Next Article in Journal
The Intervention and Mechanism of Action for Aloin against Subchronic Aflatoxin B1 Induced Hepatic Injury in Rats
Next Article in Special Issue
Histone Modification in NSCLC: Molecular Mechanisms and Therapeutic Targets
Previous Article in Journal
Biomolecular Evaluation of Piceatannol’s Effects in Counteracting the Senescence of Mesenchymal Stromal Cells: A New Candidate for Senotherapeutics?
Previous Article in Special Issue
EphB4 as a Novel Target for the EGFR-Independent Suppressive Effects of Osimertinib on Cell Cycle Progression in Non-Small Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Checkpoints Inhibitors and SRS/SBRT Synergy in Metastatic Non-Small-Cell Lung Cancer and Melanoma: A Systematic Review

by
María Rodríguez Plá
*,
Diego Dualde Beltrán
and
Eduardo Ferrer Albiach
Department of Radiation Oncology, Hospital Clinico Universitario de Valencia, 46010 Valencia, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(21), 11621; https://doi.org/10.3390/ijms222111621
Submission received: 16 September 2021 / Revised: 21 October 2021 / Accepted: 23 October 2021 / Published: 27 October 2021
(This article belongs to the Special Issue New Molecular Targets in Lung Cancer)

Abstract

:
Background: Several immunotherapy (IT) agents are FDA approved for treatment of melanoma and non-small-cell lung cancer (NSCLC). The addition of stereotactic radiosurgery (SRS) or stereotactic body radiation therapy (SBRT) to immunotherapy looks promising. A systematic review was conducted to evaluate the possible synergistic effects of immune checkpoints inhibitors (ICIs) and stereotactic radiation therapy in melanoma and NSCLC. Materials and methods: Pubmed databases from January 2010 to December 2020 were reviewed to identify English language studies reporting control of local and abscopal effect of the combination of ICI-SBRT/SRS in metastatic NSCLC and melanoma cancer. The inclusion criteria were followed according to PICO criteria. Results: Thirty-nine articles were included of the 2141 initial results. The reported rates for local control were 16.5–100% and 40–94% in brain and extracerebral metastases, respectively. Distant/abscopal response rates were 1–45% in extracerebral metastases. Abscopal effect could not be evaluated in brain metastases because it was not reported in studies. Treatments were well tolerated with few grade 4 toxicities and no grade 5. Conclusions: The combined treatment of ICI-SBRT/SRS achieves high local control and non-negligible abscopal response in patients with extracerebral metastases, with its benefit in cerebral metastases being more controversial. Clinical trials are needed to better characterize the potential synergism.

1. Introduction

Local radiation therapy (RT) is known to modulate the immune response [1,2]. The development of immunotherapeutic drugs has led to a rapid growth of publications that have attempted to elucidate the potential synergistic effects of the combined treatment of immunotherapy with radiation therapy. This combination is known as immunoradiotherapy [3,4].
It has been shown that combined immunoradiotherapy treatment can promote greater local control and an antitumor systemic response (response known in the literature as the abscopal effect) through T cell-mediated activation of the adaptive immune system [5,6]. Radiation therapy treatment induces a stress response or cell death by stimulating the production of tumor-associated antigens that activate antigen-presenting cells (APCs). Activation of APCs induces activation of specific CD8+T lymphocytes against tumor cells presenting these antigens. These circulating activated lymphocytes can be extravasated to non-irradiated tumor lesions, and can act on them [2,7].
Stereotactic radiosurgery and stereotactic body radiation therapy have undergone exponential development in recent years, as their ablative capacity has demonstrated a benefit in certain patients, such as oligometastatic or oligoprogressive patients [8]. Oligoprogression is a limited tumor progression with the rest of the disease controlled. Under ICIs, oligoprogression occurs in approximately 10–20% of cases [9]. In these patients, SRS/SBRT allows the administration of a high antitumor biologically effective dose (BED). Different fractions are used according to the anatomical location, size, and tumor histology, among other factors. In general, the most frequent fractioning schemes used in SRS/SBRT are those in which a dose per fraction >6 Gy is administered, in 1–5 fractions.
Currently, it is unknown which doses per fraction obtain a greater antitumoral immune response. However, preclinical models have shown that doses between 10 and 13 Gy seem to maximize these effects [7]. In addition, it has been shown that SBRT promotes a signaling cascade secondary to the destruction of the tumor stroma that promotes immune-mediated tumor recognition [10]. SRS/SBRT decreases repair of sublethal damage and tumor repopulation.
On the other hand, several immunotherapies are FDA approved for treatment of melanoma and NSCLC. ICIs including anti-cytotoxic T lymphocyte-associated protein 4 (CTLA4) and anti-programmed death-1 (PD1) antibodies have become the most widely used agents in this field. ICIs act by blocking checkpoint proteins from binding with their partner proteins.
T cells recognize antigens presented by the major histocompatibility complex (MHC) on the surface of cancer cells through their T cell receptor (TCR). This first signal is not sufficient to turn on a T cell response, and a second costimulatory signal consisting of the union B7 (CD80 or CD86) and CD28 is required. CTLA4 is present in the membrane of T cells, especially in regulatory T cells. It inhibits the costimulatory signal needed for T cell activation by competing with CD28 for binding B7. Anti-CTLA4 is a monoclonal antibody that acts by inhibiting CTLA4 and consequently stimulates T cell activation after antigen presentation [11].
PD-1 is expressed on activated T cells and mediates inhibitory signals upon binding to its ligand PD-L1, which is expressed on tumor cells and antigen-presenting cells. Its blockade with PD-1 or PD-L1 antibodies results in the activation of T cells against tumor cells [11].
Immunotherapies can also be used to enhance immune responses together with SBRT because both increase T cell activation and reduce cancer immune evasion [12].
Moreover, the apparent synergy between ICI and radiotherapy is potentially useful not only in stage IV tumors with oligoprogression but also with, e.g., stage III NSCLC patients not eligible for chemotherapy, who could receive immunoradiotherapy instead of the standard chemoradiotherapy [13].
Knowing the benefits of SBRT/SRS combined with ICIs and possible side effects, as well as the best sequence or timing of treatment, are key to our daily practice.

2. Results

After the search, 2141 articles were identified, of which 39 articles met the criteria initially established and were selected to carry out this review. The flowchart that explains the screening process is shown in Figure 1.
In the tables presented below, the results are separated according to the target location of the radiation therapy, that is, according to whether the local treatment with SBRT/SRS was performed on cerebral or extracerebral metastases.
It is differentiated in this way by the difficulty of the immune cells to cross the blood–brain barrier after being stimulated by a local treatment with SRS in brain lesions. The difficulty of extravasating the blood–brain barrier could lead to a potential inequality in the systemic effect produced by the brain SRS compared to that produced by the SBRT on extracerebral lesions.
Abscopal effect was evaluated as a local response in non-irradiated lesions.
In both cases (cerebral and extracerebral metastases), ICI treatment regimens were administered according to the approved clinical protocol of studies with different doses: Ipilimumab (anti-CTLA4) 3 mg/kg or 10 mg/kg every 3 weeks, Pembrolizumab (anti-PD-1) 2 mg/kg, 3 mg/kg or 10 mg/kg every 3 weeks, or Nivolumab (anti-PD-1) 2 mg/kg or 3 mg/kg every 2 weeks. Some studies did not report the dose that they used.
Table 1 presents the results of the selected articles of the ICI + SRS combination in cerebral metastases.
Table 2 presents the results of the selected articles of the ICI + SBRT/SABR combination in extracerebral metastases.

3. Discussion

3.1. ICI + SRS: Cerebral Metastases

Patients with non-small-cell lung carcinoma and melanoma have a high incidence of cerebral metastases both at diagnosis and throughout the course of the disease. An incidence of cerebral metastases >25% has been observed in both tumors [53,54]. As we know, the appearance of metastases leads to a decrease in survival and, therefore, a poor prognosis in these patients [55].
Immunotherapy has shown increased survival in certain metastatic patients with NSCLC and melanoma, leading to its approval as a first-line drug in both cases [56,57]. In addition, SRS has been shown to be effective as a local treatment for cerebral metastases in patients who are candidates for this treatment [58].
Of the 25 articles included in Table 1, 21 of them included patients with metastatic melanoma [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34], 2 with metastatic NSCLC [35,36], and 2 with heterogeneous histology (>80% patients with metastatic melanoma or NSCLC) [37,38].
All articles included were retrospective, except for one that was prospective [23].
The studies sample size varied significantly, with studies ranging from 11–260 patients and with the number of lesions treated ranging from 23–793. We need to consider these differences when evaluating the results.
First of all, it must be taken into account that the consideration of concomitant treatment differed between the different studies. Most authors considered concomitant ICI-SRS as the administration of SRS within 4 weeks before or after the start of ICI [23,25,29,30,32,33,34,38]. However, other authors considered a timeframe <2 weeks [31,37] and others up to >2 months [20,24]. When local treatment was SRS exclusively (SRS-only), some authors considered patients who had not received immune checkpoint inhibitors [14,16,22]. Nevertheless, other authors considered treatment with SRS exclusively when the last dose was applied at least 3 months [23] or 6 months before [27].
According to the results obtained, local control (LC) after SRS-only of cerebral lesions is 45–92.3%. On the other hand, the combination of ICI-SRS treatment places local control between 16.5% and 100%. Typically, the LC obtained in most articles with the ICI-SRS combination is greater than 70%, with the exception of the study by Cohen-Inbar et al. [25], who presented lower LC rates in their results.
If we focus exclusively on those studies that include a comparison of the treatment with SRS-only versus ICI-SRS, only two of them showed a significantly greater LC with the combination treatment [27,33], while in five others, there were no differences between these [14,16,22,29,37].
If we consider the systemic treatment administered (anti-PD-1 vs. anti-CTLA4), a higher LC was observed in patients with melanoma when anti-PD-1 was used with rates of 80–96% [17,20,27,28,34] versus anti-CTLA4 with rates of 16.5–100% [14,15,16,25,29]. Minniti G. et al. reported a statistically significant increase in LC when anti-PD-1 versus anti-CTLA4 was employed (85% vs. 70%, respectively) [31].
If we evaluate the treatment sequence (concomitant vs. sequential), the benefit in LC is more controversial. In those studies that included a comparison of the treatment sequence, in three of them, there were no significant differences [15,29,38]. However, in the study by Chen L. et al., a tendency to significance in favor of concomitant versus sequential treatment was observed (88% vs. 79% respectively; p = 0.08) [37]. Finally, Cohen-Inbar O. et al. did observe a statistically significant higher LC in patients treated with concomitant versus sequential SRS + ICI (54.4% vs. 16.5%; p < 0.05) [25]. Regarding the abscopal effect, none of the included studies on cerebral metastases reported rates of possible distant effects on non-irradiated lesions. Only Kiess et al. reported a possible patient with an abscopal response [15].
Regarding other secondary variables outside the scope of this review, some authors observed greater overall survival with the combination of ICI-SRS treatment versus treatment with SRS-only [22,29], as well as with its concomitant vs. sequential administration/SRS-only [15,16,25,29,37].
As a secondary analysis, regarding progression-free survival (PFS), the combination of ICI-SRS treatment versus SRS-only treatment also appears to show benefits [24,30,32,35]. In terms of toxicity rates, G3-G4 toxicity ranges from 5–24%. G5 toxicity was not reported in any of the studies.
There are currently several ongoing clinical trials, such as the MIGRAINE trial (NCT04427228) and STICk-IM-NSCLC (NCT04650490), that will provide more data in relation to the ICI-SRS combination treatment.

3.2. ICI + SBRT/SABR: Extracerebral Metastases

As mentioned above, SBRT/SABR provides a benefit in the treatment of metastatic patients [9,59].
Following the search, 13 articles that met the stated search criteria were included (Table 2). Of them, six were phase I/II clinical trials, one was a prospective study, and six were retrospective studies. The number of patients varied from 13 to 151 patients. If we consider histology, five included melanoma patients exclusively [39,40,41,42,43], three included NSCLC patients [44,45,46], and six included patients with multiple histologies [47,48,49,50,51,52]. The fractioning schemes used varied between the different studies, with multiple fractioning (3 to 10 fractions) being more frequent [39,40,41,42,43,44,45,46,47,48,49,52] compared to the single fraction [50,51].
Local control of combination treatment with SBRT/SABR + ICI ranged from 40% to 94%. However, we must take into account that the rates with an LC < 60% belong to retrospective studies and with few patients (n < 20) [39,47]. If we consider the results obtained in clinical trials, the LC increases to 75%–91% [41,42,48,49].
The response rate in non-irradiated lesions (abscopal effect) ranged from 1.3% [43] to 45% [42]. If we analyze the six clinical trials included, abscopal response rates between 10% and 45% are reported. Sundahl N. et al. obtained up to 45% of responses in non-irradiated lesions in melanoma patients, of which a full response was observed in 15% [42]. Welsh J.W. et al. observed an overall response of 26% in non-irradiated lesions, obtaining a greater response in lesions that incidentally received low doses of radiation compared to those that did not (31% vs. 5%; p < 0.05) [52]. This finding was also evidenced by Menon H. et al. in their post hoc analysis, reporting a greater response in non-irradiated lesions when receiving low doses of RT [60].
The included studies show heterogeneity in the location of the target or treatment location. In the majority of these studies, patients were compared with lesions treated in multiple locations (lung, liver, bone) that hinder an analysis of the LC or abscopal effect according to the location. Tang et al. suggested that hepatic SBRT may be associated with higher immune systemic activation than lung SBRT, given an early increase in peripheral CD8+T lymphocytes and higher PD-1 expression in CD8+T lymphocytes [48]. Luke et al. did not report differences according to the treated target but did observe a correlation between genes associated with IFN-γ expression and a greater abscopal response [49].
Four of the studies [39,40,43,51] included patients treated with SRS/cerebral radiosurgery. The study by Mowery Y. et al. [43] is the only one to report an abscopal response in a single patient when receiving cerebral SRS.
Regarding the administered systemic treatment, only one study in NSCLC evaluated differences in treatment with anti-PD-1 versus anti-CTLA4 [46]. In this study, a greater abscopal response (37% vs. 24%), overall survival (NA vs. 10.7 months), and disease-free survival (NA vs. 6.4 months) were observed in favor of treatment with anti-PD-1 in a significant way.
Based on the treatment sequence, none of the articles studied the differences in local control or abscopal effect with concomitant versus sequential ICI administration.
Finally, for toxicity ≥G3, the rate ranged from 0 to 34%.
Despite the limitations, the included clinical trials showed a high LC and an abscopal response rate (>10%) that is not inconsiderable. However, clinical trials with a larger number of patients are necessary, in which the impact of the possible abscopal effect is evaluated according to the location of treatment (in the literature, differences in immune signaling according to the irradiated organ are reported) [48,52] and the best treatment sequence.

4. Materials and Methods

4.1. ICI-SBRT/SRS Hypothesis

This systematic review aimed to evaluate the local control and systemic effect of combined ICI and stereotactic radiation therapy (SRS/SBRT) treatment in metastatic patients with NSCLC and melanoma. Overall survival (OS), progression-free survival (PFS), and toxicity of combination therapy were collected.

4.2. Search Strategy

The publications of the last 10 years were reviewed in the MEDLINE database (via PubMed) from January 2010 to December 2020. Articles in English were obtained whose object of study was the combination of stereotactic radiation therapy with immune checkpoints inhibitors in metastatic patients with non-small-cell lung cancer (NSCLC) and melanoma. Multiple terms were used for the search, including “Immunotherapy”, “Anti-PD1”, “Anti-PD-L1”, “Anti-CTLA4”, “Immune checkpoint inhibitors”, “Abscopal effect” and their combination with each of the following terms “SBRT”, “SABR”, “SRS”, “Radiosurgery”, “Stereotactic ablative radiation therapy”, “radiation therapy”, “non-small-cell lung cancer”, and “melanoma”. Non-original articles were excluded.

4.3. Selection Criteria

All articles were evaluated in a first phase according to the title and/or abstract. The articles included in the review had to be based on and comply with the previously defined PICO methodology:
(a)
Metastatic patients of melanoma or non-small-cell lung cancer,
(b)
Patients treated with concomitant/sequential SRS/SBRT to treatment with immune checkpoint inhibitors (anti-PD-L1, anti-PD-1, anti-CTLA4),
(c)
Control group studies (patients treated with ICI without radiation therapy or with SRS/SBRT without ICI) or without a control group,
(d)
Studies whose primary objective was to analyze local control and/or systemic effect (abscopal),
(e)
Clinical trials, prospective studies, and retrospective studies were included.

4.4. Exclusion Criteria

Articles that did not meet the proper design or with a low sample size were excluded. For this purpose, the following were considered as exclusion criteria:
(a)
Opinion articles, case reports, and studies with a sample size (n) less than 10 patients,
(b)
Preclinical articles: tests with murine and in vitro models.

5. Conclusions

The heterogeneity in the number and histology of patients included, in the sequence and systemic treatment administered, as well as the lack of clinical trials makes it difficult to draw robust conclusions.
The combined treatment with ICI-SBRT demonstrates high local control and non-negligible abscopal response in patients with extracerebral metastases of NSCLC and melanoma with an acceptable toxicity.
However, the benefit in local control of the ICI-SRS combination in patients with cerebral metastases is more controversial. Greater local control with anti-PD-1 versus anti-CTLA-4 was observed in cerebral metastases from melanoma patients. An abscopal effect was not reported in the included studies.
Clinical trials with a larger number of patients and more homogeneous samples are needed to obtain conclusive data. Searching for predictive markers of abscopal response in combination therapy could optimize the best sequence and treatment for these patients.

Author Contributions

Conceptualization, M.R.P., D.D.B.; methodology, M.R.P.; software, M.R.P.; validation, M.R.P., D.D.B. and E.F.A.; formal analysis, M.R.P., D.D.B.; investigation, M.R.P., D.D.B., E.F.A.; resources, M.R.P.; data curation, M.R.P.; writing—original draft preparation, M.R.P.; writing—review and editing, M.R.P., D.D.B., E.F.A.; visualization, M.R.P., D.D.B., E.F.A.; supervision, D.D.B., E.F.A.; project administration, M.R.P.; funding acquisition, E.F.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received funding by Foundation INCLIVA.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

RTRadiotherapy
SRSStereotactic radiosurgery
SABRStereotactic ablative body radiotherapy
PD-1Programmed death 1
NSCLCNon-small-cell lung cancer
BEDBiological effective dose
ICIImmune checkpoints inhibitors
SBRTStereotactic body radiotherapy
CTLA-4Cytotoxic T-Lymphocyte Antigen 4.
PD-L1Programmed death ligand 1
BMBrain metastases

References

  1. Frey, B.; Rückert, M.; Deloch, L.; Rühle, P.F.; Derer, A.; Fietkau, R.; Gaipl, U.S. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol. Rev. 2017, 280, 231–248. [Google Scholar] [CrossRef] [PubMed]
  2. Gallegos, C.E.; Michelin, S.; Dubner, D.; Carosella, E.D. Immunomodulation of classical and non-classical HLA molecules by ionizing radiation. Cell Immunol. 2016, 303, 16–23. [Google Scholar] [CrossRef]
  3. Park, B.; Yee, C.; Lee, K.-M. The effect of radiation on the immune response to cancers. Int. J. Mol. Sci. 2014, 15, 927–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mondini, M.; Levy, A.; Meziani, L.; Milliat, F.; Deutsch, E. Radiotherapy-immunotherapy combinations—Perspectives and challenges. Mol. Oncol. 2020, 14, 1529–1537. [Google Scholar] [CrossRef]
  5. Ashrafizadeh, M.; Farhood, B.; Eleojo Musa, A.; Taeb, S.; Rezaeyan, A.; Najafi, M. Abscopal effect in radioimmunotherapy. Int. Immunopharmacol. 2020, 85, 106663. [Google Scholar] [CrossRef] [PubMed]
  6. Watanabe, T.; First, E.; Scholber, J.; Gaedicke, S.; Heinrich, C.; Luo, R.; Ehrat, N.; Multhoff, G.; Schmitt-Graeff, A.; Grosu, A.L.; et al. Deep abscopal response to radiotherapy and anti-PD-1 in an oligometastatic melanoma patient with unfavorable pretreatment immune signature. Cancer Immunol. Immunother. 2020, 69, 1823–1832. [Google Scholar] [CrossRef]
  7. Poleszczuk, J.; Enderling, H. The optimal radiation dose to induce robust systemic anti-tumor immunity. Int. J. Mol. Sci. 2018, 19, 3377. [Google Scholar] [CrossRef] [Green Version]
  8. Palma, D.A.; Olson, R.; Harrow, S.; Gaede, S.; Louie, A.V.; Haasbeek, C.; Mulroy, L.; Lock, M.; Rodrigues, G.B.; Yaremko, B.P.; et al. Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): A randomised, phase 2, open-label trial. Lancet 2019, 393, 2051–2058. [Google Scholar] [CrossRef]
  9. Rheinheimer, S.; Heussel, C.P.; Mayer, P.; Gaissmaier, L.; Bozorgmehr, F.; Winter, H.; Herth, F.J.; Muley, T.; Liersch, S.; Bischoff, H.; et al. Oligoprogressive Non-Small-Cell Lung Cancer under Treatment with PD-(L)1 Inhibitors. Cancers 2020, 12, 1046. [Google Scholar] [CrossRef]
  10. Bernstein, M.B.; Krishnan, S.; Hodge, J.W.; Chang, J.Y. Immunotherapy and stereotactic ablative radiotherapy (ISABR): A curative approach? Nat. Rev. Clin. Oncol. 2016, 13, 516–524. [Google Scholar] [CrossRef]
  11. Ribas, A. Tumor immunotherapy directed at PD-1. N. Engl. J. Med. 2012, 366, 2517–2519. [Google Scholar] [CrossRef] [Green Version]
  12. Matsumura, S.; Wang, B.; Kawashima, N.; Braunstein, S.; Badura, M.; Cameron, T.O.; Babb, J.S.; Schneider, R.J.; Formenti, S.C.; Dustin, M.L.; et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J. Immunol. 2008, 181, 3099–3107. [Google Scholar] [CrossRef]
  13. Bozorgmehr, F.; Chung, I.; Christopoulos, P.; Krisam, J.; Schneider, M.A.; Brückner, L.; Mueller, D.W.; Thomas, M.; Rieken, S. Thoracic radiotherapy plus Durvalumab in elderly and/or frail NSCLC stage III patients unfit for chemotherapy—Employing optimized (hypofractionated) radiotherapy to foster durvalumab efficacy: Study protocol of the TRADE-hypo trial. BMC Cancer 2020, 20, 806. [Google Scholar] [CrossRef]
  14. Mathew, M.; Tam, M.; Ott, P.A.; Pavlick, A.C.; Rush, S.C.; Donahue, B.R.; Golfinos, J.G.; Parker, E.C.; Huang, P.P.; Narayana, A. Ipilimumab in melanoma with limited brain metastases treated with stereotactic radiosurgery. Melanoma Res. 2013, 23, 191–195. [Google Scholar] [CrossRef]
  15. Kiess, A.P.; Wolchok, J.D.; Barker, C.A.; Postow, M.A.; Tabar, V.; Huse, J.T.; Chan, T.A.; Yamada, Y.; Beal, K. 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, 368–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Patel, K.R.; Shoukat, S.; Oliver, D.E.; Chowdhary, M.; Rizzo, M.; Lawson, D.H.; Khosa, F.; Liu, Y.; Khan, M.K. Ipilimumab and stereotactic radiosurgery versus stereotactic radiosurgery alone for newly diagnosed melanoma brain metastases. Am. J. Clin. Oncol. 2017, 40, 444–450. [Google Scholar] [CrossRef] [PubMed]
  17. Ahmed, K.A.; Stallworth, D.G.; Kim, Y.; Johnstone, P.A.; Harrison, L.B.; Caudell, J.J.; Yu, H.H.; Etame, A.B.; Weber, J.S.; Gibney, G.T. Clinical outcomes of melanoma brain metastases treated with stereotactic radiation and anti-PD-1 therapy. Ann. Oncol. 2016, 27, 434–441. [Google Scholar] [CrossRef]
  18. Ahmed, K.A.; Abuodeh, Y.A.; Echevarria, M.I.; Arrington, J.A.; Stallworth, D.G.; Hogue, C.; Naghavi, A.O.; Kim, S.; Kim, Y.; Patel, B.G.; et al. Clinical outcomes of melanoma brain metastases treated with stereotactic radiosurgery and anti-PD-1 therapy, anti-CTLA-4 therapy, BRAF/MEK inhibitors, BRAF inhibitor, or conventional chemotherapy. Ann. Oncol. 2016, 27, 2288–2294. [Google Scholar] [CrossRef]
  19. Kotecha, R.; Miller, J.A.; Venur, V.A.; Mohammadi, A.M.; Chao, S.T.; Suh, J.H.; Barnett, G.H.; Murphy, E.S.; Funchain, P.; Yu, J.S.; et al. Melanoma brain metastasis: The impact of stereotactic radiosurgery, BRAF mutational status, and targeted and/or immune-based therapies on treatment outcome. J. Neurosurg. 2018, 129, 50–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Anderson, E.S.; Postow, M.A.; Wolchok, J.D.; Young, R.J.; Ballangrud, Å.; Chan, T.A.; Yamada, Y.; Beal, K. Melanoma brain metastases treated with stereotactic radiosurgery and concurrent pembrolizumab display marked regression; efficacy and safety of combined treatment. J. Immunother. Cancer 2017, 5, 76. [Google Scholar] [CrossRef] [Green Version]
  21. Choong, E.S.; Lo, S.; Drummond, M.; Fogarty, G.B.; Menzies, A.M.; Guminski, A.; Shivalingam, B.; Clarke, K.; Long, G.V.; Hong, A.M. Survival of patients with melanoma brain metastasis treated with stereotactic radiosurgery and active systemic drug therapies. Eur. J. Cancer 2017, 75, 169–178. [Google Scholar] [CrossRef] [PubMed]
  22. Kaidar-Person, O.; Zagar, T.M.; Deal, A.; Moschos, S.J.; Ewend, M.G.; Sasaki-Adams, D.; Lee, C.B.; Collichio, F.A.; Fried, D.; Marks, L.B.; et al. The incidence of radiation necrosis following stereotactic radiotherapy for melanoma brain metastases: The potential impact of immunotherapy. Anticancer Drugs 2017, 28, 669–675. [Google Scholar] [CrossRef] [PubMed]
  23. Yusuf, M.B.; Amsbaugh, M.J.; Burton, E.; Chesney, J.; Woo, S. Peri-SRS administration of immune checkpoint therapy for melanoma metastatic to the brain: Investigating efficacy and the effects of relative treatment timing on lesion response. World Neurosurg. 2017, 100, 632–640.e4. [Google Scholar] [CrossRef] [PubMed]
  24. An, Y.; Jiang, W.; Kim, B.Y.S.; Qian, J.M.; Tang, C.; Fang, P.; Logan, J.; D’Souza, N.M.; Haydu, L.E.; Wang, X.A.; et al. Stereotactic radiosurgery of early melanoma brain metastases after initiation of anti-CTLA-4 treatment is associated with improved intracranial control. Radiother. Oncol. 2017, 125, 80–88. [Google Scholar] [CrossRef]
  25. Cohen-Inbar, O.; Shih, H.-H.; Xu, Z.; Schlesinger, D.; Sheehan, J.P. The effect of timing of stereotactic radiosurgery treatment of melanoma brain metastases treated with ipilimumab. J. Neurosurg. 2017, 127, 1007–1014. [Google Scholar] [CrossRef] [Green Version]
  26. Robin, T.P.; Breeze, R.E.; Smith, D.E.; Rusthoven, C.G.; Lewis, K.D.; Gonzalez, R.; Brill, A.; Saiki, R.; Stuhr, K.; Gaspar, L.E.; et al. Immune checkpoint inhibitors and radiosurgery for newly diagnosed melanoma brain metastases. J. Neurooncol. 2018, 140, 55–62. [Google Scholar] [CrossRef]
  27. Trommer-Nestler, M.; Marnitz, S.; Kocher, M.; Rueß, D.; Schlaak, M.; Theurich, S.; von Bergwelt-Baildon, M.; Morgenthaler, J.; Jablonska, K.; Celik, E.; et al. Robotic stereotactic radiosurgery in melanoma patients with brain metastases under simultaneous anti-PD-1 treatment. Int. J. Mol. Sci. 2018, 19, 2653. [Google Scholar] [CrossRef] [Green Version]
  28. Nardin, C.; Mateus, C.; Texier, M.; Lanoy, E.; Hibat-Allah, S.; Ammari, S.; Robert, C.; Dhermain, F. Tolerance and outcomes of stereotactic radiosurgery combined with anti-programmed cell death-1 (pembrolizumab) for melanoma brain metastases. Melanoma Res. 2018, 28, 111–119. [Google Scholar] [CrossRef]
  29. Diao, K.; Bian, S.X.; Routman, D.M.; Yu, C.; Ye, J.C.; Wagle, N.A.; Wong, M.K.; Zada, G.; Chang, E.L. Stereotactic radiosurgery and ipilimumab for patients with melanoma brain metastases: Clinical outcomes and toxicity. J. Neurooncol. 2018, 139, 421–429. [Google Scholar] [CrossRef]
  30. Stera, S.; Balermpas, P.; Blanck, O.; Wolff, R.; Wurster, S.; Baumann, R.; Szücs, M.; Loutfi-Krauss, B.; Wilhelm, M.L.; Seifert, V.; et al. Stereotactic radiosurgery combined with immune checkpoint inhibitors or kinase inhibitors for patients with multiple brain metastases of malignant melanoma. Melanoma Res. 2019, 29, 187–195. [Google Scholar] [CrossRef]
  31. Minniti, G.; Anzellini, D.; Reverberi, C.; Cappellini, G.C.A.; Marchetti, L.; Bianciardi, F.; Bozzao, A.; Osti, M.; Gentile, P.C.; Esposito, V. Stereotactic radiosurgery combined with nivolumab or Ipilimumab for patients with melanoma brain metastases: Evaluation of brain control and toxicity. J. Immunother. Cancer 2019, 7, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Murphy, B.; Walker, J.; Bassale, S.; Monaco, D.; Jaboin, J.; Ciporen, J.; Taylor, M.; Dai Kubicky, C. Concurrent radiosurgery and immune checkpoint inhibition: Improving regional intracranial control for patients with metastatic melanoma. Am. J. Clin. Oncol. 2019, 42, 253–257. [Google Scholar] [CrossRef]
  33. Hadi, I.; Roengvoraphoj, O.; Bodensohn, R.; Hofmaier, J.; Niyazi, M.; Belka, C.; Nachbichler, S.B. Stereotactic radiosurgery combined with targeted/immunotherapy in patients with melanoma brain metastasis. Radiat. Oncol. 2020, 15, 37. [Google Scholar] [CrossRef] [PubMed]
  34. Carron, R.; Gaudy-Marqueste, C.; Amatore, F.; Padovani, L.; Malissen, N.; Balossier, A.; Loundou, A.; Bonnet, N.; Muracciole, X.; Régis, J.M. Stereotactic radiosurgery combined with anti-PD1 for the management of melanoma brain metastases: A retrospective study of safety and efficacy. Eur. J. Cancer 2020, 135, 52–61. [Google Scholar] [CrossRef]
  35. Ahmed, K.A.; Kim, S.; Arrington, J.; Naghavi, A.O.; Dilling, T.J.; Creelan, B.C.; Antonia, S.J.; Caudell, J.J.; Harrison, L.B.; Sahebjam, S. Outcomes targeting the PD-1/PD-L1 axis in conjunction with stereotactic radiation for patients with non-small cell lung cancer brain metastases. J. Neurooncol. 2017, 133, 331–338. [Google Scholar] [CrossRef]
  36. Singh, C.; Qian, J.M.; Yu, J.B.; Chiang, V.L. Local tumor response and survival outcomes after combined stereotactic radiosurgery and immunotherapy in non-small cell lung cancer with brain metastases. J. Neurosurg. 2019, 132, 512–517. [Google Scholar] [CrossRef]
  37. Chen, L.; Douglass, J.; Kleinberg, L.; Ye, X.; Marciscano, A.E.; Forde, P.M.; Brahmer, J.; Lipson, E.; Sharfman, W.; Hammers, H.; 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, 916–925. [Google Scholar] [CrossRef]
  38. Koenig, J.L.; Shi, S.; Sborov, K.; Gensheimer, M.F.; Li, G.; Nagpal, S.; Chang, S.D.; Gibbs, I.C.; Soltys, S.G.; Pollom, E.L. Adverse radiation effect and disease control in patients undergoing stereotactic radiosurgery and immune checkpoint inhibitor therapy for brain metastases. World Neurosurg. 2019, 126, e1399–e1411. [Google Scholar] [CrossRef]
  39. Kropp, L.M.; De Los Santos, J.F.; McKee, S.B.; Conry, R.M. Radiotherapy to control limited melanoma progression following ipilimumab. J. Immunother. 2016, 39, 373–378. [Google Scholar] [CrossRef] [Green Version]
  40. Koller, K.M.; Mackley, H.B.; Liu, J.; Wagner, H.; Talamo, G.; Schell, T.D.; Pameijer, C.; Neves, R.I.; Anderson, B.; Kokolus, K.M.; et al. Improved survival and complete response rates in patients with advanced melanoma treated with concurrent ipilimumab and radiotherapy versus ipilimumab alone. Cancer Biol. Ther. 2017, 18, 36–42. [Google Scholar] [CrossRef] [Green Version]
  41. Sundahl, N.; De Wolf, K.; Kruse, V.; Meireson, A.; Reynders, D.; Goetghebeur, E.; Van Gele, M.; Speeckaert, R.; Hennart, B.; Brochez, L.; et al. Phase 1 dose escalation trial of ipilimumab and stereotactic body radiation therapy in metastatic melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 906–915. [Google Scholar] [CrossRef] [PubMed]
  42. Sundahl, N.; Seremet, T.; Van Dorpe, J.; Neyns, B.; Ferdinande, L.; Meireson, A.; Brochez, L.; Kruse, V.; Ost, P. Phase 2 trial of nivolumab combined with stereotactic body radiation therapy in patients with metastatic or locally advanced inoperable melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2019, 104, 828–835. [Google Scholar] [CrossRef]
  43. Mowery, Y.M.; Patel, K.; Chowdhary, M.; Rushing, C.N.; Roy Choudhury, K.; Lowe, J.R.; Olson, A.C.; Wisdom, A.J.; Salama, J.K.; Hanks, B.A.; et al. Retrospective analysis of safety and efficacy of anti-PD-1 therapy and radiation therapy in advanced melanoma: A bi-institutional study. Radiother. Oncol. 2019, 138, 114–120. [Google Scholar] [CrossRef]
  44. Lesueur, P.; Escande, A.; Thariat, J.; Vauléon, E.; Monnet, I.; Cortot, A.; Lerouge, D.; Danhier, S.; Dô, P.; Dubos-Arvis, C.; et al. Safety of combined PD-1 pathway inhibition and radiation therapy for non-small-cell lung cancer: A multicentric retrospective study from the GFPC. Cancer Med. 2018, 7, 5505–5513. [Google Scholar] [CrossRef] [PubMed]
  45. Formenti, S.C.; Rudqvist, N.P.; Golden, E.; Cooper, B.; Wennerberg, E.; Lhuillier, C.; Vanpouille-Box, C.; Friedman, K.; Ferrari de Andrade, L.; Wucherpfennig, K.W.; et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat. Med. 2018, 24, 1845–1851. [Google Scholar] [CrossRef]
  46. Chen, D.; Menon, H.; Verma, V.; Guo, C.; Ramapriyan, R.; Barsoumian, H.; Younes, A.; Hu, Y.; Wasley, M.; Cortez, M.A.; et al. Response and outcomes after anti-CTLA4 versus anti-PD1 combined with stereotactic body radiation therapy for metastatic non-small cell lung cancer: Retrospective analysis of two single-institution prospective trials. J. Immunother. Cancer 2020, 8, e000492. [Google Scholar] [CrossRef] [Green Version]
  47. Ribeiro Gomes, J.; Schmerling, R.A.; Haddad, C.K.; Racy, D.J.; Ferrigno, R.; Gil, E.; Zanuncio, P.; Buzaid, A.C. Analysis of the abscopal effect with anti-PD1 therapy in patients with metastatic solid tumors. J. Immunother. 2016, 39, 367–372. [Google Scholar] [CrossRef]
  48. Tang, C.; Welsh, J.W.; de Groot, P.; Massarelli, E.; Chang, J.Y.; Hess, K.R.; Basu, S.; Curran, M.A.; Cabanillas, M.E.; Subbiah, V.; et al. Ipilimumab with stereotactic ablative radiation therapy: Phase I results and immunologic correlates from peripheral T cells. Clin. Cancer Res. 2017, 23, 1388–1396. [Google Scholar] [CrossRef] [Green Version]
  49. Luke, J.J.; Lemons, J.M.; Karrison, T.G.; Pitroda, S.P.; Melotek, J.M.; Zha, Y.; Al-Hallaq, H.A.; Arina, A.; Khodarev, N.N.; Janisch, L.; et al. Safety and clinical activity of pembrolizumab and multisite Stereotactic body radiotherapy in patients with advanced solid tumors. J. Clin. Oncol. 2018, 36, 1611–1618. [Google Scholar] [CrossRef] [PubMed]
  50. Maity, A.; Mick, R.; Huang, A.C.; George, S.M.; Farwell, M.D.; Lukens, J.N.; Berman, A.T.; Mitchell, T.C.; Bauml, J.; Schuchter, L.M.; et al. A phase I trial of pembrolizumab with hypofractionated radiotherapy in patients with metastatic solid tumours. Br. J. Cancer 2018, 119, 1200–1207. [Google Scholar] [CrossRef] [Green Version]
  51. Desideri, I.; Francolini, G.; Scotti, V.; Pezzulla, D.; Becherini, C.; Terziani, F.; Delli Paoli, C.; Olmetto, E.; Visani, L.; Meattini, I.; et al. Benefit of ablative versus palliative-only radiotherapy in combination with nivolumab in patients affected by metastatic kidney and lung cancer. Clin. Transl. Oncol. 2019, 21, 933–938. [Google Scholar] [CrossRef]
  52. Welsh, J.W.; Tang, C.; de Groot, P.; Naing, A.; Hess, K.R.; Heymach, J.V.; Papadimitrakopoulou, V.A.; Cushman, T.R.; Subbiah, V.; Chang, J.Y.; et al. Phase II trial of ipilimumab with stereotactic radiation therapy for metastatic disease: Outcomes, toxicities, and low-dose radiation-related abscopal responses. Cancer Immunol. Res. 2019, 7, 1903–1909. [Google Scholar] [CrossRef] [Green Version]
  53. Cagney, D.N.; Martin, A.M.; Catalano, P.J.; Redig, A.J.; Lin, N.U.; Lee, E.Q.; Wen, P.Y.; Dunn, I.F.; Bi, W.L.; Weiss, S.E.; et al. Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: A population-based study. Neuro Oncol. 2017, 19, 1511–1521. [Google Scholar] [CrossRef] [Green Version]
  54. Nayak, L.; Lee, E.Q.; Wen, P.Y. Epidemiology of brain metastases. Curr. Oncol. Rep. 2012, 14, 48–54. [Google Scholar] [CrossRef]
  55. Sampson, J.H.; Carter, J.H., Jr.; Friedman, A.H.; Seigler, H.F. Demographics, prognosis, and therapy in 702 patients with brain metastases from malignant melanoma. J. Neurosurg. 1998, 88, 11–20. [Google Scholar] [CrossRef] [Green Version]
  56. National Comprehensive Cancer Network. (NCCN) Non-Small-Cell Lung Cancer (Version 4.2021). Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 24 May 2021).
  57. National Comprehensive Cancer Network (NCCN). Melanoma: Cutaneous (Version 2.2021). Available online: https://www.nccn.org/professionals/physician_gls/pdf/cutaneous_melanoma.pdf (accessed on 24 May 2021).
  58. Halasz, L.M.; Uno, H.; Hughes, M.; D’Amico, T.; Dexter, E.U.; Edge, S.B.; Hayman, J.A.; Niland, J.C.; Otterson, G.A.; Pisters, K.M.; et al. Comparative effectiveness of stereotactic radiosurgery versus whole-brain radiation therapy for patients with brain metastases from breast or non-small cell lung cancer: WBRT vs SRS for Brain Metastases. Cancer 2016, 122, 2091–2100. [Google Scholar] [CrossRef] [Green Version]
  59. Gomez, D.R.; Blumenschein, G.R., Jr.; Lee, J.J.; Hernandez, M.; Ye, R.; Camidge, D.R.; Doebele, R.C.; Skoulidis, F.; Gaspar, L.E.; Gibbons, D.L.; et al. Local consolidative therapy versus maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer without progression after first-line systemic therapy: A multicentre, randomised, controlled, phase 2 study. Lancet Oncol. 2016, 17, 1672–1682. [Google Scholar] [CrossRef] [Green Version]
  60. Menon, H.; Chen, D.; Ramapriyan, R.; Verma, V.; Barsoumian, H.B.; Cushman, T.R.; Younes, A.I.; Cortez, M.A.; Erasmus, J.J.; de Groot, P.; et al. Influence of low-dose radiation on abscopal responses in patients receiving high-dose radiation and immunotherapy. J. Immunother. Cancer 2019, 7, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Selection of articles included in the review.
Figure 1. Selection of articles included in the review.
Ijms 22 11621 g001
Table 1. Results of the ICI + SRS combination in cerebral metastases in melanoma/non-small-cell lung cancer (n = 25).
Table 1. Results of the ICI + SRS combination in cerebral metastases in melanoma/non-small-cell lung cancer (n = 25).
AuthorStudy TypenNº LesionsMedian Follow up (Months)HistologyTargetDoses/Fraction (Gy/fx)ITGroupsLocal Control
(CR + PR + SR)
Abscopal
Responses
Median Survival
(Months)
Median PFS
(Months)
Toxicity ≥ Grade 3 (%)
Mathew M., et al., 2013 [14]Retrospective581986MelanomaBrain15–20 Gy/1fxAnti-CTLA4SRS
SRS + IT
65%
63%
NR5.9NRNR
Kiess, A.P., et al., 2015 [15]Retrospective46113NRMelanomaBrain15–24 Gy/1fxAnti-CTLA4SRS + IT before
SRS + IT conc
SRS + IT after
87%
100%
89%
NRNRNRG3–4: 20%
G5: 0%
Patel K., et al., 2015 [16]Retrospective54NR7.3MelanomaBrainNRAnti-CTLA4SRS
SRS + IT
92.3%
71.4%
NRNR4.2
3.1
15%
Ahmed, et al., 2015 [17]Retrospective26739.4MelanomaBrain18–24 Gy/1fx
25–30 Gy/5fx
Anti-PD-1SRS + IT89%NR124.60%
Ahmed, et al., 2016 [18]Retrospective963147.4MelanomaBrain15–24 Gy/1fxVariousSRS + IT83%NR10,53.4NR
Kotecha R., et al., 2018 [19]Retrospective1917937MelanomaBrainNRVariousSRS + IT86%NRNRNRNR
Anderson E., et al., 2017 [20]Retrospective11239.2MelanomaBrain18–21 Gy/1fx
30Gy/5fx
Anti-PD-1SRS + IT conc96%NRNDNDG3: 5%
G4–5: 0%
Choong E., et al., 2017 [21]Retrospective108NR8.6MelanomaBrainNRVariousSRS + IT78%NR14.2ND3%
Kaidar-Person O., et al., 2017 [22]Retrospective58NR21MelanomaBrain18–20 Gy/1fx
21–30 Gy/3–5fx
Anti-PD-1
Anti-CTLA4
SRS
SRS + IT
86%
52%
NR5.5
15
8
5
NR
Yusuf M., et al., 2017 [23]Prospective511677MelanomaBrain13–24 Gy/1fxAnti-PD-1
Anti-CTLA4
SRS
SRS + IT conc
75%NRNRNRNR
An Y., et al., 2017 [24]Retrospective7125715.5MelanomaBrain16–24Gy/1fxAnti-PD-1
Anti-CTLA4
Inhibitor BRAF
SRS + IT90%NR12NRNR
Cohen-Inbar O., et al., 2017 [25]Retrospective462327.9MelanomaBrain14–22 Gy/1fxAnti-CTLA4SRS + IT conc
SRS + IT seq
54.4%
16.5%
NR13.8
6.4
7.2
5
NR
Robin T., et al., 2018 [26]Retrospective38NR31.6MelanomaBrainNRAnti-PD-1
Anti-CTLA4
SRS + IT92%NRNA3.4G3: 8%
G4–5: 0%
Trommer M., et al., 2018 [27]Retrospective2648NRMelanomaBrain18–22 Gy/1fxAnti-PD-1SRS + IT
SRS
86%
80%
NRNRNR0%
AuthorStudy TypenNº LesionsMedian Follow up
(Months)
HistologyTargetDoses/FractionITGroupsLocal Control
(CR + PR + SR)
Abscopal
Responses
Median Survival
(Months)
Median PFS
(Months)
Toxicity ≥ Grade 3 (%)
Nardin C., et al., 2018 [28]Retrospective74NR14MelanomaBrain12–24 Gy/1fx
24–35 Gy/1–5fx
Anti-PD-1SRS + IT80%NR15.33G3: 12%
G4–5: 0%
Diao K., et al.,
2018 [29]
Retrospective91NRNRMelanomaBrain12–22 Gy/1fxAnti-CTLA4SRS
SRS + IT conc
SRS + IT seq
45%
60%
70%
NR7.8
11.8
18.7
NRG3–4: 5%
G5: 0%
Stera S., et al.,
2018 [30]
Retrospective452508,3MelanomaBrain15–20 Gy/1fxAnti-PD-1
Anti-CTLA4
Inh BRAF/MEK
SRS + IT89.5%NRNRNRG3: 8.33%
G4–5:0%
Minniti G., et al., 2019 [31]Retrospective8032615MelanomaBrain18–22 Gy/1fx
27 Gy/3fx
Anti-PD-1
Anti-CTLA4
SRS + anti-PD1
SRS+anti-CTLA4
85%
70%
NR22
14.7
NR24% (G3)
17% (G3)
Murphy B., et al., 2019 [32]Retrospective269018.9MelanomaBrain22 Gy/1fx
27–30 Gy/3–5 fx
Anti-PD-1
Anti-CTLA4
SRS + IT conc
SRS + IT seq
95.4%NR26.119
3.4
G3: 8% G4–5:0%
Hadi I., et al.,
2020 [33]
Retrospective305219MelanomaBrain18-24 Gy /1fxVariousSRS + IT
SRS
100%
83.3%
NR221613.5%
Carron R., et al.,
2020 [34]
Retrospective5018138.9MelanomaBrain22–26 Gy/1fxAnti-PD-1SRS + IT94%NR16.613.214.6%
Ahmed K., et al., 2017 [35]Retrospective17498.6NSCLCBrain18–24 Gy/1fx
25 Gy/5fx
Anti-PD-1
Anti- PD-L1
SRS + IT96%NR5.7NRNR
Singh C., et al., 2019 [36]Retrospective8553112NSCLCBrain12–24 Gy/1–3 fxAnti-PD-1 +/- anti-CTLA4SRS + IT
SRS + QT
97%
96.6%
NR10
11.6
4.6
6.1
NR
Chen L., et al.,
2018 [37]
Retrospective2606239.2NSCLC (60%)
Melanoma (30%)
Renal (10%)
Brain15–24 Gy/1fx
18–24 Gy/3fx
25 Gy/5fx
Anti-PD-1
Anti-CTLA4
SRS
SRS+ IT seq
SRS +IT conc
82%
79%
88%
NR12.9
14.5
24.7
NRG3: 19%
G4: 1%
G5: 0%
Koening J., et al., 2019 [38]Retrospective97580NRMelanoma (39%)
NSCLC (46%)
Others (15%)
Brain22–30 Gy/1–5 fxAnti-PD-1
Anti- PD-L1
Anti-CTLA4
SRS+ IT conc
SRS+IT seq
96%
97%
NR9.4NRG3: 7%
G4: 7%
IT: Immunotherapy; CR: Complete response; PR: Partial response; SR: Stable response; NR: Not reported; Conc: Concomitant; Seq: Sequential.
Table 2. Results of the ICI + SRS combination in extracerebral metastases in melanoma/non-small-cell lung cancer (n = 14).
Table 2. Results of the ICI + SRS combination in extracerebral metastases in melanoma/non-small-cell lung cancer (n = 14).
AuthorStudy TypenMedian Follow up (Months)HistologyTargetDoses/Fraction (Gy/fx)ITGroupsLocal Control
(CR + PR + SR)
Abscopal
Responses
Median Survival
(Months)
Median PFS
(Months)
Toxicity ≥ Grade 3 (%)
Kropp L., et al., 2016 [39]Retrospective1625.5MelanomaVisceral
11% Brain
30 Gy/5fx
36 Gy/6fx
Anti-CTLA4SBRT + IT seq56%NRNRNR0%
Koller K., et al., 2017 [40]Retrospective10119MelanomaVisceral
Bone
Brain
NR (13% SBRT)Anti-CTLA4RT + IT conc
IT
NR37.1%19
10
5
3
NR
Sundahl N., et al., 2018 [41]Phase I13NRMelanomaVisceral
Bone
24–36 Gy/3 fxAnti-CTLA4SBRT+IT conc91%23%18.5NR25%
Sundah N., et al., 2019 [42]Phase II2013.1MelanomaVisceral8 Gy/3 fxAnti-PD-1SBRT+IT conc90%45%NRNRG3: 15%
G4–5: 0%
Mowery Y., et al., 2019 [43]Retrospective15112.9MelanomaVariousNRAnti-PD-1RT + IT (SRS 26%)
IT
NR1.31%NR5
NA
G3: NR
G4–5: 0%
Lesueur P., et al., 2018 [44]Retrospective10415.8NSCLCVisceral
Bone
Brain
SBRT: 20–36 Gy/1–6fx
RT: 20–30Gy/5–10fx
Anti-PD-1RT/SBRT + IT64.4%NR112.7G3–4:
14.4%
G5: 0%
Formenti S., et al., 2018 [45]Prospective3943NSCLCVisceral
Bone
30 Gy/5fx or
27 Gy/3fx
Anti-CTLA4SBRT+ IT seqNR18%7.43.810%
Chen L., et al., 2020 [46]Retrospective33NRNSCLCVisceral60 Gy/10fx or
50 Gy/4fx
Anti-PD-1
Anti-CTLA4
SBRT+anti-PD-1
SBRT +anti-CTLA4
88%
94%
37%
24%
NA
10.7
NA
6.4
19%
29%
Ribeiro Gomes J., et al., 2016 [47]Retrospective168Melanoma (75%)
NSCLC (12.5%)
Renal (12.5%)
Visceral
Bone
24 Gy (1–40)/
3fx (1–10)
Anti-PD-1SBRT+ IT40%18.7%7.4NR0%
Tang C., et al., 2017 [48]Phase I359.3Various
(NSCLC 22.8%)
VisceralConc: 50 Gy/4fx
Seq: 50 Gy/4fx or 60 Gy/10fx
Anti-CTLA4SBRT conc
SBRT seq
90.3%10%10.23.2G3: 34%
G4–5: 0%
Luke J., et al.,
2018 [49]
Phase I797.1Various
(NSCLC 9.6%)
Visceral30–50 Gy/3–5 fxAnti-PD-1SBRT + IT seq75%13.5%9.63.19,6%
Maity A., et al., 2018 [50]Phase I24NRVarious
Melanoma (17%)
NSCLC (33%)
Visceral
Bone
24 Gy/3fx or
17 Gy/1fx
Anti-PD-1SBRT+IT concNR12.5%6.91.9G3: 33%
G4–5: 0%
Desideri I., et al., 2018 [51]Retrospective20NRNSCLC (85%)
Renal (15%)
Visceral
Brain
SBRT:18–40 Gy/1–5fx
RT: 8–30 Gy/1–10fx
Anti-PD-1SBRT+ IT conc
RT + IT conc
87.5%
NR
NR17.9
10.3
11.5
5.2
G3: 15%
G4–5: 0%
Welsh J., et al., 2019 [52]Phase II10610.5Various
(NSCL 18%)
Visceral60 Gy/10fx
50 Gy/4fx
Anti-CTLA4SBRT+IT seq
SBRT + IT conc
NR26%NA2.9G3: 33%
G4–5: 0%
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rodríguez Plá, M.; Dualde Beltrán, D.; Ferrer Albiach, E. Immune Checkpoints Inhibitors and SRS/SBRT Synergy in Metastatic Non-Small-Cell Lung Cancer and Melanoma: A Systematic Review. Int. J. Mol. Sci. 2021, 22, 11621. https://doi.org/10.3390/ijms222111621

AMA Style

Rodríguez Plá M, Dualde Beltrán D, Ferrer Albiach E. Immune Checkpoints Inhibitors and SRS/SBRT Synergy in Metastatic Non-Small-Cell Lung Cancer and Melanoma: A Systematic Review. International Journal of Molecular Sciences. 2021; 22(21):11621. https://doi.org/10.3390/ijms222111621

Chicago/Turabian Style

Rodríguez Plá, María, Diego Dualde Beltrán, and Eduardo Ferrer Albiach. 2021. "Immune Checkpoints Inhibitors and SRS/SBRT Synergy in Metastatic Non-Small-Cell Lung Cancer and Melanoma: A Systematic Review" International Journal of Molecular Sciences 22, no. 21: 11621. https://doi.org/10.3390/ijms222111621

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop