Skip to main content
Erschienen in: Targeted Oncology 2/2024

Open Access 24.02.2024 | Original Research Article

DNA Methylation Signatures Correlate with Response to Immune Checkpoint Inhibitors in Metastatic Melanoma

verfasst von: Julia Maria Ressler, Erwin Tomasich, Teresa Hatziioannou, Helmut Ringl, Gerwin Heller, Rita Silmbrod, Lynn Gottmann, Angelika Martina Starzer, Nina Zila, Philipp Tschandl, Christoph Hoeller, Matthias Preusser, Anna Sophie Berghoff

Erschienen in: Targeted Oncology | Ausgabe 2/2024

Abstract

Background

DNA methylation profiles have emerged as potential predictors of therapeutic response in various solid tumors.

Objective

This study aimed to analyze the DNA methylation profiles of patients with stage IV metastatic melanoma undergoing first-line immune checkpoint inhibitor treatment and evaluate their correlation with a radiological response according to immune-related Response Evaluation Criteria in Solid Tumors (iRECIST).

Methods

A total of 81 tissue samples from 71 patients with metastatic melanoma (27 female, 44 male) were included in this study. We utilized Illumina Methylation EPIC Beadchips to retrieve their genome-wide methylation profile by interrogating >850,000 CpG sites. Clustering based on the 500 most differentially methylated genes was conducted to identify distinct methylation patterns associated with immune checkpoint inhibitor response. Results were further aligned with an independent, previously published data set.

Results

The median progression-free survival was 8.5 months (range: 0–104.1 months), and the median overall survival was 30.6 months (range: 0–104.1 months). Objective responses were observed in 29 patients (40.8%). DNA methylation profiling revealed specific signatures that correlated with radiological response to immune checkpoint inhibitors. Three distinct clusters were identified based on the methylation patterns of the 500 most differentially methylated genes. Cluster 1 (12/12) and cluster 2 (12/24) exhibited a higher proportion of responders, while cluster 3 (39/45) predominantly consisted of non-responders. In the validation data set, responders also showed more frequent hypomethylation although differences in the data sets limit the interpretation.

Conclusions

These findings suggest that DNA methylation profiling of tumor tissues might serve as a predictive biomarker for immune checkpoint inhibitor response in patients with metastatic melanoma. Further validation studies are warranted to confirm the efficiency of DNA methylation profiling as a predictive tool in the context of immunotherapy for metastatic melanoma.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s11523-024-01041-4.
Key Points
Immune checkpoint inhibitors have revolutionized the treatment of metastatic melanoma, exhibiting an objective response rate that ranges from 40% to 58%. Despite this success, there remains a significant gap in our understanding of response-associated biomarkers.
However, the discovery of distinct methylation patterns linked to treatment response holds tremendous promise in guiding personalized therapeutic approaches and enhancing clinical outcomes for patients in this particular population observing similar trends in a validation cohort.
Here, we identified a methylation signature with a high predictive performance, characterized by an 80% sensitivity, 81% specificity, and an area under the curve value of 0.829. These findings underscore its potential as a valuable additional tool in routine clinical practice and precision medicine.

1 Introduction

Immune checkpoint inhibitors (ICIs) have revolutionized melanoma treatment. According to the EADO/EORTC/EDF and ESMO guidelines, patients with inoperable metastatic melanoma should receive first-line ICI therapy regardless of the mutation status because of prolonged long-term disease control with ICIs over targeted therapies [11, 12]. However, half of ICI-treated patients with melanoma do not benefit from treatment, underscoring the urgent need for reliable biomarkers.
Tumor mutational burden [8, 10, 30] and tumor neoantigen burden [25] are widely discussed biomarkers. However, analysis in a clinical routine is challenging, as protocols differ among papers, cut-off values are objects of discussion, and costs are rather high. Tumor tissue methylation is a well-established diagnostic biomarker for brain tumors and sarcomas [2, 22, 24]. Methylation analysis, however, including the required laboratory set-up, can be easily established, and thereby represents a promising method for biomarker evaluation in everyday clinical use. Alterations in DNA methylation are a key feature of tumorigenesis that result in dysregulated transcriptional gene activity. However, epigenetic changes in immune cells have also been reported to contribute to response and resistance to ICI-based therapies [6]. Indeed, tumor tissue methylation was previously shown to serve as a potential biomarker for ICI response in patients with sarcoma, head and neck cancer, lung cancer, and metastatic melanoma [7, 29, 39, 40]. Here, we report that DNA methylation profiling of treatment-naïve tumor samples independent of the tumor origin from patients with metastatic melanoma could be an additional useful tool for predicting response to ICIs and emphasizes that tissue-based DNA methylation profiling should be incorporated in future biomarker research studies.

2 Methods

2.1 Patients’ and Clinical Characteristics

A total of 71 patients (27 [38.0%] female and 44 [62.0%] male) with a median age of 65 years (range: 32–84 years) were retrospectively identified from the patient records of the Department of Dermatology, Medical University of Vienna, from 2015 to 2022. The inclusion criteria were as follows: (i) age ≥18 years; (ii) stage IVM1a–IVM1d metastatic melanoma according to the American Joint Committee on Cancer, 8th Edition [20]; (iii) sufficient formalin-fixed paraffin-embedded tumor tissue material prior to ICI initiation available; (iv) first-line therapy with ICIs including monotherapy with a programmed cell death protein 1 (PD-1) inhibitor (nivolumab, pembrolizumab), monotherapy with a cytotoxic T-lymphocyte associated protein 4 inhibitor (ipilimumab), or combination therapy of a PD-1 inhibitor (nivolumab) and cytotoxic T-lymphocyte associated protein 4 inhibitor (ipilimumab) and an ICI-based clinical trial study medication; (v) administration of a minimum of one cycle of ICI; (vi) radiologically measurable disease (lesion size ≥10×10 mm) prior to ICI therapy start; and (vii) availability of at least one radiological restaging under ICI therapy or the occurrence of death before the first performed radiological restaging. Patient and clinical characteristics, including age, sex, tumor mutation status (BRAF, NRAS, cKIT, or wild type), Eastern Cooperative Oncology Group (ECOG) performance status, baseline S100 and lactate dehydrogenase, and survival data (overall survival [OS], progression-free survival [PFS]) were obtained from patient records. Radiological assessment of response was defined according to the immune-based Response Evaluation Criteria in Solid Tumors (iRECIST) [38] and was performed by an independent radiologist blinded to patient outcomes. Objective response was determined by immune complete response or immune partial response, and disease control by immune complete response, immune partial response, or immune stable disease as best achieved response. Disease progression was defined as iPD and further subclassified as immune unconfirmed progressive disease and immune confirmed progressive disease. The date of immune unconfirmed progressive disease was classified as the timepoint of progression if no subsequent restaging was followed by a clear clinical progression or the occurrence of death. The date of the first radiologically confirmed progressive disease was defined as the immune confirmed progressive disease [38]. Patients with immune complete response and immune partial response were classified as responders, and patients with immune stable disease and iPD (immune unconfirmed progressive disease or immune confirmed progressive disease) were classified as non-responders. Patients who died due to early progression and received only one cycle of ICIs before the first restaging were classified as non-responders.

2.2 Genome-Wide DNA Methylation Analysis

Tumor tissue samples from patients with melanoma before to the first cycle of ICI therapy were collected retrospectively. A total of 81 tumor tissue samples from 71 patients were included in the final methylation analysis. These were categorized into four groups: 28 lymph node metastases (34.6%), 20 organ metastases (24.7%), 24 cutaneous metastases (29.6%), and nine primary tumors (11.1%) (Table 1). Of note, in a subset of patients, 8/71 (11.3%) had more than one treatment-naïve tumor sample from different organ sites available. In detail, one patient had a lymph node and an organ metastatic lesion (12.5%), two patients had a lymph node metastatic lesion and a primary tumor tissue (25%), three patients had one lymph node and one cutaneous metastatic lesion (37.5%), one patient had a cutaneous metastatic lesion and a primary tumor tissue (12.5%), and one patient had a lymph node metastatic lesion, two cutaneous metastatic lesions, and a primary tumor tissue (12.5%) [Table S1 of the Electronic Supplementary Material (ESM)]. The 5×10-μm slides were obtained from formalin-fixed paraffin-embedded tumor tissue after macrodissection with ≥70% tumor cells, as evaluated by an independent certified dermatopathologist blinded to the patients’ clinical outcomes. Genomic DNA was extracted and treated with sodium bisulfite following standard procedures, and genome-wide DNA methylation analyses were performed using Infinium MethylationEPIC BeadChip microarrays (Illumina, San Diego, CA, USA) as previously described [39]. Raw Microarray Data (.idat files) were loaded into R (version R 4.0.4, R Foundation for Statistical Computing, Vienna, Austria) using the RnBeads package (.idat files). Single nucleotide polymorphism-associated, non-specific/cross-hybridizing, and sex chromosome-specific probes were excluded from further analysis. The SWAN algorithm was applied for data normalization [26]. Calculation of differential methylation between groups was conducted using RnBeads implementation of the limma package as well as computation of a combined rank score, which depends on the difference in mean methylation levels of two groups, the mean methylation quotient, and statistical significance. For subsequent analyses, the top 500 differentially methylated CpG sites (DMPs) were selected. To evaluate the robustness of the results, we have used a validation cohort [29]. Analyses of gene ontology (GO) were conducted using the geometh function from the missMethyl package [33] and heatmaps were generated using ClustVis [27].
Table 1
Patients’ clinical and tumor characteristics
Sex (n; %)
 
 Female
27 (38.0%)
 Male
44 (62.0%)
Age (years)
 
 Median
65 (range: 32–84 years)
Metastatic melanoma, stage IV according to AJCC 8th edition (n; %)
 
 Stage IVM1aIVM1d
71 (100%)
Mutation status (n; %)
 
 BRAF
25 (35.2%)
 NRAS
15 (21.1%)
 cKIT
3 (4.2%)
 wt
28 (39.5%)
Baseline ECOG (n; %)
 
 0
64 (90.1%)
 1
7 (9.9%)
Baseline LDH (U/L)
 
 Normal
47 (66.2%)
 >ULN
20 (28.2%)
 NA
4 (5.6%)
Baseline S100 (μg/L)
 
 Normal
36 (50.7%)
 > ULN
32 (45.1%)
 NA
3 (4.2%)
Origin of treatment naïve tumor tissue for methylation profiling (n; %)
 
 Lymph node metastases
28 (34.6%)
 Organ metastases
20 (24.7%)
 Cutaneous metastases
24 (29.6%)
 Primary tumors
9 (11.1%)
First-line IO (n; %)
 
 Nivolumab monotherapy
24 (33.8%)
 Pembrolizumab monotherapy
12 (16.9%)
 Ipilimumab monotherapy
1 (1.4%)
 Nivolumab and ipilimumab
27 (38.0%)
 ICI-based study medication
7 (9.8%)
Number of cycles applied (cycles)
 
 Median
12 (range: 1–168 cycles)
Treatment duration (months)
 
 Median
11 (range: 0–80 months)
Concomitant therapy (n; %)
 
 No
44 (62.0%)
 Yes
27 (38.0%)
  Surgery
9 (12.7%)
  Radiation therapy
10 (14.1%)
  Gamma knife surgery
8 (11.3%)
PFS and OS (months) (n; %)
 
 Median PFS
8.5 (range 0–104.1 months)
 Median OS
30.6 (range: 0–104.1 months)
iBOR according to iRECIST (n; %)
 
 iCR
14 (19.7%)
 iPR
15 (21.1%)
 iSD
17 (23.9%)
 iPD (iUPD or iCPD)
25 (35.2%)
Response (n; %)
 
 Responder
29 (40.8%)
 Non-responder
42 (59.2%)
Systemic FU-therapy (n; %)
 
 Nivolumab
13 (18.3%)
 Pembrolizumab
2 (2.8%)
 Ipilimumab
9 (12.7%)
 Nivolumab and ipilimumab
12 (16.9%)
 Nivolumab and relatlimab
2 (2.8%)
 Pembrolizumab and lenvatinib
1 (1.4%)
 Dabrafenib and trametinib
7 (9.9%)
 Zelboraf and cobimetinib
1 (1.4%)
 Encorafenib and Binimetinib
7 (9.9%)
 Trametinib
1 (1.4%)
 Dacarbazine
1 (1.4%)
 Temozolomide
1 (1.4%)
 Imatinib
1 (1.4%)
Duration of FU (months)
 
 Median
17.3 (range: 0–56.6 months)
The tumor stage was defined according to the AJCC, 8th Edition. Response was classified according to immune-based Response Evaluation Criteria in Solid Tumors (iRECIST): best overall response (iBOR), immune complete response (iCR), immune partial response (iPR)
AJCC American Joint Committee on Cancer, ECOG Eastern Cooperative Oncology Group, FU follow-up, ICI immune checkpoint inhibitor, iCPD immune confirmed progressive disease, IO immunotherapy, iPD immune progressive disease, iSD immune stable disease, iUPD immune unconfirmed progressive disease, LDH lactate dehydrogenase, NA not applicable, OS overall survival, PFS progression-free survival, ULN upper limit of normal, wt wild type

2.3 Statistics

Follow-up was calculated from the start date of ICI therapy until last contact or death, whatever occurred first. Descriptive statistics were used to describe patient characteristics, such as age, sex, and mutation status. For nominally scaled variables, absolute numbers and percentages were presented, and for metric variables, the mean, standard deviation, median, minimum, and maximum were applied. The Kaplan–Meier method was used to estimate PFS and OS from the first ICI application until the occurrence of disease progression or death. Patients without disease progression were censored on the date of their last visit. Kaplan–Meier curves were compared using the log-rank test and Cox’ proportional hazards models. To test the sensitivity-specificity trade-off, receiver operating characteristic classifier curve analyses were performed. The Kruskal–Wallis test and Mann–Whitney U test were used for comparisons between the groups. Statistical significance was determined by a p-value <0.05 (two-sided), multiple testing was corrected using the Bonferroni method. Statistical analyses were performed with Statistical Package for the Social Sciences (SPSS®) 23.0 software (SPSS Inc., Chicago, IL, USA) and standard R functions (version R 4.0.4; R Foundation for Statistical Computing, Vienna, Austria). The data cut-off date was 30 August, 2022.

3 Results

3.1 DNA Methylation Signatures Correlated with Response to ICI Therapy

To investigate whether DNA methylation signatures can predict the response to ICIs, we performed Infinium MethylationEPIC microarray analyses of tumor specimens collected prior to therapy onset. Eighty-one treatment-naïve tumor tissue samples derived from 71 patients with melanoma were included in the analysis, of which 29 (40.8%) were classified as responders and 42 (59.2%) were non-responders. After quality control and exclusion of 192,085 probes owing to cross-reactivity, unreliable measurements, and sex chromosome specificity, 674,810 probes remained for a further statistical analysis. Testing for methylation differences between responders and non-responders of all tumor samples was performed using RnBeads. This resulted in a total of 58,831 DMPs with a false discovery rate <0.05 (Fig. 1a). Differentially methylated CpG sites were predominantly located within intergenic regions, gene bodies, and transcriptional start sites (Fig. 1b). Based on the top 500 DMPs, three clusters were identified revealing that hypomethylation mainly corresponded to the response to ICIs. Cluster 1 only consisted of responders (12/12), cluster 2 was evenly distributed between responders and non-responders (12/24), and cluster 3 contained mainly non-responders (39/45) (Fig. 1c). Patients in cluster 1 (12/12) and cluster 2 (23/24) had predominantly an ECOG performance status of zero, in contrast to cluster 3, where 7/45 patients had an ECOG performance status ≥1. The baseline lactate dehydrogenase (LDH) levels of most patients in cluster 1 (11/12) and cluster 2 (18/24) had baseline LDH levels within the normal range, and patients in cluster 3 had a comparably high number of patients with baseline LDH levels ≥1 upper limit of normal (13/45). A further exploratory analysis revealed that non-responders compared with responders showed a trend towards a higher ECOG status ≥1 (p = 0.0887), albeit not significant, and no differences between non-responders and responders in baseline LDH (p = 0.6646) and S100 levels (p = 0.7500) were observed. The other clusters did not differ according to factors such as the BRAF mutation status immunotherapy (IO) regimen, sex, localization of the utilized tumor tissue, mutation status, and baseline LDH and S100 values (Fig. 1c).
The predictive performance of the methylation signature was high with 80% sensitivity, 81% specificity, and an area under the curve of 0.829 (Fig. 1d). Progression-free survival and OS were significantly longer in patients from clusters 1 and 2 than in those from cluster 3 (p = 0.02 and p = 0.019, respectively) (Fig. 1e). Altogether, these results indicate that DNA methylation profiles in cutaneous metastatic melanoma correlate with the response to ICIs with high sensitivity and specificity.

3.2 Functional Characterization of Promotor Differentially Methylated Regions

A GO enrichment analysis including three sub-ontologies of biological processes (BP), cellular components (CC), and molecular functions (MF) of promotor differentially methylated regions analyzed with the missMethyl R package revealed that several pathways were significantly enriched. The most enriched pathways were related to olfactory pathways such as “detection of chemical stimulus involved in sensory perception of smell” with an enrichment false discovery rate of 6.13E−45, followed by “sensory perception of smell” and “detection of chemical stimulus involved in sensory perception” detected by GO:BP and “olfactory receptor activity” by GO:MF, which were recently demonstrated to be involved in the cell proliferation and migration processes of primary melanoma and melanoma metastasis [14]. Additionally, GO:BP and GO:MF revealed enrichments in “G protein-coupled receptor signaling pathways,” which were demonstrated to be involved in skin cancer development [32] and were shown to be engaged in melanogenesis including proliferation and migration [34]. Furthermore, GO:BP, GO:MF, and GO:CC displayed enrichments related to epidermal keratinocytes such as “keratinocyte differentiation,” “epidermis development,” “keratin filament,” and “intermediate filament cytoskeleton” (Table 2), indicative of an crosstalk between melanoma cells and the surrounding tumor microenvironment [21]. Finally, GO:BP enrichments related to B cells such as “humoral immune response” and “antimicrobial humoral immune response” were detected, which were demonstrated to promote a response to ICIs in metastatic melanoma [17].
Table 2
Pathway enrichment analysis of promoter differentially methylated regions
ID
Ontology
Term
N
DE
P.DE
FDR
GO:0050911
BP
Detection of chemical stimulus involved in sensory perception of smell
375
195
5.34E−49
6.13E−45
GO:0007608
BP
Sensory perception of smell
401
200
1.84E−46
1.41E−42
GO:0050907
BP
Detection of chemical stimulus involved in sensory perception
421
205
9.10E−46
5.22E−42
GO:0009593
BP
Detection of chemical stimulus
456
212
3.58E−43
1.64E−39
GO:0007606
BP
Sensory perception of chemical stimulus
477
218
9.53E−43
3.64E−39
GO:0050906
BP
Detection of stimulus involved in sensory perception
486
218
4.23E−41
1.39E−37
GO:0051606
BP
Detection of stimulus
613
246
3.80E−36
9.69E−33
GO:0007600
BP
Sensory perception
944
330
1.24E−33
2.85E−30
GO:0007186
BP
G protein-coupled receptor signaling pathway
1207
390
3.97E−31
8.29E−28
GO:0050877
BP
Nervous system process
1477
449
1.35E−28
2.58E−25
GO:0003008
BP
System process
2262
610
2.58E−23
3.70E−20
GO:0031424
BP
Keratinization
83
51
2.04E−17
2.75E−14
GO:0045109
BP
Intermediate filament organization
69
37
9.01E−11
8.61E−08
GO:0032501
BP
Multicellular organismal process
7555
1600
1.20E−10
1.10E−07
GO:0030216
BP
Keratinocyte differentiation
169
65
3.77E−09
3.33E−06
GO:0009913
BP
Epidermal cell differentiation
234
80
1.77E−08
1.45E−05
GO:0045104
BP
Intermediate filament cytoskeleton organization
89
39
6.52E−08
5.16E−05
GO:0045103
BP
Intermediate filament-based process
90
39
9.24E−08
7.06E−05
GO:0008544
BP
Epidermis development
361
105
1.49E−06
0.001065117
GO:0043588
BP
Skin development
301
90
2.53E−06
0.00176051
GO:0019730
BP
Antimicrobial humoral response
113
40
3.14E−05
0.018962766
GO:0006959
BP
Humoral immune response
240
70
6.25E−05
0.03583897
GO:0042221
BP
Response to chemical
4405
920
6.79E−05
0.037987162
GO:0099536
BP
Synaptic signaling
752
186
7.39E−05
0.04034171
GO:0050896
BP
Response to stimulus
8762
1761
9.57E−05
0.049913398
GO:0004984
MF
Olfactory receptor activity
373
195
1.63E−49
3.73E−45
GO:0004930
MF
G protein-coupled receptor activity
800
304
1.88E−39
5.39E−36
GO:0004888
MF
Transmembrane signaling receptor activity
1229
386
4.51E−28
7.95E−25
GO:0038023
MF
Signaling receptor activity
1448
433
2.66E−26
4.07E−23
GO:0060089
MF
Molecular transducer activity
1448
433
2.66E−26
4.07E−23
GO:0030280
MF
Structural constituent of skin epidermis
37
29
6.88E−15
7.90E−12
GO:0008528
MF
G protein-coupled peptide receptor activity
144
49
6.78E−06
0.00444656
GO:0001653
MF
Peptide receptor activity
150
49
2.30E−05
0.014240561
GO:0033691
MF
Sialic acid binding
22
13
4.57E−05
0.026900852
GO:0031406
MF
Carboxylic acid binding
170
53
7.99E−05
0.042622563
GO:0004984
MF
Olfactory receptor activity
373
195
1.63E−49
3.73E−45
GO:0004930
MF
G protein-coupled receptor activity
800
304
1.88E−39
5.39E−36
GO:0071944
CC
Cell periphery
5958
1339
6.18E−16
7.88E−13
GO:0005886
CC
Plasma membrane
5479
1242
6.63E−16
8.00E−13
GO:0045095
CC
Keratin filament
99
51
1.39E−13
1.51E−10
GO:0016021
CC
Integral component of membrane
5205
1150
1.29E−12
1.34E−09
GO:0031224
CC
Intrinsic component of membrane
5370
1180
3.01E−12
3.00E−09
GO:0005882
CC
Intermediate filament
203
72
1.03E−08
8.73E−06
GO:0045111
CC
Intermediate filament cytoskeleton
241
79
9.60E−08
7.10E−05
GO:0016020
CC
Membrane
9138
1852
3.69E−06
0.002487588
GO:0001533
CC
Cornified envelope
59
26
9.34E−06
0.005952581
GO:0071944
CC
Cell periphery
5958
1339
6.18E−16
7.88E−13
GO:0005886
CC
Plasma membrane
5479
1242
6.63E−16
8.00E−13
GO:0045095
CC
Keratin filament
99
51
1.39E−13
1.51E−10
GO:0016021
CC
Integral component of membrane
5205
1150
1.29E−12
1.34E−09
GO:0031224
CC
Intrinsic component of membrane
5370
1180
3.01E−12
3.00E−09
GO:0005882
CC
Intermediate filament
203
72
1.03E−08
8.73E−06
GO:0045111
CC
Intermediate filament cytoskeleton
241
79
9.60E−08
7.10E−05
GO:0016020
CC
Membrane
9138
1852
3.69E−06
0.002487588
GO:0001533
CC
Cornified envelope
59
26
9.34E−06
0.005952581
Pathway enrichment analysis using promoter differentially methylated regions of the whole study population. Ranked by DR (differentially ranked) using a significance cut-off FDR <0.05; total number of genes in the pathway (nGenes)
BP biological processes, CC cellular components, DE differentially expressed, FDR false discovery rate, ID identification, MF molecular functions, P.DE p-value for over-representation of the gene ontology (GO) term

3.3 Tissue Origin Did Not Affect the DNA Methylation-Based Prediction of an ICI Response

Next, we performed differential methylation analyses of tissues derived from different organ sites. Based on the top 500 DMPs, lymph node metastasis-only samples revealed two clusters and clearly separated non-responders (17/18) in cluster 1 and responders (9/10) in cluster 2 (Fig. 2). Differential methylation analyses performed on different subgroups of patients (cutaneous metastases, primary tumors, and organ metastases) also identified two clusters containing mainly responders and non-responders (Fig. 3). In detail, an analysis of exclusively cutaneous metastases, based on 500 DMPs, identified two clusters, with the majority of responders in cluster 1 (9/10) and non-responders in cluster 2 (12/13) (Fig. 3a). A differential methylation analysis of only primary tumors, based on 500 DMPs, revealed a clear separation of non-responders (10/10) and responders (2/2); however, interpretation was limited by the low number of responders in this cohort (Fig. 3b). A differential analysis of the subset of organ metastases based on 500 mostly DMPs identified two clusters, with responders being mainly in cluster 1 (6/6) and non-responders preferentially in cluster 2 (10/12) (Fig. 3c). In a subset of patients, 8/71 (11.3%), more than one treatment-naïve tumor tissue sample was available (Table S1 of the ESM), and methylation profiling of all samples was performed accordingly. In 5/8 patients, the samples from different body sites clustered together (Fig. 4), whereas in 3/8 patients, an intra-patient different methylation pattern was observed. Irrespective, in-depth analysis per tissue methylation profiling showed that all samples correlated with response to ICI, irrespective of the tumor origin (Fig. 1c). These results support that tissues derived from different organ sites do not affect the DNA methylation-based prediction of ICI responses.

3.4 Methylation Signatures of an Independent Validation Data Set

We aligned our findings to an independent data set. This validation cohort consisted of 43 patients with tumor tissue samples, of which 26 patients were classified as responders and 17 as non-responders [29]. Based on the 500 differentially methylated regions from our cohort, we identified similar trends (p = 0.146). In detail, we identified three clusters, showing responders mainly cluster 2 (15/20, 75%) and non-responders in cluster 3 (5/8, 62.5%), with hypomethylation being more prevalent in responders (Fig. S1 of the ESM).

4 Discussion

This study demonstrates that DNA methylation signatures of treatment-naïve tumor samples, regardless of the tumor tissue origin, can predict responses to ICIs in patients with stage IV metastatic melanoma, and is warranted for further analysis in prospective cohorts. It has been shown that epigenetic changes can contribute to response and failure to ICIs and DNA methylation profiling has been proposed to be a useful tool to predict responses to ICIs in different solid tumor entities [39, 40, 42]. There is still a lack of data regarding DNA methylation profiling and responses to ICIs in the field of melanoma. Newell et al. and Filipinski et al. reported that DNA methylation might be useful for predicting the response to ICIs in metastatic melanoma [7, 29]. In contrast, these studies did not evaluate whether the tumor tissue origin affected the DNA methylation signature and prediction to ICIs, nor did they use exclusively treatment-naïve tumor tissues for analysis. We have utilized the study of Newell et al. as a validation cohort, because of the biggest similarity to our cohort, although differences certainly limited the usage as a validation dataset. The Newell et al. cohort differed in the radiological response assessment, using RECIST and not iRECIST criteria, and responders were classified as complete response, partial response, and stable disease (>6 months) and non-responders as stable disease (<6 months) and progressive disease. Furthermore, no detailed information on the origin of tumor tissue used for the methylation analysis was available. The whole study cohort’s distribution of tumor tissues (n = 71) mainly consisted of subcutaneous tissue (56%), lymph nodes (30%), brain metastases (9%), and primary tumors (4%). Sixty-six percent were treatment-naïve tumor tissue samples and 34% had an intervening systemic therapy before the start of ICI treatment. Nevertheless, we observed similar trends with hypomethylation being present mainly in responders. However, the heterogenous distribution of responders and non-responders observed in these two cohorts was not unexpected because of the above-mentioned variety in the cohorts. Therefore, tumor tissue characteristics alone might be insufficient to reliably predictive ICI responses and incorporating a multimodal concept including systemic inflammatory responses in combination with tissue-based parameters might be needed for a reproducible response prediction to ICIs in metastatic melanoma.
Our study using exclusively treatment-naïve tissue samples adds to the current knowledge that there is no tissue-dependent influence on DNA methylation profiling or prediction of responses to ICIs. It has been reported that location-dependent, global DNA methylation patterns are shared between mucosal and cutaneous melanoma from primary sites, whereas uveal melanoma exhibits a different methylation signature [19]. Therefore, and because of the limited response rates of <10% to ICIs compared with cutaneous melanoma, we excluded uveal and mucosal melanomas from our analysis [1, 4, 36, 49]. Consequently, this further reduced potential batch effects when evaluating the DNA methylation signatures of different tissue sites, including primary tumors and affected body sites, by metastatic diseases of organs (including brain metastases), lymph nodes, and skin. To our knowledge, no such comparison regarding the DNA methylation signature and response to ICIs have been performed, and our results indicate that tissue samples from different sites may likewise predict the response to ICIs to a similar degree.
In addition, our pathway analysis provides further insights into the potentially differentially regulated pathways between responders and non-responders. Enrichments were detected in olfactory and G-protein coupled receptor signaling pathways, which were recently associated with migration and proliferation processes in melanoma [14, 34], as well as enrichments in the humoral response, which were shown to be supportive for ICI responses in metastatic melanoma [17].
Furthermore, our data imply that DNA methylation profiling has a higher sensitivity and specificity than established clinical markers such as LDH [47] and ECOG performance status [44]. An LDH ≥1 upper limit of normal is a well-established negative prognostic marker for reduced PFS and OS but has a low predictive value for a beneficial treatment outcome [47]. The ECOG performance status was initially established and used as a predictor of response and toxicity in patients receiving chemotherapy [31]. Regarding ICIs, a baseline ECOG performance status of zero has been associated with disease control [44], whereas other studies did not find a correlation between an ECOG score of zero and ICI-based survival benefit [48]. This confirms our findings that non-responders compared to responders showed a trend towards a higher ECOG status ≥1 (p = 0.0887), albeit not significant. However, we did not detect differences between non-responders and responders in baseline LDH (p = 0.6646) and S100 levels (p = 0.7500). We further observed that hypomethylation was associated with a beneficial response to ICIs. It was shown that global hypomethylation was associated with the expression of programmed death-ligand 1 (PD-L1) on melanoma cells in vitro [6, 15]. This is important because immunosuppressive receptors such as PD-L1 and PD-1 can inhibit the function of effector T cells [18]. However, the role of PD-L1 expression and response to ICIs in melanoma is discussed controversially and could not predict a positive clinical outcome [3, 28], Robert et al. 2015; [46], which led to the approval of PD-1 inhibitors for melanoma in metastatic [23, 45] and adjuvant [43] settings by the US Food and Drug Administration irrespective of the PD-L1 expression status of the tumor tissue.
Several limitations must be considered in the current analysis, such as the heterogeneity of the patient cohort in terms of patient characteristics and applied treatments. In comparison with other studies, we could demonstrate that tumor tissues of different origins can be used for DNA methylation profiling, which will potentially contribute to the clinical routine as a useful and easily applicable biomarker in the near future. Tissue-based DNA methylation profiling together with for example, systemic inflammatory signatures, should be addressed by future studies.

5 Conclusions

Our predictive DNA methylation signatures showed promising sensitivity and specificity; however, preferentially prospective validation cohorts are needed, possibly by using a combinational tissue-based and systemic inflammatory approach. Nevertheless, given the current lack of reliable and robust biomarkers for ICI responses in metastatic melanoma, the current study is of certain relevance. Indeed, methylation analysis is of high interest for everyday clinical use, as its analytical performance is robust, the costs are within a reasonable range, and a methylation analysis has already been established for diagnostic purposes in central nervous system malignancies [2, 24]. In summary, this study indicates that DNA methylation profiling is a useful tool for predicting the response to ICIs in patients with metastatic melanoma with high sensitivity and specificity, independent of the tissue origin.

Acknowledgments

We thank the Department of Pathology, Medical University of Vienna for providing tissue samples for the DNA methylation profiling analysis.

Declarations

Funding

Open access funding provided by Medical University of Vienna. This study was supported by the Christian Doppler Research Association, the Austrian Federal Ministry for Digital and Economic Affairs, and the National Foundation for Research, Technology, and Development.

Conflicts of Interest

Julia Maria Ressler received honoraria for lectures from Bristol-Myers Squibb, Roche, Amgen, and Novartis and travel support from Sanofi, Roche, and Bristol-Myers Squibb. Angelika Martina Starzer received honoraria for lectures from AstraZeneca and travel and congress support from PharmaMar, MSD, and Lilly. Christoph Höller reports speaker honoraria from Amgen, BMS, MSD, Novartis, and Roche, participation in advisory boards for Amgen, Astra Zeneca, BMS, Incyte, MSD, Novartis, Pierre Fabre, and Roche, and research funding by Amgen to the institution. Matthias Preusser has received honoraria for lectures, consultation, and advisory board participation from the following for-profit companies: Bayer, Bristol-Myers Squibb, Novartis, Gerson Lehrman Group (GLG), CMC Contrast, GlaxoSmithKline, Mundipharma, Roche, BMJ Journals, MedMedia, Astra Zeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dome, Tocagen, Adastra, Gan & Lee Pharmaceuticals, and Servier. Anna Sophie Berghoff has research support from Daiichi Sankyo and Roche, honoraria for lectures, consultation, and advisory board participation from Roche Bristol-Meyers Squibb, Merck, Daiichi Sankyo, AstraZeneca, and CeCaVa, and travel support from Roche, Amgen, and AbbVie. Philipp Tschandl received honoraria from Silverchair, unrestricted grants for education projects from Lilly, and honoraria for lectures from AbbVie, Lilly, FotoFinder, and Novartis. Erwin Tomasich, Teresa Hatziioannou, Helmut Ringl, Gerwin Heller, Rita Silmbrod, Lynn Gottmann, and Nina Zila have no conflicts of interest that are directly relevant to the content of this article.

Ethics Approval

This study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Ethics committee of the Medical University of Vienna (Number 1312/2021).
Written informed consent from the study participants was obtained.
Not applicable.

Availability of Data and Material

Raw data of DNA methylation analysis are publicly available at the Gene Expression Omnibus database respiratory under the accession number GSE235122. Further data will be shared upon reasonable request to the corresponding author. The relevant data set of the study is included in the article or uploaded as supplementary information. Additional data will be shared upon request by the authors.

Code Availability

Not applicable.

Authors’ Contributions

JMR: conceptualization, resources, data curation, formal analysis, validation, writing of the original draft. ET: formal analysis, validation, writing. TH: formal analysis, validation. HR: radiological assessment, validation. GW: bioinformatical analysis, visualization, validation, writing. RS: data curation, formal analysis. LG: data curation, formal analysis. AMS: data curation, formal analysis. NZ data curation, formal analysis. PT: resources, formal analysis. CH: resources, validation, supervision. MP: conceptualization, resources, validation, writing, supervision. ASB: conceptualization, resources, formal analysis, validation, writing. All authors read and approved the final manuscript.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
19.
Zurück zum Zitat Jurmeister P, Wrede N, Hoffmann I, Vollbrecht C, Heim D, Hummel M, et al. Mucosal melanomas of different anatomic sites share a common global DNA methylation profile with cutaneous melanoma but show location-dependent patterns of genetic and epigenetic alterations. J Pathol. 2022;256(1):61–70. https://doi.org/10.1002/path.5808.CrossRefPubMed Jurmeister P, Wrede N, Hoffmann I, Vollbrecht C, Heim D, Hummel M, et al. Mucosal melanomas of different anatomic sites share a common global DNA methylation profile with cutaneous melanoma but show location-dependent patterns of genetic and epigenetic alterations. J Pathol. 2022;256(1):61–70. https://​doi.​org/​10.​1002/​path.​5808.CrossRefPubMed
Metadaten
Titel
DNA Methylation Signatures Correlate with Response to Immune Checkpoint Inhibitors in Metastatic Melanoma
verfasst von
Julia Maria Ressler
Erwin Tomasich
Teresa Hatziioannou
Helmut Ringl
Gerwin Heller
Rita Silmbrod
Lynn Gottmann
Angelika Martina Starzer
Nina Zila
Philipp Tschandl
Christoph Hoeller
Matthias Preusser
Anna Sophie Berghoff
Publikationsdatum
24.02.2024
Verlag
Springer International Publishing
Erschienen in
Targeted Oncology / Ausgabe 2/2024
Print ISSN: 1776-2596
Elektronische ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-024-01041-4

Weitere Artikel der Ausgabe 2/2024

Targeted Oncology 2/2024 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.