The presence of tumor-infiltrating immune cells is associated with longer survival and a better response to immunotherapy in early-stage melanoma, but a comprehensive study of the in situ immune microenvironment in stage IV melanoma has not been performed. We investigated the combined influence of a series of immune factors on survival and response to adoptive cell transfer (ACT) in stage IV melanoma patients. Metastases of 73 stage IV melanoma patients, 17 of which were treated with ACT, were studied with respect to the number and functional phenotype of lymphocytes and myeloid cells as well as for expression of galectins-1, -3, and -9. Single factors associated with better survival were identified using Kaplan–Meier curves and multivariate Cox regression analyses, and those factors were used for interaction analyses. The results were validated using The Cancer Genome Atlas database. We identified four parameters that were associated with a better survival: CD8+ T cells, galectin-9+ dendritic cells (DC)/DC-like macrophages, a high M1/M2 macrophage ratio, and the expression of galectin-3 by tumor cells. The presence of at least three of these parameters formed an independent positive prognostic factor for long-term survival. Patients displaying this four-parameter signature were found exclusively among patients responding to ACT and were the ones with sustained clinical benefit. Cancer Immunol Res; 5(2); 170–9. ©2017 AACR.

Melanoma is the most aggressive form of skin cancer and has long been recognized as a highly immunogenic tumor and a good target for immunotherapy (1). In different types of cancer, including melanoma, the presence of type I cytokine–oriented tumor-infiltrating lymphocytes (TIL) has been associated with improved survival (2). Indeed, a strong ongoing immune response was linked to spontaneous regression in about half of the primary melanomas (3) and longer survival of patients with stage I–III primary and regionally metastasized melanoma (4–6). More recently, a large study in patients with stage IV (distant metastases) melanoma revealed that even at this stage, intratumoral T-cell content was associated with improved survival (7). However, the predictive value for survival was not so strong, indicating that other immune-related factors previously studied in primary melanoma may also play a role (8–12); this involvement of other immune parameters was also suggested by studies at the gene expression level (13, 14). In parallel, studies showing that a strong intratumoral T-cell infiltrate fosters a better response to PD-1 checkpoint therapy (15) and autologous tumor cell vaccination (11), but also that intratumoral macrophages can hamper CTLA-4 checkpoint therapy (16), suggest that the tumor's immune contexture may also influence the response to immunotherapy.

In this study, we have expanded on earlier studies (4, 7) by assessing the influence of a series of immune factors in the metastatic tumor microenvironment in a large group of stage IV melanoma patients with up to 10 years of follow-up since metastasis. We identified four parameters, each of which was associated with better survival. These parameters comprised CD8 T cells, the presence of galectin-9+ dendritic cells (DC)/DC-like macrophages, a higher M1/M2 macrophage ratio, and galectin-3 expression by tumor cells. The presence of at least three parameters was an independent prognostic factor for survival, which was validated by analysis of these parameters in stage IV melanoma patients in The Cancer Genome Atlas (TCGA) database. Furthermore, with the introduction of targeted therapies and checkpoint inhibitors, adoptive cell transfer (ACT) has mostly become a salvage therapy (17) for treatment of stage IV melanoma patients. Analysis of the predictive value of this signature for the response to ACT revealed that the pretreatment tumors of patients without clinical benefit (CB) predominantly display two or fewer of the beneficial immune parameters, whereas the presence of three or four of these parameters was most frequent in patients showing sustained CB after ACT.

Patient material

Formalin-fixed, paraffin-embedded tissue blocks from 73 stage IV metastatic melanoma patients undergoing surgery were collected at Leiden University Medical Center (LUMC, Leiden, the Netherlands) and at the Netherlands Cancer Institute (NCI, Amsterdam, the Netherlands). The patients were included in clinical studies that were approved by a local ethical committee (LUMC study P04.085, NCI study EudraCT 2010-021885-31), and all patients gave written informed consent. All specimens were from metastases, and biopsies were taken before any immunotherapeutic treatment. Classification of metastases was done by tumor–node–metastasis (TNM) staging criteria (18), and information on the concentration of lactate dehydrogenase (LDH) at the moment of sampling was collected. Included in the cohort of 73 patients were 17 patients that were treated with ACT in ongoing clinical studies in the LUMC and NCI. Of these patients, 7 were classified as patients without CB [progressive disease (PD)] and 10 patients with CB (stable disease (SD), partial response (PR), and complete response (CR)] according to RECIST1.1 criteria.

Immunofluorescence and IHC

The presence of T-cell and macrophage infiltrate in the tumor area and the expression of galectin-1, galectin-3, and galectin-9 by the tumor was analyzed using previously determined optimal antibody concentrations and immunofluorescence staining protocols as described before (19, 20). Briefly, T-cell infiltrates were stained with antibodies to CD3, CD8, and FoxP3. Macrophages were identified using antibodies to CD14 and CD163. To examine which cells expressed galectin-9, a small part of the cohort received triple immunofluorescence staining with antibodies to galectin-9, CD68, and CD11c. All secondary antibodies were isotype-specific antibodies labeled with the fluorochromes Alexa Fluor 488, 546, or 647. The expression of Tbet was analyzed by IHC as described before (19) with the exception that after incubation with the primary Tbet antibody and incubation with BrightVision poly-HRP anti-mouse/rabbit/rat IgG, the antigen-antibody reactions were visualized using the NovaRED Substrate Kit for peroxidase (Vector Laboratories). For all antibody labeling, negative controls and controls omitting the primary or secondary antibody were performed. Positive control tissue slides were included for all antibody labeling, using tonsil for T cells, colon for galectin, and placenta for macrophage controls. Images were captured using a confocal microscope (LSM15, Zeiss) for the immunofluorescence labeling and a spectral microscope (Leica DM4000 B, Leica Microsystems) for the immunohistochemical stains. Random images (five per slide) were taken for analysis. Analysis of the images was done using ImageJ. Intratumoral T cells, macrophages, galectin-9+ cells, and Tbet+ cells were manually counted using the “cell counter” plugin of ImageJ and presented as number of cells/mm2 (average of five images). Galectin-1 and galectin-3 expression by tumor cells were analyzed using an immunoreactive score (IRS; ref. 21), taking into account the percentage of positive cells and the intensity of the staining (Supplementary Table S1A).

TCGA analysis

For validation of our results of the IHC and immunofluorescence experiments, we analyzed data from the publicly available TCGA database (5). To reconstruct our parameters, we used gene expression profiles of the subset of patients with stage IV melanoma. CD8+ T cells were identified by taking the average of CD8A and CD8B expression, galectin-9+ DC-like macrophages by GALS9 expression, M1/M2 macrophage ratio by the ratio of CD86 and CD163 expression, and expression of galectin-3 by tumor cells by LGALS3 expression. This approach assumes that the genes we used are expressed preferably by the target cells for the parameter, and not in the other cell types in the sample. We used the z-scores as available in the TCGA data. This will yield a more comparable weight of all parameters. For each parameter, a high and a low group was defined by splitting the samples on the median value for the parameter.

Statistical analysis

The differences between different patient groups were analyzed using the nonparametric Mann–Whitney U test for comparison of continuous variables between two groups and the one-way ANOVA or Kruskal–Wallis test for the comparison of three groups. For comparison of categorical data, the χ2 test or the Fisher exact test was used. Correlation analysis was done using Spearman ρ correlation. Correlation between immune parameters and overall survival (OS) since metastasis was calculated by the Kaplan–Meier method and statistically analyzed by the log-rank test. Univariate and multivariate Cox proportional hazards models were used to determine the HR that represents the relative risk of death among patients in the different indicated groups. In the multivariate Cox regression models, analysis was corrected for age, serum LDH, and the pattern of visceral metastases (TNM staging), the latter two being established prognostic factors for stage IV melanoma. Interaction analyses were performed on the parameters that were identified as prognostic for survival. For all tests, P values <0.05 were considered statistically significant. For statistical analysis, the software package SPSS statistics 20.0.0 was used (SPSS Inc.).

Patient characteristics

To study the immune markers that relate to survival in stage IV metastatic melanoma, tumor biopsies from a group of 73 patients were investigated (Supplementary Table S1B). The mean age at the moment of sampling was 52 (range, 25–74; 41 males and 32 females). The patients were divided in tertiles (<9 months, 9–20 months, >20 months) based on the survival of stage IV disease. The mean age of the group of patients with the longest survival cohort was a bit lower, but this difference was not significant (one-way ANOVA, P = 0.09). Maximal follow-up of the patients since collection of the metastatic sample was 120 months, and the median survival since metastasis was 13 months (range, 1–120). Within the cohort of 73 patients, 17 patients were included that were treated with ACT. Of these 17 patients, 6 patients received T cells that were generated by mixed lymphocyte tumor cultures of autologous tumor cells and peripheral blood mononuclear cells (22). The other 11 patients received T cells that were generated by rapid expansion of TILs (23). Within the ACT-treated group of 17 patients, 10 were classified as having CB (3× SD, 4× PR, and 3× CR). The CB patients showed a significantly better survival compared with the patients with no CB when calculated by the Kaplan–Meier method and analyzed by the log-rank test (PD; P < 0.001; Supplementary Fig. S1). The different methods used to generate T-cell batches for ACT did not influence clinical outcome (Fisher exact test, P = 0.64).

T cells, macrophages, galectin-3, and galectin-9 are prognostic factors for survival

To study the immune signature in each tumor, the numbers of T cells, macrophages, and galectin-9–positive immune cells were quantified per square millimeter of tumor area, whereas the expression of galectin-1 and galectin-3 by tumor cells was determined via the IRS (Fig. 1; Supplementary Table S2A). CD8+ T cells were defined as CD3+CD8+FoxP3, CD4+ T cells were defined as CD3+CD8FoxP3, and regulatory T cells (Treg) were defined as CD3+CD8FoxP3+ (Supplementary Fig. S2A). Two types of macrophages were defined: CD14+CD163 (M1) and CD14+CD163+ (M2) macrophages (Fig. 2B). Galectin-9 was, based on morphology, expressed by cells of myeloid origin (Fig. 2C). Additional costaining with CD68 and CD11c revealed that a majority (70%) of these galectin-9–expressing cells coexpressed CD11c, and part of these cells also expressed CD68, indicating that the galectin-9+ myeloid cells were predominantly DCs or DC-like macrophages (Fig. 3A and B).

Figure 1.

Cell counts of immune infiltrate and expression of galectin-1 and galectin-3 in the short-, medium-, and long-term survival cohorts. A, Cell counts (cells/mm2) and the median are depicted from the number of CD8+ T cells, CD4+ T cells, FoxP3+ T cells, and Total T cells (Tc) that infiltrated the tumor. B, Cell counts and median from the intratumoral M1 (CD14+CD163) and M2 (CD14+CD163+) macrophages as well as the ratio of M1/M2. C, Cell counts (cells/mm2) for galectin-9+ cells and the IRS of galectin-1 and galectin-3 on the tumor cells is shown. Statistical analysis of the differences between the three patient groups was performed with the nonparametric Kruskal–Wallis test.

Figure 1.

Cell counts of immune infiltrate and expression of galectin-1 and galectin-3 in the short-, medium-, and long-term survival cohorts. A, Cell counts (cells/mm2) and the median are depicted from the number of CD8+ T cells, CD4+ T cells, FoxP3+ T cells, and Total T cells (Tc) that infiltrated the tumor. B, Cell counts and median from the intratumoral M1 (CD14+CD163) and M2 (CD14+CD163+) macrophages as well as the ratio of M1/M2. C, Cell counts (cells/mm2) for galectin-9+ cells and the IRS of galectin-1 and galectin-3 on the tumor cells is shown. Statistical analysis of the differences between the three patient groups was performed with the nonparametric Kruskal–Wallis test.

Close modal
Figure 2.

CD8, galectin-3, galectin-9, and the M1/M2 ratio are associated with a longer survival. A, Kaplan–Meier curves showing the cumulative survival since metastasis for the patients with high and low number of CD8+ T cells, high and low ratios of M1/M2 macrophages, high and low numbers of galectin-9+ (Gal9) cells, and high and low galectin-3 (Gal3) expression. B, Kaplan–Meier curves for the patient groups that show a combination of the indicated two parameters as depicted in the graphs compared with all other patient groups. The depicted P values are from the log-rank test.

Figure 2.

CD8, galectin-3, galectin-9, and the M1/M2 ratio are associated with a longer survival. A, Kaplan–Meier curves showing the cumulative survival since metastasis for the patients with high and low number of CD8+ T cells, high and low ratios of M1/M2 macrophages, high and low numbers of galectin-9+ (Gal9) cells, and high and low galectin-3 (Gal3) expression. B, Kaplan–Meier curves for the patient groups that show a combination of the indicated two parameters as depicted in the graphs compared with all other patient groups. The depicted P values are from the log-rank test.

Close modal
Figure 3.

Immune signatures of patients with short-, medium-, and long-term survival. A, The fraction of tumors displaying the number of positive immune parameters (0/4, 1/4, 2/4, 3/4, or 4/4) was plotted for the three survival groups. The fraction of tumors displaying a certain immune signature differed significantly between the three survival cohorts (χ2 test, P < 0.001). B, The correlation between the immune signature and survival was analyzed by the Kaplan–Meier method. C, The correlation between immune signature was analyzed in an independent patient cohort of stage IV melanoma patients from the TCGA database by the Kaplan–Meier method.

Figure 3.

Immune signatures of patients with short-, medium-, and long-term survival. A, The fraction of tumors displaying the number of positive immune parameters (0/4, 1/4, 2/4, 3/4, or 4/4) was plotted for the three survival groups. The fraction of tumors displaying a certain immune signature differed significantly between the three survival cohorts (χ2 test, P < 0.001). B, The correlation between the immune signature and survival was analyzed by the Kaplan–Meier method. C, The correlation between immune signature was analyzed in an independent patient cohort of stage IV melanoma patients from the TCGA database by the Kaplan–Meier method.

Close modal

Galectin-1 and galectin-3 were expressed by the tumor cells, but with different intensities between samples (Fig. 2C). To determine the type I orientation of the TIL, part of the cohort was stained for Tbet (Fig. 3C). A strong positive correlation between the number of T cells and the number of Tbet+ cells was observed (Spearman ρ correlation coefficient 0.720, P < 0.001; Supplementary Table S2), suggesting a type I immune contexture in strongly T-cell–infiltrated tumors (24). In addition, the number of galectin-9+ DCs/DC-like macrophages was also strongly related to that of intratumoral T cells and Tbet+ cells (Supplementary Table S2B), fitting with the notion that galectin-9 is expressed on immune cells upon exposure to proinflammatory mediators (25). No correlation was found between OS and any of the investigated markers (Supplementary Table S2B).

Quantification of the T cells revealed huge variability in the number of CD8, CD4, and Treg cells, as well as tumor-infiltrating M1 and M2 macrophages, within each survival group. Patients with high T-cell counts and a higher ratio of M1/M2 macrophages were more often found in the long- and medium-term survival groups (Fig. 1A and B). The number of patients with dense galectin-9+ DCs/DC-like macrophages was higher in the groups of patients with medium- and long-term survival (Fig. 1C). The expression of galectin-1 and -3 by tumor cells was not so different between the patient groups, albeit that in the long-term survival group, galectin-1 expression was somewhat lower and galectin-3 higher (Fig. 1C). Notably, none of the observed differences were statistically significant between the three groups (Supplementary Table S2).

To investigate the potential influence of all these parameters on survival since metastasis, a univariate and multivariate Cox analysis was performed, and this revealed that high numbers of CD8+ T cells and galectin-9+ DCs/DC-like macrophages, a high M1/M2 ratio, and tumors expressing galectin-3 were prognostic factors for survival (Table 1). For calculation of the correlation between these parameters and survival using the Kaplan–Meier method, the patients were stratified on the basis of the median cell counts or IRS. The presence of these four parameters was also related in this analysis to longer survival (Fig. 2A). Because these four different elements in the tumor immune microenvironment were prognostic for survival, we performed interaction analyses of each combination of two parameters. Patients with a combination of two of any of these parameters (CD8+ high/M1/M2 high/galectin-9+ high/galectin-3 high) showed better survival compared with the other patients, resulting in decreased HRs in the multivariate Cox regression analysis (Table 1) as well as higher survival rates when the Kaplan–Meier analysis was used (Fig. 2B). Each component positively contributed to the other (Fig. 4).

Table 1.

Univariate and multivariate analysis of survival since metastasis

VariableCrude HR (95% CI)PAdjusted HRa (95% CI)P
Gender 0.867 (0.507–1.484) 0.603   
Age 1.027 (1.005–1.049) 0.017   
LDH level high 4.513 (2.080–9.793) 0.000   
TNM stage M1c 2.280 (1.275–4.075) 0.005   
CD4+ infiltration 0.681 (0.340–1.160) 0.158 0.758 (0.421–1.364) 0.356 
CD8+ infiltration 0.561 (0.328–0.961) 0.035 0.583 (0.325–1.044) 0.058 
FoxP3+ infiltration 0.620 (0.362–1.062) 0.081 0.795 (0.443–1.428) 0.443 
Total T-cell infiltration 0.608 (0.356–1.037) 0.068 0.590 (0.334–1.043) 0.069 
Tbet 0.668 (0.378–1.181) 0.165 0.833 (0.446–1.557) 0.567 
CD8/Treg ratio 1.637 (0.956–2.801) 0.072 1.129 (0.631–2.020) 0.683 
Galectin-1 expression by tumor 1.245 (0.731–2.123) 0.420 1.461 (0.800–2.669) 0.217 
Galectin-3 expression by tumor 0.481 (0.275–0.839) 0.010 0.432 (0.239–0.782) 0.006 
Galectin-9 infiltration 0.575 (0.334–0.991) 0.046 0.712 (0.377–1.347) 0.297 
M1 infiltration 0.723 (0.425–1.230) 0.232 0.836 (0.460–1.521) 0.557 
M2 infiltration 1.307 (0.763–2.242) 0.330 1.374 (0.761–2.481) 0.293 
M1/M2 ratio 0.518 (0.297–0.903) 0.020 0.426 (0.227–0.802) 0.008 
CD8 high & Gal3 high 0.316 (0.142–0.704) 0.005 0.278 (0.112–0.686) 0.005 
CD8 high & Gal9 high 0.468 (0.254–0.862) 0.015 0.600 (0.296–1.214) 0.156 
CD8 high & M1/M2 high 0.444 (0.203–0.867) 0.017 0.409 (0.191–0.876) 0.021 
Gal3 high & Gal9 high 0.331 (0.616–0.684) 0.003 0.324 (0.145–0.723) 0.006 
Gal3 high & M1/M2 high 0.339 (0.172–0.668) 0.002 0.289 (0.142–0.589) 0.001 
Gal9 high & M1/M2 high 0.357 (0.717.0.722) 0.004 0.434 (0.212–0.892) 0.023 
3 or more parameters high 0.328 (0.177–0.608) 0.000 0.273 (0.134–0.555) 0.000 
VariableCrude HR (95% CI)PAdjusted HRa (95% CI)P
Gender 0.867 (0.507–1.484) 0.603   
Age 1.027 (1.005–1.049) 0.017   
LDH level high 4.513 (2.080–9.793) 0.000   
TNM stage M1c 2.280 (1.275–4.075) 0.005   
CD4+ infiltration 0.681 (0.340–1.160) 0.158 0.758 (0.421–1.364) 0.356 
CD8+ infiltration 0.561 (0.328–0.961) 0.035 0.583 (0.325–1.044) 0.058 
FoxP3+ infiltration 0.620 (0.362–1.062) 0.081 0.795 (0.443–1.428) 0.443 
Total T-cell infiltration 0.608 (0.356–1.037) 0.068 0.590 (0.334–1.043) 0.069 
Tbet 0.668 (0.378–1.181) 0.165 0.833 (0.446–1.557) 0.567 
CD8/Treg ratio 1.637 (0.956–2.801) 0.072 1.129 (0.631–2.020) 0.683 
Galectin-1 expression by tumor 1.245 (0.731–2.123) 0.420 1.461 (0.800–2.669) 0.217 
Galectin-3 expression by tumor 0.481 (0.275–0.839) 0.010 0.432 (0.239–0.782) 0.006 
Galectin-9 infiltration 0.575 (0.334–0.991) 0.046 0.712 (0.377–1.347) 0.297 
M1 infiltration 0.723 (0.425–1.230) 0.232 0.836 (0.460–1.521) 0.557 
M2 infiltration 1.307 (0.763–2.242) 0.330 1.374 (0.761–2.481) 0.293 
M1/M2 ratio 0.518 (0.297–0.903) 0.020 0.426 (0.227–0.802) 0.008 
CD8 high & Gal3 high 0.316 (0.142–0.704) 0.005 0.278 (0.112–0.686) 0.005 
CD8 high & Gal9 high 0.468 (0.254–0.862) 0.015 0.600 (0.296–1.214) 0.156 
CD8 high & M1/M2 high 0.444 (0.203–0.867) 0.017 0.409 (0.191–0.876) 0.021 
Gal3 high & Gal9 high 0.331 (0.616–0.684) 0.003 0.324 (0.145–0.723) 0.006 
Gal3 high & M1/M2 high 0.339 (0.172–0.668) 0.002 0.289 (0.142–0.589) 0.001 
Gal9 high & M1/M2 high 0.357 (0.717.0.722) 0.004 0.434 (0.212–0.892) 0.023 
3 or more parameters high 0.328 (0.177–0.608) 0.000 0.273 (0.134–0.555) 0.000 

NOTE: Cox regression analyses. Crude HRs and adjusted HRs and the 95% confidence intervals for high versus low numbers of CD4 T cells, CD8 T cells, FoxP3 T cells, total T cells, and Tbet+ cells, high versus low CD8/Treg ratio, high versus low expression of galectin-1 and galectin-3 by the tumor, high versus low numbers of galectin-9+ myeloid cells, M1 macrophages, and M2 macrophages cells, and a high versus low ratio of M1/M2 macrophages since metastasis are shown. HRs for patient groups showing a combination of markers versus all other patients are shown. Significant HRs are depicted in bold an italic numbers.

aAdjusted for age, gender, LDH level, and TNM stage.

Figure 4.

Cell counts of immune infiltrate and expression of galectin-1 and galectin-3 in patients that were treated with ACT. Cell counts (cells/mm2) and the median are depicted of infiltrated immune cells [CD8+ T cells, CD4+ T cells, FoxP3+ T cells, total T cells, M1 (CD14+CD163), and M2 (CD14+CD163+) macrophages], the ratio of M1/M2, and expression (IRS) of galectin-1 and galectin-3 on tumors of patients showing either PD or CB after treatment with ACT. Differences between the two groups were statistically analyzed by a Mann–Whitney U test (*, P < 0.05; **, P < 0.01).

Figure 4.

Cell counts of immune infiltrate and expression of galectin-1 and galectin-3 in patients that were treated with ACT. Cell counts (cells/mm2) and the median are depicted of infiltrated immune cells [CD8+ T cells, CD4+ T cells, FoxP3+ T cells, total T cells, M1 (CD14+CD163), and M2 (CD14+CD163+) macrophages], the ratio of M1/M2, and expression (IRS) of galectin-1 and galectin-3 on tumors of patients showing either PD or CB after treatment with ACT. Differences between the two groups were statistically analyzed by a Mann–Whitney U test (*, P < 0.05; **, P < 0.01).

Close modal

To investigate whether it would be possible to define a certain immune signature for each of the three survival cohorts, the fraction of tumors displaying combinations of immune parameters was plotted (Figs. 3A and 5). This revealed that the fraction of tumors displaying a certain immune signature differed significantly between the three survival cohorts (χ2 test, P < 0.001). The group with the longest survival contained the largest fraction of patients with a tumor that was positive for four beneficial parameters, whereas this immune phenotype was not present in the short survival cohort. We then questioned how many beneficial parameters were required to have a major impact on survival. The group of patients with metastases displaying two or less of the beneficial immune parameters showed a low survival rate. The survival rate increased when patients displayed three or four of the immune parameters, with a 100% survival for patients with a tumor that displayed all four immune parameters (Fig. 3B). In addition, a multivariate Cox analysis was performed with age, gender, LDH level, and TNM stage as covariates. This showed that an immune signature consisting of more than three parameters was an independent prognostic factor for survival (Table 1).

Figure 5.

Immune signature of ACT-treated patients. A, The immune signatures for the patients showing either PD (n = 7) or CB (n = 10) after treatment with ACT. Plotted are the fractions of patients showing tumors that were positive for 0/4, 1/4, 2/4, 3/4, or 4/4 relevant immune parameters. The type of response is depicted for the patients showing CB. B, The correlation between the immune signature and survival was analyzed by the Kaplan–Meier method for the complete group of ACT-treated patients. The patient group that was positive for 3 to 4 of 4 parameters versus the group of patients that was positive for 0 to 2 of 4 parameters was plotted. The depicted P value is from the log-rank test.

Figure 5.

Immune signature of ACT-treated patients. A, The immune signatures for the patients showing either PD (n = 7) or CB (n = 10) after treatment with ACT. Plotted are the fractions of patients showing tumors that were positive for 0/4, 1/4, 2/4, 3/4, or 4/4 relevant immune parameters. The type of response is depicted for the patients showing CB. B, The correlation between the immune signature and survival was analyzed by the Kaplan–Meier method for the complete group of ACT-treated patients. The patient group that was positive for 3 to 4 of 4 parameters versus the group of patients that was positive for 0 to 2 of 4 parameters was plotted. The depicted P value is from the log-rank test.

Close modal

To validate our results, this immune signature was analyzed in a set of stage IV melanoma patients in the TCGA database, which contained 23 patients. In this cohort, patients with a tumor that displayed three or four of the immune parameters had better long-term survival (P < 0.001) compared with the rest of the patients (Fig. 3C).

More patients with a beneficial immune signature among those with CB from ACT

We analyzed 17 pre-ACT treatment biopsies of metastases. Their immune microenvironment showed that the tumors from patients with CB showed higher numbers of intratumoral T cells compared with the patients showing PD (Fig. 4; Supplementary Table S2A). CB patients showed a significantly higher ratio of M1/M2 macrophages, which was mostly the result of a low M2 infiltration (Fig. 4). In addition, the number of intratumoral galectin-9+ DCs/DC-like macrophages and tumors expressing galectin-1 was lower in the CB patients. Next, the fraction of tumors displaying the combination of immune parameters linked to better survival was plotted (Fig. 5A). Thirty percent of the CB patients displayed an immune signature positive for four of our identified beneficial immune parameters, whereas none of the PD patients showed this immune phenotype. Of the 5 CB patients that showed an immune signature of two or less beneficial immune parameters, 4 showed a low infiltrate of CD8+ T cells (Supplementary Table S3), which potentially was corrected by the ACT. Analysis of the effect of this immune signature on the survival after ACT revealed that sustainable CB was only found in those patients whose tumor immune signature included three to four of the identified parameters before treatment with ACT (Fig. 5B).

In this study, we investigated the tumor immune contexture in stage IV metastatic melanoma and analyzed its association to survival. We found that an infiltrate of CD8+ T cells, galectin-9+ DCs or DC-like macrophages, a high M1/M2 ratio, and a high expression of galectin-3 by the tumor were associated with survival. The groups of patients with an immune signature consisting of a combination of three or four immune contexture–defining elements displayed the longest survival. Notably, most of the patients in our cohort were treated in the preimmunotherapy era when the OS generally was low (26). Since the introduction of immunotherapies, the OS of stage IV melanoma patients has increased considerably. Independent validation of our results on a more recent cohort of 23 stage IV melanoma patients with a higher median OS in the TCGA database (5) sustained our observation that patients with tumors that expressed at least three of the defined immune parameters survived longer than patients with tumors not displaying three to four of these immune parameters. Notably, the immune signature is also prognostic for longer survival after ACT treatment and may be predictive for response after ACT, as treatment of stage IV melanoma patients by adoptive T-cell transfer was more likely to be successful and durable when the metastases of these patients displayed at least three of the identified four beneficial immune parameters prior to treatment. However, a firm statement about the predictive value of the immune signature requires validation in a prospectively ACT-treated patient cohort.

Our study clearly showed that not only T cells but multiple elements defining the tumor immune contexture are of importance for the prognosis of stage IV metastatic melanoma patients and their response to ACT. Rather than the numbers of several types of infiltrating immune cells, which did not differ among the short-, medium, and long-term survival groups, it is their combined presence and the processes they reflect that bears an impact on outcome. We showed that there were strong correlations between the Tbet+ lymphocyte counts and CD4 and CD8 T-cell counts, suggesting an ongoing type I cytokine intratumoral T-cell response. The notion of such a proinflammatory signature is sustained by the presence of galectin-9+ myeloid cells, the number of which was strongly correlated to the number of Tbet+ cells and T cells. Galectin-9 is known to be expressed by myeloid cells after exposure to proinflammatory cues in the microenvironment (25, 27). On the basis of the coexpression of CD68 and CD11c, we conclude that these galectin-9+ cells predominantly are DCs and DC-like macrophages that can be crucial for the induction of strong antitumor immunity similar to their published role as crucial mediators of anticancer immune responses after anthracycline-based chemotherapy (28). It is not clear whether galectin-9 is also secreted by these myeloid cells. If so, this could also contribute to the antitumor response, as galectin-9 was shown to increase the numbers and cytolytic capacity of CD8 T cells and natural killer (NK) cells in the Meth-A and B16F10 mouse tumor models (29, 30). The presence and activity of NK cells was not assessed in our study but may form an additional parameter associated with survival, as highly activated NK cells have been found in tumor-infiltrating lymph nodes of melanoma patients (31). In addition, the presence of a proinflammatory environment was reflected by the higher M1 to M2 macrophage ratio found in long-term survivors. Previous studies in melanoma did not find associations between macrophage infiltrate and survival, most likely because these studies did not distinguish between functionally different macrophages and did not relate the presence of macrophages to other immune cells (4, 8, 32). In other types of cancer, however, the presence of proinflammatory (M1) macrophages and, in particular, a high M1 to M2 ratio was shown to be related to improved survival (33, 34).

In our study, a high expression of galectin-3 by the tumor was associated with long-term survival, which was also reported for primary melanoma (12). This is counterintuitive as existing literature mostly describes an immunosuppressive role for galectin-3 (35). Galectin-3 can hamper T-cell activation by interfering with T-cell receptor signaling (36) and by binding to the checkpoint inhibitor LAG-3 (37). In addition, it has been shown to increase apoptosis in effector T cells (38, 39). The positive effect of galectin-3 expression on survival is possibly explained by its direct effect on the tumor cell and might be a result of a higher susceptibility to oxidative damage and apoptotic death of galectin-3–expressing melanoma cells (40).

The presence of an at least three-parameter beneficial immune signature was also found in the patients displaying sustained CB after ACT. The other 5 CB patients, lacking this signature, displayed only short-term benefit from ACT. None of the patients that were unresponsive to ACT showed an immune signature with the presence of all four parameters. We hypothesize that the quality of the ACT T-cell product determines whether a patient responds to therapy. Indeed, T-cell batches that were infused into CB patients comprised tumor-specific T cells with predominantly a Th1 cytokine profile, whereas the T-cell batches that were infused into the patients with PD showed a non-Th1 cytokine profile (22). We speculate that even if good-quality ACT is given to patients whose tumor displays a less desirable immune signature, in particular those showing a failure to recruit large numbers of T cells, only a short period of CB can be achieved. It can be envisaged that an initial failure to recruit T cells, potentially due to a lack of T-cell–attracting chemokines secreted by these tumors (13), will also hamper ACT. Unfortunately, we do not have the tumor material after ACT treatment to test our hypothesis.

Our study is limited by the relatively low number of patients that we could analyze, in particular the patients treated with tumor-reactive T cells and the stage IV melanoma patients available for analysis in the TCGA cohort. Validation of our results in other cohorts will be important to understand the true value of our findings.

We conclude that a tumor immune contexture of at least three of the four parameters described in our study is prognostic to long-term survival of stage IV melanoma patients. Moreover, this same immune signature is required for sustainable responsiveness to ACT of these patients. It would be of interest to validate the predictive value of this immune signature in a prospective cohort of patients treated with ACT as well as to study whether this signature may predict the response to immune checkpoint blockers. If so, the use of this immune signature will be important for personalized therapy, that is, development of tailor-made therapy with higher predicted treatment benefit.

No potential conflicts of interest were disclosed.

Conception and design: S.M. Melief, V.T.H.B.M. Smit, S.H. van der Burg, E.M.E. Verdegaal

Development of methodology: S.M. Melief, V.V. Visconti, E.M.E. Verdegaal

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.M. Melief, E.H.W. Kapiteijn, E.M.E. Verdegaal

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.M. Melief, M. Visser, M. van Diepen, J. Oosting, S.H. van der Burg, E.M.E. Verdegaal

Writing, review, and/or revision of the manuscript: S.M. Melief, M. van Diepen, E.H.W. Kapiteijn, J.H. van den Berg, J.B.A.G. Haanen, V.T.H.B.M. Smit, S.H. van der Burg, E.M.E. Verdegaal

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.M. Melief, M. Visser, J.H. van den Berg

Study supervision: S.H. van der Burg, E.M.E. Verdegaal

We would like to acknowledge the NKI-AVL Core Facility Molecular Pathology & Biobanking (CFMPB) for supplying NKI-AVL Biobank material and/or laboratory support.

This study was supported by the Landsteiner Foundation for Blood Transfusion Research (project number 1207) and the Dutch Cancer Society (KWF UL 2012-5544).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Rosenberg
SA
,
Yannelli
JR
,
Yang
JC
,
Topalian
SL
,
Schwartzentruber
DJ
,
Weber
JS
, et al
Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2
.
J Natl Cancer Inst
1994
;
86
:
1159
66
.
2.
Fridman
WH
,
Pagès
F
,
Sautès-Fridman
C
,
Galon
J
. 
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
2012
;
12
:
298
306
.
3.
Kalialis
LV
,
Drzewiecki
KT
,
Klyver
H
. 
Spontaneous regression of metastases from melanoma: review of the literature
.
Melanoma Res
2009
;
19
:
275
82
.
4.
Erdag
G
,
Schaefer
JT
,
Smolkin
ME
,
Deacon
DH
,
Shea
SM
,
Dengel
LT
, et al
Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma
.
Cancer Res
2012
;
72
:
1070
80
.
5.
Akbani
R
,
Akdemir Kadir
C
,
Aksoy
BA
,
Albert
M
,
Ally
A
,
Amin Samirkumar
B
, et al
Genomic classification of cutaneous melanoma
.
Cell
2015
;
161
:
1681
96
.
6.
Ladanyi
A
. 
Prognostic and predictive significance of immune cells infiltrating cutaneous melanoma
.
Pigment Cell Melanoma Res
2015
;
28
:
490
500
.
7.
Kluger
HM
,
Zito
CR
,
Barr
ML
,
Baine
MK
,
Chiang
VLS
,
Sznol
M
, et al
Characterization of PD-L1 expression and associated T-cell infiltrates in metastatic melanoma samples from variable anatomic sites
.
Clin Cancer Res
2015
;
21
:
3052
60
.
8.
Emri
E
,
Egervari
K
,
Varvolgyi
T
,
Rozsa
D
,
Miko
E
,
Dezso
B
, et al
Correlation among metallothionein expression, intratumoural macrophage infiltration and the risk of metastasis in human cutaneous malignant melanoma
.
J Eur Acad Dermatol Venereol
2013
;
27
:
e320
7
.
9.
Jensen
TO
,
Schmidt
H
,
Moller
HJ
,
Hoyer
M
,
Maniecki
MB
,
Sjoegren
P
, et al
Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma
.
J Clin Oncol
2009
;
27
:
3330
7
.
10.
Ji
RR
,
Chasalow
SD
,
Wang
L
,
Hamid
O
,
Schmidt
H
,
Cogswell
J
, et al
An immune-active tumor microenvironment favors clinical response to ipilimumab
.
Cancer Immunol Immunother
2012
;
61
:
1019
31
.
11.
Tjin
EP
,
Krebbers
G
,
Meijlink
KJ
,
van de Kasteele
W
,
Rosenberg
EH
,
Sanders
J
, et al
Immune-escape markers in relation to clinical outcome of advanced melanoma patients following immunotherapy
.
Cancer Immunol Res
2014
;
2
:
538
46
.
12.
Brown
ER
,
Doig
T
,
Anderson
N
,
Brenn
T
,
Doherty
V
,
Xu
Y
, et al
Association of galectin-3 expression with melanoma progression and prognosis
.
Eur J Cancer
2012
;
48
:
865
74
.
13.
Harlin
H
,
Meng
Y
,
Peterson
AC
,
Zha
Y
,
Tretiakova
M
,
Slingluff
C
, et al
Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment
.
Cancer Res
2009
;
69
:
3077
85
.
14.
Gajewski
T
,
Zha
Y
,
Thurner
B
,
Schuler
G
. 
Association of gene expression profile in metastatic melanoma and survival to a dendritic cell-based vaccine
.
J Clin Oncol
2009
;
27
.
15.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJ
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
16.
Romano
E
,
Kusio-Kobialka
M
,
Foukas
PG
,
Baumgaertner
P
,
Meyer
C
,
Ballabeni
P
, et al
Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients
.
Proc Natl Acad Sci
2015
;
112
:
6140
5
.
17.
Zikich
D
,
Schachter
J
,
Besser
MJ
. 
Predictors of tumor-infiltrating lymphocyte efficacy in melanoma
.
Immunotherapy
2016
;
8
:
35
43
.
18.
Balch
CM
,
Gershenwald
JE
,
Soong
S-j
,
Thompson
JF
,
Atkins
MB
,
Byrd
DR
, et al
Final version of 2009 AJCC melanoma staging and classification
.
J Clin Oncol
2009
;
27
:
6199
206
.
19.
van Esch
EM
,
van Poelgeest
MI
,
Kouwenberg
S
,
Osse
EM
,
Trimbos
JB
,
Fleuren
GJ
, et al
Expression of coinhibitory receptors on T cells in the microenvironment of usual vulvar intraepithelial neoplasia is related to proinflammatory effector T cells and an increased recurrence-free survival
.
Int J Cancer
2015
;
136
:
E95
106
.
20.
van Esch
EM
,
van Poelgeest
MI
,
Trimbos
JB
,
Fleuren
GJ
,
Jordanova
ES
,
van der Burg
SH
. 
Intraepithelial macrophage infiltration is related to a high number of regulatory T cells and promotes a progressive course of HPV-induced vulvar neoplasia
.
Int J Cancer
2015
;
136
:
E85
94
.
21.
Specht
E
,
Kaemmerer
D
,
Sanger
J
,
Wirtz
RM
,
Schulz
S
,
Lupp
A
. 
Comparison of immunoreactive score, HER2/neu score and H score for the immunohistochemical evaluation of somatostatin receptors in bronchopulmonary neuroendocrine neoplasms
.
Histopathology
2015
;
67
:
368
77
.
22.
Verdegaal
EM
,
Visser
M
,
Ramwadhdoebe
TH
,
van der Minne
CE
,
van Steijn
JA
,
Kapiteijn
E
, et al
Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha
.
Cancer Immunol Immunother
2011
;
60
:
953
63
.
23.
Donia
M
,
Larsen
SM
,
Met
O
,
Svane
IM
. 
Simplified protocol for clinical-grade tumor-infiltrating lymphocyte manufacturing with use of the Wave bioreactor
.
Cytotherapy
2014
;
16
:
1117
20
.
24.
Sullivan
BM
,
Juedes
A
,
Szabo
SJ
,
von Herrath
M
,
Glimcher
LH
. 
Antigen-driven effector CD8 T cell function regulated by T-bet
.
Proc Natl Acad Sci U S A
2003
;
100
:
15818
23
.
25.
Gieseke
F
,
Kruchen
A
,
Tzaribachev
N
,
Bentzien
F
,
Dominici
M
,
Muller
I
. 
Proinflammatory stimuli induce galectin-9 in human mesenchymal stromal cells to suppress T-cell proliferation
.
Eur J Immunol
2013
;
43
:
2741
9
.
26.
Korn
EL
,
Liu
PY
,
Lee
SJ
,
Chapman
JA
,
Niedzwiecki
D
,
Suman
VJ
, et al
Meta-analysis of phase II cooperative group trials in metastatic stage IV melanoma to determine progression-free and overall survival benchmarks for future phase II trials
.
J Clin Oncol
2008
;
26
:
527
34
.
27.
Imaizumi
T
,
Kumagai
M
,
Sasaki
N
,
Kurotaki
H
,
Mori
F
,
Seki
M
, et al
Interferon-gamma stimulates the expression of galectin-9 in cultured human endothelial cells
.
J Leukoc Biol
2002
;
72
:
486
91
.
28.
Ma
Y
,
Adjemian
S
,
Mattarollo
SR
,
Yamazaki
T
,
Aymeric
L
,
Yang
H
, et al
Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells
.
Immunity
2013
;
38
:
729
41
.
29.
Nobumoto
A
,
Oomizu
S
,
Arikawa
T
,
Katoh
S
,
Nagahara
K
,
Miyake
M
, et al
Galectin-9 expands unique macrophages exhibiting plasmacytoid dendritic cell-like phenotypes that activate NK cells in tumor-bearing mice
.
Clin Immunol
2009
;
130
:
322
30
.
30.
Nagahara
K
,
Arikawa
T
,
Oomizu
S
,
Kontani
K
,
Nobumoto
A
,
Tateno
H
, et al
Galectin-9 increases Tim-3+ dendritic cells and CD8+ T cells and enhances antitumor immunity via galectin-9-Tim-3 interactions
.
J Immunol
2008
;
181
:
7660
9
.
31.
Ali
TH
,
Pisanti
S
,
Ciaglia
E
,
Mortarini
R
,
Anichini
A
,
Garofalo
C
, et al
Enrichment of CD56(dim)KIR + CD57 + highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients
.
Nat Commun
2014
;
5
:
5639
.
32.
Taube
JM
,
Anders
RA
,
Young
GD
,
Xu
H
,
Sharma
R
,
McMiller
TL
, et al
Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape
.
Sci Transl Med
2012
;
4
:
127ra37
.
33.
de Vos van Steenwijk
PJ
,
Ramwadhdoebe
TH
,
Goedemans
R
,
Doorduijn
EM
,
van Ham
JJ
,
Gorter
A
, et al
Tumor-infiltrating CD14-positive myeloid cells and CD8-positive T-cells prolong survival in patients with cervical carcinoma
.
Int J Cancer
2013
;
133
:
2884
94
.
34.
Ohri
CM
,
Shikotra
A
,
Green
RH
,
Waller
DA
,
Bradding
P
. 
Macrophages within NSCLC tumour islets are predominantly of a cytotoxic M1 phenotype associated with extended survival
.
Eur Respir J
2009
;
33
:
118
26
.
35.
Braeuer
RR
,
Shoshan
E
,
Kamiya
T
,
Bar-Eli
M
. 
The sweet and bitter sides of galectins in melanoma progression
.
Pigment Cell Melanoma Res
2012
;
25
:
592
601
.
36.
Demotte
N
,
Wieers
G
,
Van Der Smissen
P
,
Moser
M
,
Schmidt
C
,
Thielemans
K
, et al
A galectin-3 ligand corrects the impaired function of human CD4 and CD8 tumor-infiltrating lymphocytes and favors tumor rejection in mice
.
Cancer Res
2010
;
70
:
7476
88
.
37.
Kouo
T
,
Huang
L
,
Pucsek
AB
,
Cao
M
,
Solt
S
,
Armstrong
T
, et al
Galectin-3 shapes antitumor immune responses by suppressing CD8+ T cells via LAG-3 and inhibiting expansion of plasmacytoid dendritic cells
.
Cancer Immunol Res
2015
;
3
:
412
23
.
38.
Zubieta
MR
,
Furman
D
,
Barrio
M
,
Bravo
AI
,
Domenichini
E
,
Mordoh
J
. 
Galectin-3 expression correlates with apoptosis of tumor-associated lymphocytes in human melanoma biopsies
.
Am J Pathol
2006
;
168
:
1666
75
.
39.
Chen
HY
,
Fermin
A
,
Vardhana
S
,
Weng
IC
,
Lo
KF
,
Chang
EY
, et al
Galectin-3 negatively regulates TCR-mediated CD4+ T-cell activation at the immunological synapse
.
Proc Natl Acad Sci U S A
2009
;
106
:
14496
501
.
40.
Borges
BE
,
Teixeira
VR
,
Appel
MH
,
Steclan
CA
,
Rigo
F
,
Filipak Neto
F
, et al
De novo galectin-3 expression influences the response of melanoma cells to isatin-Schiff base copper (II) complex-induced oxidative stimulus
.
Chem Biol Interact
2013
;
206
:
37
46
.