Purpose:

The tumor immune microenvironment determines clinical outcome. Whether the original tissue in which a primary tumor develops influences this microenvironment is not well understood.

Experimental Design:

We applied high-dimensional single-cell mass cytometry [Cytometry by Time-Of-Flight (CyTOF)] analysis and functional studies to analyze immune cell populations in human papillomavirus (HPV)–induced primary tumors of the cervix (cervical carcinoma) and oropharynx (oropharyngeal squamous cell carcinoma, OPSCC).

Results:

Despite the same etiology of these tumors, the composition and functionality of their lymphocytic infiltrate substantially differed. Cervical carcinoma displayed a 3-fold lower CD4:CD8 ratio and contained more activated CD8+CD103+CD161+ effector T cells and less CD4+CD161+ effector memory T cells than OPSCC. CD161+ effector cells produced the highest cytokine levels among tumor-specific T cells. Differences in CD4+ T-cell infiltration between cervical carcinoma and OPSCC were reflected in the detection rate of intratumoral HPV-specific CD4+ T cells and in their impact on OPSCC and cervical carcinoma survival. The peripheral blood mononuclear cell composition of these patients, however, was similar.

Conclusions:

The tissue of origin significantly affects the overall shape of the immune infiltrate in primary tumors.

Translational Relevance

The tumor immune contexture determines therapy responsiveness and survival. It develops as a consequence of the interaction between the tumor cells and the host. Although there is accumulating evidence on the contribution of tumor-intrinsic factors, the impact of the tissue microenvironment in which the tumor develops is largely unknown. Our study of the immune cell populations present in human papillomavirus–induced primary tumors of the cervix and oropharynx, 2 tumors that share the same virus-driven oncogenic pathway but arise in different anatomical locations, allowed us to address this question. We show that the microenvironment of the original tissue has an influence on the constitution of the intratumoral lymphocytic infiltrate, the efficiency of tumor-specific T cells to infiltrate the tumor, and as such the response and survival of patients after therapy. These results will fuel the discussion on therapeutic approaches aiming to treat tumors driven by common oncogenic pathways.

The tumor microenvironment is involved in tumorigenesis and tumor progression (1). Analyses of many tumor types revealed that a strong T helper 1 (Th1) cytotoxic microenvironment is associated with a more favorable prognosis and therapy responsiveness (2, 3). When the tumor metastasizes to other anatomical locations, the original immune contexture of the primary tumor is maintained (4), indicating that the immune contexture of the primary tumor determines the therapy response of later metastases (5). However, whether the microenvironment of the original tissue in which the primary tumor arises has an impact on the shape of the tumor microenvironment has largely been neglected. Perhaps because primary tumors of the same type can have different mutations and activated oncogenic pathways, some of which are known to affect the constitution of the tumor microenvironment. For instance, overexpression of BRAF, loss of PTEN, and activation of the WNT/β-catenin signaling pathway modulate the extent of the lymphocytic tumor infiltrate (6–8). In this context, it is hard to dissect the potential influence of the original tissue on the tumor microenvironment. An answer to what extent the immune contexture can be imprinted by the site or origin may come from studies on human papillomavirus (HPV)–induced tumors. Although these tumors can arise in different anatomical locations (cervix, vagina, vulva, anus, and oropharynx), they share the same virus-driven oncogenic pathway.

HPV is strictly epitheliotropic and infects basal epithelial cells in skin and mucous membranes. Integration of the viral genome into the host DNA leads to overexpression of the E6 and E7 oncoproteins and finally to transformation of the epithelial basal cells into cancer cells (9). Within the cervix, the cells close to the squamocolumnar junction, in what is called the transformation zone, are susceptible to HPV infection, transformation, and progression to cancer (10). Notably, within the genital tract it is the normal uterine cervix, including the transformation zone that contains the highest numbers of immune cells (11, 12). The oropharynx comprises the palatine tonsils, the soft palate, the tongue base, and the posterior pharyngeal wall, but HPV almost exclusively infects and transforms the highly specialized lymphoepithelium lining the tonsillar crypts (13). A direct comparison of the lymphocytic infiltrate in routinely removed nondiseased fresh tonsils and cervical tissue revealed differences in the CD4:CD8 T-cell ratio and the distribution of central memory (Tcm) and effector memory (Tem) CD4+ T cells between the 2 tissue types (14). The indication that these 2 issues at different anatomical locations already display a different immune contexture under noncancerous conditions provides an opportunity to assess if the original tissue microenvironment also bears impact on the tumor immune microenvironment.

To study the potential impact of the original tissue on the intratumoral immune contexture, we analyzed immune cell populations in a series of primary tumors, tumor-draining lymph nodes (TDLNs), and peripheral blood mononuclear cell (PBMC) samples, obtained from patients with either HPV-driven cervical carcinoma or oropharyngeal squamous cell carcinoma (OPSCC), by high-dimensional single-cell mass cytometry (CyTOF) and different functional analyses, including the assessment of HPV-specific T cells. Our data show that primary tumors of the same etiology, but arising in different anatomical locations, are infiltrated with distinct lymphocytic populations, and that the anatomical location affects the efficiency of tumor-specific T cells to infiltrate the tumor.

The authors acknowledge the reporting of Minimal Information About T-cell Assays.

Patients

Patients included in this study were part of 2 larger observational studies on cervical carcinoma and OPSCC. Women with histologically proven cervical carcinoma (International Federation of Gynecology and Obstetrics 1a2, 1b1/2) were included in the CIRCLE study investigating cellular immunity against anogenital lesions (15, 16). Patients with histology-confirmed OPSCC were included in the P07-112 study investigating the circulating and local immune response in patients with head and neck cancer (17, 18). Patients were included after signing informed consent. Both studies were conducted in accordance with the Declaration of Helsinki and approved by the local medical ethical committee of the Leiden University Medical Center (LUMC) and in agreement with the Dutch law. The patients received standard-of-care treatment consisting of surgery, radiotherapy, chemotherapy, treatment with monoclonal antibody, or combinations hereof. HPV typing and p16ink4a IHC staining were performed on formalin-fixed paraffin-embedded tumor sections at the LUMC Department of Pathology as described (19). Tumor staging was done according to the National Comprehensive Cancer Network (https://www.nccn.org/professionals). An overview of patient characteristics and treatment is given in Supplementary Table S1.

TP53 mutational status OPSCC tumors

We performed an IHC staining for p53 on the tissue sections of the OPSCC tumors as described previously (20). An experienced pathologist reviewed all slides and scored the specimen as “wild-type” (p53wt) when nuclei of the tumor cells stained weak to moderate, comparable with adjacent normal epithelium.

In addition, next-generation sequencing was performed as described previously (21). In brief, formalin-fixed paraffin-embedded tissue blocks of the OPSCC tumors were microdissected to get a tumor percentage of >70%. Tumor DNA was isolated using the Tissue Preparation System with VERSANT Tissue Preparation Reagents (Siemens Healthcare Diagnostics) after which somatic variant analysis of the TP53 gene was performed using the AmpliSeq Cancer Hotspot Panel 4 (Thermo Fisher Scientific) with a sequence coverage of 90%. Results of this analysis are shown in Supplementary Table S1.

Blood, LN, and tumor cell isolation and culturing

Venous blood samples were drawn prior to surgery, and PBMCs were isolated using Ficoll density gradient centrifugation as described previously (18, 22). Cervical carcinoma and OPSCC tumor and cervical carcinoma TDLN material or biopsies were obtained and handled as described (16, 18). First, tumor material was cut into small pieces. One-third of the tumor pieces were incubated for 60 minutes at 37°C in Iscove's Modified Dulbecco's Medium (IMDM, Lonza) with 10% human AB serum (Capricorn Scientific) and supplemented with high dose of antibiotics [50 μg/mL Gentamycin (Gibco/Thermo Fisher Scientific, TFS), 25 μg/mL Fungizone (Invitrogen/TFS), 100 IU/mL penicillin (pen; Gibco/TFS), and 100 μg/mL streptomycin (strep; Gibco/TFS)], after which the tumor pieces were put in culture in IMDM supplemented with 10% human AB serum, 100 IU/mL penicillin, 100 μg/mL streptomycin, 2 mmol/L l-glutamin (Lonza; IMDM complete), and 1,000 IU/mL human recombinant IL2 (Aldesleukin, Novartis). Cultures were replenished every 2 to 3 days with fresh IMDM complete and IL2 to a final concentration of 1,000 IU/mL. When there were sufficient T cells, the cells were cryopreserved and stored in liquid nitrogen until use. Approximately two-thirds of the tumor pieces were incubated for 30 minutes at 37°C in IMDM dissociation mixture containing 10% human AB serum, high dose of antibiotics (as above) and 50 μg/mL DNAse I (Roche), and 1 mg/mL collagenase D (Roche), after which the tumor was dispersed to single cells using the GentleMACS dissociator (Miltenyi) with a company-installed program (h_tumor_02). Following centrifugation, cells were resuspended in PBS (B. Braun), put on a 70-μm cell strainer (Falcon) to obtain a single-cell suspension, counted using trypan blue exclusion (Sigma), and cryopreserved at approximately 2 million cells/vial. All tumor samples from OPSCC and part of the cervical carcinoma samples were dissociated as above. From patient C1016 onward, the approach to prepare single-cell suspensions was adjusted. The DNAse I/collagenase D enzymes were replaced by 0.38 mg/mL of the commercially available Liberase enzymes (Liberase TL, research grade, Roche), the incubation period was reduced to 15 minutes, and the GentleMACS dissociator was no longer used.

Single-cell suspensions from cervical carcinoma TDLN material were prepared either through DNAse I and collagenase D dissociation as described above with or without the use of the GentleMACS dissociator, or by scraping the cutting surface of the LN with a surgical scalpel blade, after which they were cryopreserved until further analysis. To generate TDLN-derived T-cell batches, ex vivo TDLN were expanded as described previously (22). In brief, ex vivo TDLN were thawed, washed, and seeded at a density of 0.4 to 1 × 10e6 cells per well in a 24-well plate in 1 mL of IMDM complete medium and stimulated with 5 μg/mL HPV16 E6 and E7 clinical grade long peptide pools in the presence of 150 U/mL recombinant human IL2. Cultures were replenished every 2 to 3 days with fresh IMDM complete and IL2, and after 22 to 28 days, the expanded T cells were cryopreserved and stored in the vapor phase of liquid nitrogen until further use.

Mass cytometry analysis

Blood and matched tumor and/or LN samples from 20 patients with cervical carcinoma and 9 patients with OPSCC were analyzed by mass cytometry (CyTOF). Details on antibodies used are listed in Supplementary Table S2. Conjugation of the purified antibodies with the isotopic tags was done using the MaxPar X8 antibody labeling Kit (Fluidigm Sciences) according to the manufacturer's instructions, and following conjugation, the antibodies were stored at 4°C in Candor PBS Antibody Stabilization Buffer (Candor Bioscience GmbH). Mass cytometry antibody staining and acquisition procedures were performed as described (23). In brief, cell samples were thawed according to standard operation procedures and stained with 1 μmol/L Cell-ID intercalator-103Rh (Fluidigm Sciences) for 15 minutes at room temperature (RT) to identify dead cells. Following incubation, the cells were washed with MaxPar Cell stain buffer (Fluidigm Sciences) and incubated with Human TruStain FcX Fc Receptor Blocking Solution (Biolegend) for at least 10 minutes at RT. Next, cells were incubated with the metal-conjugated antibodies for 45 minutes at RT. After the cells were washed twice with the MaxPar Cell stain buffer, the cells were stained overnight at 4°C with 125 nmol/L Cell-ID intercalator-Ir in MaxPar Fix and Perm Buffer (Fluidigm Sciences). The next day, cells were washed and acquired by CyTOF 2 or helios-upgraded CyTOF2 mass cytometer (Fluidigm Sciences). Data were normalized by using EQ Four Element Calibration Beads (Fluidigm Sciences) with the reference EQ passport P13H2302.

Mass cytometry data analysis

Gating for single, live CD45+ cells for each PBMC, LN or tumor sample was done using the cloud-based Cytobank software (Fluidigm Sciences; example in Supplementary Fig. S1). The high-dimensional single-cell data were analyzed by analysis by tSNE analysis in Cytosplore with default parameters (perplexity, 30; iterations, 1,000; ref. 24) and by the fully automated hierarchical clustering (unsupervised) tool CITRUS using the cytobank software. The different cell populations were visualized and quantified.

Tumor-specific T-cell reactivity analysis

To determine the specificity of T cells infiltrating the tumor and/or TDLN, cultured TIL and TDLN T-cell batches from HPV16+ cervical carcinoma and OPSCC tumors were analyzed for the presence of HPV16-specific T cells using 5-day [3H]-thymidine–based proliferation assay as described (18). T-cell responses against autologous HPV16 E6/E7 synthetic long peptide (SLP; 22-mers with 14 amino acids overlap) loaded monocytes were tested in triplicate. PHA (0.5 μg/mL; HA16 Remel; Thermo Fisher Scientific) was taken along as positive control, whereas unloaded monocytes served as negative control. At days 1.5 and 4, supernatant (50 μL/well) was harvested to determine cytokine production. During the last 16 hours of culture, 0.5 μCi/well of [3H]-thymidine was added to measure proliferation. The stimulation index was calculated as the average of test wells divided by the average of the medium control wells. A positive response was defined as a stimulation index of at least 3.

Antigen-specific cytokine production was determined by cytometric bead array (Th1/Th2 kit, BD Bioscience) according to the manufacturer's instructions. The cutoff value for cytokine production was 20 pg/mL, except for IFNγ for which it was 100 pg/mL. Positive cytokine production was defined as at least twice above that of the unstimulated cells.

The HPV16 reactivity of TIL was also determined by intracellular cytokine staining (ICS). ICS for the markers CD3, CD4, CD8, CD137, CD154, CD161, CD103, IFNγ, and TNFα was performed as described previously (18) following stimulation with HPV16 E6/E7 SLP-loaded autologous monocytes or Epstein-Barr virus (EBV)-transformed B-lymphoblastoid cell lines (BLCL). Unloaded autologous monocytes or BLCL cells were used as negative and Staphylococcal Enterotoxin B (SEB; 2 μg/mL; Sigma) was used as positive control. A positive response was defined as at least twice the value of the negative control and at least 10 events in the gate. Acquisition of cells was done on a LSRII Fortessa (BD Biosciences). Data were analyzed using DIVA software (version 6.2 or 8.02 BD Biosciences).

Statistical analysis

Nonparametric (Wilcoxon signed-rank or Mann–Whitney test for 2 samples and Friedman or Kruskal–Wallis with Dunn multiple comparison test for multiple samples) and parametric (paired or unpaired t test for 2 samples or repeated measures one-way ANOVA or ordinary one-way ANOVA with Tukey multiple comparison test for multiple samples) tests were performed as appropriate. For survival analysis, patients were grouped into 2 groups according to the median (i.e., grouped into below or above the median of the total group for each parameter), after which survival was tested using the Kaplan–Meier method, and statistical significance of the survival distribution was analyzed by the log-rank testing. All statistical tests were performed at the 0.05 significance level, and differences were considered significant when P < 0.05, as indicated with an asterisk (#, P < 0.1; *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001). Statistical analyses were performed using GraphPad Prism 7.1.

Cervical tumors but not TDLN are highly infiltrated by CD8+CD103+ T cells

First, the lymphocyte content was studied in a series of tumors, TDLN, and PBMC samples of patients with HPV-induced primary cervical carcinoma (Supplementary Table S1) using 36-parameter mass cytometry (Supplementary Table S2). tSNE analyses of all samples revealed different clusters of B cells, natural killer (NK) cells, and several populations of CD4+ and CD8+ T cells (Fig. 1A and B; Supplementary Fig. S1). Clearly, the naïve (Tn), Tcm, and Tem populations of CD4+ and the Tcm/Tem populations of CD8+ T cells did not differ between PBMC and LN (Fig. 1C). In contrast, the tumor showed a strong enrichment of CD8+ T cells, which were mainly effector T cells (Teff; Fig. 1B and C). Further analysis of the tSNE populations revealed 8 distinctive (groups of) lymphocyte populations (Fig. 1D) and demonstrated that cervical carcinoma TDLN contained more total B cells (both IgM and IgM+ B cells), whereas more NK cells were found in PBMC (Fig. 1D). Strikingly, the percentage of total CD4+ T cells was lower in tumors, albeit that those of CD4+ Treg-like cells was higher. CD8+ T cells were enriched in tumors, in particular the CD8+CD103+ T cells, also known as long-lived tissue-resident memory cells (25). Thus, although the T-cell populations in cervical carcinoma TDLN more or less resembled that of PBMC, cervical carcinoma tumors displayed a more prominent CD8+CD103+ T-cell infiltrate, with the CD8+ T cells in ratio of about 1:1 to CD4+ T cells.

Figure 1.

HPV-induced cervical carcinoma tumors are highly infiltrated by CD8+CD103+ effector T cells. PBMCs, TDLN, and tumors of patients with cervical carcinoma were analyzed by 36-parameter mass cytometry analysis. A, A tSNE density plot (left) and corresponding cluster partitions (right) visualizing the high-dimensional CyTOF data in 2 dimensions for the collective immune cells derived from 20 cervical carcinoma patients' PBMC, TDLN, and tumors (n = 19, n = 12, and n = 9 patients, respectively). The identified cell subsets are indicated in the plots by numbers, and groups of B cells, myeloid cells, NK cells, and CD4+ and CD8+ T cells are specified by different colors. B, Pie charts showing the composition and relative contribution of the immune cells in cervical carcinoma PBMC (left), TDLN (middle), and tumor (right). C, The subdivision of the CD4+ and CD8+ T-cell frequencies (mean ± SEM) into Tn (CCR7+CD45RA+CD127), Tcm (CCR7+CD45RACD127+), and Tem (CCR7CD45RACD127+) T cells for CD4 (left; orange) or into Tn, Tcm/em, effector (Teff; CCR7CD45RACD127), and CD45RA+ effector memory T cells (Temra; CCR7CD45RA+CD127) for CD8 (right; pink). Significant differences in the effector/memory distribution of CD4+ and CD8+ T cells between PBMC, TDLN, and tumors were found. D, Box-and-whiskers (plus min–max) plots depicting 8 distinctive (groups of) lymphocyte populations within cervical carcinoma PBMC, TDLN, and tumor samples. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Figure 1.

HPV-induced cervical carcinoma tumors are highly infiltrated by CD8+CD103+ effector T cells. PBMCs, TDLN, and tumors of patients with cervical carcinoma were analyzed by 36-parameter mass cytometry analysis. A, A tSNE density plot (left) and corresponding cluster partitions (right) visualizing the high-dimensional CyTOF data in 2 dimensions for the collective immune cells derived from 20 cervical carcinoma patients' PBMC, TDLN, and tumors (n = 19, n = 12, and n = 9 patients, respectively). The identified cell subsets are indicated in the plots by numbers, and groups of B cells, myeloid cells, NK cells, and CD4+ and CD8+ T cells are specified by different colors. B, Pie charts showing the composition and relative contribution of the immune cells in cervical carcinoma PBMC (left), TDLN (middle), and tumor (right). C, The subdivision of the CD4+ and CD8+ T-cell frequencies (mean ± SEM) into Tn (CCR7+CD45RA+CD127), Tcm (CCR7+CD45RACD127+), and Tem (CCR7CD45RACD127+) T cells for CD4 (left; orange) or into Tn, Tcm/em, effector (Teff; CCR7CD45RACD127), and CD45RA+ effector memory T cells (Temra; CCR7CD45RA+CD127) for CD8 (right; pink). Significant differences in the effector/memory distribution of CD4+ and CD8+ T cells between PBMC, TDLN, and tumors were found. D, Box-and-whiskers (plus min–max) plots depicting 8 distinctive (groups of) lymphocyte populations within cervical carcinoma PBMC, TDLN, and tumor samples. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Close modal

The populations of tumor-infiltrating lymphocytes differ between cervical carcinoma and OPSCC

Next, the lymphocyte content of the cervical carcinoma samples was compared with a series of tumor and blood samples from patients with HPV-induced OPSCC. Two-dimensional principal component analysis (2D-PCA; Fig. 2A) revealed clear phenotypic diversity between tumor, TDLN, and PBMC samples as well as between the groups of cervical carcinoma and OPSCC tumor samples (Fig. 2A). Combined tSNE analysis of paired blood and tumor samples resulted in the identification of 29 distinct immune populations (Fig. 2B and C). Subsequent comparison of the immune cell content showed that the lymphocytic composition of cervical carcinoma and OPSCC greatly differed with respect to the percentages of IgM+ B cells, CD4+ T cells, and CD8+ T cells (Fig. 2D). The lymphocytic composition of PBMC samples was quite similar for cervical carcinoma and OPSCC (Fig. 2E).

Figure 2.

Cervical carcinoma (CxCa) and OPSCC tumors show different populations of infiltrating lymphocytes. PBMC, TDLN, and tumor samples from patients with HPV-induced CxCa and OPSCC were analyzed by combined tSNE analysis. A, 2D-PCA depicting the collective t-SNE dimensionality reduced cell percentage data (as percentage of CD45+ cells) of 30 subsets for 63 samples (n = 28, n = 17, and n = 18 for PBMC, TDLN, and tumor samples, respectively). Every dot represents a single sample. The color and shape of the sample show the corresponding clinical information (CxCa vs. OPSCC and tumor vs. TDLN vs. PBMC). B, A tSNE density plot (left) and corresponding cluster partitions (right) of collective total CD45+ immune cells of 35 samples (n = 9 for CxCa and OPSCC tumor, n = 9 and n = 8 for CxCa and OPSCC PBMC). The identified cell subsets are indicated in the plots by numbers, and groups of B cells, myeloid cells, NK cells, and CD4+ and CD8+ T cells are specified by different colors. C, Heatmap visualizing the 29 identified cell clusters for the collective total CD45+ immune cells derived from 35 samples. Shown are ArcSinh5-transformed values of marker expression (green to pink scale) and hierarchical clustering of markers and subsets. The groups of NK cells, myeloid cells, B cells, CD4+, and CD8+ T cells are indicated underneath the map. D and E, top, tSNE density plots of collective CxCa (left) and OPSCC (right) total CD45+ immune cells of (D) 18 tumor samples (n = 9 for patients with CxCa and OPSCC) and (E) 17 PBMC samples (n = 9 for patients with CxCa and n = 8 for patients with OPSCC). D and E, bottom, Pie charts showing composition and relative contribution of the identified immune cell subsets in CxCa (left; purple) and OPSCC (right; green) in tumor (D) and PBMC (E).

Figure 2.

Cervical carcinoma (CxCa) and OPSCC tumors show different populations of infiltrating lymphocytes. PBMC, TDLN, and tumor samples from patients with HPV-induced CxCa and OPSCC were analyzed by combined tSNE analysis. A, 2D-PCA depicting the collective t-SNE dimensionality reduced cell percentage data (as percentage of CD45+ cells) of 30 subsets for 63 samples (n = 28, n = 17, and n = 18 for PBMC, TDLN, and tumor samples, respectively). Every dot represents a single sample. The color and shape of the sample show the corresponding clinical information (CxCa vs. OPSCC and tumor vs. TDLN vs. PBMC). B, A tSNE density plot (left) and corresponding cluster partitions (right) of collective total CD45+ immune cells of 35 samples (n = 9 for CxCa and OPSCC tumor, n = 9 and n = 8 for CxCa and OPSCC PBMC). The identified cell subsets are indicated in the plots by numbers, and groups of B cells, myeloid cells, NK cells, and CD4+ and CD8+ T cells are specified by different colors. C, Heatmap visualizing the 29 identified cell clusters for the collective total CD45+ immune cells derived from 35 samples. Shown are ArcSinh5-transformed values of marker expression (green to pink scale) and hierarchical clustering of markers and subsets. The groups of NK cells, myeloid cells, B cells, CD4+, and CD8+ T cells are indicated underneath the map. D and E, top, tSNE density plots of collective CxCa (left) and OPSCC (right) total CD45+ immune cells of (D) 18 tumor samples (n = 9 for patients with CxCa and OPSCC) and (E) 17 PBMC samples (n = 9 for patients with CxCa and n = 8 for patients with OPSCC). D and E, bottom, Pie charts showing composition and relative contribution of the identified immune cell subsets in CxCa (left; purple) and OPSCC (right; green) in tumor (D) and PBMC (E).

Close modal

In order to automatically discover stratifying biological signatures within the OPSCC and cervical carcinoma blood and tumor samples, we made use of the automated and data-driven CITRUS platform, as an unbiased and thorough correlation-based tool for mining and inspection of cell subsets nested within high-dimensional datasets (26). The CITRUS analysis resulted in 30 distinctive (groups of) lymphocyte populations (Fig. 3A and B; Supplementary Fig. S2; and Supplementary Table S3). Closer inspection of the types of lymphocytes in cervical carcinoma and OPSCC revealed that the percentage of IgM+ B cells is higher in OPSCC and comprised 2 different populations (subsets 3 and 4; Fig. 3C) that differed with respect to surface levels of CD27 and HLA-DR (Supplementary Fig. S2). Major differences were observed with respect to infiltrating T cells (Fig. 3E and F; Supplementary Fig. S2; and Supplementary Table S3). This was not biased by the higher frequency of B cells in OPSCC, as similar differences were also observed within the total CD3+ T-cell population (Supplementary Fig. S3A and S3B). Cervical carcinoma displayed a slightly lower infiltration with CD8+ Tcm/Tem (subset 15) and a more dense infiltration of CD8+ effector memory RA+ T cells (Temra; subset 17). In addition, cervical carcinomas were highly infiltrated with CD8+CD103+CD161 Teff (subsets 8 and 11) and CD8+CD103+CD161+ Teff (subsets 9 and 10) cells. Subset 8 Teff cells (CD27-HLADR-CD38dimPD1-) seem less activated based on their profile. A fifth population of CD8+CD103+ Teff (subset 12) and a population of CD8+ Teff, expressing intermediate levels of CD103, HLA-DR, CD27, CD38, and PD-1 (subset 14), were more prominent in OPSCC. At the CD4+ T-cell level, OPSCC contained more CD4+ Tn (subsets 19–21) and potentially CD4+CD161+ Tcm (subset 28). Levels of CD4+CD161+ Tem (subsets 22–24) were clearly higher in OPSCC and comprised populations of CD27+ and CD27 cells. We also observed 2 populations of CD4+ cells with a Treg-like phenotype (subsets 26 and 27), but their levels were similar in cervical carcinoma and OPSCC. Notably, there were no gross differences in the composition of CD8+ and CD4+ T cells in the blood of patients with cervical carcinoma and OPSCC.

Figure 3.

Clustering analysis using CITRUS revealed 30 distinctive populations of B cells, NK cells, and T cells. Automatic discovery of stratifying biological signatures within tumor and blood samples was performed using the CITRUS algorithm to identify significantly different cell populations in 35 tumor and PBMC samples (n = 9 for cervical carcinoma and OPSCC tumor and n = 9 and n = 8 for cervical carcinoma and OPSCC PBMC). Every cell population represented by a node is divided on basis of median level of expression into 2 new nodes (cellular subsets) going from the center (all cells) to the periphery of the plot. A and B, CITRUS analysis visualizing (A) 6 distinctive populations of B and NK cells within the total CD45+ immune population and (B) 24 distinctive populations of CD8+ and CD4+ T cells within the total CD3+ immune population. The parental (total CD45 or total CD3) nodes are depicted in black. The total CD45 node divides into total T-cell (depicted in red) and non–T-cell nodes with B-cell (blue), NK-cell (pink), and myeloid cell (orange) nodes. The total CD3 node divides into CD8+ T-cell (depicted in pink) and CD4+ T-cell (depicted in orange) nodes. CF, Box-and-whiskers (plus min–max) plots displaying frequencies of (C) B cells (subsets 1 to 4), (D) NK cells (subsets 5 and 6), (E) CD8+ T cells (subsets 7–18), and (F) CD4+ T cells (subsets 19–30) as % of lymphocytes for cervical carcinoma (purple) and OPSCC (green) tumors and PBMC. #, P < 0.1; *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Figure 3.

Clustering analysis using CITRUS revealed 30 distinctive populations of B cells, NK cells, and T cells. Automatic discovery of stratifying biological signatures within tumor and blood samples was performed using the CITRUS algorithm to identify significantly different cell populations in 35 tumor and PBMC samples (n = 9 for cervical carcinoma and OPSCC tumor and n = 9 and n = 8 for cervical carcinoma and OPSCC PBMC). Every cell population represented by a node is divided on basis of median level of expression into 2 new nodes (cellular subsets) going from the center (all cells) to the periphery of the plot. A and B, CITRUS analysis visualizing (A) 6 distinctive populations of B and NK cells within the total CD45+ immune population and (B) 24 distinctive populations of CD8+ and CD4+ T cells within the total CD3+ immune population. The parental (total CD45 or total CD3) nodes are depicted in black. The total CD45 node divides into total T-cell (depicted in red) and non–T-cell nodes with B-cell (blue), NK-cell (pink), and myeloid cell (orange) nodes. The total CD3 node divides into CD8+ T-cell (depicted in pink) and CD4+ T-cell (depicted in orange) nodes. CF, Box-and-whiskers (plus min–max) plots displaying frequencies of (C) B cells (subsets 1 to 4), (D) NK cells (subsets 5 and 6), (E) CD8+ T cells (subsets 7–18), and (F) CD4+ T cells (subsets 19–30) as % of lymphocytes for cervical carcinoma (purple) and OPSCC (green) tumors and PBMC. #, P < 0.1; *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Close modal

Thus, HPV-driven cervical carcinoma and OPSCC differ considerably with respect to their lymphocytic infiltrate. In OPSCC, more intratumoral IgM+ B cells as well as CD4+ Tn and CD4+CD161+ Tem were found, whereas cervical carcinoma contained higher numbers of CD8+CD103+ Teff and in particular CD8+CD103+CD161+ cells. Importantly, lymph node involvement or absence thereof in cervical cancer did not account for these observed differences between the cervical carcinoma and OPSCC tumor immune environment (Supplementary Fig. S3C and S3D).

CD161 identifies tumor-specific T cells with the strongest effector function

Because the large majority of CD8+CD103+ Tem and CD4+ Tem coexpressed CD161, we next examined their functional properties. To this end, tumor-infiltrating lymphocyte (TIL) cultures containing HPV-specific T cells were stimulated with cognate antigen, and cytokine production was analyzed in CD103 and CD161 double-negative, single-positive, and double-positive CD4 (n = 28) or CD8 (n = 9) responding T-cell populations (Fig. 4A–C). CD8 reactivity was predominantly found in CD103+CD161+ and CD103+CD161 T cells, with more than half of the responding CD8+ T cells expressing CD161. About 40% of the cytokine-producing CD4+ T cells expressed CD161 (Fig. 4B and C). Moreover, the expression of CD103 and/or CD161 within the CD4+ and CD8+ T-cell populations is not biased by changes in their expression following antigenic stimulation, as CD103 and CD161 expression was shown stable following HPV16 and SEB stimulation (Supplementary Fig. S4). Importantly, the level of cytokine production—based on the mean fluorescence intensity of cytokine staining (27, 28)—was higher in CD161+CD4+ or CD8+ T cells (Fig. 4A and D), suggesting that CD161+ T cells are highly activated. To substantiate this notion, the staining intensity of surface markers expressed by CD161+ and CD161CD4+ and CD8+ T cells infiltrating cervical carcinoma and OPSCC tumors was analyzed (Fig. 4E). In both types of tumors, CD4+CD161+ T cells expressed higher levels of CD103, PD-1, and CD127, whereas levels of CD25, CD27, and CD45RA were lower. CD8+CD161+ T cells displayed higher levels of CD103, HLA-DR, and PD-1, whereas levels of CD45RA and CCR7 were lower (Fig. 4E). Collectively, this classifies CD4+CD161+ and CD8+CD103+CD161+ TIL as highly activated effector T cells.

Figure 4.

CD161 identifies HPV16-specific CD4+ and CD8+ T cells with the strongest effector function. Three cervical carcinoma (CxCa) and 3 OPSCC TIL cultures containing HPV-specific T cells (as determined by [3H]-thymidine–based proliferation assay) were selected and analyzed for HPV16 reactivity by ICS following stimulation with pools of HPV16 E6/E7 SLPs or protein-loaded autologous monocytes or BLCL. Expression of CD3, CD4, CD8, CD137, CD154, CD161, CD103, IFNg, and TNFa following overnight stimulation in the presence of Brefeldin A was determined by flow cytometry. A, A representative example of an HPV16-reactive CD4+ and CD8+ T-cell response within TIL of a patient with OPSCC is shown. The TILs were first gated for viable and single cells, and further gated for CD3, CD4, and CD8. Within the CD4+ (top) and CD8+ (bottom) T-cell population, the cells expressing CD103 and/or CD161 were gated, and IFNγ and TNFα are depicted. The red dotted lines indicate the mean TNFα signal observed in CD103CD161 cells. B and C, The distribution of the different cell populations within the total cytokine-producing HPV-specific population (i.e., IFNγ+TNFα, IFNγ+TNFα+, and IFNγTNFα+ cells) is shown for CD4+ (left) and CD8+ (right) T cells in TIL of (B) a representative patient with OPSCC and (C) for all 28 CD4+ T-cell and 9 CD8 T-cell–mediated responses detected in all 6 HPV16-reactive patients with CxCa and OPSCC. D, The mean fluorescence intensity of TNFα and IFNγ within the CD103CD161, CD103+CD161, CD103+CD161+, and CD103CD161+ populations is depicted for CD4+ T cells and CD8+ T cells. E, Expression ratio of CD161+ versus CD161CD4+ and CD8+ T cells for each marker for 9 CxCa (purple) and 9 OPSCC (green) tumors is given on a log10 scale. Each symbol represents an individual tumor sample. Markers with an increased expression ratios within CD4+ or CD8 + T cells are indicated with an arrow. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Figure 4.

CD161 identifies HPV16-specific CD4+ and CD8+ T cells with the strongest effector function. Three cervical carcinoma (CxCa) and 3 OPSCC TIL cultures containing HPV-specific T cells (as determined by [3H]-thymidine–based proliferation assay) were selected and analyzed for HPV16 reactivity by ICS following stimulation with pools of HPV16 E6/E7 SLPs or protein-loaded autologous monocytes or BLCL. Expression of CD3, CD4, CD8, CD137, CD154, CD161, CD103, IFNg, and TNFa following overnight stimulation in the presence of Brefeldin A was determined by flow cytometry. A, A representative example of an HPV16-reactive CD4+ and CD8+ T-cell response within TIL of a patient with OPSCC is shown. The TILs were first gated for viable and single cells, and further gated for CD3, CD4, and CD8. Within the CD4+ (top) and CD8+ (bottom) T-cell population, the cells expressing CD103 and/or CD161 were gated, and IFNγ and TNFα are depicted. The red dotted lines indicate the mean TNFα signal observed in CD103CD161 cells. B and C, The distribution of the different cell populations within the total cytokine-producing HPV-specific population (i.e., IFNγ+TNFα, IFNγ+TNFα+, and IFNγTNFα+ cells) is shown for CD4+ (left) and CD8+ (right) T cells in TIL of (B) a representative patient with OPSCC and (C) for all 28 CD4+ T-cell and 9 CD8 T-cell–mediated responses detected in all 6 HPV16-reactive patients with CxCa and OPSCC. D, The mean fluorescence intensity of TNFα and IFNγ within the CD103CD161, CD103+CD161, CD103+CD161+, and CD103CD161+ populations is depicted for CD4+ T cells and CD8+ T cells. E, Expression ratio of CD161+ versus CD161CD4+ and CD8+ T cells for each marker for 9 CxCa (purple) and 9 OPSCC (green) tumors is given on a log10 scale. Each symbol represents an individual tumor sample. Markers with an increased expression ratios within CD4+ or CD8 + T cells are indicated with an arrow. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Close modal

The general lymphocyte composition of primary tumors is comparable with the tissue of origin

A recent publication on flow cytometric analyses of the percentages of CD4+ and CD8+ subpopulations in routinely removed nondiseased fresh tonsils and cervical tissue (14) prompted us to compare this with the composition of these populations in OPSCC and cervical carcinoma.

Interestingly, the CD4:CD8 ratios in cervical carcinoma tumors resembled that of normal cervical epithelium, whereas the CD4:CD8 ratios in cervical carcinoma TDLN and OPSCC were more similar to normal tonsil (Fig. 5A). These data are consistent with our calculations showing that the median CD4:CD8 tumor-infiltrating T-cell ratio in cervical carcinoma is not different from normal cervical epithelium (0.41 vs. 0.51, respectively) as measured by IHC (29) and is 3-fold lower than the CD4:CD8 ratio (1.56) in OPSCC (30). Furthermore, the composition of CD8+ Tn, Tcm, Tem, and Temra within cervical carcinoma and cervical carcinoma TDLN showed similarity to normal cervical tissue and tonsils, respectively. This was not the case for OPSCC and normal tonsils (Fig. 5B). The composition of CD4+ T cells was different in OPSCC and cervical carcinoma compared with normal tissue (Fig. 5C). The CD4+ and CD8+ T-cell populations in cervical carcinoma TDLN paralleled that of PBMC (Fig. 5B and C). Thus, cervical carcinoma and OPSCC strongly differ with respect to the composition of CD8+ and CD4+ T-cell subsets and CD4:CD8 ratio, the latter of which is more comparable with the tissue of origin. The observation that the CD4:CD8 ratio and composition of CD4 and CD8 Tn, Tcm, Tem, and Temra of HPV-driven OPSCC is similar to that of OPSCC with a nonviral etiology (Supplementary Fig. S5) also suggests an impact of the location on the tumor immune microenvironment.

Figure 5.

CD4:CD8 ratios in cervical carcinoma (CxCa) and OPSCC tumors resemble those found in the original tissue. Frequencies of CD4 and CD8 T cells, and effector/memory distribution (based on CD45RA and CCR7 expression) within these populations, were determined in TDLN and/or PBMC and tumor samples of CxCa and OPSCC samples by mass cytometry after manual gating. CD4:CD8 ratios, and effector/memory distribution within CD4+ and CD8+ T cells of normal cervical epithelium and healthy tonsils, measured by flow cytometry, were obtained from Saba and colleagues (14). A, Bar graphs depicting the CD4:CD8 ratio in the indicated samples. B and C, The distribution of Tn (CCR7+CD45RA+), Tcm (CCR7+CD45RA), Tem (CCR7CD45RA), and effector memory CD45RA+ (Temra: CCR7CD45RA+) cells within CD8+ (B) and CD4+ (C) T cells is given for indicated samples of patients with CxCa (left), patients with OPSCC (middle), and normal cervical epithelium and healthy tonsil (right). *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Figure 5.

CD4:CD8 ratios in cervical carcinoma (CxCa) and OPSCC tumors resemble those found in the original tissue. Frequencies of CD4 and CD8 T cells, and effector/memory distribution (based on CD45RA and CCR7 expression) within these populations, were determined in TDLN and/or PBMC and tumor samples of CxCa and OPSCC samples by mass cytometry after manual gating. CD4:CD8 ratios, and effector/memory distribution within CD4+ and CD8+ T cells of normal cervical epithelium and healthy tonsils, measured by flow cytometry, were obtained from Saba and colleagues (14). A, Bar graphs depicting the CD4:CD8 ratio in the indicated samples. B and C, The distribution of Tn (CCR7+CD45RA+), Tcm (CCR7+CD45RA), Tem (CCR7CD45RA), and effector memory CD45RA+ (Temra: CCR7CD45RA+) cells within CD8+ (B) and CD4+ (C) T cells is given for indicated samples of patients with CxCa (left), patients with OPSCC (middle), and normal cervical epithelium and healthy tonsil (right). *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Close modal

Tumor-specific CD4+ T cells were more often found in OPSCC and cervical carcinoma TDLN than cervical carcinoma

To determine whether the differences in CD4+ T-cell infiltration between cervical carcinoma and OPSCC were also visible at the tumor-specific T-cell level, we compared the detection rate of HPV16-specific CD4+ T-cell responses in cervical carcinoma (n = 48), TDLN of cervical carcinoma (n = 18), and OPSCC (n = 53) as analyzed and reported by us earlier (15–18, 22). Indeed, HPV16-specific T-cell reactivity significantly differed between the groups and was detected in 35% of cervical carcinoma, 100% of cervical carcinoma TDLN, and 63% of OPSCC (Fig. 6). The detection of HPV16-specific CD4+ T cells in all cervical carcinoma TDLN, including those of patients in which the matched TIL samples did not show reactivity (n = 2), and in only 35% of HPV16+ cervical carcinoma tumors suggests that the infiltration of cervical carcinoma by tumor-specific CD4+ T cells is hampered.

Figure 6.

HPV16-specific T cells are more frequently found in OPSCC and cervical carcinoma (CxCa) LN than CxCa. HPV16 specificity of tumor and LN-expanded T cells was analyzed by 5-day [3H]-thymidine–based proliferation assay. Proliferation of tumor or LN-expanded T-cell cultures was tested in triplicate against autologous HPV16 E6/E7 peptide-loaded monocytes, and CxCa and OPSCC tumors and LN were considered immune response positive (IR+) when the stimulation index (SI), calculated as the average proliferation of test wells divided by the average of the medium control wells, was >3. Depicted is the HPV16 IR status (IR+ or IR) of 18 CxCa LN, 49 CxCa, and 53 OPSCC samples. Statistical significance in HPV16-specific IR detection between CxCa LN, CxCa, and OPSCC samples was calculated by 2-tailed 2 × 3 Fisher exact probability test with Freeman–Halton extension. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Figure 6.

HPV16-specific T cells are more frequently found in OPSCC and cervical carcinoma (CxCa) LN than CxCa. HPV16 specificity of tumor and LN-expanded T cells was analyzed by 5-day [3H]-thymidine–based proliferation assay. Proliferation of tumor or LN-expanded T-cell cultures was tested in triplicate against autologous HPV16 E6/E7 peptide-loaded monocytes, and CxCa and OPSCC tumors and LN were considered immune response positive (IR+) when the stimulation index (SI), calculated as the average proliferation of test wells divided by the average of the medium control wells, was >3. Depicted is the HPV16 IR status (IR+ or IR) of 18 CxCa LN, 49 CxCa, and 53 OPSCC samples. Statistical significance in HPV16-specific IR detection between CxCa LN, CxCa, and OPSCC samples was calculated by 2-tailed 2 × 3 Fisher exact probability test with Freeman–Halton extension. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Close modal

Differences in CD4+ T-cell infiltration are reflected in survival of OPSCC and patients with cervical carcinoma

Previously, we showed that a strong infiltration with CD4+ T cells and CD4+CD161+ T cells is positively associated with survival in patients with HPV+ OPSCC (18). Consequently, the lower CD4+ T-cell infiltration observed in cervical carcinoma would predict that here CD4+ T-cell infiltration would have less impact on clinical outcome. First, we analyzed the effect of CD4 gene expression in a group of 214 squamous cervical carcinoma patients within the publicly available cancer genomic atlas (TCGA) database (31). Patients with higher than median CD4 expression in cervical carcinoma tumors displayed no survival benefit (Fig. 7A), irrespective of molecular classification as CD4+ Tcm or Tem (32). In addition, we performed follow-up analysis of a group of 38 patients with cervical carcinoma from whom we had previously analyzed the tumors with respect to T-cell infiltration by IHC (19). This analysis confirmed that there is no significant impact on survival when the patients were divided based on the median number of tumor-infiltrating CD4+ T cells in cervical carcinoma (Fig. 7B). Thus, the number of CD4+ T cells is generally lower in cervical carcinoma than OPSCC, and this is reflected by a different impact on survival of CD4+ T cells in these 2 tumor types.

Figure 7.

Differences in the magnitude of CD4+ T-cell infiltration between OPSCC and cervical carcinoma (CxCa) LN patients are reflected in their impact on survival. A, Kaplan–Meier survival plots of 214 squamous CxCa patients in the TCGA database grouped according to high and low expression of CD4 (left), CD4 Tcm (middle), and CD4 Tem (right) in their tumor. B, Kaplan–Meier survival plot of 38 HPV16/18+ CxCa patients from the LUMC from whom T-cell infiltration had been analyzed previously by us with IHC staining and were grouped on basis of the median number of tumor-infiltrating CD4 T cells (29). C and D, Kaplan–Meier survival curves showing the outcome of 42 tested HPV16-positive CxCa (C) and 51 HPV16-positive patients with OPSCC (D) that were grouped based on the presence (immune response positive, IR+) or absence (immune response negative, IR) of detectable HPV16-specific T cells in their tumors. E, Box-and-whiskers (plus min–max) plots displaying percentages of CD4+CD161+ Tem (subset 22) detected in the tumor of HPV16 IR and IR+ patients with CxCa (purple) and OPSCC (green). F, Kaplan–Meier survival plots of 214 patients with squamous CxCa in the TCGA database grouped according to high median expression of CD4 and CD161 (KLRB1) versus all others. For all Kaplan–Meier plots, the HR with the 95% confidence interval (CI) as well as the log-rank test P value is given. NS, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Figure 7.

Differences in the magnitude of CD4+ T-cell infiltration between OPSCC and cervical carcinoma (CxCa) LN patients are reflected in their impact on survival. A, Kaplan–Meier survival plots of 214 squamous CxCa patients in the TCGA database grouped according to high and low expression of CD4 (left), CD4 Tcm (middle), and CD4 Tem (right) in their tumor. B, Kaplan–Meier survival plot of 38 HPV16/18+ CxCa patients from the LUMC from whom T-cell infiltration had been analyzed previously by us with IHC staining and were grouped on basis of the median number of tumor-infiltrating CD4 T cells (29). C and D, Kaplan–Meier survival curves showing the outcome of 42 tested HPV16-positive CxCa (C) and 51 HPV16-positive patients with OPSCC (D) that were grouped based on the presence (immune response positive, IR+) or absence (immune response negative, IR) of detectable HPV16-specific T cells in their tumors. E, Box-and-whiskers (plus min–max) plots displaying percentages of CD4+CD161+ Tem (subset 22) detected in the tumor of HPV16 IR and IR+ patients with CxCa (purple) and OPSCC (green). F, Kaplan–Meier survival plots of 214 patients with squamous CxCa in the TCGA database grouped according to high median expression of CD4 and CD161 (KLRB1) versus all others. For all Kaplan–Meier plots, the HR with the 95% confidence interval (CI) as well as the log-rank test P value is given. NS, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.

Close modal

Detection of HPV-specific T cells in OPSCC is a strong predictor for patient survival in OPSCC (18). Hence, we analyzed the value of this in patients with cervical carcinoma from whom we cultured TIL and were able to test HPV specificity. In contrast to a group of 51 patients with OPSCC, the detection of HPV-specific T cells among ex vivo–expanded TIL was not able to predict survival in a set of 42 patients with cervical carcinoma (Fig. 7C and D). Most likely, because the total CD4+ T-cell number and thus also that of HPV-specific T cells—before the in vitro expansion—are too low to mediate any strong effect in vivo. Interestingly, whenever an HPV-specific CD4+ T-cell response was detected in the tumor, the percentage of CD4+CD161+ Tem was higher in both OPSCC and cervical carcinoma (Fig. 7E). As we showed that the intratumoral CD4+CD161+ T-cell population is highly activated, we assessed whether high numbers of CD4+CD161+ effector cells would have the same positive association with survival in cervical carcinoma as previously reported by us for OPSCC (18). Indeed, the group of patients with cervical carcinoma with high expression levels of CD4 and CD161 in the tumor showed a better outcome (Fig. 7F).

We exploited the fact that HPV16 can cause tumors to arise in different tissues to study the extent to which the anatomical location contributes to the immune contexture of a developing primary tumor. Application of our high-dimensional single-cell mass cytometry–based approach with 36 markers in freshly digested tumor samples and PBMC of patients with cervical carcinoma or OPSCC identified 30 distinctive clusters of lymphocytes. These findings are the first line of evidence showing that tumors of the same etiology, but arising in a different tissue, have a different immune contexture. In a direct comparison, OPSCC contained more B cells and showed a specific enrichment with subpopulations of CD4+CD161+ Tem, whereas several subpopulations of CD8+CD103+ Teff were enriched in cervical carcinoma. In addition, the CD4:CD8 ratio was 3-fold higher in OPSCC than cervical carcinoma. Finally, HPV-specific TILs were more frequently detected in OPSCC than cervical carcinoma.

Interestingly, the CD4:CD8 ratio in OPSCC closely resembled the ratio found in normal tonsils (14), and the CD4:CD8 ratio in cervical carcinoma is highly similar to that found in normal cervical epithelium as measured by flow cytometry (14, 33) and IHC (11, 12, 29). Furthermore, the percentages of CD8+ Tn, Tcm, Tem, and Temra found in cervical carcinoma are comparable with previous reported findings in normal cervical tissue (14). Moreover, the numbers of cervical carcinoma resident (CD8+CD103+) T cells display the same mean and variability as found in normal cervix (34), suggesting that these cells are already present in normal tissue and thus not appear due to malignant transformation. It seems that the different immune contextures developed in primary OPSCC and cervical carcinoma reflect the immune contexture found in the tissue of origin. This notion is sustained by the observation that OPSCC of nonviral etiology displays a similar general (CD4:CD8 ratio and percentages of Tn, Tcm, Tem, and Temra) immune infiltration as HPV-driven OSPCC. Evidently, differences in the distribution of naïve and effector memory CD4+ and CD8+ T cells between healthy tonsil (and TDLN) and cervical tissue are explained by the fact that tonsil and TDLN are secondary lymphoid organs where naïve T cells recirculate (35).

In contrast to CD8+ T cells, the composition of the CD4+ T-cell population in OPSCC and cervical carcinoma was different from healthy tissue. A strong infiltration of OPSCC with CD4+ T cells was found to be beneficial for survival in 3 independent cohorts, and this was correlated to their functional activity, as indicated by strong expression of HLA-DR, CD38, and PD-1 (36) with or without CD161 and by expression of Tbet (18). Activated CD4+ Teff cells producing IFNγ and TNFα were shown to induce permanent growth arrest in the Simian virus 40 large T antigen–driven β-cancer cell model (37, 38). CD4+ T cells were also responsible for control of spontaneous cervical tumor outgrowth in genetically engineered K14-HPV16 transgenic mice. Within this model, a strong reduction of progressive precursor lesions was found when the HPV-specific CD4+ T-cell levels and activity were boosted by vaccination (39). Hence, tumor-specific CD4+ T cells actively participate in tumor control. However, here we showed that the relative CD4+ T-cell numbers are lower in cervical carcinoma than OPSCC. In addition, comparison of median CD4+ T-cell infiltration numbers in 2 previous studies in cervical carcinoma and 1 in OPSCC performed with similar techniques (18, 19, 29) showed that the absolute median number of CD4+ T-cell infiltration per square millimeter of tumor was 2-fold lower in cervical carcinoma than OPSCC. Moreover, this difference was also reflected in their association with patient's survival. Although division of patients with OPSCC based on their median infiltration with CD4+ T cells showed a beneficial clinical effect, this was not observed in patients with cervical carcinoma. Similar to our previous findings in OPSCC (18), CD4+ T-cell–mediated survival benefit is found for patients with cervical carcinoma with high levels of highly active CD4+ T cells, which are identified by CD161. Moreover, CD161+ Teff produced the highest amounts of type 1 cytokines upon activation with their cognate tumor antigen. Intermediate expression (as compared with MAIT cells) of CD161 has identified CD4+ and CD8+ mucosal Teff and Tcm cells as highly functional type 1/17 cytokine-producing cell (40–44). Acute Graft-versus-Host Disease (GvHD) is associated with high CD4+CD161+ to CD8+CD161+ ratio (45) validating a role in tissue rejection specifically for the CD4+ T-cell population. However, high levels of CD4 and CD161 were only observed in a small group of cervical carcinoma patients. The numerical and relative lower abundance of CD4+ T cells in cervical carcinoma, which is also observed in normal cervical tissue, suggests that the lack of sufficient attraction of CD4+ T cells is intrinsic to the location. This notion is sustained by our observation that in all patients tested, HPV-specific CD4+ T-cell responses are detected in the TDLN. However, HPV-specific CD4+ T cells were only found in a minor fraction of the large set of patients with cervical carcinoma analyzed.

Thus, the strong differences in lymphocytic infiltrate between oncogenic HPV-driven primary cervical carcinoma and OPSCC indicate a role for the originating tissue in shaping the immune contexture. Our results imply that the problem of CD4+ T-cell attraction in cervical carcinoma will continue to exist throughout the progression of disease and suggests that it should form a point of focus for future immunotherapeutic approaches aiming to treat progressive cervical carcinoma.

No potential conflicts of interest were disclosed.

Conception and design: S.J. Santegoets, S.H. van der Burg

Development of methodology: S.J. Santegoets, M.J.P. Welters, S.H. van der Burg

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.J. Santegoets, V.J. van Ham, I. Ehsan, L.-A. van der Velden, S.L. van Egmond, K.E. Kortekaas, P.J. de Vos van Steenwijk, M.I.E. van Poelgeest

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.J. Santegoets, V.J. van Ham, I. Ehsan, P. Charoentong, C.L. Duurland, V. van Unen, K.E. Kortekaas, M.J.P. Welters, S.H. van der Burg

Writing, review, and/or revision of the manuscript: S.J. Santegoets, C.L. Duurland, L.-A. van der Velden, K.E. Kortekaas, P.J. de Vos van Steenwijk, M.I.E. van Poelgeest, M.J.P. Welters, S.H. van der Burg

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): T. Höllt, M.J.P. Welters

Study supervision: M.J.P. Welters, S.H. van der Burg

We thank the patients for participating in this study.

This study was financially supported by grants from the Dutch Cancer Society 2014-6696 (to S.H. van der Burg, L.-A. van der Velden, and M.J.P. Welters) and 2016-10726 (to S.H. van der Burg, M.J.P. Welters, and S.J. Santegoets).

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.
Witz
IP
. 
The tumor microenvironment: the making of a paradigm
.
Cancer Microenviron
2009
;
2
:
9
17
.
2.
Galon
J
,
Angell
HK
,
Bedognetti
D
,
Marincola
FM
. 
The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures
.
Immunity
2013
;
39
:
11
26
.
3.
Fridman
WH
,
Pages
F
,
Sautes-Fridman
C
,
Galon
J
. 
The immune contexture in human tumours: impact on clinical outcome
.
Nat Rev Cancer
2012
;
12
:
298
306
.
4.
Remark
R
,
Alifano
M
,
Cremer
I
,
Lupo
A
,
Dieu-Nosjean
MC
,
Riquet
M
, et al
Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin
.
Clin Cancer Res
2013
;
19
:
4079
91
.
5.
Giraldo
NA
,
Becht
E
,
Remark
R
,
Damotte
D
,
Sautes-Fridman
C
,
Fridman
WH
. 
The immune contexture of primary and metastatic human tumours
.
Curr Opin Immunol
2014
;
27
:
8
15
.
6.
Liu
C
,
Peng
W
,
Xu
C
,
Lou
Y
,
Zhang
M
,
Wargo
JA
, et al
BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice
.
Clin Cancer Res
2013
;
19
:
393
403
.
7.
Peng
W
,
Chen
JQ
,
Liu
C
,
Malu
S
,
Creasy
C
,
Tetzlaff
MT
, et al
Loss of PTEN promotes resistance to T cell-mediated immunotherapy
.
Cancer Discov
2016
;
6
:
202
16
.
8.
Spranger
S
,
Bao
R
,
Gajewski
TF
. 
Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
.
9.
zur Hausen
H
. 
Papillomaviruses and cancer: from basic studies to clinical application
.
Nat Rev Cancer
2002
;
2
:
342
50
.
10.
Doorbar
J
,
Quint
W
,
Banks
L
,
Bravo
IG
,
Stoler
M
,
Broker
TR
, et al
The biology and life-cycle of human papillomaviruses
.
Vaccine
2012
;
30
:
F55
70
.
11.
Pudney
J
,
Quayle
AJ
,
Anderson
DJ
. 
Immunological microenvironments in the human vagina and cervix: mediators of cellular immunity are concentrated in the cervical transformation zone
.
Biol Reprod
2005
;
73
:
1253
63
.
12.
Poppe
WA
,
Drijkoningen
M
,
Ide
PS
,
Lauweryns
JM
,
Van Assche
FA
. 
Lymphocytes and dendritic cells in the normal uterine cervix. An immunohistochemical study
.
Eur J Obstet Gynecol Reprod Biol
1998
;
81
:
277
82
.
13.
Gelwan
E
,
Malm
IJ
,
Khararjian
A
,
Fakhry
C
,
Bishop
JA
,
Westra
WH
. 
Nonuniform distribution of high-risk human papillomavirus in squamous cell carcinomas of the oropharynx: rethinking the anatomic boundaries of oral and oropharyngeal carcinoma from an oncologic HPV perspective
.
Am J Surg Pathol
2017
;
41
:
1722
8
.
14.
Saba
E
,
Grivel
JC
,
Vanpouille
C
,
Brichacek
B
,
Fitzgerald
W
,
Margolis
L
, et al
HIV-1 sexual transmission: early events of HIV-1 infection of human cervico-vaginal tissue in an optimized ex vivo model
.
Mucosal Immunol
2010
;
3
:
280
90
.
15.
de Vos van Steenwijk
PJ
,
Heusinkveld
M
,
Ramwadhdoebe
TH
,
Lowik
MJ
,
van der Hulst
JM
,
Goedemans
R
, et al
An unexpectedly large polyclonal repertoire of HPV-specific T cells is poised for action in patients with cervical cancer
.
Cancer Res
2010
;
70
:
2707
17
.
16.
Piersma
SJ
,
Welters
MJ
,
van der Hulst
JM
,
Kloth
JN
,
Kwappenberg
KM
,
Trimbos
BJ
, et al
Human papilloma virus specific T cells infiltrating cervical cancer and draining lymph nodes show remarkably frequent use of HLA-DQ and -DP as a restriction element
.
Int J Cancer
2008
;
122
:
486
94
.
17.
Heusinkveld
M
,
Goedemans
R
,
Briet
RJ
,
Gelderblom
H
,
Nortier
JW
,
Gorter
A
, et al
Systemic and local human papillomavirus 16-specific T-cell immunity in patients with head and neck cancer
.
Int J Cancer
2012
;
131
:
E74
85
.
18.
Welters
MJP
,
Ma
W
,
Santegoets
S
,
Goedemans
R
,
Ehsan
I
,
Jordanova
ES
, et al
Intratumoral HPV16-specific T cells constitute a type I-oriented tumor microenvironment to improve survival in HPV16-driven oropharyngeal cancer
.
Clin Cancer Res
2018
;
24
:
634
47
.
19.
Piersma
SJ
,
Jordanova
ES
,
van Poelgeest
MI
,
Kwappenberg
KM
,
van der Hulst
JM
,
Drijfhout
JW
, et al
High number of intraepithelial CD8+ tumor-infiltrating lymphocytes is associated with the absence of lymph node metastases in patients with large early-stage cervical cancer
.
Cancer Res
2007
;
67
:
354
61
.
20.
Nooij
LS
,
Ter Haar
NT
,
Ruano
D
,
Rakislova
N
,
van Wezel
T
,
Smit
V
, et al
Genomic characterization of vulvar (Pre)cancers identifies distinct molecular subtypes with prognostic significance
.
Clin Cancer Res
2017
;
23
:
6781
9
.
21.
Ten Broeke
SW
,
van Bavel
TC
,
Jansen
AML
,
Gomez-Garcia
E
,
Hes
FJ
,
van Hest
LP
, et al
Molecular background of colorectal tumors from patients with lynch syndrome associated with germline variants in PMS2
.
Gastroenterology
2018
;
155
:
844
51
.
22.
van Poelgeest
MI
,
Visconti
VV
,
Aghai
Z
,
van Ham
VJ
,
Heusinkveld
M
,
Zandvliet
ML
, et al
Potential use of lymph node-derived HPV-specific T cells for adoptive cell therapy of cervical cancer
.
Cancer Immunol Immunother
2016
;
65
:
1451
63
.
23.
van Unen
V
,
Li
N
,
Molendijk
I
,
Temurhan
M
,
Höllt
T
,
van der Meulen-de
Jong AE
, et al
Mass cytometry of the human mucosal immune system identifies tissue- and disease-associated immune subsets
.
Immunity
2016
;
44
:
1227
39
.
doi: 10.1016/j.immuni.2016.04.014. Epub 2016 May 10.
24.
Höllt
T
,
Pezzotti
N
,
van Unen
V
,
Koning
F
,
Eisemann
E
,
Lelieveldt
B
, et al
Cytosplore: interactive immune cell phenotyping for large single-cell datasets
.
Computer Graphics Forum
2016
;
35
:
171
80
.
25.
Topham
DJ
,
Reilly
EC
. 
Tissue-resident memory CD8(+) T cells: from phenotype to function
.
Front Immunol
2018
;
9
:
515
.
26.
Bruggner
RV
,
Bodenmiller
B
,
Dill
DL
,
Tibshirani
RJ
,
Nolan
GP
. 
Automated identification of stratifying signatures in cellular subpopulations
.
Proc Natl Acad Sci U S A
2014
;
111
:
E2770
7
.
27.
Darrah
PA
,
Hegde
ST
,
Patel
DT
,
Lindsay
RW
,
Chen
L
,
Roederer
M
, et al
IL-10 production differentially influences the magnitude, quality, and protective capacity of Th1 responses depending on the vaccine platform
.
J Exp Med
2010
;
207
:
1421
33
.
28.
Darrah
PA
,
Patel
DT
,
De Luca
PM
,
Lindsay
RW
,
Davey
DF
,
Flynn
BJ
, et al
Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major
.
Nat Med
2007
;
13
:
843
50
.
29.
Jordanova
ES
,
Gorter
A
,
Ayachi
O
,
Prins
F
,
Durrant
LG
,
Kenter
GG
, et al
Human leukocyte antigen class I, MHC class I chain-related molecule A, and CD8+/regulatory T-cell ratio: which variable determines survival of cervical cancer patients?
Clin Cancer Res
2008
;
14
:
2028
35
.
30.
Wansom
D
,
Light
E
,
Thomas
D
,
Worden
F
,
Prince
M
,
Urba
S
, et al
Infiltrating lymphocytes and human papillomavirus-16–associated oropharyngeal cancer
.
Laryngoscope
2012
;
122
:
121
7
.
31.
Cancer Genome Atlas Network.
Comprehensive genomic characterization of head and neck squamous cell carcinomas
.
Nature
2015
;
517
:
576
82
.
32.
Charoentong
P
,
Finotello
F
,
Angelova
M
,
Mayer
C
,
Efremova
M
,
Rieder
D
, et al
Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade
.
Cell Rep
2017
;
18
:
248
62
.
33.
Trifonova
RT
,
Lieberman
J
,
van Baarle
D
. 
Distribution of immune cells in the human cervix and implications for HIV transmission
.
Am J Reprod Immunol
2014
;
71
:
252
64
.
34.
Rodriguez-Garcia
M
,
Fortier
JM
,
Barr
FD
,
Wira
CR
. 
Aging impacts CD103(+) CD8(+) T cell presence and induction by dendritic cells in the genital tract
.
Aging Cell
2018
;
e12733
.
35.
Butcher
EC
,
Picker
LJ
. 
Lymphocyte homing and homeostasis
.
Science
1996
;
272
:
60
6
.
36.
Badoual
C
,
Hans
S
,
Merillon
N
,
Van Ryswick
C
,
Ravel
P
,
Benhamouda
N
, et al
PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer
.
Cancer Res
2013
;
73
:
128
38
.
37.
Braumuller
H
,
Wieder
T
,
Brenner
E
,
Assmann
S
,
Hahn
M
,
Alkhaled
M
, et al
T-helper-1-cell cytokines drive cancer into senescence
.
Nature
2013
;
494
:
361
5
.
38.
Wieder
T
,
Braumuller
H
,
Brenner
E
,
Zender
L
,
Rocken
M
. 
Changing T-cell enigma: cancer killing or cancer control?
Cell Cycle
2013
;
12
:
3146
53
.
39.
Daniel
D
,
Chiu
C
,
Giraudo
E
,
Inoue
M
,
Mizzen
LA
,
Chu
NR
, et al
CD4+ T cell-mediated antigen-specific immunotherapy in a mouse model of cervical cancer
.
Cancer Res
2005
;
65
:
2018
25
.
40.
Cohavy
O
,
Shih
DQ
,
Doherty
TM
,
Ware
CF
,
Targan
SR
. 
CD161 defines effector T cells that express LIGHT and respond to TL1A-DR3 signaling
.
Eur J Microbiol Immunol (Bp)
2011
;
1
:
70
9
.
41.
Fergusson
JR
,
Huhn
MH
,
Swadling
L
,
Walker
LJ
,
Kurioka
A
,
Llibre
A
, et al
CD161(int)CD8+ T cells: a novel population of highly functional, memory CD8+ T cells enriched within the gut
.
Mucosal Immunol
2016
;
9
:
401
13
.
42.
Gonzalez
Y
,
Herrera
MT
,
Juarez
E
,
Salazar-Lezama
MA
,
Bobadilla
K
,
Torres
M
. 
CD161 expression defines a Th1/Th17 polyfunctional subset of resident memory T lymphocytes in bronchoalveolar cells
.
PLoS One
2015
;
10
:
e0123591
.
43.
Takahashi
T
,
Dejbakhsh-Jones
S
,
Strober
S
. 
Expression of CD161 (NKR-P1A) defines subsets of human CD4 and CD8 T cells with different functional activities
.
J Immunol
2006
;
176
:
211
6
.
44.
Duurland
CL
,
Brown
CC
,
O'Shaughnessy
RF
,
Wedderburn
LR
. 
CD161(+) Tconv and CD161(+) treg share a transcriptional and functional phenotype despite limited overlap in TCRbeta Repertoire
.
Front Immunol
2017
;
8
:
103
.
45.
Lee
SE
,
Lim
JY
,
Yoon
JH
,
Shin
SH
,
Cho
BS
,
Eom
KS
, et al
CD161(+) T cells as predictive markers for acute graft-versus-host disease
.
Biol Blood Marrow Transplant
2015
;
21
:
421
8
.