiYXRvbSI7czoxODoiL2NibS8xNy80LzkxMC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 93. 93. Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK, et al. Regional delivery of chimeric antigen receptor-engineered t cells effectively targets HER2(+) breast cancer metastasis to the brain. Clin Cancer Res. 2018; 24: 95–105. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6NzoiMjQvMS85NSI7czo0OiJhdG9tIjtzOjE4OiIvY2JtLzE3LzQvOTEwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 94. 94. Margolin K, Ernstoff MS, Hamid O, Lawrence D, McDermott D, Puzanov I, et al. Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial. Lancet Oncol. 2012; 13: 459–65. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S1470-2045(12)70090-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22456429&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000303508700037&link_type=ISI) 95. 95. Vosoughi E, Lee JM, Miller JR, Nosrati M, Minor DR, Abendroth R, et al. Survival and clinical outcomes of patients with melanoma brain metastasis in the era of checkpoint inhibitors and targeted therapies. BMC Cancer. 2018; 18: 490. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1186/s12885-018-4374-x&link_type=DOI) 96. 96. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015; 372: 320–30. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa1412082&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25399552&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000348204500007&link_type=ISI) 97. 97. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015; 372: 2521–32. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa1503093&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25891173&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 98. 98. Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017; 377: 1345–56. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1056/NEJMoa1709684&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28889792&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 99. 99. van Opijnen MP, Dirven L, Coremans IEM, Taphoorn MJB, Kapiteijn EHW. The impact of current treatment modalities on the outcomes of patients with melanoma brain metastases: a systematic review. Int J Cancer. 2020; 146: 1479–89. 100.100. Williams NL, Wuthrick EJ, Kim H, Palmer JD, Garg S, Eldredge-Hindy H, et al. Phase 1 study of ipilimumab combined with whole brain radiation therapy or radiosurgery for melanoma patients with brain metastases. Int J Radiat Oncol Biol Phys. 2017; 99: 22–30. 101.101. Stokes WA, Binder DC, Jones BL, Oweida AJ, Liu AK, Rusthoven CG, et al. Impact of immunotherapy among patients with melanoma brain metastases managed with radiotherapy. J Neuroimmunol. 2017; 313: 118–22. 102.102. Fang P, Jiang W, Allen P, Glitza I, Guha N, Hwu P, et al. Radiation necrosis with stereotactic radiosurgery combined with CTLA-4 blockade and PD-1 inhibition for treatment of intracranial disease in metastatic melanoma. J Neurooncol. 2017; 133: 595–602. 103.103. Nardin C, Mateus C, Texier M, Lanoy E, Hibat-Allah S, Ammari S, et al. Tolerance and outcomes of stereotactic radiosurgery combined with anti-programmed cell death-1 (pembrolizumab) for melanoma brain metastases. Melanoma Res. 2018; 28: 111–9. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2F17%2F4%2F910.atom) 104.104. Trommer-Nestler M, Marnitz S, Kocher M, Ruess D, Schlaak M, Theurich S, et al. Robotic stereotactic radiosurgery in melanoma patients with brain metastases under simultaneous anti-PD-1 treatment. Int J Mol Sci. 2018; 19: 2653. 105.105. Petrelli F, De Stefani A, Trevisan F, Parati C, Inno A, Merelli B, et al. Combination of radiotherapy and immunotherapy for brain metastases: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2019; 144: 102830.