Next Article in Journal
Hybrid Formation and Fusion of Cancer Cells In Vitro and In Vivo
Previous Article in Journal
The Expression Profiles and Deregulation of UDP-Glycosyltransferase (UGT) Genes in Human Cancers and Their Association with Clinical Outcomes
Previous Article in Special Issue
Immunotherapy in Non-Small Cell Lung Cancer Patients with Brain Metastases: Clinical Challenges and Future Directions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Intracranial Treatment in Melanoma Patients with Brain Metastasis Is Associated with Improved Survival in the Era of Immunotherapy and Anti-BRAF Therapy

1
Radiation Oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France
2
Dermato-Oncology Department, Institut Universitaire du Cancer, CEDEX 9, 31059 Toulouse, France
3
Biostatistics Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France
4
Gamma Knife Unit, CHU–Toulouse-Purpan, 31000 Toulouse, France
5
Neuro-Surgery Department, CHU de Toulouse–Purpan, 31000 Toulouse, France
6
Radiation Oncology Department, Oncorad, Clinique Pasteur, 31000 Toulouse, France
7
Dermatology Department, CHU de Toulouse, Hôpital Larrey, CEDEX 9, 31059 Toulouse, France
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(17), 4493; https://doi.org/10.3390/cancers13174493
Submission received: 11 June 2021 / Revised: 19 August 2021 / Accepted: 1 September 2021 / Published: 6 September 2021
(This article belongs to the Special Issue Immunotherapy in Cancer Metastasis)

Abstract

:

Simple Summary

Melanoma is one of the top three causes of brain metastases, which is still a poor prognostic factor of overall survival. Novel systemic therapies have changed the prognosis of patients and the place of local intracranial treatment, surgery and/or radiotherapy in this era remains unclear. We evaluated the incidence of brain metastasis in melanoma patients in a large retrospective French cohort who received immunotherapy and/or targeted therapy for and identified prognostic factors. Local intracranial treatment is statistically significantly associated with improved overall survival through comparable groups in terms of age, number of BM, BRAF status and systemic treatment. The question of administering local treatment, even for more than one metastasis, such as stereotactic radiotherapy, should be addressed at the diagnosis of brain metastasis while introducing systemic treatment such as immunotherapy. Three prospective trials evaluating additional SRT in combination to ipilimumab and nivolumab association are ongoing.

Abstract

Metastatic melanoma patients are at high risk of brain metastases (BM). Although intracranial control is a prognostic factor for survival, impact of local (intracranial) treatment (LT), surgery and/or radiotherapy (stereotactic or whole brain) in the era of novel therapies remains unknown. We evaluated BM incidence in melanoma patients receiving immune checkpoint inhibitors (ICI) or anti-BRAF therapy and identified prognostic factors for overall survival (OS). Clinical data and treatment patterns were retrospectively collected from all patients treated for newly diagnosed locally advanced or metastatic melanoma between May 2014 and December 2017 with available BRAF mutation status and receiving systemic therapy. Prognostic factors for OS were analyzed with univariable and multivariable survival analyses. BMs occurred in 106 of 250 eligible patients (42.4%), 64 of whom received LT. Median OS in patients with BM was 7.8 months (95% CI [5.4–10.4]). In multivariable analyses, LT was significantly correlated with improved OS (HR 0.21, p < 0.01). Median OS was 17.3 months (95% CI [8.3–22.3]) versus 3.6 months (95% CI [1.4–4.8]) in patients with or without LT. LT correlates with improved OS in melanoma patients with BM in the era of ICI and anti-BRAF therapy. The use of LT should be addressed at diagnosis of BM while introducing systemic treatment.

1. Introduction

Immune checkpoint inhibitors (ICI) and BRAF + MEK inhibitors combinations have dramatically changed the management of metastatic melanoma patients and improved outcomes [1,2]. However, melanoma is still the third most common cancer type associated with brain metastasis (BM), after lung and breast carcinomas, even in cases of complete extracranial disease control [3]. It has been estimated that a proportion of 20% present brain metastasis at baseline and 50% of patients display intra-cranial progression in the course of metastatic disease (up to 80% of patients in autopsy series) [4,5].
To date, several phases II studies of targeted therapy or immunotherapy have been reported in melanoma patients with central nervous system metastases. [3,6,7,8,9].
In the case of asymptomatic BMs, ipilimumab plus nivolumab is considered as the standard treatment leading to high overall response rate (ORR) in the brain (51–54%) [6,9]. Although dabrafenib plus trametinib shows clear activity in patients with BRAF V600-mutated asymptomatic BMs (ORR 58%), the durability of these responses appears shorter, with a 1-year PFS of 19% [3]. Therefore, there is consensus to use combination immunotherapy in patients with BMs irrespective of BRAF mutational status [10].
Despite recent advances in the management of systemic disease, the occurrence of BMs remains associated with a significantly worse prognosis [11,12,13,14]. BM melanoma patients are a heterogeneous subgroup with a variable prognosis. Prognosis is associated with risk factors including ECOG ≥ 1, presence > 3 BM, extracranial tumour load and age >70 years [13,15,16]. Given the small size of the lesions in both BM trials, the impact of local treatment in the era of novel therapies remains unclear [17,18,19]. We conducted a retrospective study to assess potential prognostic factors, especially outcomes according to administration of local (intracranial) treatment (LT) with surgery and/or radiotherapy for BM in melanoma patients receiving ICI or anti-BRAF therapy.

2. Materials and Methods

2.1. Study Design and Data Collection

As part of an institutional board-approved study, all consecutive patients treated at the Institut Universitaire du Cancer and CHU de Toulouse, France between May 2014 and December 2017 for newly diagnosed, locally advanced or metastatic melanoma with available tumour status for BRAF mutation were identified by the treatments’ database which excluded patients who did not received systemic treatment. Data on demographics, clinical pathology and molecular analyses, including prognostic factors according to the updated melanoma-molGPA score [15,20] were retrospectively extracted from prospectively collected medical records.

Treatments

For each patient, therapeutic strategy was discussed by the institutional melanoma tumour board. Choice of systemic therapy was based on tumour BRAF status and previously received systemic therapies. Regimens administered included a BRAF + MEK inhibitor (BRAFi + MEKi) combination consisting of dabrafenib 150 mg twice daily plus trametinib 2 mg daily. Patients receiving the BRAFi + MEKi regimen with concurrent radiotherapy were instructed to interrupt treatment for three days before and during radiotherapy. Drug intake was resumed the day after. Anti-PD-1 therapy consisted of nivolumab (3 mg/kg, every 2 weeks) or pembrolizumab (2 mg/kg, every 3 weeks). The anti-CTLA-4 therapy ipilimumab was administered as four infusions of 3 mg/kg, every 3 weeks. Both anti-PD-1 and anti-CTLA-4 were administered concomitantly with radiation therapy. In the event of progression after ICI or BRAF therapy, chemotherapy consisting of dacarbazine, fotemustine or temozolomide was administered. Patients with BM received either systemic therapy with or without LT, consisting of surgery, radiation therapy (whole brain radiation therapy [WBRT], or definitive or adjuvant stereotactic radiation therapy [SRT]) taking into account the size and number of BM, the medical condition of the patient and the status of the extra-cranial disease. Surgery was planned in cases of voluminous (>3 cm) or symptomatic BM, particularly in case of inaugural BM present at metastatic diagnosis. SRT was selected in cases of limited intracranial disease (≤3 BM) and was performed at a dose of 20 Gy delivered in a single fraction, or 27 Gy delivered in three fractions for BM with a diameter >2 cm (Gamma Knife, Elekta® or Novalis, Varian®) prescribed to the 80% isodose. Adjuvant SRT was delivered to the tumour resection’s bed at a dose of 27 Gy or 30 Gy delivered in three or five fractions, respectively (Novalis, Varian®). Alternatively, WBRT was administered at a dose of 30 Gy or 37.5 Gy in 10 to 15 fractions in the event of multiple simultaneous BM (≥4). Combined treatment was defined as immunotherapy or BRAFi delivered within the three months preceding or 11 days following LT respectively (corresponding to five half-lives) [18,21]. All patients underwent systematic follow-up including brain CT scan or Gadolinium-enhanced MRI every 3 months after the first metastasis was diagnosed or, in the event of neurological symptoms, during the course of the disease. Toxicity related to LT was reported according to the Common Terminology Criteria for Adverse Events (CTCAE), version 4.3.

2.2. Statistical Analysis

Patient characteristics were presented with descriptive statistics. Qualitative variables were summarized using the frequency and percentage for each category. Continuous variables were summarized using median and range. Differences between groups were assessed using Chi-square or Fisher’s exact test for categorical variables and Kruskal–Wallis test for continuous variables. Overall survival (OS) was defined as the time from the first BM to death or last follow-up (censored data) and was estimated with the Kaplan-Meier method and presented with 95% confidence intervals (CI).
Univariable analyses were performed using the log-rank test. Qualitative ssignificant variables on univariable analyses and clinically relevant variables were included in the multivariable analysis, which was performed using the Cox proportional hazards model and presented with hazard ratios (HR) and the 95% CI. For all statistical tests, differences were considered significant at the 5% level. All statistical analyses were performed using STATA 13.0 software.

2.3. Ethics

All of the regulatory procedures required to comply with the laws in force in France were complied with (declaration to the Health Data Hub under MR-004).

3. Results

3.1. Patient Population

Of the 330 patients screened for this study, 80 were excluded from analyses (27 of whom did not receive systemic therapy and 53 who were lost to follow-up), resulting in 250 eligible patients. With a median follow-up of 28 months (95% CI 25–33), 106 (42.4%) patients presented BMs during the course of their disease. Demographic, disease and treatment characteristics of the 250 patients are presented in Table 1 according to the presence of BM or not. Among the 106 patients with BM, 29.2% presented symptoms at time of BM diagnosis, including intracranial hypertension (n = 13), sensitive-motor deficit (n = 12) and epilepsy (n = 7).

3.2. BM and Survival

The 106 patients with BM showed a median OS of 7.8 months (95% CI [5.4–10.4]). In this population, age < 70 years at first BM, LDH levels ≤ 1 ULN (upper limit of normal), three or fewer BM and LT were significantly correlated with favourable prognosis in the univariable analysis (Table 2). Multivariable analysis showed that LT (HR = 0.21, 95% CI [0.10–0.43], p < 0.01), ECOG PS ≤ 2 (HR = 0.40% CI [0.18–0.88], p = 0.02) and LDH ≤ 1 ULN (HR = 0.44, 95% CI [0.21–0.95], p = 0.04) were significantly associated with better OS (Table 3). Among patients who had not received LT, median OS was 3.6 months (95% CI [1.4–4.8]) and the 12-month OS rate was 9.6% (95% CI [2.8–21.7]), compared to 17.3 months (95% CI [8.3–22.3]) and 56.1% (95% CI [42.8–67.5]), respectively, in patients who had undergone LT (Figure 1; p < 0.001). As previously reported, the addition of fractioned SRT to anti-PD-1 treatment after initial progression led to a complete extracranial and intracranial response in one patient via an abscopal effect [22].

3.3. BM Patients with or without LT

Sixty-four of the 106 patients with BM (60.4%) received LT, with a median of follow-up of 28.5 months (95% CI [25.2;33.2]) versus 27.8 months (95% CI [1 0.7;NR]) for patients who did not receive LT. Local treatment consisted of definitive SRT (n = 28, 43.8%), WBRT (n = 21, 32.8%) or surgery (n = 16, 25%), which was followed by radiation therapy of the operating bed using SRT for eight patients and WBRT for one patient. Concurrent systemic therapy was administered to 44 of the 64 (68.8%) patients, including ipilimumab (n = 6, 13.6%), anti-PD-1 (n = 19, 43.2%), targeted therapy (n = 15, 34.1%), or chemotherapy (n = 4, 9.1%). After the diagnosis of BM, 61.9%, 53.6% and 57.1% of patients who respectively underwent surgery, SRT or WBRT received at least one line of ICI during the process of the disease, and 33.3%, 35.7% and 42.9% of patients who respectively underwent surgery, SRT and WBRT received at least one line of targeted therapy during the process of the disease (Table 4).
Seventeen patients underwent LT after a 2 month interval; meanwhile they received systemic treatment.
Patients with BM who underwent LT were more likely to present with neurological symptoms at diagnosis of BM (p < 0.01), normal LDH (p < 0.01) and be systemic treatment naïve (inaugural BM) than patients who did not (p = 0.04) (Table 3). Patients who underwent LT were less likely to receive immunotherapy before BM appearance than pateints who did not have LT (p = 0.03). There was no difference in receiving previous iBRAF and/or iMEK for BRAF-mutated patients before BM.

3.4. Safety of LT

No grade 4 or 5 radiation or surgical related toxicities were reported in the 64 patients who underwent LT. No events associated with the surgical procedure were reported. Details of radiation-induced side effects are provided on Table 5.

4. Discussion

We herein report on outcomes from a large retrospective single-institution cohort of metastatic melanoma patients treated during the era of novel systemic therapies. Of the 250 patients with metastatic melanoma, 42.4% presented BMs during the course of their disease, associated with a poor median OS (7.8 months), which is consistent with previously reported rates [3,7,12,15]. The median OS of 3.6 months (95% CI [1.4–4.8]) and a 12-month OS rate of 9.6% (95% CI [2.8–21.7]) in patients who had not received LT compared to median OS of 17.3 months (95% CI [8.3–22.3]) and a 12-month survival rate of 56.1% (95% CI [42.8–67.5]) in patients who received LT (p < 0.001), shows that LT significantly favours survival. In multivariable analyses, LT remained significantly associated with improved OS (HR 0.21, 95% CI [0.1–0.43]; p < 0.001). Despite major therapeutic advances, BM remained associated with a poorer prognosis. Moreover, only a few data are available on the treatment of melanoma BM, as these patients were systematically excluded from clinical trials [23]. The rare clinical trials addressing the treatment of melanoma patients with BM did not evaluate combined strategies including LTs. These studies also illustrated the heterogeneity of the melanoma BM subpopulation and the major prognostic differences between BM patients. This makes it difficult to extrapolate results from prospective trials in relation to highly selected patients and strengthens the need of additional local treatment strategies to improve intracranial control even in the era of recent effective systemic treatment. In our study, using prognostic factors from the melanoma-molGPA score, we found comparable survival rates to those previously reported by Sperduto et al., and patients were well balanced according to melanoma molGPA criteria whether they received LT or not [15,20]. So far LT has not been used to define the criteria of the MolGPA score.
Survival of our unselected patients with BM was similar to that reported in the phase II COMBI-MB study (median OS ranged from 11 months for asymptomatic patients without LT to 24 months for asymptomatic ones with previous LT) and in line with the non-reached median OS in BM patients receiving nivolumab plus ipilimumab in two phase II studies [3,6]. Interestingly, in another phase II study, BREAK-MB, evaluating dabrafenib as a single agent therapy for patients with melanoma BM, the subgroup of BM patients who did not receive LT was also associated with lower survival rates than those receiving LT (median of OS 4 vs. 8 months respectively) [7]. Vosoughi et al. also reported in a retrospective analysis that LT (craniotomy or SRT) was independently associated with improved OS (17 months after LT versus 12 months for the entire cohort from time to BM diagnosis) [12]. Together these studies highlight that intracranial control including local treatment is associated with improved overall survival [3,6,12].
The role of local treatment for single BM patients has been investigated across various tumours. Even in cases necessitating initial surgery (i.e., symptomatic lesions or inaugural diagnosis), adjuvant irradiation, either with WBRT or fractioned-SRT, can add benefit to local control [24,25,26]. SRT has shown better results in terms of quality of life whereas WBRT brings cognitive deterioration which is no more compatible with prolonged survival [27,28,29]. In a recent randomized phase III study, WBRT after local treatment for one to three melanoma BMs did not provide any clinical benefit, distant intracranial control, survival or preservation of quality of life, which differs from other histologies, highlighting the need for adapted studies [30]. Median of OS was 16 months in the entire population, which is comparable to our results.
In the latest German guidelines, LT (but not WBRT) is recommended for all BM in case of oligometastatic disease [31]. Definitive or adjuvant SRT delivered in single or multiple fractions, depending on the size of the BM, is recommended in the first management step when technically feasible [19,26,32,33]. In the randomised phase II COMET trial, ablative SRT of oligo-metastatic patients, including those with BM derived from various tumours, was associated with improved OS (13 months) [33].
Many retrospective studies reported enhanced local control when radiotherapy was delivered in combination with immunotherapy, providing further evidence for a synergistic effect of ICIs and radiation therapy [34,35,36].
Several preclinical studies reported that the combination of low-dose fractionated radiation therapy and PD-1/PD-L1 axis inhibitors activates cytotoxic T cells infiltrating and immediately surrounding the tumour [37]. Recent studies reported clinical results of this synergy, leading to an immunotherapy-mediated abscopal effect [9,17,34,35,36]. In a retrospective analysis of patients with BM from various tumours, patients who received immunotherapy in combination with fractionated SRT had significantly better 1-year local control (96% vs. 78%, p = 0.02) and improved distant intracranial disease-free survival (62% vs. 51%, p = 0.007) than those who did not [35]. In a retrospective studies of 46 patients, better results in OS and local recurrence were reported if immunotherapy was delivered during (or before) SRT than after [37]. In another retrospective series of 25 patients, SRT delivered in less than 30 days after immunotherapy yielded a trend toward a better brain control [38]. Nevertheless, these small series lack the power to be included in the melanoma BM strategy.
For Braf-mutated tumours, the combination of iBraf + iMek in the first line is challenged by the bi-immunotherapy anti CTLA4 + antiPD1/PDL1, especially for an asymptomatic and slow progressive disease. Either with ICI, or with targeted therapies, bitherapy is superior to monotherapy [31]. The ongoing clinical trials SECOMBIT and IMMUNOCOBIVEN will help to define the best therapeutic sequence in Braf-mutated patients [39,40].
We observed very few adverse events when LT was combined with systemic therapy, confirming the previously reported safety of such a therapeutic combination [22,41]. Safety of combination therapy in the literature is heterogeneous; however almost all authors agree that the putative slightly increased risk is manageable, particularly in light of the likely benefit on efficacy and survival [42,43]. In our study, 10% of the patients developed radionecrosis, which was, in most cases, reversible and manageable with transient steroid therapy and consistent with previously reported results [35]. However given this non-negligible risk, and the difficulty of distinguishing it from local failure, multimodal MRI is strongly recommended during follow-up [44]. Interestingly some authors previously reported that fractionated SRT can reduce the incidence of radionecrosis compared to a single fraction, without compromising local benefit [45].
In addition to its retrospective nature, our study presents a number of limitations. Although the two treatment subgroups (LT vs. no LT) were well balanced with respect to age, number of BM, BRAF status and systemic treatment, patients receiving LT presented significantly more symptomatic BM. Interestingly, despite this negative clinical feature, this subgroup had a better prognosis, supporting our hypothesis that LT is associated with improved survival. Furthermore, a large proportion of our patients (32%) receiving LT was treated with WBRT, which has been reported to be associated with worse local control compared with SRT [28,32], which may have lowered the potential survival benefit of LT. In recent years, a growing body of evidence supports adjuvant or definitive SRT as an alternative to WBRT as an initial therapy approach [10].

5. Conclusions

We report on a large series of 106 consecutive melanoma patients with BM receiving systemic therapy of ICI or anti-BRAF inhibitors. Intracranial control has been a recognised prognostic factor of the utmost importance in BM patients for some time, including in the current era of immunotherapy and targeted therapy. Our multivariable analysis confirmed that intracranial treatment was significantly associated with improved OS. The combination of radiation therapy and immunotherapy was well tolerated, avoiding delay for systemic treatment initiation. The question of administering LT such as stereotactic radiotherapy should be addressed at diagnosis of BM, while introducing systemic treatment such as immunotherapy. Three prospective trials; NCT03340129, NCT0210775 and NCT03728465 evaluating additional SRT in combination to ipilimumab and nivolumab association are ongoing [46,47].

Author Contributions

Conceptualization, A.M., N.M., C.D.; methodology, A.M., C.D., L.C.; software, L.C.; formal analysis, A.M., C.D., N.M., L.C.; investigation, C.D.; resources, C.P., C.C., V.S., E.M., J.C.S., I.L., N.M., A.M.; data curation, C.D., L.C.; writing—original draft preparation, C.D., A.M.; writing—review and editing, C.D., C.P., L.C., C.C., V.S., E.M., J.C.S., I.L., N.M., A.M.; visualization, C.D., A.M.; supervision, A.M.; project administration, A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, was approved by the Institutional Review Board (or Ethics Committee) of Institut Universitaire de Toulouse and was declared to the French Data protection authority “CNIL” on Health Data Hub under MR-004.

Informed Consent Statement

According to the regulatory procedures required to comply with the laws in force in France, an informative document has been sent to living patients with the possibility for them to forbid the use of personal data even if anonymized.

Data Availability Statement

Data available on request due to restrictions e.g., privacy or ethical. The data presented in this study are available on request from the corresponding author. The data are not publicly available due to restrictions e.g., privacy.

Acknowledgments

Disclosure: Modesto reports for honoraria Bristol-Myer Squibb and Bayer; for consulting or advisory roles Merk Serono and Astra Zeneca; for research funding Astra Zeneca, and for travel grants Bristol-Myer Squibb. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. Meyer reports for consulting or advisory role Bristol-Myer Squibb, MSD Oncology, Genentech, Novartis and Pierre Fabre. For Research funding Bristol-Myer Squibb, (inst) and MSD Oncology (inst). The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. Sibaud reports for advisory role and as speaker Bristol-Myer Squibb, Novartis, MSD and Pierre Fabre. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. Pagès reports for consulting or advisory role Bristol-Myer Squibb, MSD Oncology, Genentech, Novartis and Pierre Fabre. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Conflicts of Interest

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References

  1. Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in Previously Untreated Melanoma without BRAF Mutation. N. Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.-J.; et al. Dabrafenib and Trametinib versus Dabrafenib and Placebo for Val600 BRAF-Mutant Melanoma: A Multicentre, Double-Blind, Phase 3 Randomised Controlled Trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  3. Davies, M.A.; Saiag, P.; Robert, C.; Grob, J.-J.; Flaherty, K.T.; Arance, A.; Chiarion-Sileni, V.; Thomas, L.; Lesimple, T.; Mortier, L.; et al. Dabrafenib plus Trametinib in Patients with BRAFV600-Mutant Melanoma Brain Metastases (COMBI-MB): A Multicentre, Multicohort, Open-Label, Phase 2 Trial. Lancet Oncol. 2017, 18, 863–873. [Google Scholar] [CrossRef]
  4. 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] [PubMed] [Green Version]
  5. Schouten, L.J.; Rutten, J.; Huveneers, H.A.M.; Twijnstra, A. Incidence of Brain Metastases in a Cohort of Patients with Carcinoma of the Breast, Colon, Kidney, and Lung and Melanoma. Cancer 2002, 94, 2698–2705. [Google Scholar] [CrossRef] [PubMed]
  6. Long, G.V.; Atkinson, V.; Lo, S.; Sandhu, S.; Guminski, A.D.; Brown, M.P.; Wilmott, J.S.; Edwards, J.; Gonzalez, M.; Scolyer, R.A.; et al. Combination Nivolumab and Ipilimumab or Nivolumab Alone in Melanoma Brain Metastases: A Multicentre Randomised Phase 2 Study. Lancet Oncol. 2018, 19, 672–681. [Google Scholar] [CrossRef]
  7. Long, G.V.; Trefzer, U.; Davies, M.A.; Kefford, R.F.; Ascierto, P.A.; Chapman, P.B.; Puzanov, I.; Hauschild, A.; Robert, C.; Algazi, A.; 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, 1087–1095. [Google Scholar] [CrossRef]
  8. Margolin, K.; Ernstoff, M.S.; Hamid, O.; Lawrence, D.; McDermott, D.; Puzanov, I.; Wolchok, J.D.; Clark, J.I.; Sznol, M.; Logan, T.F.; et al. Ipilimumab in Patients with Melanoma and Brain Metastases: An Open-Label, Phase 2 Trial. Lancet Oncol. 2012, 13, 459–465. [Google Scholar] [CrossRef]
  9. Tawbi, H.A.-H.; Forsyth, P.A.J.; Hodi, F.S.; Lao, C.D.; Moschos, S.J.; Hamid, O.; Atkins, M.B.; Lewis, K.D.; Thomas, R.P.; Glaspy, J.A.; et al. Efficacy and Safety of the Combination of Nivolumab (NIVO) plus Ipilimumab (IPI) in Patients with Symptomatic Melanoma Brain Metastases (CheckMate 204). JCO 2019, 37, 9501. [Google Scholar] [CrossRef]
  10. Keilholz, U.; Ascierto, P.A.; Dummer, R.; Robert, C.; Lorigan, P.; van Akkooi, A.; Arance, A.; Blank, C.U.; Chiarion Sileni, V.; Donia, M.; et al. ESMO Consensus Conference Recommendations on the Management of Metastatic Melanoma: Under the Auspices of the ESMO Guidelines Committee. Ann. Oncol. 2020, 31, 1435–1448. [Google Scholar] [CrossRef]
  11. Long, G.V.; Grob, J.-J.; Nathan, P.; Ribas, A.; Robert, C.; Schadendorf, D.; Lane, S.R.; Mak, C.; Legenne, P.; Flaherty, K.T.; et al. Factors Predictive of Response, Disease Progression, and Overall Survival after Dabrafenib and Trametinib Combination Treatment: A Pooled Analysis of Individual Patient Data from Randomised Trials. Lancet Oncol. 2016, 17, 1743–1754. [Google Scholar] [CrossRef]
  12. Vosoughi, E.; Lee, J.M.; Miller, J.R.; Nosrati, M.; Minor, D.R.; Abendroth, R.; Lee, J.W.; Andrews, B.T.; Leng, L.Z.; Wu, M.; 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. [Google Scholar] [CrossRef] [PubMed]
  13. Badakhshi, H.; Engeling, F.; Budach, V.; Ghadjar, P.; Zschaeck, S.; Kaul, D. Are Prognostic Indices for Brain Metastases of Melanoma Still Valid in the Stereotactic Era? Radiat. Oncol. 2018, 13, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Buchsbaum, J.C.; Suh, J.H.; Lee, S.-Y.; Chidel, M.A.; Greskovich, J.F.; Barnett, G.H. Survival by Radiation Therapy Oncology Group Recursive Partitioning Analysis Class and Treatment Modality in Patients with Brain Metastases from Malignant Melanoma: A Retrospective Study. Cancer 2002, 94, 2265–2272. [Google Scholar] [CrossRef]
  15. Sperduto, P.W.; Jiang, W.; Brown, P.D.; Braunstein, S.; Sneed, P.; Wattson, D.A.; Shih, H.A.; Bangdiwala, A.; Shanley, R.; Lockney, N.A.; et al. Estimating Survival in Melanoma Patients With Brain Metastases: An Update of the Graded Prognostic Assessment for Melanoma Using Molecular Markers (Melanoma-MolGPA). Int. J. Radiat. Oncol. Biol. Phys. 2017, 99, 812–816. [Google Scholar] [CrossRef] [Green Version]
  16. Frinton, E.; Tong, D.; Tan, J.; Read, G.; Kumar, V.; Kennedy, S.; Lim, C.; Board, R.E. Metastatic Melanoma: Prognostic Factors and Survival in Patients with Brain Metastases. J. Neurooncol. 2017, 135, 507–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. 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] [Green Version]
  18. Tallet, A.V.; Dhermain, F.; Le Rhun, E.; Noël, G.; Kirova, Y.M. Combined Irradiation and Targeted Therapy or Immune Checkpoint Blockade in Brain Metastases: Toxicities and Efficacy. Ann. Oncol. 2017, 28, 2962–2976. [Google Scholar] [CrossRef]
  19. Modesto, A.; Chira, C.; Sol, J.-C.; Lubrano, V.; Boulinguez, S.; Pagès, C.; Sibaud, V.; Gomez-Roca, C.; Moyal, É.; Meyer, N. Prise En Charge Des Patients Atteints de Métastases Cérébrales de Mélanome. Cancer/Radiothérapie 2019, 23, 147–150. [Google Scholar] [CrossRef]
  20. Dalmasso, C.; Pagès, C.; Chaltiel, L.; Brun, A.; Sibaud, V.; Boulinguez, S.; Chira, C.; Moyal, E.; Lubrano, V.; Meyer, N.; et al. Survival Estimation of Melanoma Patients with Brain Metastasis Using the Melanoma-MolGPA Score: External Validation from a French Cohort. Melanoma Res. 2020, 30, 472–476. [Google Scholar] [CrossRef]
  21. Magnuson, W.J.; Lester-Coll, N.H.; Wu, A.J.; Yang, T.J.; Lockney, N.A.; Gerber, N.K.; Beal, K.; Amini, A.; Patil, T.; Kavanagh, B.D.; et al. Management of Brain Metastases in Tyrosine Kinase Inhibitor–Naïve Epidermal Growth Factor Receptor–Mutant Non–Small-Cell Lung Cancer: A Retrospective Multi-Institutional Analysis. JCO 2017, 35, 1070–1077. [Google Scholar] [CrossRef]
  22. Aboudaram, A.; Modesto, A.; Chaltiel, L.; Gomez-Roca, C.; Boulinguez, S.; Sibaud, V.; Delord, J.-P.; Chira, C.; Delannes, M.; Moyal, E.; et al. Concurrent Radiotherapy for Patients with Metastatic Melanoma and Receiving Anti-Programmed-Death 1 Therapy: A Safe and Effective Combination. Melanoma Res. 2017, 27, 485–491. [Google Scholar] [CrossRef]
  23. Donia, M.; Kimper-Karl, M.L.; Høyer, K.L.; Bastholt, L.; Schmidt, H.; Svane, I.M. The Majority of Patients with Metastatic Melanoma Are Not Represented in Pivotal Phase III Immunotherapy Trials. Eur. J. Cancer 2017, 74, 89–95. [Google Scholar] [CrossRef]
  24. Patchell, R.A.; Regine, W.F.; Loeffler, J.S.; Sawaya, R.; Andrews, D.W.; Chin, L.S. Radiosurgery Plus Whole-Brain Radiation Therapy for Brain Metastases. JAMA 2006, 296, 2089–2091. [Google Scholar] [CrossRef]
  25. Soffietti, R.; Kocher, M.; Abacioglu, U.M.; Villa, S.; Fauchon, F.; Baumert, B.G.; Fariselli, L.; Tzuk-Shina, T.; Kortmann, R.-D.; Carrie, C.; 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. [Google Scholar] [CrossRef]
  26. Mahajan, A.; Ahmed, S.; McAleer, M.F.; Weinberg, J.S.; Li, J.; Brown, P.; Settle, S.; Prabhu, S.S.; Lang, F.F.; Levine, N.; 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, 1040–1048. [Google Scholar] [CrossRef]
  27. Sperduto, P.W.; Shanley, R.; Luo, X.; Andrews, D.; Werner-Wasik, M.; Valicenti, R.; Bahary, J.-P.; Souhami, L.; Won, M.; Mehta, M. Secondary Analysis of RTOG 9508, a Phase 3 Randomized Trial of Whole-Brain Radiation Therapy Versus WBRT Plus Stereotactic Radiosurgery in Patients With 1-3 Brain Metastases; Poststratified by the Graded Prognostic Assessment (GPA). Int. J. Radiat. Oncol. Biol. Phys. 2014, 90, 526–531. [Google Scholar] [CrossRef] [Green Version]
  28. Mulvenna, P.; Nankivell, M.; Barton, R.; Faivre-Finn, C.; Wilson, P.; McColl, E.; Moore, B.; Brisbane, I.; Ardron, D.; Holt, T.; 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, 2004–2014. [Google Scholar] [CrossRef] [Green Version]
  29. Kocher, M.; Soffietti, R.; Abacioglu, U.; Villà, S.; Fauchon, F.; Baumert, B.G.; Fariselli, L.; Tzuk-Shina, T.; Kortmann, R.-D.; Carrie, C.; 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. JCO 2011, 29, 134–141. [Google Scholar] [CrossRef] [Green Version]
  30. Hong, A.M.; Fogarty, G.B.; Dolven-Jacobsen, K.; Burmeister, B.H.; Lo, S.N.; Haydu, L.E.; Vardy, J.L.; Nowak, A.K.; Dhillon, H.M.; Ahmed, T.; et al. Adjuvant Whole-Brain Radiation Therapy Compared With Observation After Local Treatment of Melanoma Brain Metastases: A Multicenter, Randomized Phase III Trial. J. Clin. Oncol. 2019, 37, 3132–3141. [Google Scholar] [CrossRef]
  31. Gutzmer, R.; Vordermark, D.; Hassel, J.C.; Krex, D.; Wendl, C.; Schadendorf, D.; Sickmann, T.; Rieken, S.; Pukrop, T.; Höller, C.; et al. Melanoma Brain Metastases—Interdisciplinary Management Recommendations 2020. Cancer Treat. Rev. 2020, 89, 102083. [Google Scholar] [CrossRef]
  32. Brown, P.D.; Ballman, K.V.; Cerhan, J.H.; Anderson, S.K.; Carrero, X.W.; Whitton, A.C.; Greenspoon, J.; Parney, I.F.; Laack, N.N.I.; Ashman, J.B.; 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, 1049–1060. [Google Scholar] [CrossRef]
  33. 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]
  34. Petrelli, F.; De Stefani, A.; Trevisan, F.; Parati, C.; Inno, A.; Merelli, B.; Ghidini, M.; Bruschieri, L.; Vitali, E.; Cabiddu, M.; et al. Combination of Radiotherapy and Immunotherapy for Brain Metastases: A Systematic Review and Meta-Analysis. Crit. Rev. Oncol./Hematol. 2019, 144, 102830. [Google Scholar] [CrossRef]
  35. Guénolé, M.; Lucia, F.; Bourbonne, V.; Dissaux, G.; Reygagne, E.; Goasduff, G.; Pradier, O.; Schick, U. Impact of Concomitant Systemic Treatments on Toxicity and Intracerebral Response after Stereotactic Radiotherapy for Brain Metastases. BMC Cancer 2020, 20, 991. [Google Scholar] [CrossRef]
  36. Gatterbauer, B.; Hirschmann, D.; Eberherr, N.; Untersteiner, H.; Cho, A.; Shaltout, A.; Göbl, P.; Fitschek, F.; Dorfer, C.; Wolfsberger, S.; et al. Toxicity and Efficacy of Gamma Knife Radiosurgery for Brain Metastases in Melanoma Patients Treated with Immunotherapy or Targeted Therapy—A Retrospective Cohort Study. Cancer Med. 2020, 9, 4026–4036. [Google Scholar] [CrossRef] [Green Version]
  37. Dovedi, S.J.; Cheadle, E.J.; Popple, A.L.; Poon, E.; Morrow, M.; Stewart, R.; Yusko, E.C.; Sanders, C.M.; Vignali, M.; Emerson, R.O.; 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, 5514–5526. [Google Scholar] [CrossRef] [Green Version]
  38. 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] [Green Version]
  39. Skrepnik, T.; Sundararajan, S.; Cui, H.; Stea, B. Improved Time to Disease Progression in the Brain in Patients with Melanoma Brain Metastases Treated with Concurrent Delivery of Radiosurgery and Ipilimumab. OncoImmunology 2017, 6, e1283461. [Google Scholar] [CrossRef] [Green Version]
  40. Fondazione Melanoma Onlus. A Three Arms Prospective, Randomized Phase II Study to Evaluate the Best Sequential Approach with Combo Immunotherapy (Ipilimumab/Nivolumab) and Combo Target Therapy (LGX818/MEK162) in Patients with Metastatic Melanoma and BRAF Mutation. Available online: Clinicaltrials.gov (accessed on 5 September 2021).
  41. Schadendorf, D. A Phase II, Open-Label, Randomized-Controlled Trial Evaluating the Efficacy and Safety of a Sequencing Schedule of Cobimetinib Plus Vemurafenib Followed by Immunotherapy with an Anti- PD-L1 Antibody Atezolizumab for the Treatment in Patients with Unresectable or Metastatic BRAF V600 Mutant Melanoma. Available online: Clinicaltrials.gov (accessed on 5 September 2021).
  42. 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]
  43. 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] [PubMed]
  44. Glitza Oliva, I.; Tawbi, H.; Davies, M.A. Melanoma Brain Metastases: Current Areas of Investigation and Future Directions. Cancer J. 2017, 23, 68–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Minniti, G.; Scaringi, C.; Paolini, S.; Lanzetta, G.; Romano, A.; Cicone, F.; Osti, M.; Enrici, R.M.; Esposito, V. Single-Fraction Versus Multifraction (3 × 9 Gy) Stereotactic Radiosurgery for Large (>2 Cm) Brain Metastases: A Comparative Analysis of Local Control and Risk of Radiation-Induced Brain Necrosis. Int. J. Radiat. Oncol. Biol. Phys. 2016, 95, 1142–1148. [Google Scholar] [CrossRef] [PubMed]
  46. Melanoma Institute Australia. A Phase II, Open Label, Randomised, Controlled Trial of Ipilimumab and Nivolumab with Concurrent Intracranial Stereotactic Radiotherapy versus Ipilimumab and Nivolumab Alone in Patients with Melanoma Brain Metastases. Available online: Clinicaltrials.gov (accessed on 5 September 2021).
  47. University Hospital Tuebingen. An Open Label Phase II Study to Evaluate Safety and Efficacy of Combined Treatment With Ipilimumab and Nivolumab in Patients With Four and More Symptomatic Brain Metastases of Melanoma. Available online: Clinicaltrials.gov (accessed on 30 May 2021).
Figure 1. Kaplan-Meier estimate of overall survival according to local intracranial treatment for melanoma brain metastasis in patients with brain metastases.
Figure 1. Kaplan-Meier estimate of overall survival according to local intracranial treatment for melanoma brain metastasis in patients with brain metastases.
Cancers 13 04493 g001
Table 1. Patient characteristics according to the presence of BM, at the diagnosis of 1st metastasis.
Table 1. Patient characteristics according to the presence of BM, at the diagnosis of 1st metastasis.
Total
n = 250
Patients with BM n = 106Patients without BM n = 144
Age at 1st metastasis in years
Median666566
(range)(27–90)(27–89)(28–90)
Sex
Male141 (56.4%)64 (60.4%)77 (53.5%)
female109 (43.6%)42 (39.6%)67 (46.5%)
Primary site of metastasis
Limbs88 (35.2%)43 (40.6%)45 (31.3%)
Trunk66 (26.4%)27 (25.5%)39 (27.1%)
Head or neck34 (13.6%)15 (14.2%)19 (13.2%)
Mucosa21 (8.4%)4 (3.8%)17 (11.8%)
Unknown31 (12.4%)15 (14.2%)16 (11.1%)
Other10 (4.0%)2 (1.9%)8 (5.6%)
Molecular characteristics
BRAF105 (42.0%)52 (49.1%)53 (36.8%)
NRAS38 (15.2%)15 (14.2%)23 (16.0%)
BRAF + NRAS2 (0.8%)0 (0.0%)2 (1.4%)
cKIT11 (4.4%)6 (5.7%)5 (3.5%)
BRAF + cKIT1 (0.4%)1 (0.9%)0 (0.0%)
Wildtype88 (35.2%)31 (29.2%)57 (36.9%)
LDH
≤1 ULN79 (52.7%)32 (50%)47 (54.7%)
>1 ULN71 (47.3%)32 (50%)39 (45.3%)
Missing1004258
BM: brain metastasis, LDH: lactate dehydrogenase, ULN: upper limit of normal. p value statistically significant, <0.05.
Table 2. Univariable and multivariable analysis of OS in patients with BM.
Table 2. Univariable and multivariable analysis of OS in patients with BM.
UnivariableMultivariable
12-Month Survival[95% CI]p ValueHR[95% CI]p Value
Age at first BM
<70 years49.6%[36.6; 61.3] 1
≥70 years20.1%[9.1; 34.1]0.041.07[0.52–2.22]0.86
ECOG PS
2/3/425.0%[9.1; 44.9%] 1
0/141.8%[30.5; 52.7]0.210.40[0.18–0.88]0.02
LDH
>1 ULN16.2%[5.9; 30.9] 1
≤1 ULN57.8%[37.6; 73.5]<0.010.44[0.21–0.95]0.04
Intracranial local treatment
No9.6%[2.8; 21.7] 1
Yes56.1%[42.8; 67.5]<0.010.21[0.10–0.43]<0.01
Number of BM
≤349.6%[36.4; 61.5] 1
>322.8%[11.2; 36.8]<0.011.61[0.81;3.21]0.17
Maximal diameter
≤10 mm50.8%[30.6; 67.8]
>10 mm41.6%[27.4; 55.3]0.35
BRAF mutation
Absent31.2%[19.1; 44.2]
present45.6%[31.4; 58.8]0.70
Gender
Male37.0%[25.2; 48.8]
Female41.1%[25.2; 56.0]0.94
BM: brain metastases, CI: confidence interval, HR: Hazard ratio, ECOG PS: Eastern-Cooperative-Oncology-Group Performance Status, LDH: lactate dehydrogenase. Bold police: p value statistically significant, <0.05.
Table 3. Characteristics in patients with BM, according to intracranial treatment.
Table 3. Characteristics in patients with BM, according to intracranial treatment.
Patients with BM
n = 106
Patients without Local Treatment n = 42Patients with Local Treatment n = 64p Value
Age at first BM in years66.068.565.00.62
Median (range)(27.0–90.0)(27.0–90.0)(28.0–89.0)
Gender
Male64 (60.4%)28 (66.7%)36 (56.3%)0.28
Female42 (39.6%)14 (33.3%)28 (43.8%)
Molecular characteristics
BRAF53 (50%)19 (45.2%)34 (53.1%)0.43
NRAS15 (14.2%)5 (11.9%)10 (15.6%)0.59
c-KIT7 (6.6%)3 (7.1%)4 (6.3%)1.0
Wildtype31 (29.2%)16 (38.1%)15 (23.4%)0.10
LDH
≤132 (50%)8 (29.6%)24 (64.9%)<0.01
>132 (50%)19 (70.4%)13 (35.1%)
Missing421527
ECOG PS (n = 101)
0/180 (79.2%)31 (77.5%)49 (80.3%)0.7320
2/3/421 (20.8%)9 (22.5%)12 (19.7%)
missing523
Extracranial disease at first metastasis
No5 (4.7%)05 (7.8%)
Yes101 (95.3%)42 (100%)59 (92.2%)0.15
Node61 (57.5%)26 (61.9%)35 (54.7%)0.46
Liver27 (25.5%)16 (38.1%)11 (17.2%)0.02
Lung51 (48.1%)20 (47.6)31 (48.4%)0.93
Bone 20 (18.9%)10 (23.8%)10 (15.6%)0.29
Inaugural * BM14 (13.2%)2 (4.8%)12 (18.8%)0.04
Metachronous ** BM92 (86.8%)40 (95.2%)52 (81.3%)
Neurologic symptoms at diagnosis of BM
Yes31 (29.2%)4 (9.5%)27 (42.2%)<0.01
No75 (70.8%)38 (90.5%)37 (57.8)
Number of BM
145 (42.9%)15 (35.7%)30 (47.6%)0.45
2–425 (23.8%)12 (28.6%)13 (20.6%)
>435 (33.3%)15 (35.7%)20 (31.7%)
Missing101
Maximal diameter (mm)
Median (range)
13.0 (1.0–60.0)11 (1.0–43.0)14.5 (1.0–60.0)0.08
Previous systemic treatment
No49 (46.2%)12 (28.6%)37 (57.8%)<0.01
Yes57 (53.8%)30 (71.4%)27 (42.2%)
Previous immunotherapy
No66 (62.3%)21 (50%)45 (70.3%)0.03
Yes40 (37.7%)21 (50%)19 (29.7%)
For BRAF patients: previous iBRAF/iMEK
No28 (52.8%)7 (36.8%)21 (61.8%)0.08
Yes25 (47.2%)12 (63.2%)13 (38.2%)
Melanoma-molGPA (n = 100)
0–123 (23.0%)9 (22.5%)14 (23.3%)0.15
1.5–251 (51.0%)25 (62.5%)26 (43.3%)
2.5–324 (24.0%)6 (15.0%)18 (30.0%)
3.5–42 (2.0%)0 2 (3.3%)
Missing624
BM: brain metastases, LDH: lactate dehydrogenase, ECOG PS: Eastern-Cooperative-Oncology-Group Performance Status, Melanoma-molGPA = updated Graded Prognostic Assessment index. Bold police: p value statistically significant, < 0.05. *: Inaugural BM means presence of BM at the time of the melanoma’s diagnosis. **: Metachronous BM means absence of BM at the time of the melanoma’s diagnosis, differed apparition of BM.
Table 4. Systemic treatment received at least one time after diagnosis of brain metastasis according to the local treatment.
Table 4. Systemic treatment received at least one time after diagnosis of brain metastasis according to the local treatment.
No LT.
(n = 42)
SRT
(n = 28)
WBRT
(n = 21)
Surgery (+/−RT)
(n = 16)
No systemic treatment 12 (28.6%)5 (17.9%)4 (19.0%)2 (12.5%)
At least one line of ICI21 (50.0%)15 (53.6%)12 (57.1%)13 (61.9%)
At least one line of anti-BRAF and/or anti-Mek12 (28.6%)10 (35.7%)9 (42.9%)7 (33.3%)
At least one line of chemotherapy5 (11.9%)7 (25.0%)5 (23.8%)3 (14.2%)
Only chemotherapy2 (4.8%)2 (7.1%)2 (9.5%)0 (0.0%)
LT: local treatment, WBRT: whole brain radiation therapy, SRT: stereotatctic radiosurgery, RT: radiation therapy (i.e., SRT or WBRT), ICI: immune checkpoint inhibitor.
Table 5. Radiation therapy related side-effects, grades and corresponding treatment.
Table 5. Radiation therapy related side-effects, grades and corresponding treatment.
Total Radiation Therapy Patients
n = 58
Late toxicities
Radionecrosis
All grade 6
Grade 16
Grade 22
Grade 33
Treatment for radionecrosis
Steroids4
Surgery1
Radio-induced Oedema
All grade9
Grade 10
Grade 24
Grade 35
Treatment for radio-induced Oedema
Steroids 9
Anticonvulsant9
Haemorrhagic transformation
All grades0
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dalmasso, C.; Pagès, C.; Chaltiel, L.; Sibaud, V.; Moyal, E.; Chira, C.; Sol, J.C.; Latorzeff, I.; Meyer, N.; Modesto, A. Intracranial Treatment in Melanoma Patients with Brain Metastasis Is Associated with Improved Survival in the Era of Immunotherapy and Anti-BRAF Therapy. Cancers 2021, 13, 4493. https://doi.org/10.3390/cancers13174493

AMA Style

Dalmasso C, Pagès C, Chaltiel L, Sibaud V, Moyal E, Chira C, Sol JC, Latorzeff I, Meyer N, Modesto A. Intracranial Treatment in Melanoma Patients with Brain Metastasis Is Associated with Improved Survival in the Era of Immunotherapy and Anti-BRAF Therapy. Cancers. 2021; 13(17):4493. https://doi.org/10.3390/cancers13174493

Chicago/Turabian Style

Dalmasso, Céline, Cécile Pagès, Léonor Chaltiel, Vincent Sibaud, Elisabeth Moyal, Ciprian Chira, Jean Christophe Sol, Igor Latorzeff, Nicolas Meyer, and Anouchka Modesto. 2021. "Intracranial Treatment in Melanoma Patients with Brain Metastasis Is Associated with Improved Survival in the Era of Immunotherapy and Anti-BRAF Therapy" Cancers 13, no. 17: 4493. https://doi.org/10.3390/cancers13174493

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