Next Article in Journal
Clinical Outcomes of Online Adaptive Magnetic Resonance-Guided Stereotactic Body Radiotherapy of Adrenal Metastases from a Single Institution
Previous Article in Journal
Distance of Biopsy-Confirmed High-Risk Breast Lesion from Concurrently Identified Breast Malignancy Associated with Risk of Carcinoma at the High-Risk Lesion Site
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Predictive and Prognostic Factors in Melanoma Central Nervous System Metastases—A Cohort Study

1
Dermatology Department, Hospital Clinic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
2
Melanoma and Skin Cancer Unit, Dermatology Department, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320165, Chile
3
Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
4
Radiology Service, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
5
Institut d’Investigacions Biomediques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
6
CIBER on Rare Disease, Instituto de Salud Carlos III, 28029 Barcelona, Spain
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(12), 2272; https://doi.org/10.3390/cancers16122272
Submission received: 17 May 2024 / Revised: 8 June 2024 / Accepted: 13 June 2024 / Published: 19 June 2024
(This article belongs to the Section Cancer Metastasis)

Abstract

:

Simple Summary

We conducted a study at the Melanoma Unit of the Hospital Clinic of Barcelona to investigate brain metastases in patients with cutaneous melanoma. We collected data from patients diagnosed between January 1998 and September 2023. Patients with melanoma in situ or those with prior lung or breast cancer were excluded. Our aim was to identify factors associated with the development and survival outcomes of brain metastases. We analyzed patient demographics, tumor characteristics, and survival data. The diagnosis of brain metastases was confirmed using imaging techniques, and biopsies were performed when feasible. Our study followed strict guidelines for reporting observational studies. We found that younger age and larger primary tumor thickness increased the risk of developing brain metastases. Additionally, the presence of ulceration and microscopic satellitosis in the primary tumor were associated with a higher risk. Melanomas located on the trunk had a higher risk compared to those on the extremities. Patients with brain metastases had a median survival of around six months. Neurological symptoms and leptomeningeal involvement were associated with poorer survival outcomes. Higher number of brain lesions and elevated levels of lactate dehydrogenase (LDH) also predicted worse survival. Our findings highlight the importance of early detection and monitoring of melanoma patients, especially those at higher risk of brain metastases. Understanding these factors can aid in personalized treatment approaches and improving patient outcomes.

Abstract

Background: Melanoma is the cancer with the highest risk of dissemination to the central nervous system (CNS), one of the leading causes of mortality from this cancer. Objective: To identify patients at higher risk of developing CNS metastases and to evaluate associated prognostic factors. Methods: A cohort study (1998–2023) assessed patients who developed CNS melanoma metastases. Multivariate logistic regression was used to identify predictive factors at melanoma diagnosis for CNS metastasis. Cox regression analysis evaluated the CNS-independent metastasis-related variables impacting survival. Results: Out of 4718 patients, 380 (8.05%) developed CNS metastases. Multivariate logistic regression showed that a higher Breslow index, mitotic rate ≥ 1 mm2, ulceration, and microscopic satellitosis were significant risk factors for CNS metastasis development. Higher patient age and the location of the primary tumor in the upper or lower extremities were protective factors. In survival analysis, post-CNS metastasis, symptomatic disease, prior non-CNS metastases, CNS debut with multiple metastases, elevated LDH levels, and leptomeningeal involvement correlated with poorer survival. Conclusion: Predictive factors in the primary tumor independently associated with brain metastases include microscopic satellitosis, ulceration, higher Breslow index, and trunk location. Prognostic factors for lower survival in CNS disease include symptomatic disease, multiple CNS metastases, and previous metastases from different sites.

1. Introduction

Cutaneous melanoma represents 5% of all skin cancers, yet it causes 90% of skin cancer mortality [1]. Furthermore, melanoma is the third most common cancer that leads to brain metastases, after lung and breast cancer [2,3,4]. Further, the percentage of patients with metastatic melanoma who will develop brain metastases at some point in their disease has been calculated as between 40% and 60%, and central nervous system (CNS) metastases have been detected in up to 80% of autopsies of patients with metastatic melanoma [4,5,6,7].
Previous studies aimed to identify risk factors for CNS metastasis at the time of diagnosis. Notably, male sex, age (40–60), Breslow > 4 mm, ulceration, head and neck location, nodular subtype, and higher T stage were identified as key factors for the risk of developing brain metastases [8].
Historically, brain metastases have been associated with a poor prognosis, with median overall survival estimated to be approximately 4–6 months [7,8,9,10,11,12,13,14]. More recent studies have reported an improvement in median overall survival from 14 months to 23 months for metastatic melanoma since the introduction of targeted therapies and immune checkpoint inhibitors [15,16,17]. The CheckMate 204 trial reported that the 3-year overall survival rate was 71.9% for patients with asymptomatic brain metastases who received ipilimumab plus nivolumab [18].
The aim of our study was to describe risk factors associated with the development of brain metastases and to identify prognostic factors associated with survival once the patient develops brain metastasis.

2. Materials and Methods

A single-center cohort study was performed in the Melanoma Unit of the Hospital Clinic of Barcelona between January 1998 and September 2023. The data were collected from the electronic medical records of patients with a diagnosis of cutaneous melanoma and brain metastases developed at the time of diagnosis or during follow-up.
Patients with in situ melanoma and patients with brain metastasis who presented with a prior diagnosis of lung or breast cancer were excluded (Flowchart 1). To standardize AJCC in different time periods, a restaging of all primary melanomas was performed according to the AJCC 8th edition.
Since not all brain metastases could be confirmed histologically, diagnosis was performed by compatible magnetic resonance imaging or computed tomography, and in those cases where it was possible to perform a biopsy, the histopathologic report was used. Follow-up protocols were carried out at our center by a multidisciplinary tumor board of specialists in skin cancer and melanoma. The STROBE guidelines for the conducting and reporting of this observational study were followed.

2.1. Independent Variables

Collected data encompassed patient demographics and histological characteristics. CNS metastases characteristics were analyzed, covering number, size, bleeding, affected organs, symptoms, location, leptomeningeal involvement, and previous metastatic disease. Metastasis was categorized as deriving from 1, 2–5, or >5 lesions, and organ involvement as one, two-three, or >three. The largest metastasis size was reported as <10 mm, 10–25 mm, or >25 mm. Location was classified as supratentorial, infratentorial, multiple locations, or leptomeninges only. Additionally, leptomeningeal involvement alone was noted. The presence of metastases (debut or previous) and serum lactate dehydrogenase (LDH) concentration (≤ULN or >ULN) were recorded.

2.2. Dependent Variables

For the first predictive analysis, the presence of CNS metastases was coded as a dichotomous categorical variable.
In the post-metastasis survival analysis, the first dependent variable was melanoma-specific survival, calculated from the time of CNS melanoma metastases to the time of death from melanoma or the last follow-up visit. The second was overall survival, which was calculated from the time of CNS melanoma metastases to the time of death from any cause or the last follow-up visit.

2.3. Statistical Analysis

The distributions of categorical variables were reported as frequencies and percentages, and the distributions of continuous variables were reported as median and interquartile range (IQR). Pearson’s Chi-squared test was applied for categorical variables, and the trend test was applied for ordinal variables. The Wilcoxon rank sum test was used to compare continuous independent variables. The median follow-up of the cohort was calculated using the reverse Kaplan–Meier estimator applying the “Hist” functions in the ‘‘prodlim’’ package (v 2023.3.31) in R.
We utilized Sankey plots to represent the migratory patterns of AJCC stages in melanoma patients, leading to the development of brain metastasis. We also calculated the proportion of patients within each AJCC group who progressed. To describe the percentage of patients who will develop CNS metastases over time, we utilized the cumulative plot of survival.
To evaluate the association of covariables with brain metastasis development, we utilized multivariable logistic regression analysis.
After the development of brain metastasis, our objective was to assess the factors associated with poorer survival. Therefore, we performed univariate and multivariate analyses using Cox proportional hazard models for melanoma-specific survival. Models were fitted using the ‘‘coxph’’ function in the ‘‘survival’’ package (v 3.5.5) in R. Hazard ratio (HR) estimates were calculated to assess the effect of brain metastasis on melanoma-specific survival (MSS) adjusted for age at diagnosis of the brain metastasis, sex, primary tumor T score (AJCC 8th), presence of symptoms, levels of serum LDH, brain hemorrhage, number of brain metastasis, major diameter of metastasis, metastasis location, leptomeningeal involvement, and previous metastasis in other organs. We accounted for the changes in the therapeutic landscape by including a variable for treatment era in our survival analyses. Moreover, patients were grouped by treatment period: pre-2011 and post-2011, marking the introduction of targeted therapies and immune checkpoint inhibitors. This classification was included in the Cox proportional hazards model to adjust for the impact of different treatment regimens on survival outcomes.
Missing data were evaluated, classified as missing at random, and excluded from the analysis. To prevent immortal time bias in the setting of multiple primary melanomas, we used a worst-case analysis. To account for multiple comparisons, we applied the False Discovery Rate (FDR) method. This adjustment ensures that the likelihood of type I errors is minimized when performing multiple statistical tests.
All statistical analyses were performed using the computing environment R version 4.3.1 (16 June 2023) and RStudio (v 2023.6.1.524). A p-value of <0.05 was considered statistically significant.

3. Results

A total of 7010 patients with a histopathological diagnosis of melanoma were assessed for eligibility. After applying the inclusion and exclusion criteria, a total of 4718 patients were analyzed, of which 380 (8.05%) developed CNS melanoma metastases (Figure 1). The median follow-up time of the cohort using the inverse Kaplan–Meier estimator was 8.2 years (IQR 3.4–14.1). Clinicopathological characteristics of our study population at the time of initial diagnosis are summarized in Table 1.
The Sankey plot in Figure 2 illustrates the migration of patients who developed CNS metastases according to their initial AJCC 8th edition stage. The plot shows the percentage of patients from each stage who developed CNS metastases and those who did not. The highest proportion of CNS metastases originates from patients initially staged as IIID (53.8%) and IV (33.7%). Stages IIA and IIIA show similar percentages of CNS metastases, with approximately 7.7% and 7.3%, respectively. Stage IIC has a similar proportion to stage IIIB, with 15.1% and 18.5%, respectively. This visualization highlights the substantial risk of CNS metastases in later-stage melanoma patients. Additionally, it underscores that early-stage melanomas also contribute significantly in absolute numbers to the development of brain metastases.
In Figure 3, the cumulative plot of survival shows the percentage of patients who will develop CNS metastases over the years. At 5 years, the incidence rate in stages IIC was 17.7% (95% CI 10.5–24.4%), which was similar to stages IIIB with 20.6% (95% CI 13.6–27%) and IIIC with 22.7% (95% CI 17.9–27.3%). Furthermore, stage IIIA had an even lower incidence rate than stages IIA and IIB, at only 4.6% (95% CI 0.9–8.2%).
Multivariable logistic regression analysis to evaluate the association of covariables with brain metastasis development showed that CNS metastases decrease significantly in older patients. We noticed a reduced risk of metastasis development in the age group of 47.5–66 years and even lower in patients over 66 years. The odds ratios (OR) for these groups were 0.78 (95%CI 0.58–1.04; p < 0.001) and 0.47 (95%CI 0.35–0.64; p < 0.001), respectively, when compared to patients under 47.5 years of age. Moreover, a higher Breslow index correlated with an exponentially increased risk of developing CNS metastases with an OR of 2.7 (95%CI 1.9–4.1, p < 0.001) for T2, 3.7 (95%CI 2.4–5.6, p < 0.001) for T3, and 6.2 (95%CI 4.1–9.5, p < 0.001) when compared with T1. More importantly, the presence of ulceration and microscopic satellitosis was associated with a statistically significant elevation in this risk, with an OR of 2.4 (95%CI 1.8–3.2, p < 0.001) and 1.9 (95%CI 1.2–3.0, p < 0.007). Furthermore, when considering tumor location, primary tumors located on both the upper and lower extremities exhibited a decreased risk of CNS metastasis in comparison to those located on the trunk (Table 2).
Basal characteristics of the cohort at the time of CNS metastasis are summarized in Table 3. The median MSS of CNS metastatic patients was 5.73 months (95% Confidence Interval (CI): 5.06–6.53). For individuals experiencing CNS metastases at the onset of metastatic disease, the median time of survival was 7.46 months, while for patients whose metastases had spread to other organs before CNS, the median survival time was 5.39 months. Survival analysis using multivariate Cox regression models after the diagnosis of CNS metastasis showed that the presence of neurological symptoms is linked to higher specific mortality with an HR of 2.34 (95%CI 1.67–3.29; p < 0.001), as well as leptomeningeal involvement with an HR of 1.97 (95% IC 0.99–3.93; p < 0.054), however, it was not statistically significative. In addition, the presence of 2–5 lesions is associated with a worse MSS prognosis than a single metastatic lesion, with an HR of 1.53 (95% IC 1.05–2.22; p < 0.025) and more than 5 lesions had an HR of 2.45 (95% IC 1.65–3.65; p < 0.001), respectively, when compared with single lesions. Furthermore, another factor that influences survival negatively is an elevated level of LDH with an HR of 1.85 (95% IC 1.32–2.58; p < 0.001), while an indicator of better prognosis, with an HR of 0.66 (95% IC 0.44–0.99, p < 0.045), is the location of the metastasis as other than supratentorial (Table 4).

4. Discussion

Our study included 4718 melanoma patients, of whom 380 (8.05%) developed central nervous system metastases. We observed variations in CNS metastasis incidence rates across AJCC stages, with stage IIIC-D posing the highest risk. Moreover, when evaluating predictive risk factors for the development of brain metastasis, we observed that the risk of CNS metastases decreased with age, while an elevated Breslow index, ulceration, and microscopic satellitosis increased this risk, while tumors located on extremities rather than the trunk were associated with a lower risk. These findings provide new insights into the metastatic patterns of melanoma, which can be crucial for early detection and intervention.
Analyzing the baseline characteristics of the cohort, we observed that patients who developed central nervous system (CNS) metastases initially exhibited unfavorable tumor features: specifically, a median Breslow index exceeding a median of 3 mm, with over half presenting ulceration, aligning with findings from prior studies [2,19,20,21,22,23,24,25]. Notably, microsatellites were also seen in our study to be more prevalent in patients who subsequently developed CNS metastasis, occurring in 8.7% of cases vs. 2.3% in the control group. In a recent study by our research group, conducted by Riquelme-McLoughlin et al., we also identified microsatellites as an independent risk factor linked to poorer survival outcomes [26].
Examining metastasis occurrence across all AJCC substages, we find that stages IIIB through IV constitute about 50% of the CNS metastatic cohort. However, early stages, particularly stage IA, represent a significant portion of CNS progression cases (10.5%), attributed to their higher absolute numbers. Despite lower proportions, the substantial volume of individuals in early stages contributes significantly to metastatic cases. Moreover, risk analysis by AJCC groups aligns with expectations, revealing higher-risk stages with earlier metastasis development. Notably, AJCC Stage IIID stands out as the group at highest risk, with 53.8% developing CNS disease. Interestingly, many AJCC stages overlap; for example, stage IIIA behaves similarly to stage IIA, comprising 7.3% and 7.7% of total patients, respectively, showing that the AJCC is not a linear risk classification. Our results are in line with the study by Johannet et al., where patients with stage III melanoma were at a higher risk of developing brain metastases compared to those with stage II melanoma (21.4% vs. 14.0%, p = 0.002). However, individuals with stage IIC melanoma had a significantly higher rate of isolated first recurrences in the CNS compared to those with stage III disease (12.1% vs. 3.6%, p = 0.002). The risk of ever developing brain metastases was elevated for patients with stage IIC, IIIB, and IIIC disease, with the highest risk observed in patients with stage IIID disease (HR, 8.59; 95%CI: 4.11–17.97) [17].
Early identification of patients at high risk of CNS metastases is challenging. Notably, primary tumor factors such as microscopic satellitosis, ulceration, higher Breslow index, and trunk location serve as predictive indicators. Independent risk analysis shows younger age of onset correlates with higher risk, while those aged 66 or older are at a reduced risk. Breslow index, especially in tumors larger than 4 mm, amplifies risk six-fold, underscoring its prognostic significance. Davies et al. observed that the majority of patients who developed brain metastases had a primary tumor with an advanced Breslow index, with only 18% having an index < 1 mm [7]. Moreover, Zhang et al. and Gardner et al. identified male gender, ages between 40 and 60, Breslow thickness > 4 mm, ulceration, head and neck primary site, nodular subtype, and higher T stage as key risk factors associated with the development of brain metastases [8,27]. Interestingly, tumors situated in the upper and lower extremities exhibited a lower rate of progression to CNS compared to primary trunk tumors, suggesting a potential protective factor. This characteristic aligns with the findings described by Gardner et al. [27]. While some studies link the occurrence of brain metastases to male gender, in our study, male sex was not identified as an independent factor increasing the risk of CNS metastases [14,25,28,29]. Furthermore, the presence of microscopic satellitosis, a factor not previously reported, has emerged as an independent risk factor for the development of CNS disease, with an odds ratio of 1.90 (95%CI: 1.20 to 2.95). Our study becomes more significant after recent research by Riquelme-Mc Loughlin et al., which highlights microsatellites as an independent risk factor for reduced overall survival in melanoma [26].
In post-CNS metastasis survival analysis, factors contributing to a poorer prognosis include neurological symptoms and a higher lesion count, particularly when exceeding five, which is associated with diminished MSS. Elevated LDH levels were also linked to worse outcomes. However, the presence of hemorrhage and metastasis diameter did not significantly impact melanoma-specific survival. These findings align with a previous study by Glitza et al., where active disease outside the CNS, over three brain metastases, poor patient functional status, and leptomeningeal involvement were associated with poor survival [7]. Conversely, a more favorable prognosis was observed for metastases located other than supratentorial. Furthermore, in line with our findings, Vecchio et al. noted that asymptomatic patients exhibit significantly better survival [30]. On the other hand, other variables associated with the primary tumor were not associated with poor survival after central nervous system metastasis.
We believe that regular follow-up is essential for detecting metastasis in the early stages of relapse, particularly to identify asymptomatic patients, as our study identified a better outcome in individuals who exhibited no symptoms and presented with smaller tumor volumes. Looking ahead, it will be crucial to identify patients at risk of CNS relapse based on their basal characteristics and potentially through tumoral genetic tests such as gene expression profiling [31]. This could enable the selection of high-risk patients for a more intensive and targeted follow-up approach. The data from our study can be used to develop risk models for predicting CNS metastases, informing personalized follow-up strategies, and improving patient outcomes.
Our study’s retrospective nature introduces inherent heterogeneities in patient follow-up and treatment. Additionally, we were not able to analyze the molecular status of primary tumors due to the unavailability of comprehensive molecular panels, such as those for BRAF mutations, during the earlier years of our study period. Evaluating survival is challenging due to the diverse therapies applied to metastatic melanoma over the past decade. To address this, we grouped patients into two survival periods: pre-2011 and post-2011, marking the introduction of targeted therapies and immune checkpoint inhibitors. This classification helps to adjust for the impact of different treatment regimens on survival outcomes. Despite being a single-center study, our strength lies in analyzing a substantial cohort of 4718 patients, identifying 380 with CNS metastases.

5. Conclusions

In conclusion, our study identifies key risk factors for predicting CNS metastases in melanoma patients, with microsatellitosis being particularly significant. We also found that demographic and clinical factors such as advanced age, primary tumor thickness, and ulceration are associated with increased risk. Our analysis of prognostic factors influencing survival after CNS metastasis highlights the impact of neurological symptoms, the number of brain lesions, and elevated LDH levels on patient outcomes. These findings have important clinical implications for guiding neuroimaging follow-up strategies and tailoring treatment plans. By understanding both predictive and prognostic factors, healthcare providers can make more informed decisions in the management of melanoma, ultimately improving patient outcomes.

Author Contributions

E.S.: Conceptualization, Methodology, Data curation, Writing—Original draft preparation; Á.A.-A.: Conceptualization, Methodology, Data curation, Writing—Original draft preparation; R.M.-H. and A.A.: Writing—Review and Editing; F.A.: Writing—Review and Editing; M.L.O.: Writing—Review and Editing; D.R.-P.: Writing—Review and Editing; J.M.: Conceptualization, Methodology, Writing—Review and Editing; S.P. (Susana Puig): Conceptualization, Methodology, Writing—Review and Editing; C.C.: Conceptualization, Methodology, Writing—Original draft preparation, Writing—Review and Editing; S.P. (Sebastian Podlipnik): Conceptualization, Methodology, Writing—Original draft preparation, Supervision, Formal analysis, Writing—Reviewing and Editing, Visualization. All authors have read and agreed to the published version of the manuscript.

Funding

The research at the Melanoma group in Hospital Clinic Barcelona is supported by the CIBER de Enfermedades Raras of the Instituto de Salud Carlos III, Spain; AGAUR 2017_SGR_1134 and CERCA Programme by Generalitat de Catalunya, Spain; a Research Grant from “Fundación Científica de la Asociación Española Contra el Cáncer” GCB15152978SOEN, Spain; European Commission under the sixth Framework Programme, Contract No. LSHC-CT-2006-018702 (GenoMEL), by the European Commission under the Seventh Framework Programme, Diagnoptics; the European Commission under the HORIZON2020 Framework Programme, iTobos and Qualitop; and European Commission under the Horizon Europe Programme, HORIZON-MISS-2021-CANCER-02, MELCAYA (reference 101096667). This research was in part supported by grants from Fondo de Investigaciones Sanitarias P.I. 18/00419 and 22/01467 Spain. Part of the work was carried out at the Esther Koplowitz Center, Barcelona.

Institutional Review Board Statement

This study has been approved by the institutional review board. All procedures performed in studies involving human participants are in accordance with the ethical standards of the institutional research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Reviewed and approved by the Research Ethics Committee of Hospital Clinic Barcelona IRB; approval HCB/2015/0298.

Informed Consent Statement

The patient has provided voluntary consent to participate in this study. Consent for the publication of recognizable patient photographs or other identifiable material was obtained by the authors and included at the time of article submission to the journal, stating that all patients gave consent with the understanding that this information may be publicly available.

Data Availability Statement

The data presented in this study are available in this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Garbe, C.; Amaral, T.; Peris, K.; Hauschild, A.; Arenberger, P.; Basset-Seguin, N.; Bastholt, L.; Bataille, V.; del Marmol, V.; Dréno, B.; et al. European Consensus-Based Interdisciplinary Guideline for Melanoma. Part 2: Treatment—Update 2022. Eur. J. Cancer 2022, 170, 256–284. [Google Scholar] [CrossRef] [PubMed]
  2. Gumusay, O.; Coskun, U.; Akman, T.; Ekinci, A.S.; Kocar, M.; Erceleb, Ö.B.; Yazici, O.; Kaplan, M.A.; Berk, V.; Cetin, B.; et al. Predictive Factors for the Development of Brain Metastases in Patients with Malignant Melanoma: A Study by the Anatolian Society of Medical Oncology. J. Cancer Res. Clin. Oncol. 2014, 140, 151–157. [Google Scholar] [CrossRef]
  3. Taillibert, S. Le Rhun Epidemiology of Brain Metastases. Cancer/Radiother. 2015, 19, 3–9. [Google Scholar] [CrossRef] [PubMed]
  4. Cohen, J.V.; Tawbi, H.; Margolin, K.A.; Amravadi, R.; Bosenberg, M.; Brastianos, P.K.; Chiang, V.L.; de Groot, J.; Glitza, I.C.; Herlyn, M.; et al. Melanoma Central Nervous System Metastases: Current Approaches, Challenges, and Opportunities. Pigment Cell Melanoma Res. 2016, 29, 627–642. [Google Scholar] [CrossRef] [PubMed]
  5. Glitza, I.C.; Heimberger, A.B.; Sulman, E.P.; Davies, M.A. Prognostic Factors for Survival in Melanoma Patients with Brain Metastases. In Brain Metastases from Primary Tumors: Epidemiology, Biology, and Therapy of Melanoma and Other Cancers; Elsevier Inc.: Amsterdam, The Netherlands, 2016; Volume 3, pp. 267–297. ISBN 9780128035597. [Google Scholar]
  6. Diaz, M.J.; Mark, I.; Rodriguez, D.; Gelman, B.; Tran, J.T.; Kleinberg, G.; Levin, A.; Beneke, A.; Root, K.T.; Tran, A.X.V.; et al. Melanoma Brain Metastases: A Systematic Review of Opportunities for Earlier Detection, Diagnosis, and Treatment. Life 2023, 13, 828. [Google Scholar] [CrossRef] [PubMed]
  7. Davies, M.A.; Liu, P.; McIntyre, S.; Kim, K.B.; Papadopoulos, N.; Hwu, W.J.; Hwu, P.; Bedikian, A. Prognostic Factors for Survival in Melanoma Patients with Brain Metastases. Cancer 2011, 117, 1687–1696. [Google Scholar] [CrossRef]
  8. Zhang, D.; Wang, Z.; Shang, D.; Yu, J.; Yuan, S. Incidence and Prognosis of Brain Metastases in Cutaneous Melanoma Patients: A Population-Based Study. Melanoma Res. 2019, 29, 77–84. [Google Scholar] [CrossRef]
  9. Nishino, M.; Dahlberg, S.E.; Fulton, L.E.; Digumarthy, S.R.; Hatabu, H.; Johnson, B.E.; Sequist, L.V. Volumetric Tumor Response and Progression in EGFR-Mutant NSCLC Patients Treated with Erlotinib or Gefitinib. Acad. Radiol. 2016, 23, 329–336. [Google Scholar] [CrossRef] [PubMed]
  10. Fife, K.M.; Colman, M.H.; Stevens, G.N.; Firth, I.C.; Moon, D.; Shannon, K.F.; Harman, R.; Petersen-Schaefer, K.; Zacest, A.C.; Besser, M.; et al. Determinants of Outcome in Melanoma Patients with Cerebral Metastases. J. Clin. Oncol. 2004, 22, 1293–1300. [Google Scholar] [CrossRef]
  11. Staudt, M.; Lasithiotakis, K.; Leiter, U.; Meier, F.; Eigentler, T.; Bamberg, M.; Tatagiba, M.; Brossart, P.; Garbe, C. Determinants of Survival in Patients with Brain Metastases from Cutaneous Melanoma. Br. J. Cancer 2010, 102, 1213–1218. [Google Scholar] [CrossRef]
  12. Bordia, R.; Zhong, H.; Lee, J.; Weiss, S.; Han, S.W.; Osman, I.; Jain, R. Melanoma Brain Metastases: Correlation of Imaging Features with Genomic Markers and Patient Survival. J. Neurooncol. 2017, 131, 341–348. [Google Scholar] [CrossRef] [PubMed]
  13. 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]
  14. Ostheimer, C.; Bormann, C.; Fiedler, E.; Marsch, W.; Vordermark, D. Malignant Melanoma Brain Metastases: Treatment Results and Prognostic Factors—A Single-Center Retrospective Study. Int. J. Oncol. 2015, 46, 2439–2448. [Google Scholar] [CrossRef] [PubMed]
  15. Iorgulescu, J.B.; Harary, M.; Zogg, C.K.; Ligon, K.L.; Reardon, D.A.; Hodi, F.S.; Aizer, A.A.; Smith, T.R. Improved Risk-Adjusted Survival for Melanoma Brain Metastases in the Era of Checkpoint Blockade Immunotherapies: Results from a National Cohort. Cancer Immunol. Res. 2018, 6, 1039–1045. [Google Scholar] [CrossRef] [PubMed]
  16. Kluger, H.M.; Chiang, V.; Mahajan, A.; Zito, C.R.; Sznol, M.; Tran, T.; Weiss, S.A.; Cohen, J.V.; Yu, J.; Hegde, U.; et al. Long-Term Survival of Patients with Melanoma with Active Brain Metastases Treated with Pembrolizumab on a Phase II Trial. J. Clin. Oncol. 2018, 37, 52–60. [Google Scholar] [PubMed]
  17. Johannet, P.; Simons, M.; Qian, Y.; Azmy, N.; Mehnert, J.M.; Weber, J.S.; Zhong, J.; Osman, I. Risk and Tropism of Central Nervous System (CNS) Metastases in Patients with Stage II and III Cutaneous Melanoma. Cancer 2022, 128, 3620–3629. [Google Scholar] [CrossRef] [PubMed]
  18. Tawbi, H.A.; Forsyth, P.A.; Hodi, F.S.; Algazi, A.P.; Hamid, O.; Lao, C.D.; Moschos, S.J.; Atkins, M.B.; Lewis, K.; Postow, M.A.; et al. Long-Term Outcomes of Patients with Active Melanoma Brain Metastases Treated with Combination Nivolumab plus Ipilimumab (CheckMate 204): Final Results of an Open-Label, Multicentre, Phase 2 Study. Lancet Oncol. 2021, 22, 1692–1704. [Google Scholar] [CrossRef] [PubMed]
  19. Raizer, J.J.; Hwu, W.J.; Panageas, K.S.; Wilton, A.; Baldwin, D.E.; Bailey, E.; Von Althann, C.; Lamb, L.A.; Alvarado, G.; Bilsky, M.H.; et al. Brain and Leptomeningeal Metastases from Cutaneous Melanoma: Survival Outcomes Based on Clinical Features. Neuro Oncol. 2008, 10, 199–207. [Google Scholar] [CrossRef] [PubMed]
  20. Qian, M.; Ma, M.W.; Fleming, N.H.; Lackaye, D.J.; Hernando, E.; Osman, I.; Shao, Y. Clinicopathological Characteristics at Primary Melanoma Diagnosis as Risk Factors for Brain Metastasis. Melanoma Res. 2013, 23, 461–467. [Google Scholar] [CrossRef]
  21. Zakrzewski, J.; Geraghty, L.N.; Rose, A.E.; Christos, P.J.; Mazumdar, M.; Polsky, D.; Shapiro, R.; Berman, R.; Darvishian, F.; Hernando, E.; et al. Clinical Variables and Primary Tumor Characteristics Predictive of the Development of Melanoma Brain Metastases and Post-Brain Metastases Survival. Cancer 2011, 117, 1711–1720. [Google Scholar] [CrossRef]
  22. Gorka, E.; Fabó, D.; Gézsi, A.; Czirbesz, K.; Liszkay, G. Distance from Primary Tumor Is the Strongest Predictor for Early Onset of Brain Metastases in Melanoma. Anticancer Res. 2016, 36, 3065–3069. [Google Scholar] [PubMed]
  23. Frankel, T.L.; Bamboat, Z.M.; Ariyan, C.; Coit, D.; Sabel, M.S.; Brady, M.S. Predicting the Development of Brain Metastases in Patients with Local/Regional Melanoma. J. Surg. Oncol. 2014, 109, 770–774. [Google Scholar] [CrossRef] [PubMed]
  24. Bedikian, A.Y.; Wei, C.; Detry, M.; Kim, K.B.; Papadopoulos, N.E.; Hwu, W.-J.; Homsi, J.; Davies, M.; McIntyre, S.; Hwu, P. Predictive Factors for the Development of Brain Metastasis in Advanced Unresectable Metastatic Melanoma. Am. J. Clin. Oncol. 2011, 34, 603–610. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed]
  26. Riquelme-Mc Loughlin, C.; Sandoval-Clavijo, A.; Blanco de Tord, M.; Boada, A.; Alos, L.; García, A.; Carrera, C.; Malvehy, J.; Puig, S.; Toll, A.; et al. Prognostic Role of Microsatellites in Melanoma and Implications in the American Joint Committee on Cancer Classification System: A Cohort Study. J. Am. Acad. Dermatol. 2023, 88, 338–347. [Google Scholar] [CrossRef] [PubMed]
  27. Gardner, L.J.; Ward, M.; Andtbacka, R.H.I.; Boucher, K.M.; Bowen, G.M.; Bowles, T.L.; Cohen, A.L.; Grossmann, K.; Hitchcock, Y.J.; Holmen, S.L.; et al. Risk Factors for Development of Melanoma Brain Metastasis and Disease Progression: A Single-Center Retrospective Analysis. Melanoma Res. 2017, 27, 477–484. [Google Scholar] [CrossRef] [PubMed]
  28. Gugger, A.; Barnhill, R.L.; Seifert, B.; Dehler, S.; Moch, H.; Lugassy, C.; Marques-Maggio, E.; Rushing, E.J.; Mihic-Probst, D. Cutaneous Melanoma with Brain Metastasis: Report of 193 Patients with New Observations. PLoS ONE 2016, 11, e0156115. [Google Scholar] [CrossRef] [PubMed]
  29. Jackson, J.E.; Burmeister, B.H.; Burmeister, E.A.; Foote, M.C.; Thomas, J.M.; Meakin, J.A.; Smithers, B.M. Melanoma Brain Metastases: The Impact of Nodal Disease. Clin. Exp. Metastasis 2014, 31, 81–85. [Google Scholar] [CrossRef] [PubMed]
  30. Vecchio, S.; Spagnolo, F.; Merlo, D.F.; Signori, A.; Acquati, M.; Pronzato, P.; Queirolo, P. The Treatment of Melanoma Brain Metastases before the Advent of Targeted Therapies: Associations between Therapeutic Choice, Clinical Symptoms and Outcome with Survival. Melanoma Res. 2014, 24, 61–67. [Google Scholar] [CrossRef]
  31. Podlipnik, S.; Potrony, M.; Puig, S. Genetic Markers for Characterization and Prediction of Prognosis of Melanoma Subtypes: A 2021 Update. Ital. J. Dermatol. Venereol. 2021, 156, 322–330. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the cohort.
Figure 1. Flowchart of the cohort.
Cancers 16 02272 g001
Figure 2. Sankey plot illustrating the migration of patients from initial AJCC 8th edition stages to CNS metastasis status. The width of each flow corresponds to the proportion of patients progressing to brain metastases or remaining metastasis-free. Higher stages, particularly IIID and IV, show a greater proportion of CNS metastases.
Figure 2. Sankey plot illustrating the migration of patients from initial AJCC 8th edition stages to CNS metastasis status. The width of each flow corresponds to the proportion of patients progressing to brain metastases or remaining metastasis-free. Higher stages, particularly IIID and IV, show a greater proportion of CNS metastases.
Cancers 16 02272 g002
Figure 3. Cumulative plot for the incidence of central nervous system metastases.
Figure 3. Cumulative plot for the incidence of central nervous system metastases.
Cancers 16 02272 g003
Table 1. Basal characteristics of the cohort.
Table 1. Basal characteristics of the cohort.
CharacteristicOverall, N = 4715Stratified Groupsp-Valueq-Value
No Brain Metastasis, N = 4335Brain Metastasis, N = 380
Gender, n (%) <0.001<0.001
       Female2410 (51.1%)2249 (51.9%)161 (42.4%)
       Male2305 (48.9%)2086 (48.1%)219 (57.6%)
Age, Median (IQR)56.8 (42.8–70.3)56.8 (42.9–70.6)56.6 (41.3–67.8)0.1140.110
       Missing values990
8th version AJCC, n (%) --
       IA1889 (41.8%)1852 (44.5%)37 (10.5%)
       IB717 (15.9%)690 (16.6%)27 (7.6%)
       IIA426 (9.4%)393 (9.4%)33 (9.3%)
       IIB328 (7.3%)294 (7.1%)34 (9.6%)
       IIC199 (4.4%)169 (4.1%)30 (8.5%)
       IIIA165 (3.7%)153 (3.7%)12 (3.4%)
       IIIB195 (4.3%)159 (3.8%)36 (10.2%)
       IIIC457 (10.1%)368 (8.8%)89 (25.2%)
       IIID39 (0.9%)18 (0.4%)21 (5.9%)
       IV101 (2.2%)67 (1.6%)34 (9.6%)
       Missing values19917227
Melanoma of unknown primary, n (%)130 (2.8%)91 (2.1%)39 (10.3%)<0.001<0.001
       Missing values990
Breslow index (mm), Median (IQR)1.2 (0.7–2.7)1.1 (0.6–2.5)3.2 (1.5–6.0)<0.001<0.001
       Missing values1238439
Ulceration, n (%) <0.001<0.001
       Absent3360 (75.7%)3220 (78.3%)140 (42.6%)
       Present1081 (24.3%)892 (21.7%)189 (57.4%)
       Missing values27422351
Mitotic index mm2, n (%) <0.001<0.001
       Zero Mitosis1738 (44.7%)1681 (46.7%)57 (20.1%)
       One or more mitosis2146 (55.3%)1919 (53.3%)227 (79.9%)
       Missing values83173596
Histological subtype, n (%) <0.001<0.001
       Superficial spreading2916 (65.5%)2773 (67.1%)143 (44.0%)
       Lentiginous malignant289 (6.5%)273 (6.6%)16 (4.9%)
       Nodular695 (15.6%)582 (14.1%)113 (34.8%)
       Other299 (6.7%)279 (6.8%)20 (6.2%)
       Acral lentiginous256 (5.7%)223 (5.4%)33 (10.2%)
       Missing values26020555
Satellitosis, n (%) <0.001<0.001
       Absent4581 (97.2%)4234 (97.7%)347 (91.3%)
       Present134 (2.8%)101 (2.3%)33 (8.7%)
Regression, n (%) <0.001<0.001
       <50%966 (31.5%)915 (32.0%)51 (24.6%)
       >50%369 (12.0%)361 (12.6%)8 (3.9%)
       None1734 (56.5%)1586 (55.4%)148 (71.5%)
       Missing values16461473173
Location, n (%) --
       Trunk2044 (44.9%)1867 (44.4%)177 (51.0%)
       Head and neck644 (14.2%)579 (13.8%)65 (18.7%)
       Lower limbs916 (20.1%)879 (20.9%)37 (10.7%)
       Upper limbs546 (12.0%)522 (12.4%)24 (6.9%)
       Acral333 (7.3%)297 (7.1%)36 (10.4%)
       Mucosa64 (1.4%)56 (1.3%)8 (2.3%)
       Other3 (0.1%)3 (0.1%)0 (0.0%)
       Missing values16513233
Legend Table 1: q-value: Adjusted p-value using the False Discovery Rate (FDR) method, which controls for the number of tests being performed to reduce false positives.
Table 2. Multiple Binomial Logistic Regression.
Table 2. Multiple Binomial Logistic Regression.
CharacteristicNOR (95% CI) 1p-Valueq-Value 2
Age4401 <0.001<0.001
      <47.5
      47.5–66 0.78 (0.58 to 1.04)
      >66 0.47 (0.35 to 0.64)
Gender4401 0.280.28
      female
      male 1.14 (0.89 to 1.47)
T score4401 <0.001<0.001
      pT1
      pT2 2.77 (1.87 to 4.13)
      pT3 3.67 (2.42 to 5.60)
      pT4 6.19 (4.07 to 9.53)
Ulceration4401 <0.001<0.001
      absent
      present 2.41 (1.83 to 3.19)
Location4401 <0.001<0.001
      trunk
      head and neck 1.00 (0.72 to 1.39)
      lower limbs 0.45 (0.30 to 0.65)
      upper limbs 0.52 (0.32 to 0.82)
      acral 0.84 (0.55 to 1.26)
      mucosa 0.62 (0.24 to 1.37)
Satellitosis4401 0.0070.009
      absent
      present 1.90 (1.20 to 2.95)
1 OR = Odds Ratio, CI = Confidence Interval. 2 False discovery rate correction for multiple testing.
Table 3. Basal characteristics of the cohort at the time of metastasis.
Table 3. Basal characteristics of the cohort at the time of metastasis.
CharacteristicN = 380
Gender, n (%)
       Female161 (42.4%)
       Male219 (57.6%)
Age at metastasis, Median (IQR)59.5 (46.5–70.6)
Brain metastasis location, n (%)
       Supratentorial249 (71.3%)
       Multiple locations81 (23.2%)
       Infratentorial14 (4.0%)
       Leptomeninges Exclusively5 (1.4%)
       Missing values31
Leptomeningeal involvement, n (%)
       No leptomeningeal involvement326 (93.4%)
       Leptomeningeal metastasis23 (6.6%)
       Missing values31
Brain hemorrage, n (%)
       Absent256 (72.3%)
       Present98 (27.7%)
       Missing values26
Previous metastasis, n (%)
       Debut82 (22.5%)
       Previous metastasis282 (77.5%)
       Missing values16
Number of other organs affected, n (%)
       None77 (21.4%)
       1103 (28.7%)
       2–3114 (31.8%)
       >365 (18.1%)
       Missing values21
Symptoms, n (%)
       Asymptomatic169 (47.2%)
       Symptomatic189 (52.8%)
       Missing values22
Diametre of largest brain metastasis, n (%)
       <10 mm103 (33.9%)
       10–25 mm113 (37.2%)
       >25 mm88 (28.9%)
       Missing values76
Lactate dehydrogenase level (LDH), n (%)
       ≤ULN186 (61.6%)
       >ULN116 (38.4%)
       Missing values78
Table 4. Univariate and multivariate Cox regression tables for melanoma-specific and overall survival.
Table 4. Univariate and multivariate Cox regression tables for melanoma-specific and overall survival.
Melanoma-Specific SurvivalOverall Survival
VariableValuesN (%)HR (Univariable)HR (Multivariable)HR (Univariable)HR (Multivariable)
Sexfemale
male161 (42.8)1.00 (0.80–1.24, p = 0.978)0.96 (0.71–1.30, p = 0.782)1.00 (0.81–1.25, p = 0.967)0.96 (0.71–1.29, p = 0.773)
Age<47.5215 (57.2)
47.5–66130 (34.6)1.27 (0.98–1.64, p = 0.069)1.08 (0.76–1.55, p = 0.658)1.28 (0.99–1.65, p = 0.061)1.08 (0.75–1.54, p = 0.684)
>66135 (35.9)1.31 (0.99–1.72, p = 0.055)1.18 (0.81–1.72, p = 0.391)1.30 (0.99–1.71, p = 0.058)1.14 (0.78–1.66, p = 0.487)
T scorepT1111 (29.5)
pT244 (13.1)0.80 (0.54–1.17, p = 0.249)1.01 (0.61–1.68, p = 0.967)0.79 (0.54–1.17, p = 0.246)1.00 (0.60–1.66, p = 0.993)
pT375 (22.3)0.81 (0.55–1.17, p = 0.262)0.91 (0.55–1.51, p = 0.717)0.84 (0.58–1.21, p = 0.346)0.94 (0.57–1.56, p = 0.818)
pT487 (25.8)0.82 (0.58–1.17, p = 0.276)0.93 (0.58–1.51, p = 0.778)0.84 (0.59–1.19, p = 0.317)0.93 (0.57–1.50, p = 0.751)
SymptomsAsymptomatic131 (38.9)
Symptomatic169 (47.5)2.33 (1.86–2.92, p < 0.001)2.34 (1.67–3.29, p < 0.001)2.31 (1.85–2.90, p < 0.001)2.31 (1.65–3.25, p < 0.001)
LDH levels≤ULN187 (52.5)
>ULN186 (61.6)1.94 (1.51–2.48, p < 0.001)1.85 (1.32–2.58, p < 0.001)1.92 (1.50–2.46, p < 0.001)1.82 (1.30–2.54, p < 0.001)
Brain hemorrhageabsent116 (38.4)
present256 (72.7)1.44 (1.13–1.83, p = 0.003)1.25 (0.86–1.82, p = 0.242)1.42 (1.11–1.80, p = 0.005)1.25 (0.86–1.81, p = 0.249)
Number of brain metastasis1 lesion96 (27.3)
2–5 lesions121 (35.3)2.07 (1.57–2.72, p < 0.001)1.53 (1.05–2.22, p = 0.025)2.09 (1.59–2.75, p < 0.001)1.55 (1.07–2.25, p = 0.020)
>5 lesions125 (36.4)2.38 (1.77–3.20, p < 0.001)2.45 (1.65–3.65, p < 0.001)2.43 (1.81–3.26, p < 0.001)2.44 (1.64–3.63, p < 0.001)
Diameter of largest brain metastasis<10 mm97 (28.3)
10–25 mm103 (34.0)1.52 (1.14–2.04, p = 0.005)0.99 (0.69–1.43, p = 0.953)1.50 (1.13–2.01, p = 0.005)0.97 (0.67–1.39, p = 0.861)
>25 mm112 (37.0)1.80 (1.32–2.45, p < 0.001)0.91 (0.59–1.42, p = 0.686)1.76 (1.30–2.39, p < 0.001)0.89 (0.57–1.39, p = 0.611)
LocationSupratentorial88 (29.0)
Other location248 (71.5)1.21 (0.94–1.55, p = 0.140)0.66 (0.44–0.99, p = 0.045)1.20 (0.94–1.54, p = 0.139)0.66 (0.44–0.98, p = 0.041)
Leptomeningeal involvementNo leptomeningeal involvement99 (28.5)
Leptomeningeal metastasis324 (93.4)1.94 (1.27–2.97, p = 0.002)1.97 (0.99–3.93, p = 0.054)1.92 (1.25–2.93, p = 0.003)1.90 (0.96–3.79, p = 0.067)
Previous metastasisDebut23 (6.6)
Previous metastasis79 (21.9)1.60 (1.22–2.10, p = 0.001)1.87 (1.26–2.78, p = 0.002)1.63 (1.24–2.14, p < 0.001)1.92 (1.30–2.86, p = 0.001)
Year of diagnosis<2011281 (78.1)
≥2011114 (30.3)0.74 (0.59–0.93, p = 0.010)0.71 (0.52–0.98, p = 0.040)0.73 (0.58–0.92, p = 0.008)0.70 (0.51–0.97, p = 0.031)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Serra, E.; Abarzua-Araya, Á.; Arance, A.; Martin-Huertas, R.; Aya, F.; Olondo, M.L.; Rizo-Potau, D.; Malvehy, J.; Puig, S.; Carrera, C.; et al. Predictive and Prognostic Factors in Melanoma Central Nervous System Metastases—A Cohort Study. Cancers 2024, 16, 2272. https://doi.org/10.3390/cancers16122272

AMA Style

Serra E, Abarzua-Araya Á, Arance A, Martin-Huertas R, Aya F, Olondo ML, Rizo-Potau D, Malvehy J, Puig S, Carrera C, et al. Predictive and Prognostic Factors in Melanoma Central Nervous System Metastases—A Cohort Study. Cancers. 2024; 16(12):2272. https://doi.org/10.3390/cancers16122272

Chicago/Turabian Style

Serra, Estefania, Álvaro Abarzua-Araya, Ana Arance, Roberto Martin-Huertas, Francisco Aya, María Lourdes Olondo, Daniel Rizo-Potau, Josep Malvehy, Susana Puig, Cristina Carrera, and et al. 2024. "Predictive and Prognostic Factors in Melanoma Central Nervous System Metastases—A Cohort Study" Cancers 16, no. 12: 2272. https://doi.org/10.3390/cancers16122272

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