Purpose: The tumor immune microenvironment plays a crucial role in the development and progression of cancer. Sarcomas are a group of heterogeneous soft tissue malignancies that are often treated with radiotherapy as a part of the treatment concept. There is increasing evidence that radiotherapy leads to alterations in the tumor microenvironment, particularly with respect to the immune infiltrate. This study has been carried out to develop a better understanding of such changes following radiotherapy.

Experimental Design: We retrospectively analyzed the expression of 35 immune response-related genes by quantitative reverse transcription PCR analysis and immunohistochemistry on paired formalin-fixed paraffin-embedded tumor samples from 38 sarcoma patients before and after radiotherapy.

Results: We observed that radiotherapy results in a significant upregulation of several immune effectors and cancer-testis antigens and a concomitant downregulation of immune suppressors, indicating that radiotherapy may support the immune defense in sarcomas.

Conclusions: These novel findings may have implications for the design of therapeutic regimens which exploite the immune system in sarcoma patients by combining standard radiotherapy with immunotherapeutic strategies. Clin Cancer Res; 19(17); 4843–53. ©2013 AACR.

This article is featured in Highlights of This Issue, p. 4545

Translational Relevance

Sarcomas are cancers with low incidence but high mortality, which are often treated with radiotherapy. However, very little is known about the impact of radiotherapy on the quality of the tumor-associated immune infiltrate in human sarcomas. This study set out to investigate such changes using paired tumor samples before and after radiotherapy from a cohort of 38 sarcoma patients. Our findings show that radiotherapy may groom the local tumor microenvironment by promoting an intratumoral immune effector signature in patients who survived for 3 years or more following radiotherapy. Furthermore, radiotherapy resulted in upregulated expression of MHC class I molecules and cancer-testis antigen. This knowledge argues for combining radiotherapy with immunotherapy as a novel modality for the treatment of sarcoma.

Sarcomas are a heterogeneous group of malignant tumors of mesenchymal origin. They can arise anywhere in the bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue (1). Sarcomas comprise 1% of all adult cancers and about 15% of all childhood cancers there, with high mortality and metastatic potential (2). There is clear evidence from patients and preclinical models that the immune system can control the development of malignancies, a concept termed tumor immunoediting and immunosurveillance (3). Moreover, spontaneous tumor-specific immunity can be detected in cancer patients and in tumor-bearing mice (4, 5). The targets of such responses are usually tumor-associated antigens, of which the group of cancer-testis (CT) antigens is a prominent representative (6). Cancer-testis antigens are especially interesting because they are highly immunogenic and as their name suggests, their expression is restricted to germ cells and cancer cells. More than 100 cancer-testis genes are known to date (7, 8). However, when tumor-specific immunity fails to eliminate cancer, it has proven very difficult to boost this response such that tumor control is achieved. The adaptive arm of the immune response consists of T and B lymphocytes, including antibodies, CD8+ T cells that recognize tumor antigens presented by MHC class I (MHC-I) molecules on tumor cells (9) are thought to be major effectors. Natural killer (NK) cells, which represent the innate immune system, may also mediate antitumor effects by direct cytotoxicity toward malignant cells that express low levels of MHC-I and by producing immunostimulatory cytokines such as IFN-γ (10). Immune effector functions, however, are hampered by an immunosuppressive environment, which is created by the tumor and to which FoxP3+ regulatory T cells as well as myeloid cells contribute through a variety of mechanisms including TGF-β, arginase-1, and interleukin-10 (IL-10; refs. 11–13). The tumor microenvironment presumably significantly influences the outcome of conventional cancer therapies (14, 15). Human sarcomas are treated by radiotherapy, chemotherapy, and/or surgery (16). Recent evidence suggests that both radio- and chemotherapy result in the mobilization of innate and adaptive immunity (17–19), but very little is known about the impact of radiotherapy on the immune infiltrate in human sarcomas. We investigated the effect of radiotherapy on the tumor microenvironment in sarcoma patients using paired biopsies that were collected before and after radiotherapy from 38 sarcoma patients. Upon radiotherapy we found an increase of many cell types and molecules that are characteristic for protective immunity, whereas those associated with immunoregulation were often downregulated. Furthermore, we found a relatively high expression of immune effectors and low expression of immune suppressors after radiotherapy in those patients who survived for 3 years or more after radiotherapy in comparison to those who survived for less than 3 years and died. Our results justify further investigating whether a combination of radiotherapy and immune stimulation results in a better clinical outcome than either therapy alone.

Patient material

Formalin-fixed, paraffin-embedded (FFPE) paired tumor samples were collected from a cohort of 38 sarcoma patients before and after radiotherapy. Tumor samples were retrieved from the archives of the Department of Pathology, University Hospital Zurich, Switzerland. The samples after radiotherapy were obtained from planned resection and the time points of collection following radiotherapy varied between 1 and 89 days (Supplementary Table S1). Patients with nonmetastatic disease and only those patients who received neo-adjuvant radiotherapy before surgery as a part of their standard treatment regimen were included in the study. The patients were treated by standard photon-radiotherapy and received a cumulative dose of 40 to 60 Gy (2 Gy per treatment). An overview of the sarcoma subtypes, cumulative radiation dose, percentage of dead cells following radiotherapy and clinical outcome is given in Table 1. The ethics committee “Ethical Committee of the Canton of Zurich” specifically approved this study (EK-2011-0224/0). For this study, all relevant data were retrieved from the computerized database of the Department of Pathology, University Hospital Zurich, and transferred into a separate anonymous database. Fifteen patient samples from our previous work overlapped in this study (17), but were specifically approved under the study (EK-2011-0224/0). All patients signed the informed consent form in accordance with the declaration of Helsinki and were given a unique code, thus protecting the patient identity.

Table 1.

Patient information

N
Diagnosis 
Angiosarcoma 
Leiomyosarcoma 
MPNST 
Myxofibrosarcoma 
Myxoid liposarcoma 10 
Pleomorphic liposarcoma 
Pleomorphic sarcoma NOS 10 
Pleomorphic sarcoma with Rhabdomyosarcoma 
Rhabdomyosarcoma 
Synovial sarcoma 
Sclerosing epithelial fibrosarcoma 
Spindle cell sarcoma with pleomorphic Liposarcoma 
RT dose (Gy) 
60 
50 25 
48 
46.4 
40 
Unknown 
% cells alive following RT 
<5 
5—10 
11—20 
21—30 
31—40 
41—50 
51—60 
61—70 
71—80 
81—90 
> 90 
Unknown 
Survival status following RT 
Alive 20 
Dead 10 
Unknown 
N
Diagnosis 
Angiosarcoma 
Leiomyosarcoma 
MPNST 
Myxofibrosarcoma 
Myxoid liposarcoma 10 
Pleomorphic liposarcoma 
Pleomorphic sarcoma NOS 10 
Pleomorphic sarcoma with Rhabdomyosarcoma 
Rhabdomyosarcoma 
Synovial sarcoma 
Sclerosing epithelial fibrosarcoma 
Spindle cell sarcoma with pleomorphic Liposarcoma 
RT dose (Gy) 
60 
50 25 
48 
46.4 
40 
Unknown 
% cells alive following RT 
<5 
5—10 
11—20 
21—30 
31—40 
41—50 
51—60 
61—70 
71—80 
81—90 
> 90 
Unknown 
Survival status following RT 
Alive 20 
Dead 10 
Unknown 

NOTE: The table summarizes patient diagnosis, cumulative dose of radiation, percentage of cells alive following radiotherapy, and the survival status. All patients received only radiotherapy (RT) and underwent surgery as part of their standard treatment regimen.

Abbreviations: MPNST, malignant peripheral nerve sheath tumor; N, number of patients; NOS, not otherwise specified.

RNA isolation and reverse transcription

Three punches (0.6-mm diameter) were randomly taken from each of the FFPE blocks using a tissue arrayer MTA-1 (Beecher Instruments). Deparaffinization was conducted in 300 μL elution buffer [1 M Tris pH 8, 0.5 M EDTA pH 8, 20% SDS (all Ambion), ultrapure water (Sigma)] at 95°C for 10 minutes on a shaker followed by centrifugation at 4°C, 14,000 rpm for 10 minutes, followed by digestion with 3 μL of Proteinase K (18 ± 4 mg/mL; Roche) at 55°C for 72 hours. The samples were then centrifuged at 4°C, 14,000 rpm for 2 minutes. The supernatants (250 μL) were transferred into fresh tubes and 750 μL TriZol LS reagent (Invitrogen) was added to each sample while vortexing followed by homogenization by centrifugation at 4°C, 14,000 rpm for 2 minutes using QIA-shredder columns. This was followed by RNA purification using phenol and chloroform. Two hundred microliters of chloroform was added to each flow-through with mild vortexing, followed by incubation at room temperature for 5 minutes until the 2 phases separated, followed by centrifugation at 4°C, 14,000 rpm for 15 minutes. The upper aqueous phase containing the RNA was transferred into a new tube, 20 μg glycogen (Invitrogen) was added followed by the precipitation of RNA by adding 0.5 mL isopropanol [99.9% (v/v); Kantonsapotheke Zürich]. Samples were incubated at room temperature for 15 minutes and then centrifuged at 4°C, 14,000 rpm for 20 minutes. The supernatant was removed and the pellet was washed with 1 mL 75% ethanol (Merck), air-dried, dissolved in Rnase-free water (Sigma), and digested with 80 U/mL DNAse I (New England Biolabs) at room temperature for 15 minutes, followed by inactivation using 2 mmol/L EDTA (Ambion) at 65°C for 10 minutes. The concentration and purity was evaluated using the NanoDrop ND-1000 spectrophotometer (NanoDrop Technologies). Three hundred nanograms of RNA was reverse transcribed using the high-capacity cDNA Reverse Transcription Kit (Applied Biosciences). The cDNA was either used immediately for quantitative reverse transcription PCR (qRT-PCR) reactions or stored at −20°C until use. Because of limited material availability the cDNA was preamplified. Preamplification was conducted for 14 cycles using TaqMan PreAmp Master Mix Kit (Applied Biosystems). All kits were used according to the manufacturer's instructions.

Real-time quantitative reverse transcription-PCR

The expression of target genes was analyzed using TaqMan gene expression assays containing commercially available predeveloped TaqMan reagents with optimized primer and probe concentrations and TaqMan 1× universal master mix (Applied Biosystems; Supplementary Table S2), on a RotoGene Q real-time PCR cycler (Qiagen). The reaction mixture (10 μL) consisted of 1 μL cDNA, 3.5 μL water, 0.5 μL primer, and 5 μL TaqMan 1× universal master mix. The following cycle conditions were used: initial hold for 2 minutes at 50°C and 10 minutes at 95°C, the probes were cycled 45 times at 95°C for 15 seconds and at 60°C for 1 minute. All reactions were conducted in at least triplicates. Threshold cycle (Ct) values were determined using the Rotor-Gene Q Series software 1.7. ΔCt levels of the transcripts were calculated by normalization to the endogenous control (β-actin). The change in expression (ΔΔCt) levels of the target genes following radiotherapy is given as the fold change in expression (2− (ΔΔCt)) and is determined as (ΔCt before radiotherapy − ΔCt after radiotherapy). Ct values >38 cycles were interpreted as negative for gene expression. Fold changes in the range of a fold increase of >0.5 and <2 was considered as not significant. This range is marked as shaded area in the figures.

Immunohistochemistry

Immunohistochemistry (IHC) was conducted on FFPE-paired tissue sections obtained from a cohort of 37 to 38 sarcoma patients. Sections were stained with mouse antihuman monoclonal antibodies against CD4 (clone 1F6, 1:30; ZYMED Laboratories Inc.), CD8 (clone C8/114B, 1:100; DAKO A/S), MHC-I (clone C21, 1:1000; RDI Research Diagnostics, Inc.), CD3 (clone F7.2.38, 1:50; Dako), CT7 (clone CT7-33, 1:80; DAKO A/S), NY-ESO-1 (clone E978, 1:50; ZYMED). Sections were counterstained with hematoxylin, dehydrated, and mounted. All sections were processed with the Ventana Benchmark automated staining system (Ventana Medical Systems) using Ventana reagents for NY-ESO-1, CT7, CD4, CD8, and CD3 and the BondMax (Vison BioSystems) for MHC-I. UView (Ventana) or Refine DAB (Vision BioSystems) were used as chromogens against the primary antibodies. The slides were scanned on a NDP.view2 software (Hamamatsu), and analyzed using the IHC analysis software (NDP.analyze; Hamamatsu). The fields were observed at a magnification of 200× (field of view = 493.1 × 368.5 μm−1). The IHC analysis was conducted by counting 10 random fields. The staining was scored on a scale of 0 to 5 for the expression of NY-ESO-1 and MAGE-C1/CT7 as a percentage, based on the number of positive cells expressing the antigen to the total number of cells in a high-power field (HPF). The tumor infiltrates were calculated as the number of cells expressing CD3, CD4, and CD8 per HPF. The MHC-I sections were acquired on Zeiss-Axiovert 200M (Carl Zeiss Light Microscopy) inverse microscope using Carl Zeiss Axiovision CD28 imaging system. The scoring for MHC-I expression was done based on the intensity of staining (details in Supplementary Table S3). Two individuals conducted the scoring blindly and independently for MHC-I and 1 individual conducted the scoring blindly for the other stainings. The IHC staining was interpreted in conjunction with a single hematoxylin and eosin (H&E) stained slide by a pathologist (B. Bode). The malignant nature of the tumors for the IHC evaluation and qRT-PCR analysis was apparent from the H&E-stained slides.

Statistical analysis and heatmaps

Correlations between the different immune response related transcripts was assessed using the Spearman's rho correlation coefficient (ρ) and the significance two-tailed analysis (P value) was used to analyze the statistical significance (IBM SPSS statistics software). The dot plots for all significant correlations were generated using the Graphpad-Prism software. Statistical significance was defined as P < 0.05.

For visualizing the gene expression profile in the sarcoma patients following radiotherapy, heatmaps were generated using software: R, Bioconductor library “gplots,” heatmap.2 software. Subtracting the mean expression value across patients centered the values for each transcript. No gene-specific scaling (standardization) was done, so that the information about the relative signal strength between probes remained intact. The color tone in the heatmaps was calibrated, so that saturated red (upregulation) and saturated green (downregulation) were reached at values equal to 3 times the standard deviation of the expression values of the entire matrix. Heatmaps were created for all the patients using the ΔΔCt values (ΔCt before radiotherapy − ΔCt after radiotherapy). For the purpose of clustering the genes, the de novo expression was assigned a ΔΔCt value of −10 and the loss of expression was assigned a ΔΔCt value of +10. A hierarchical clustering method using software: R, Bioconductor library “gplots,” heatmap.2 software was used to group genes on the basis of similar expression patterns, in relation to their survival status (green indicates similar behavior of genes and red indicates the reverse). Statistical analysis was conducted for each transcript between the cohort before and after radiotherapy using the two-tailed bivariate analysis (P value; Graphpad-Prism) on the ΔCt values after normalization to β-actin.

Radiotherapy results in upregulation of transcripts of several immune effector molecules and CT-antigens and downregulation of transcripts associated with immune suppression in sarcoma patients

To understand the immunomodulatory effects of radiotherapy in sarcomas, we retrospectively analyzed the expression of several immune response-related transcripts and CT-antigens (Supplementary Table S2) by qRT-PCR analysis on paired FFPE tumor tissues from 37 sarcoma patients before and after radiotherapy (Table 1). We observed that radiotherapy resulted in upregulated transcripts specific for several immune effector cells or proteins including CD45, CD3, CD4, CD8, MHC-I, MHC-II, perforin, granzyme B, IFN-γ, IL-12, and NKG2D in most patients. In addition, we observed de novo expression of perforin, NKG2D, and iNOS in 2 or more patients. TNF-α transcripts were upregulated in several patients but downregulated in others. Concomitantly, the expression of various transcripts related to immune suppression (FoxP3, CD68, CD163, CTLA-4, arginase-1, IDO, PD-1, PD-L1, IL-10, TGF-β, and BTLA) was downregulated in some patients but upregulated or unchanged in others. At least in one of the patients the expression of FoxP3, PD-1, CTLA-4, BTLA, or IL-10 was lost following radiotherapy (Fig. 1A and B, Table 2). In addition, radiotherapy resulted in a substantial upregulation or de novo expression of one or more CT-antigens (CT7, LAGE-A1, MAGE-A9, and NY-ESO-1; Fig. 1C, Table 2). As the fold change in expression in terms of de novo or loss of gene expression cannot be computed, the grand mean was calculated excluding these values. Our data suggest that radiotherapy results in a favorable upregulation of transcripts associated with effector function when compared to those associated with immune suppression, therefore indicating that radiotherapy may presumably promotes protective immunity in the tumor (Table 2 and Supplementary Fig. S4).

Figure 1.

Radiotherapy-induced changes of immune response-related gene expression in human sarcoma. Thirty-seven paired FFPE sarcoma samples before/after radiotherapy were subjected to qRT-PCR analysis for 35 different immune response-related genes. A, immune cells; B, cytokines; and C, cancer testis antigens. ΔCt levels of the transcripts were calculated by normalization to the endogenous control (β-actin). Results are presented as fold change in expression (2−(ΔΔCt)), which was determined as (ΔCtbefore radiotherapy − ΔCtafter radiotherapy). Genes displayed on the left side of the vertical dotted line are associated with immune effectors and those on the right with immune suppressors. Fold differences in expression within the shaded area are considered as not significant. The bold horizontal line for each transcript represents the grand mean. Each symbol represents an individual patient.

Figure 1.

Radiotherapy-induced changes of immune response-related gene expression in human sarcoma. Thirty-seven paired FFPE sarcoma samples before/after radiotherapy were subjected to qRT-PCR analysis for 35 different immune response-related genes. A, immune cells; B, cytokines; and C, cancer testis antigens. ΔCt levels of the transcripts were calculated by normalization to the endogenous control (β-actin). Results are presented as fold change in expression (2−(ΔΔCt)), which was determined as (ΔCtbefore radiotherapy − ΔCtafter radiotherapy). Genes displayed on the left side of the vertical dotted line are associated with immune effectors and those on the right with immune suppressors. Fold differences in expression within the shaded area are considered as not significant. The bold horizontal line for each transcript represents the grand mean. Each symbol represents an individual patient.

Close modal
Table 2.

Summary of radiotherapy-induced changes in immune response-related transcripts in human sarcomas

Upregulation (N)%Downregulation (N)%De novo (N)%Loss of expression (N)%No change (N)%ND (N)%N
Immune cells 
 CD45 19 51.4 8.1 0.0 0.0 15 40.5 0.0 37 
 CD3 19 51.4 8.1 0.0 0.0 15 40.5 0.0 37 
 CD4 21 56.8 8.1 0.0 0.0 13 35.1 0.0 37 
 CD8 18 48.6 5.4 0.0 0.0 17 45.9 0.0 37 
 MHC-I 20 54.1 0.0 0.0 0.0 17 45.9 0.0 37 
 MHC-II 19 51.4 5.4 0.0 0.0 16 43.2 0.0 37 
 β2 Microglobulin 20 54.1 5.4 0.0 0.0 15 40.5 0.0 37 
 Perforin 21 56.8 10.8 5.4 0.0 24.3 2.7 37 
 Granzyme B 13 35.1 10.8 0.0 0.0 24.3 11 29.7 37 
 iNOS 16.2 21.6 5.4 0.0 16.2 15 40.5 37 
 NKG2D 16 43.2 10.8 8.1 0.0 10 27.0 10.8 37 
 Arginase 1 10.8 10.8 0.0 0.0 8.1 26 70.3 37 
 BTLA 2.7 5.4 0.0 2.7 12 32.4 21 56.8 37 
 CTLA-4 5.4 10.8 0.0 2.7 18.9 23 62.2 37 
 FOXP3 13.5 13 35.1 0.0 2.7 10 27.0 21.6 37 
 IDO 10 27.0 21.6 0.0 0.0 18.9 12 32.4 37 
 PD-1 18.9 16.2 0.0 2.7 16.2 17 45.9 37 
 PD-L1 18.9 16.2 0.0 0.0 16 43.2 21.6 37 
Cytokines 
 IFN-Y 11 29.7 0.0 13.5 0.0 21.6 13 35.1 37 
 TNF-α 11 29.7 21.6 0.0 0.0 16 43.2 5.4 37 
 IL-12 13.5 2.7 5.4 0.0 13.5 24 64.9 37 
 IL-10 10.8 11 29.7 0.0 2.7 15 40.5 16.2 37 
 TGF-β 15 40.5 10 27.0 0.0 0.0 11 29.7 2.7 37 
Transcription factors 
 STAT4 12 32.4 10.8 0.0 0.0 15 40.5 16.2 37 
 STAT6 15 40.5 10.8 0.0 0.0 15 40.5 8.1 37 
 STAT3 15 40.5 13.5 0.0 0.0 17 45.9 0.0 37 
CT-antigens 
 CT7 11 29.7 2.7 0.0 0.0 8.1 17 45.9 37 
 CT10 13.5 2.7 0.0 0.0 5.4 29 78.4 37 
 LAGE-A1 16 43.2 0.0 0.0 0.0 5.4 18 48.6 37 
 MAGE-A9 10 27.0 0.0 0.0 0.0 8.1 23 62.2 37 
 NY-ESO-1 20 54.1 10.8 0.0 0.0 24.3 8.1 37 
 PRAME 18 48.6 13.5 0.0 0.0 11 29.7 8.1 37 
 SSX-2 0.0 0.0 0.0 0.0 5.4 35 94.6 37 
Upregulation (N)%Downregulation (N)%De novo (N)%Loss of expression (N)%No change (N)%ND (N)%N
Immune cells 
 CD45 19 51.4 8.1 0.0 0.0 15 40.5 0.0 37 
 CD3 19 51.4 8.1 0.0 0.0 15 40.5 0.0 37 
 CD4 21 56.8 8.1 0.0 0.0 13 35.1 0.0 37 
 CD8 18 48.6 5.4 0.0 0.0 17 45.9 0.0 37 
 MHC-I 20 54.1 0.0 0.0 0.0 17 45.9 0.0 37 
 MHC-II 19 51.4 5.4 0.0 0.0 16 43.2 0.0 37 
 β2 Microglobulin 20 54.1 5.4 0.0 0.0 15 40.5 0.0 37 
 Perforin 21 56.8 10.8 5.4 0.0 24.3 2.7 37 
 Granzyme B 13 35.1 10.8 0.0 0.0 24.3 11 29.7 37 
 iNOS 16.2 21.6 5.4 0.0 16.2 15 40.5 37 
 NKG2D 16 43.2 10.8 8.1 0.0 10 27.0 10.8 37 
 Arginase 1 10.8 10.8 0.0 0.0 8.1 26 70.3 37 
 BTLA 2.7 5.4 0.0 2.7 12 32.4 21 56.8 37 
 CTLA-4 5.4 10.8 0.0 2.7 18.9 23 62.2 37 
 FOXP3 13.5 13 35.1 0.0 2.7 10 27.0 21.6 37 
 IDO 10 27.0 21.6 0.0 0.0 18.9 12 32.4 37 
 PD-1 18.9 16.2 0.0 2.7 16.2 17 45.9 37 
 PD-L1 18.9 16.2 0.0 0.0 16 43.2 21.6 37 
Cytokines 
 IFN-Y 11 29.7 0.0 13.5 0.0 21.6 13 35.1 37 
 TNF-α 11 29.7 21.6 0.0 0.0 16 43.2 5.4 37 
 IL-12 13.5 2.7 5.4 0.0 13.5 24 64.9 37 
 IL-10 10.8 11 29.7 0.0 2.7 15 40.5 16.2 37 
 TGF-β 15 40.5 10 27.0 0.0 0.0 11 29.7 2.7 37 
Transcription factors 
 STAT4 12 32.4 10.8 0.0 0.0 15 40.5 16.2 37 
 STAT6 15 40.5 10.8 0.0 0.0 15 40.5 8.1 37 
 STAT3 15 40.5 13.5 0.0 0.0 17 45.9 0.0 37 
CT-antigens 
 CT7 11 29.7 2.7 0.0 0.0 8.1 17 45.9 37 
 CT10 13.5 2.7 0.0 0.0 5.4 29 78.4 37 
 LAGE-A1 16 43.2 0.0 0.0 0.0 5.4 18 48.6 37 
 MAGE-A9 10 27.0 0.0 0.0 0.0 8.1 23 62.2 37 
 NY-ESO-1 20 54.1 10.8 0.0 0.0 24.3 8.1 37 
 PRAME 18 48.6 13.5 0.0 0.0 11 29.7 8.1 37 
 SSX-2 0.0 0.0 0.0 0.0 5.4 35 94.6 37 

NOTE: A retrospective qRT-PCR analysis of transcripts for (A) immune cells (B) cytokines (C) transcription factors and (D) CT-antigens.

Abbreviations: N, number of patients; ND, not determined.

Radiotherapy results in increased expression of CT-antigens, MHC-I, and in lymphocyte infiltration at the protein level

To validate our findings at the protein level we compared the expression of CT-antigens, MHC-I as well as the infiltration of lymphocytes by immunohistochemistry in 37 to 38 paired FFPE sections obtained from sarcoma patients before and after radiotherapy. Radiotherapy upregulated the expression of MHC-I in 30 of 38 patients (Fig. 2D) and resulted in an increased infiltration by CD4+ and CD8+ T lymphocytes (Fig. 2A–C, Table 3). Furthermore, radiotherapy resulted in a substantial upregulation or de novo expression of the CT-antigens NY-ESO-1 in 13 of 38 patients (Fig. 2E, Table 3) and of CT7 in 17 of 38 patients (Fig. 2F, Table 3). Interestingly, most of the patients with an upregulated expression of one or more CT-antigens had a concomitant upregulation of MHC-I and/or increased infiltration of T cells (patient number: 2, 3, 5, 7, 8, 9,10, 11, 15, 17,19, 20, 22, 23 25, 27, 31, 33, 34, 35, and 36), which approximately comprises 55.3% of the patients. Those changes following radiotherapy were significant when compared to the untreated samples (Supplementary Fig. S5). Because of limited availability of material, we could not conduct staining for additional CT-antigens. We know from our previous work, that the expression of CT-antigens is upregulated upon irradiation in different tumor types (17). Representative sections (Fig. 2G) illustrate that there is a difference in the degree of radiotherapy-induced changes between individual sarcoma patients. A particular gene may be homogeneously expressed after radiotherapy (lower panels), whereas its expression may be upregulated, but still heterogeneous in others (upper panels). The concomitant upregulated expression of MHC-I and CT-antigens upon radiotherapy may increase the visibility of the tumor for CD8+ T cells.

Figure 2.

Radiotherapy of human sarcoma results in increased expression of cancer testis antigens and MHC-I and promotes infiltration by lymphocytes. Paired FFPE tissue sections from sarcoma patients (n = 37–38) before and after radiotherapy were analyzed by IHC for (A) expression of MHC-I, infiltration of (B) CD3+, (C) CD4+, (D) CD8+ cells, expression of (E) NY-ESO-1 and (F) CT7. Scores of individual patients before (open symbols) and after (closed symbols) radiotherapy are connected with a line. Details to scoring are described in Materials and Methods and are listed in Supplementary Table S3. G, representative staining of paired samples before (NR) and after (RT) radiotherapy for MHC-I, CD3, CD4, CD8, CT7, and NY-ESO-1.

Figure 2.

Radiotherapy of human sarcoma results in increased expression of cancer testis antigens and MHC-I and promotes infiltration by lymphocytes. Paired FFPE tissue sections from sarcoma patients (n = 37–38) before and after radiotherapy were analyzed by IHC for (A) expression of MHC-I, infiltration of (B) CD3+, (C) CD4+, (D) CD8+ cells, expression of (E) NY-ESO-1 and (F) CT7. Scores of individual patients before (open symbols) and after (closed symbols) radiotherapy are connected with a line. Details to scoring are described in Materials and Methods and are listed in Supplementary Table S3. G, representative staining of paired samples before (NR) and after (RT) radiotherapy for MHC-I, CD3, CD4, CD8, CT7, and NY-ESO-1.

Close modal
Table 3.

Summary of radiotherapy-induced changes in infiltration by T cells and expression of MHC-I, CT7, and NY-ESO-1 in human sarcomas

Upregulation (N)%Downregulation (N)%De novo expression (N)%Loss of expression (N)%No change (N)%ND (N)%N
Immune cells 
 CD3 26 70.3 2.7 0.0 0.0 10 27.0 37 
 CD4 34 89.5 5.3 0.0 0.0 5.3 38 
 CD8 26 68.4 2.6 0.0 0.0 11 28.9 38 
MHC-I 30 78.9 0.0 0.0 0.0 21.1 38 
CT-antigens 
 CT7 21.1 2.6 23.7 0.0 21.1 12 31.6 38 
 NY-ESO-1 21.1 2.6 13.2 0.0 21.1 16 42.1 38 
Upregulation (N)%Downregulation (N)%De novo expression (N)%Loss of expression (N)%No change (N)%ND (N)%N
Immune cells 
 CD3 26 70.3 2.7 0.0 0.0 10 27.0 37 
 CD4 34 89.5 5.3 0.0 0.0 5.3 38 
 CD8 26 68.4 2.6 0.0 0.0 11 28.9 38 
MHC-I 30 78.9 0.0 0.0 0.0 21.1 38 
CT-antigens 
 CT7 21.1 2.6 23.7 0.0 21.1 12 31.6 38 
 NY-ESO-1 21.1 2.6 13.2 0.0 21.1 16 42.1 38 

NOTE: IHC analysis of a cohort of paired tumor samples before/after radiotherapy of 37 to 38 sarcoma patients for T cells, MHC-I, CT7, and NY-ESO-1.

Abbreviations: N, number of patients; ND, not determined.

Radiotherapy results in clustered changes in expression of genes associated with immune suppression or immune effector function

To investigate whether the changes we observed upon radiotherapy are part of a coordinated program, we plotted our data as a heatmap by computing the ΔΔCt values of each of the transcripts against the ΔΔCt value of the other transcripts. Transcripts associated with immune suppression including IDO, BTLA, FoxP3, PD-L1, IL-10, TGF-β, STAT-3, CT10, TNF-α, and iNOS seem to follow a similar pattern of downregulated expression after radiotherapy. Furthermore, transcripts associated with immune effector function such as NKG2D, CD45, CD3, CD4, CD8, MHC-II, β2M, perforin, granzyme B, the CT-antigen CT7 and macrophages (CD68, CD163) seem to be upregulated in a coordinated fashion following radiotherapy (Fig. 3A). To determine the significance of correlation between 2 target genes, the ΔΔCt values of each of the target genes were computed against the other using a two-tailed bivariate analysis and the Spearman's ρ correlation coefficient. For mathematical reasons, we excluded patients with de novo expression or loss of expression following radiotherapy from the analysis. We found a positive correlation between several immune effectors and an inverse correlation between the transcripts of several immune effectors and immune suppressors. The scattered dot plots for the significant correlations are represented in Supplementary Fig. S6. The Spearman's ρ correlation coefficient for all the correlations is listed in the Supplementary Table S7.

Figure 3.

Correlation between different immune response-related transcripts following radiotherapy in sarcoma patients. Heatmap showing the correlation pattern of immune response-related transcripts in a cohort of paired 37 sarcoma patients following radiotherapy. The histogram indicates gene expression patterns from similar (green) to opposite (red). The white line demarks genes with a similar pattern of expression.

Figure 3.

Correlation between different immune response-related transcripts following radiotherapy in sarcoma patients. Heatmap showing the correlation pattern of immune response-related transcripts in a cohort of paired 37 sarcoma patients following radiotherapy. The histogram indicates gene expression patterns from similar (green) to opposite (red). The white line demarks genes with a similar pattern of expression.

Close modal

Radiotherapy results in downregulation of genes associated with immune suppression in sarcoma patients with prolonged survival

To correlate the gene expression profile in sarcoma patients following radiotherapy in relation to their survival status, we plotted the change in expression (ΔΔCt) levels of the target genes following radiotherapy in a heatmap. We excluded the CT-antigen SSX-2 from this analysis, because it was expressed in only 2 tumor samples. All patients who survived for 3 or more years after radiotherapy, irrespective of the clinical outcome at the end of the follow up, formed one group. Those who survived for less than 3 years after radiotherapy and were dead at the end of the follow up formed the other group. The heatmap reveals a clear downregulation of genes associated with immune suppression such as arginase-1, BTLA, CTLA-4, IDO, iNOS, PD-1, PD-L1, IL-10, TGF-β, TNF-α, FoxP3, macrophages (CD68, CD163), and CT10 in samples of patients who survived for a period of 3 years or more after radiotherapy, but not in those who died. The immune effector genes followed a more homogenous pattern of expression independent of the survival status following radiotherapy (Fig. 4).

Figure 4.

Correlation between radiotherapy-induced immunological changes in sarcoma patients and their survival status. Heatmap showing the expression pattern of immune response-related transcripts in sarcoma patients following radiotherapy. The histogram indicates expression level (ΔΔCt values) from low (green) to high (red). The patients are divided into 2 groups (alive or dead), depending on the 3-year survival status. Patients who were alive at the end of the follow up but did not reach the 3-year survival mark yet were excluded. The yellow box demarcates a clear difference in gene expression between the alive and dead patients.

Figure 4.

Correlation between radiotherapy-induced immunological changes in sarcoma patients and their survival status. Heatmap showing the expression pattern of immune response-related transcripts in sarcoma patients following radiotherapy. The histogram indicates expression level (ΔΔCt values) from low (green) to high (red). The patients are divided into 2 groups (alive or dead), depending on the 3-year survival status. Patients who were alive at the end of the follow up but did not reach the 3-year survival mark yet were excluded. The yellow box demarcates a clear difference in gene expression between the alive and dead patients.

Close modal

Radiotherapy is an integrated treatment option for sarcoma patients. For considerable time it was thought that radiotherapy may have immunosuppressive effects (20), however evidence from recent animal studies suggests that radiotherapy supports local antitumor immunity (18, 21, 22). This study shows in human sarcoma patients that radiotherapy indeed changes the local tumor microenvironment from immunosuppressive into one that is reminiscent of protective immunity. We compared the immune infiltrate in paired biopsies from 38 human sarcoma patients collected before and after radiotherapy and found that radiotherapy induced remarkable changes, all of which suggest a skewing toward protective effector immunity. The activation of CD8+ T and CD4+ T cells contributes to antitumor immunity (23, 24) and is also known to facilitate the efficacy of radiotherapy (25, 26); this is clearly supported by a higher tumor incidence in immunosuppressed patients (27). We observed a higher infiltration of both CD4+ and CD8+ T cells in most of the sarcoma patients after radiotherapy. In addition, we observed that radiotherapy resulted in an upregulated expression of MHC-I and of different CT-antigens in most sarcoma patients, which makes tumors more visible to CD8+ T cells. These data are in accordance with published data showing that nonlethal doses of radiation induce a dose-dependent increase of MHC-I (28) and CT-antigens resulting in better recognition by specific CD8+ T cells in vitro (17). Importantly, radiotherapy sometimes changed the heterogeneous expression of MHC-I and/or CT-antigens, which is thought to result from immune escape (3, 29), to a homogeneous expression, thus enabling a more complete surveillance of the tumor. In addition to changes that improve immunological visibility of tumor cells, we also detected de novo expression of the highly immunogenic CT-antigens in some patients as a result of radiotherapy; this is well in line with our previous data (17). The observed changes are relevant as the loss of MHC-I expression in the equilibrium phase is thought to contribute to escape from immune control leading to tumor progression (30).

The tumor microenvironment is often immunosuppressive, thereby failing to activate local immune responses (31). Several studies have described the roles of immunosuppressive cytokines such as TGF-β and IL-10 in aiding tumor progression and that of stimulatory cytokines such as TNF-α, IL-12, and IFN-γ in supporting antitumor effects (32–36). We observed a higher expression of immune effector molecules, including perforin, granzyme B, NKG2D, IFN-γ, TNF-α, and IL-12, some of which were only expressed after radiotherapy in particular patients. At the same time many molecules and cell types associated with immunosuppression, including arginase-1, BTLA, CTLA-4, PD-1, PD-L1, IDO, FoxP3, macrophages, TGF-β, and IL-10 were expressed at lower level or even disappeared completely after radiotherapy. In search for immune parameters that predict the response to radiotherapy, we found that radiotherapy-induced downregulation of several immune suppressors correlated with survival status. In addition, the correlation analysis revealed that the transcripts associated with immune suppression (IDO, BTLA, FoxP3, PD-L1, IL-10, TGF-β, and macrophages) have a similar pattern of expression. Also immune effectors such as NKG2D, CD45, CD3, CD4, CD8, MHC-II, β2M, perforin, and granzyme B respond in a similar and presumably coordinated fashion upon radiotherapy. Despite the clinical heterogeneity in the cohort of sarcoma patients in our study, the response to radiotherapy was similar as were the changes in the immune infiltrate.

The apparent immunostimulation of radiotherapy per se argues for combining this therapy with treatments that interfere with immunoregulation and/or enhance immune stimulation. There is evidence in humans and mice that local radiotherapy under CTLA-4 blockade results in tumor regression of nonirradiated tumors as well (abscopal effect; refs. 37, 38). Similar results may be obtained when radiotherapy is combined with blockade of other coinhibitory molecules, such as Tim-3, PD-1, or BTLA (39–41). A recent study has shown that anti-PD-1 antibody produced objective responses, 1 in 4 to 1 in 5 patients with advanced non-small cell lung cancer, melanoma, or renal-cell cancer (42). This obviously is a promising combination especially in the case of micro-metastases. Another option may be combination radiotherapy with the targeted delivery of immunostimulatory cytokines, which has been shown to synergize with chemotherapy in human and murine cancers (43, 44). A previous study reported that a combination of radiotherapy with IL-12 improves both local and distant tumor control compared to either therapy alone (45). In addition, radiotherapy may create a favorable environment for adoptively transferred tumor-specific T cells (18, 46), which argues for combining adoptive T-cell therapy and radiotherapy. Preclinical studies in carcinoembryonic antigen (CEA) transgenic mice and a murine carcinoma cell line transfected with CEA, on combined treatment with low-dose radiation and vaccine expressing CEA and costimulatory molecules, could show a 50% reduction in tumor growth and a massive trafficking of T cells, when compared to either treatment alone (47). A clinical trial in cancer patients, vaccinated with NY-ESO-1 DNA vaccine showed induction of antigen-specific T-cell responses in 93% of vaccinated patients, however this did not lead to a clinical benefit (48). A recent study also showed that NY-ESO-1 OLP vaccine could rapidly induce integrated immune responses (antibody, CD8+, and CD4+) in nearly all vaccinated patients with advanced ovarian cancer, when administered with appropriate adjuvants (49). A phase I clinical trial in HLA-A2+ melanoma patients using immunization with autologous PBMCs pulsed with a MAGE-3 or a Melan-A peptide, increased specific CD8+ T-cell responses, and 6 of 8 patients had evidence of clinical activity (50). These studies show that on vaccination with a relevant CT-antigen the peripheral responses could be boosted in cancer patients, however they clearly do not show the desired antitumor efficacy. Therefore, combining radiotherapy with immune stimulation may support the immune modulating potential of radiotherapy by interfering with the local immunosuppressive milieu. Future trials with a large cohort of patients are required to evaluate the question of clinical outcome and the safety of this combination. This may lead to new concepts in the clinical management of cancer.

M. Okoniewski is employed (other than primary affiliation; e.g., consulting) as a Data integration specialist, bioinformatician in University Hospital Zurich, Department of Neuroimmunology and MS Research. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A. Sharma, A. Knuth, L. von Boehmer, M. van den Broek

Development of methodology: A. Sharma, B. Bode, H. Moch, M. van den Broek

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Sharma, B. Bode, A. Knuth, L. von Boehmer

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Sharma, B. Bode, M. Okoniewski, A. Knuth, L. von Boehmer, M. van den Broek

Writing, review, and/or revision of the manuscript: A. Sharma, H. Moch, A. Knuth, L. von Boehmer, M. van den Broek

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Sharma, B. Bode, H. Moch, A. Knuth, L. von Boehmer

Study supervision: H. Moch, A. Knuth, M. van den Broek

Other: Radiation treatment planning/realization/surveillance and provision of all related RT data of the patients: G. Studer

The authors thank M. Storz, M. Bieri (Department of Pathology, University Hospital Zurich, Switzerland) and S. Behnke (Sophistolab, Eglisau, Switzerland) for expert immunohistochemistry.

This work was supported by the Cancer Research Institute (Cancer Vaccine Collaborative), the Arigon Generalunternehmung AG, Zurich and medalumni Zurich, the Hartmann Müller Foundation, and the Dr. Leopold and Carmen Ellinger Foundation.

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.
Martin
Liberal J
,
Lagares-Tena
L
,
Sainz-Jaspeado
M
,
Mateo-Lozano
S
,
Garcia Del Muro
X
,
Tirado
OM
. 
Targeted therapies in sarcomas: challenging the challenge
.
Sarcoma
2012
;
2012
:
626094
.
2.
Howlader
N
,
Noone
AM
,
Krapcho
M
,
Neyman
N
,
Aminou
R
,
Altekruse
SF
, et al
(
eds
). 
SEER cancer statistics review, 1975–2009 (vintage 2009 populations)
.
Bethesda, MD:
National Cancer Institute
. Available from http://seer.cancer.gov/csr/1975_2009_pops09/sections.html,
based on November 2011 SEER data submission, posted to the SEER web site; April
2012
.
3.
Schreiber
RD
,
Old
LJ
,
Smyth
MJ
. 
Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion
.
Science
2011
;
331
:
1565
70
.
4.
Knuth
A
,
Danowski
B
,
Oettgen
HF
,
Old
LJ
. 
T-cell-mediated cytotoxicity against autologous malignant melanoma: analysis with interleukin 2-dependent T-cell cultures
.
Proc Natl Acad Sci U S A
1984
;
81
:
3511
5
.
5.
van der Bruggen
P
,
Traversari
C
,
Chomez
P
,
Lurquin
C
,
De Plaen
E
,
Van den Eynde
B
, et al
A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma
.
Science
1991
;
254
:
1643
7
.
6.
Parmigiani
RB
,
Bettoni
F
,
Vibranovski
MD
,
Lopes
MH
,
Martins
WK
,
Cunha
IW
, et al
Characterization of a cancer/testis (CT) antigen gene family capable of eliciting humoral response in cancer patients
.
Proc Natl Acad Sci U S A
2006
;
103
:
18066
71
.
7.
Hofmann
O
,
Caballero
OL
,
Stevenson
BJ
,
Chen
YT
,
Cohen
T
,
Chua
R
, et al
Genome-wide analysis of cancer/testis gene expression
.
Proc Natl Acad Sci U S A
2008
;
105
:
20422
7
.
8.
Scanlan
MJ
,
Simpson
AJ
,
Old
LJ
. 
The cancer/testis genes: review, standardization, and commentary
.
Cancer Immun
2004
;
4
:
1
.
9.
Parish
IA
,
Kaech
SM
. 
Diversity in CD8(+) T cell differentiation
.
Curr Opin Immunol
2009
;
21
:
291
7
.
10.
Lanier
LL
. 
NK cell recognition
.
Annu Rev Immunol
2005
;
23
:
225
74
.
11.
Marigo
I
,
Dolcetti
L
,
Serafini
P
,
Zanovello
P
,
Bronte
V
. 
Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells
.
Immunol Rev
2008
;
222
:
162
79
.
12.
Bilate
AM
,
Lafaille
JJ
. 
Induced CD4+Foxp3+ regulatory T cells in immune tolerance
.
Annu Rev Immunol
2012
;
30
:
733
58
.
13.
Fan
H
,
Wang
J
,
Zhou
X
,
Liu
Z
,
Zheng
SG
. 
Induction of antigen-specific immune tolerance by TGF-beta-induced CD4+Foxp3+ regulatory T cells
.
Int J Clin Exp Med
2009
;
2
:
212
20
.
14.
Tan
PH
,
Lota
AS
. 
Interaction of current cancer treatments and the immune system: implications for breast cancer therapeutics
.
Expert Opin Pharmacother
2008
;
9
:
2639
60
.
15.
Zitvogel
L
,
Apetoh
L
,
Ghiringhelli
F
,
Kroemer
G
. 
Immunological aspects of cancer chemotherapy
.
Nat Rev Immunol
2008
;
8
:
59
73
.
16.
Van De Voorde
L
,
Delrue
L
,
van Eijkeren
M
,
De Meerleer
G
. 
Radiotherapy and surgery—an indispensable duo in the treatment of retroperitoneal sarcoma
.
Cancer
2011
;
117
:
4355
64
.
17.
Sharma
A
,
Bode
B
,
Wenger
RH
,
Lehmann
K
,
Sartori
AA
,
Moch
H
, et al
gamma-Radiation promotes immunological recognition of cancer cells through increased expression of cancer-testis antigens in vitro and in vivo
.
PLoS One
2011
;
6
:
e28217
.
18.
Gupta
A
,
Probst
HC
,
Vuong
V
,
Landshammer
A
,
Muth
S
,
Yagita
H
, et al
Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation
.
J Immunol
2012
;
189
:
558
66
.
19.
Gupta
A SA
,
von Boehmer
L
,
Surace
L
,
Knuth
A
,
van den Broek
M
. 
Radiotherapy supports protective tumor-specific immunity
.
Oncoimmunology
2012
;
1
:
21
2
.
20.
Merrick
A
,
Errington
F
,
Milward
K
,
O'Donnell
D
,
Harrington
K
,
Bateman
A
, et al
Immunosuppressive effects of radiation on human dendritic cells: reduced IL-12 production on activation and impairment of naive T-cell priming
.
Br J Cancer
2005
;
92
:
1450
8
.
21.
Demaria
S
,
Bhardwaj
N
,
McBride
WH
,
Formenti
SC
. 
Combining radiotherapy and immunotherapy: a revived partnership
.
Int J Radiat Oncol Biol Phys
2005
;
63
:
655
66
.
22.
Lugade
AA
,
Sorensen
EW
,
Gerber
SA
,
Moran
PJ
,
Frelinger
JG
,
Lord
EM
. 
Radiation-induced IFN-γ production within the tumor microenvironment influences antitumor immunity
.
J Immunol
2008
;
180
:
3132
9
.
23.
Takeshima
T
,
Chamoto
K
,
Wakita
D
,
Ohkuri
T
,
Togashi
Y
,
Shirato
H
, et al
Local radiation therapy inhibits tumor growth through the generation of tumor-specific CTL: its potentiation by combination with Th1 cell therapy
.
Cancer Res
2010
;
70
:
2697
706
.
24.
Gerloni
M
,
Zanetti
M
. 
CD4 T cells in tumor immunity
.
Springer Semin Immunopathol
2005
;
27
:
37
48
.
25.
Rubner
Y
,
Wunderlich
R
,
Rühle
PF
,
Kulzer
L
,
Werthmöller
N
,
Frey
B
, et al
How does ionizing irradiation contribute to the induction of anti-tumor immunity?
Front Oncol
2012
;
2
:
75
.
26.
Burnette
BC
,
Liang
H
,
Lee
Y
,
Chlewicki
L
,
Khodarev
NN
,
Weichselbaum
RR
, et al
The efficacy of radiotherapy relies upon induction of type I interferon-dependent innate and adaptive immunity
.
Cancer Res
2011
;
71
:
2488
96
.
27.
Schulz
TF
. 
Cancer and viral infections in immunocompromised individuals
.
Int J Cancer
2009
;
125
:
1755
63
.
28.
Reits
EA
,
Hodge
JW
,
Herberts
CA
,
Groothuis
TA
,
Chakraborty
M
,
Wansley
EK
, et al
Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy
.
J Exp Med
2006
;
203
:
1259
71
.
29.
Jager
E
,
Ringhoffer
M
,
Altmannsberger
M
,
Arand
M
,
Karbach
J
,
Jager
D
, et al
Immunoselection in vivo: independent loss of MHC class I and melanocyte differentiation antigen expression in metastatic melanoma
.
Int J Cancer
1997
;
71
:
142
7
.
30.
Kim
R
,
Emi
M
,
Tanabe
K
. 
Cancer immunoediting from immune surveillance to immune escape
.
Immunology
2007
;
121
:
1
14
.
31.
Leroy
P
,
Slos
P
,
Homann
H
,
Erbs
P
,
Poitevin
Y
,
Regulier
E
, et al
Cancer immunotherapy by direct in vivo transfer of immunomodulatory genes
.
Res Immunol
1998
;
149
:
681
4
.
32.
Joshi
A
,
Cao
D
. 
TGF-beta signaling, tumor microenvironment and tumor progression: the butterfly effect
.
Front Biosci
2010
;
15
:
180
94
.
33.
Wang
R
,
Lu
M
,
Zhang
J
,
Chen
S
,
Luo
X
,
Qin
Y
, et al
Increased IL-10 mRNA expression in tumor-associated macrophage correlated with late stage of lung cancer
.
J Exp Clin Cancer Res
2011
;
30
:
62
.
34.
van Horssen
R
,
Ten Hagen
TL
,
Eggermont
AM
. 
TNF-alpha in cancer treatment: molecular insights, antitumor effects, and clinical utility
.
Oncologist
2006
;
11
:
397
408
.
35.
Barth
RJ
 Jr
,
Mulé
JJ
,
Spiess
PJ
,
Rosenberg
SA
. 
Interferon gamma and tumor necrosis factor have a role in tumor regressions mediated by murine CD8+ tumor-infiltrating lymphocytes
.
J Exp Med
1991
;
173
:
647
58
.
36.
Papatriantafyllou
M
. 
Tumour immunology: how does IL-12 enhance antitumour responses?
Nat Rev Immunol
2011
;
11
:
801
.
37.
Dewan
MZ
,
Galloway
AE
,
Kawashima
N
,
Dewyngaert
JK
,
Babb
JS
,
Formenti
SC
, et al
Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody
.
Clin Cancer Res
2009
;
15
:
5379
88
.
38.
Stamell
EF
,
Wolchok
JD
,
Gnjatic
S
,
Lee
NY
,
Brownell
I
. 
The abscopal effect associated with a systemic anti-melanoma immune response
.
Int J Radiat Oncol Biol Phys
2013
;
85
:
293
95
.
39.
Wu
YL
,
Liang
J
,
Zhang
W
,
Tanaka
Y
,
Sugiyama
H
. 
Immunotherapies: the blockade of inhibitory signals
.
Int J Biol Sci
2012
;
8
:
1420
30
.
40.
Sierro
SR
,
Donda
A
,
Perret
R
,
Guillaume
P
,
Yagita
H
,
Levy
F
, et al
Combination of lentivector immunization and low-dose chemotherapy or PD-1/PD-L1 blocking primes self-reactive T cells and induces anti-tumor immunity
.
Eur J Immunol
2011
;
41
:
2217
28
.
41.
Ngiow
SF
,
von Scheidt
B
,
Akiba
H
,
Yagita
H
,
Teng
MW
,
Smyth
MJ
. 
Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors
.
Cancer Res
2011
;
71
:
3540
51
.
42.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
43.
Moschetta
M
,
Pretto
F
,
Berndt
A
,
Galler
K
,
Richter
P
,
Bassi
A
, et al
Paclitaxel enhances therapeutic efficacy of the F8-IL2 immunocytokine to EDA-fibronectin-positive metastatic human melanoma xenografts
.
Cancer Res
2012
;
72
:
1814
24
.
44.
Steiner
M
,
Neri
D
. 
Antibody-radionuclide conjugates for cancer therapy: historical considerations and new trends
.
Clin Cancer Res
2011
;
17
:
6406
16
.
45.
Seetharam
S
,
Staba
MJ
,
Schumm
LP
,
Schreiber
K
,
Schreiber
H
,
Kufe
DW
, et al
Enhanced eradication of local and distant tumors by genetically produced interleukin-12 and radiation
.
Int J Oncol
1999
;
15
:
769
73
.
46.
Berrien-Elliott
MM
,
Jackson
SR
,
Meyer
JM
,
Rouskey
CJ
,
Nguyen
TL
,
Yagita
H
, et al
Durable adoptive immunotherapy for leukemia produced by manipulation of multiple regulatory pathways of CD8+ T cell tolerance
.
Cancer Res
2013
;
73
:
605
16
.
47.
Chakraborty
M
,
Abrams
SI
,
Coleman
CN
,
Camphausen
K
,
Schlom
J
,
Hodge
JW
. 
External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing
.
Cancer Res
2004
;
64
:
4328
37
.
48.
Gnjatic
S
,
Altorki
NK
,
Tang
DN
,
Tu
SM
,
Kundra
V
,
Ritter
G
, et al
NY-ESO-1 DNA vaccine induces T-cell responses that are suppressed by regulatory T cells
.
Clin Cancer Res
2009
;
15
:
2130
9
.
49.
Sabbatini
P
,
Tsuji
T
,
Ferran
L
,
Ritter
E
,
Sedrak
C
,
Tuballes
K
, et al
Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients
.
Clin Cancer Res
2012
;
18
:
6497
508
.
50.
Gajewski
TF
,
Fallarino
F
,
Ashikari
A
,
Sherman
M
. 
Immunization of HLA-A2+ melanoma patients with MAGE-3 or MelanA peptide-pulsed autologous peripheral blood mononuclear cells plus recombinant human interleukin 12
.
Clin Cancer Res
2001
;
7
:
895s
901s
.

Supplementary data