Head and neck cancers positive for human papillomavirus (HPV) have a more favorable clinical outcome than HPV-negative cancers, but it is unknown why this is the case. We hypothesized that prognosis was affected by intrinsic features of HPV-infected tumor cells or differences in host immune response. In this study, we focused on a comparison of regulatory Foxp3+ T cells and programmed death-1 (PD-1)+ T cells in the microenvironment of tumors that were positive or negative for HPV, in two groups that were matched for various clinical and biologic parameters. HPV-positive head and neck cancers were more heavily infiltrated by regulatory T cells and PD-1+ T cells and the levels of PD-1+ cells were positively correlated with a favorable clinical outcome. In explaining this paradoxical result, we showed that these PD-1+ T cells expressed activation markers and were functional after blockade of the PD-1–PD-L1 axis in vitro. Approximately 50% of PD-1+ tumor-infiltrating T cells lacked Tim-3 expression and may indeed represent activated T cells. In mice, administration of a cancer vaccine increased PD-1 on T cells with concomitant tumor regression. In this setting, PD-1 blockade synergized with vaccine in eliciting antitumor efficacy. Our findings prompt a need to revisit the significance of PD-1–infiltrating T cells in cancer, where we suggest that PD-1 detection may reflect a previous immune response against tumors that might be reactivated by PD-1/PD-L1 blockade. Cancer Res; 73(1); 128–38. ©2012 AACR.

Recent studies have detected oncogenic HPV in 25.9% of head and neck cancers (mostly HPV16) and this prevalence increases to more than 50% for cancers of the oropharynx (tonsil, base of tongue, etc.) with a growing worldwide incidence (1).

Although these tumors frequently have aggressive histopathological features (2), the presence of HPV DNA is a favorable prognostic factor with regard to recurrence and survival (3–5). The mechanisms underlying the more favorable outcome of HPV-associated head and neck cancer have not been clearly elucidated.

These tumors are more responsive to chemotherapy and radiotherapy, but even patients with HPV-positive oropharyngeal cancers treated by surgery alone have a better prognosis than those with HPV-negative tumors after adjustment for tumor stage (6, 7).

HPV E6-E7 expressing tumors grew slower in immunocompetent mice but not in nude mice, than HPV-negative tumors suggesting a role of adaptive immunity and not simply an intrinsic biologic property of HPV-positive tumor cells to explain this difference (8).

In human, T cells directed against HPV derived proteins (E2, E6, and E7) have been described in the blood and in the tumor of patients with head and neck cancers (9–12). However, the capacity of tumor-infiltrating lymphocytes (TIL) to act as effector cells may be affected by the tumor microenvironment. Immunosuppressive cells or molecules (13, 14) or chronic inflammation in head and neck cancer predispose to a tumor promoting microenvironment (15, 16). Analysis of differences in the type of immunity within the tumor microenvironment of HPV-associated head and neck cancer compared with HPV-negative tumors may, therefore, provide new clues to explain the more favorable clinical behavior of this type of cancer. In the present study, we addressed the clinical significance of regulatory Foxp3+T cells and programmed death (PD-1)-positive T-cell infiltration in the tumor microenvironment of HPV-positive and HPV-negative head and neck cancers. These immunosuppressive cells were selected because regulatory CD4+CD25+Foxp3+T cells have emerged as the dominant T-cell population governing peripheral tolerance by inhibiting effector T cells. We and other authors have shown that the frequency of intratumoral Treg is increased in head and neck cancer patients and in HPV-associated cervical carcinoma (17, 18). PD-1, a member of the CD28 receptor family, is expressed by activated lymphocytes and inhibits their proliferation and effector functions after binding to PD-1 ligands such as B7-H1 (PD-L1; ref. 19). PD-1 expression is indicative of chronic antigen stimulation and contributes to T-cell dysfunction (exhaustion; ref. 19). Interference with PD-1–PD-L1 signaling, via either antibody blockade or PD-1 deficiency has been shown to improve clinical outcome and restore functional T-cell responses in chronic viral diseases and in several types of cancer (20). Preliminary results documented that more than 60% of freshly isolated human head and neck tumors express PD-L1 (21) and in early stage tongue cancer, intraepithelial T cells frequently express PD-1 (22). All these data, therefore, support a comprehensive analysis of PD-1–PD-L1 expression in head and neck cancers associated with chronic viral infection.

As expected, HPV-associated head and neck cancers were found to be associated with a good prognosis compared with HPV-negative tumors. HPV-positive tumors were more heavily infiltrated by regulatory T cells and PD-1 expressing T cells than HPV-negative tumors. Surprisingly, levels of PD-1+ T-cell infiltration positively correlated with a favorable clinical outcome in HPV-associated cancers and, in a preclinical HPV tumor model, predicted the clinical response to anti–PD-L1 antibody.

Patients

Sixty-four newly diagnosed untreated patients with primary histologically proven head and neck squamous cell carcinoma (HNSCC) were included in this study. Patient characteristics are presented in Supplementary Table S1. Each patient's disease was staged according to the 7th edition of the International Union Against Cancer/American Joint Committee on Cancer system for head and neck cancer. Treatment modalities consisted of chemoradiotherapy without surgery (organ preservation) or surgery combined or not with radiotherapy and chemotherapy. This study was conducted in accordance with French laws and after approval by the local ethics committee (ID RCB 2007-A01128-45).

Patients were divided in 2 groups depending on the presence or absence of oncogenic HPV. These 2 groups were matched for various parameters (gender, primary tumor site, tumor staging, lymph node involvement, presence of metastasis, and treatment modalities; Supplementary Table S1).

Mice

Six- to eight-week-old female C57BL/6 (H-2b; B6) mice were purchased from Charles River Laboratories (L'Arbresle, France). All mice were kept under specific pathogen-free conditions at the INSERM U970 animal facility. Experiments were conducted according to institutional guidelines after acceptance by the Veterinary School of Maisons-Alfort ethics committee.

Cells

TC-1 cells are transformed murine (H2b) epithelial cells cotransfected with HPV-16 E6/E7 genes and the activated human Ha-ras (G12V) oncogene DNA. They were obtained from Dr. TC Wu's laboratory (Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, USA). Cells were grown in RPMI 1640 medium supplemented with 10% fetal calf serum, 2 mmol/L L-Glutamine, antibiotics and 0.4 mg/mL G418 at 37°C with 5%CO2.

Peptide, vaccine, and antibodies

Pool of 15 mer peptides spanning the entire HPV16 E7 protein were designed with a 4 mer overlap between each peptide. They were obtained from PolyPeptide Laboratories (Strasbourg, France), reconstituted in PBS and stored at −20°C.

The STxB-E743–57 vaccine was produced by chemical coupling between the N-bromoacetylated E743–57 peptide and the sulfhydryl group of STxB-Cys recombinant protein, as previously described (23). After purification, endotoxin concentrations determined by the Limulus assay test (Lonza) were less than 0.5 EU/mg.

The invariant natural killer T-cell ligand α-GalCer (KRN7000) was purchased from Funakoshi (Tokyo, Japan).

CT-011 is a humanized monoclonal anti-PD-1 antibody manufactured by CureTech LTD. This antibody has already been validated in ex vivo functional assays in humans (24).

Anti-mouse PD-L1 and isotype control antibodies were purchased from BioXcell. Mice received intraperitoneal injections of 200 μg mAb/mouse as previously described (25).

For immunofluorescence analysis, an anti-PD-1 mAb generated by D Olive's group was selected and used (see Supplementary Table S2) as previously described (26).

HPV detection

The presence of HPV in head and neck cancer biopsies was detected using the INNO-LIPA genotyping Extra assay (Innogenetics).

Immunofluorescence staining and flow cytometry analysis

Immunofluorescence staining and flow cytometry analysis were conducted as previously described (16) and detailed in Supplementary Data. The antibodies used for the various immunofluorescence stainings are described in Supplementary Table S2.

Immunization of mice and in vivo tumor protection

C57BL/6 mice were immunized via the intraperitoneal route at days 0 and 14 with the STxB-E7 vaccine (20 μg) combined with 1 μg of αGalCer during the first immunization.

In a therapeutic setting experiment, 105 TC-1 tumor cells were injected subcutaneously (s.c.) in the right flank of B6 mice. They were then vaccinated or not at days 9 and 18 after tumor graft in combination with anti-PD-L1 or isotype control antibodies. Mice were monitored every 48 to 72 hours for tumor growth.

Statistical analysis

Survival variables were estimated using the Kaplan–Meier method and compared by the log-rank test for categorical variables and the Cox model and associated Wald ×2 statistic for quantitative variables.

Overall survival was defined as the time from initial diagnosis until death or until last follow-up (right censored data).

Locoregional control was calculated from the end of treatment and defined as the absence or either persistent or recurrent disease at the primary site or in the cervical lymph nodes. Patients with persistent disease at the end of treatment were considered to have experienced failure at time 0. Patients with no signs of relapse were censored at the time of last-follow up or death. The median follow-up for the overall population was 26 months.

The Mann–Whitney U test was used to assess whether 2 samples of observations were derived from the same distribution. The χ2 test with Yates correction was used to analyze the relationship between tumor cell infiltration and the tumor HPV status.

HPV-associated head and neck cancer has a good prognosis

In a retrospective study, we divided 64 head and neck cancer patients into 2 groups differing according to presence or absence of oncogenic HPV (30 HPV16, 1 HPV18 and 1 HPV33, and 32 HPV-negative) in their tumors. These 2 groups were balanced for gender, primary tumor sites, tumor (T) staging, lymph node involvement, and treatment modalities (Supplementary Table S1). On the basis of Kaplan–Meier estimates, the overall survival for patients with HPV-positive cancer was significantly better (median survival not reached) than for patients with HPV-negative cancer (median survival: 16 months; P = 0.001; log rank test; Fig. 1). Patients with HPV-positive tumors also had a significantly better disease-free survival (P = 0.003; log rank test) than patients with HPV-negative cancer (Fig. 1).

Figure 1.

HPV-associated head and neck cancers are associated with a good prognosis. Relationships between the presence or absence of oncogenic HPV in head and neck tumors and overall survival (A) or locoregional control (B).

Figure 1.

HPV-associated head and neck cancers are associated with a good prognosis. Relationships between the presence or absence of oncogenic HPV in head and neck tumors and overall survival (A) or locoregional control (B).

Close modal

Intratumoral Foxp3+ T cells and PD-1+ T cells differ between HPV-positive and HPV-negative head and neck cancers

HPV-positive head and neck cancers were more heavily infiltrated by CD8+ cells (P = 0.01), PD-1+CD4+ T cells (P = 0.045), and the total number of PD-1+CD4+ and PD-1+CD8+ T cells (P = 0.045). A trend was also observed between high levels of total PD-1+ cell (P = 0.079), PD-1+CD8+ cell (P = 0.078), and Foxp3+CD4+ T-cell (P = 0.059) infiltration and the HPV detection in the tumors (Fig. 2 and Table 1). PD-1 expression was higher in CD4+ T cells (median of PD-1+CD4+ cells = 5.5), than in CD8+ T cells (median of PD-1+CD8+ = 1).

Figure 2.

PD-1+ and Foxp3+ T cells infiltrate HPV-associated head and neck cancer. Tissue derived from biopsies of HPV-positive head and neck cancers were stained with antibodies to human CD4, CD8, Foxp3, and PD-1. Left and second column from the left, results of simple immunofluorescence acquisition with each antibody. Third column from the left, double immunofluorescence staining. Isotype control antibodies were also included in each experiment (right). Arrows indicate colocalization between the markers recognized by the specific antibodies. (Original magnification, ×400).

Figure 2.

PD-1+ and Foxp3+ T cells infiltrate HPV-associated head and neck cancer. Tissue derived from biopsies of HPV-positive head and neck cancers were stained with antibodies to human CD4, CD8, Foxp3, and PD-1. Left and second column from the left, results of simple immunofluorescence acquisition with each antibody. Third column from the left, double immunofluorescence staining. Isotype control antibodies were also included in each experiment (right). Arrows indicate colocalization between the markers recognized by the specific antibodies. (Original magnification, ×400).

Close modal
Table 1.

Correlation between HPV status and T-cell infiltration and PD-L1 expression by tumor cells

HPV-positive head and neck cancerHPV-negative head and neck cancer
CD8 High 17  
 Low 15 25 P = 0.01 
CD4 High 22 16  
 Low 10 16 P = 0.12 
PD-1+CD4+ High 20 12  
 Low 12 20 P = 0.045 
PD-1+CD8+ High 18 11  
 Low 14 21 P = 0.078 
PD-1+CD4+ and PD-1+CD8+ High 20 12  
 Low 12 20 P = 0.045 
PD-1 High 19 12  
 Low 13 20 P = 0.079 
Foxp3+ CD4+ High 20 12  
 Low 12 19 P = 0.059 
PD-L1 High 20 13  
 Low 12 19 P = 0.08 
HPV-positive head and neck cancerHPV-negative head and neck cancer
CD8 High 17  
 Low 15 25 P = 0.01 
CD4 High 22 16  
 Low 10 16 P = 0.12 
PD-1+CD4+ High 20 12  
 Low 12 20 P = 0.045 
PD-1+CD8+ High 18 11  
 Low 14 21 P = 0.078 
PD-1+CD4+ and PD-1+CD8+ High 20 12  
 Low 12 20 P = 0.045 
PD-1 High 19 12  
 Low 13 20 P = 0.079 
Foxp3+ CD4+ High 20 12  
 Low 12 19 P = 0.059 
PD-L1 High 20 13  
 Low 12 19 P = 0.08 

NOTE: For quantitative parameters, low and high levels of each cell type were defined by using the median level of tumor infiltration by these cells as cut-off. For semiquantitative parameters (CD4, CD8, and PD-L1), a score of 0 or + corresponds to low levels, whereas a score of ++/+++ corresponds to high levels. All parameters were measured on 64 head and neck tumor biopsies (32 HPV-positive and 32 HPV-negative) except for Foxp3+CD4+ cells, which were counted on 63 biopsies.

As PD-1+CD4+ cells and PD-1+CD8+ cells could also represent macrophages and NK cells respectively, tumor dissociation was conducted for 14 tumors and cytometry analysis confirmed that more than 95% of PD-1+CD45+ cells were CD3+ T cells (Supplementary Fig. S1).

Thirty-three (51.5%) of the 64 head and neck tumors expressed significant levels of PD-L1 (score ++/+++) with homogeneous staining (Table 1 and Supplementary Fig. S2), but no correlation was observed between PD-L1 expression and HPV status of these head and neck cancer patients (Table 1).

PD-1–positive infiltrating T cells are associated with better overall survival in HPV-positive head and neck cancer

Surprisingly, we showed that HPV-positive tumors, infiltrated by large numbers of PD-1–positive cells or total number of PD-1+CD4 and PD-1+CD8 T cells, were correlated with better overall survival of these patients. Indeed, patients with PD-1–positive tumor-infiltrating T cells above the median value (15 PD-1 cells/fields) had a 93.9% 60-month overall survival, whereas patients with low PD-1–positive cell infiltration had a 63.6% 60-month overall survival [P = 0.025; HR, 0.13; 95% confidence interval (CI), 0.02–0.067] (Fig. 3A). In line with these results and with the previous demonstration that most PD-1 cells are T cells (Supplementary Fig. S1), we showed that tumors infiltrated by high levels of PD-1+CD4+ and PD-1+CD8+ cells had a better survival than tumors with low PD-1+ T-cell infiltration (P = 0.045; HR 0.15; 95% CI, 0.03–0.7). Patients with high infiltration (>median value) by PD-1+CD4+ T cells (P = 0.046) but not by PD-1+CD8+ T cells also had a better overall survival.

Figure 3.

PD-1–positive infiltrating T cells positively correlate with survival in both HPV-positive head and neck cancer and in the overall head and neck cancer population. A, tissues derived from biopsies of HPV-positive head and neck cancers were stained with antibodies to human CD8, CD4, PD-1 by simple immunofluorescence analysis. Double immunofluorescence stainings for CD4 and PD-1, CD8 and PD-1, CD4 and Foxp3 were also conducted. For overall survival analysis, high and low levels of these various populations were defined using the median of tumor infiltration for these cells as cut-off values. B, PD-1+ cells and total number of PD-1+CD4+ and PD-1+CD8+ cells were measured in biopsies derived from all head and neck cancer patients, regardless of their HPV status. The relationship between the total number of these cells selected as a quantitative variable and the 18-month survival is shown. The blue line corresponds to this relationship, whereas the red line represents the upper or lower limits of the 95% CI.

Figure 3.

PD-1–positive infiltrating T cells positively correlate with survival in both HPV-positive head and neck cancer and in the overall head and neck cancer population. A, tissues derived from biopsies of HPV-positive head and neck cancers were stained with antibodies to human CD8, CD4, PD-1 by simple immunofluorescence analysis. Double immunofluorescence stainings for CD4 and PD-1, CD8 and PD-1, CD4 and Foxp3 were also conducted. For overall survival analysis, high and low levels of these various populations were defined using the median of tumor infiltration for these cells as cut-off values. B, PD-1+ cells and total number of PD-1+CD4+ and PD-1+CD8+ cells were measured in biopsies derived from all head and neck cancer patients, regardless of their HPV status. The relationship between the total number of these cells selected as a quantitative variable and the 18-month survival is shown. The blue line corresponds to this relationship, whereas the red line represents the upper or lower limits of the 95% CI.

Close modal

A correlation was also shown between the total number of PD-1+CD4+ and PD-1+CD8+ infiltrating T cells and 18-month overall survival in the whole population of head and neck patients whatever their HPV status (P = 0.009; Fig. 3B).

In contrast, the levels of infiltration by CD8+ T cells or CD4+ T cells or Foxp3 regulatory CD4+ T cells infiltration were not correlated with overall survival in HPV-positive head and neck tumors (Fig. 3A). However, as previously reported by us and other authors (17, 27), the number of Foxp3 regulatory T cells was correlated with better 18-month overall survival in the whole population of head and neck cancer patients (P = 0.0033; Supplementary Fig. S3).

PD-L1 expression was not correlated with overall survival or disease free survival in either the whole head and neck cancer cohort or in HPV-positive patients (Supplementary Fig. S2). In bivariate analysis including PD-L1 and various subpopulation of PD-1 cells each analyzed separately, we showed that only PD-1+CD4+ T cells, the total number of PD-1+CD4+ and PD-1+CD8+ T cells and the total number of PD-1 cells remained significant (data not shown).

Phenotypic and functional characterization of PD-1+ T cells infiltrating head and neck cancer

PD-1+ T cells infiltrated both the stroma and the nest of tumor cells. As shown in Fig. 4A, close contact was shown between PD-L1 tumor cells and PD-1+ cells strongly suggesting that a negative signal was delivered to these PD-1+ tumor–infiltrating T cells. Interestingly, in the whole population, a correlation was observed between PD-L1 expression and the number of tumor infiltrating CD4+PD-1+ T cells (P = 0.029). A trend was also observed between PD-L1 expression and the number of PD-1+ CD4+ T cells and PD1+CD8+ T cells (P = 0.07) and also the total number of PD-1+ T cells (P = 0.089; Supplementary Fig. S4).

Figure 4.

Phenotypic analysis of PD-1–positive T cells in head and neck cancer patients. A, head and neck cancer biopsies were stained with anti–PD-1 (second image from the right) and anti–PD-L1 antibodies (second image from the left). Double immunofluorescence staining for PD-1 and PD-L1 are shown on the right. Isotype control antibodies for anti–PD-L1 (left) and PD-1 (shown in Fig. 2) were included in each experiment. B, head and neck cancer biopsies were dissociated by DNAse and collagenase and cells were stained with CD45, Dye780, and CD3. Live T cells were then stained with CD4, CD8, PD-1, HLA-DR, and CD38. After gating on PD-1+CD4+ or PD-1CD4+ or PD-1+CD8+ or CD8+PD-1 cells, expression of the activation markers (HLA-DR and CD38) were compared on the various PD-1–positive and PD-1–negative T-cell populations. A representative experiment is shown in B. Mean (± SD) expression of HLA-DR and CD38 on CD3+ T cells, CD4+ T cells, and CD8+ T cells from 14 patients is shown in C. D, twenty PD-1+ T cells were selected at random from 16 HPV-positive (black square) or HPV-negative (white square) head and neck cancers. The number of cells costained with Tim-3 is shown with the mean indicated for the PD-1+Tim-3 and PD-1+Tim-3+ groups.

Figure 4.

Phenotypic analysis of PD-1–positive T cells in head and neck cancer patients. A, head and neck cancer biopsies were stained with anti–PD-1 (second image from the right) and anti–PD-L1 antibodies (second image from the left). Double immunofluorescence staining for PD-1 and PD-L1 are shown on the right. Isotype control antibodies for anti–PD-L1 (left) and PD-1 (shown in Fig. 2) were included in each experiment. B, head and neck cancer biopsies were dissociated by DNAse and collagenase and cells were stained with CD45, Dye780, and CD3. Live T cells were then stained with CD4, CD8, PD-1, HLA-DR, and CD38. After gating on PD-1+CD4+ or PD-1CD4+ or PD-1+CD8+ or CD8+PD-1 cells, expression of the activation markers (HLA-DR and CD38) were compared on the various PD-1–positive and PD-1–negative T-cell populations. A representative experiment is shown in B. Mean (± SD) expression of HLA-DR and CD38 on CD3+ T cells, CD4+ T cells, and CD8+ T cells from 14 patients is shown in C. D, twenty PD-1+ T cells were selected at random from 16 HPV-positive (black square) or HPV-negative (white square) head and neck cancers. The number of cells costained with Tim-3 is shown with the mean indicated for the PD-1+Tim-3 and PD-1+Tim-3+ groups.

Close modal

PD-1 is a hallmark of both activated and exhausted T cells (28). We found that PD-1 positive T cells, either CD4 or CD8+ T cells, expressed higher levels of HLA-DR and CD38 than PD-1 negative T cells (Fig. 4B and C). No difference in this activation state appeared to occur between HPV-positive or HPV-negative tumors (data not shown). Tim-3 expression was also assessed, as it has been associated with an exhausted phenotype of PD-1+ T cells (29, 30). One-half of PD-1+ T cells in head and neck cancer were found to express Tim-3 (Fig. 4D and Supplementary Fig. S5A). The same number of PD-1+ T cells in HPV-positive and HPV-negative tumors presented hallmarks of exhausted T cells (Fig. 4D).

We also showed that PD-1–positive cells did not express the Foxp3 marker of regulatory T cells, despite the fact that, in some cases, PD-1–positive cells and Foxp3 regulatory T cells were in close contact (Supplementary Fig. S5B).

To assess whether the functionality of these T cells could be enhanced, fresh TIL were cocultured with tumor cells in the presence or absence of anti-PD-1 mAb and a pool of E7 peptides. In 3 (2 HPV negative and 1 HPV positive) of 4 patients, a significant increase of IFNγ positive T cells was shown by Elispot in the presence of CT-011, an anti–PD-1 mAb (Fig. 5). An isotype control antibody had no effect on enhanced IFNγ production (Fig. 5).

Figure 5.

Functional analysis of intratumoral PD-1+ T cells after blockade of the PD-1/PD-L1 pathway. Head and neck cancer biopsies from 4 head and neck cancer patients (3 HPV-negative and 1 HPV-positive) were dissociated by DNAse and collagenase, resulting in cell suspensions containing both tumor cells and stromal cells including T cells. These cells (2 × 105) were transferred to IFNγ Elispot plates incubated with medium, anti-PD-1, or isotype control antibodies in the presence or absence of a pool of E7 peptides for 48 hours. All tests were conducted in triplicate. The number of spots (mean ± standard deviation) is shown. *, P < 0.05.

Figure 5.

Functional analysis of intratumoral PD-1+ T cells after blockade of the PD-1/PD-L1 pathway. Head and neck cancer biopsies from 4 head and neck cancer patients (3 HPV-negative and 1 HPV-positive) were dissociated by DNAse and collagenase, resulting in cell suspensions containing both tumor cells and stromal cells including T cells. These cells (2 × 105) were transferred to IFNγ Elispot plates incubated with medium, anti-PD-1, or isotype control antibodies in the presence or absence of a pool of E7 peptides for 48 hours. All tests were conducted in triplicate. The number of spots (mean ± standard deviation) is shown. *, P < 0.05.

Close modal

In 1 HPV-positive patient, E7 peptides slightly increased the frequency of CT-011 induced IFNγ secretion (Fig. 5).

Vaccination increases PD-1 and PD-1 blockade synergizes with the vaccine antitumor effect

To explain this paradoxical good prognostic value of PD-1+ T-cell infiltration in HPV-associated head and neck cancer, we modelized this phenomenon in a preclinical model.

When mice were grafted with an epithelial tumor cell line (TC-1) expressing the E7 protein derived from HPV, tumors were poorly infiltrated by CD8+ T cells that expressed low levels of PD-1 (6%; Fig. 6A).

Figure 6.

PD-1 induced on CD8+ T cells after vaccination is required for the efficacy of PD-1–PD-L1 blockade. A, mice were s.c. grafted with the E7-expressing TC-1 epithelial cell line. At day 9, mice were vaccinated twice at a 9-day interval with a vaccine composed of a nonreplicative delivery vector, the B subunit of Shiga toxin, coupled to an E7 polypeptide (STxB-E7) used at 20 μg. αGalCer (1 μg) was mixed with the vaccine during the prime. PD-1 expression on total CD8 or E749–57 specific CD8+ T cells was determined on dissociated cells by cytometry analysis from biopsies of mice tumors (previously vaccinated or not) at a time corresponding to 5 to 6 days after the second vaccination. B, PD-1 expression on total CD8+ T cells and E749–57–specific CD8+ T cells in mice vaccinated or not with STxB-E7. PD-1 expression on E749–57–specific CD8+ T cells from the nonvaccinated group is not shown, as we could not detect these cells in this group. This experiment is representative of 3 experiments with 4 mice per group. C, the epithelia tumor cell line, TC-1, was stained in vitro with anti–PD-L1 or isotype control antibodies. D, mice were s.c. grafted with the E7-expressing TC-1 epithelial cell line. Mice were then either vaccinated or not vaccinated at day 9 and day 18 with STxB-E7 mixed during the prime with αGalCer. The vaccine was combined with anti–PD-L1 or isotype control antibodies (200 μg/injection) that were administered 1, 4, and 9 days following the vaccine injection. A group of mice receiving the anti–PD-L1 mAb without the vaccine was also included. Tumor growth was monitored every 3 days. Results shown are representative of 3 experiments with 7 to 10 mice per group. For statistical analysis, each group was compared with the control group. *, P < 0.05; **, P < 0.01; ***, P < 0.005.

Figure 6.

PD-1 induced on CD8+ T cells after vaccination is required for the efficacy of PD-1–PD-L1 blockade. A, mice were s.c. grafted with the E7-expressing TC-1 epithelial cell line. At day 9, mice were vaccinated twice at a 9-day interval with a vaccine composed of a nonreplicative delivery vector, the B subunit of Shiga toxin, coupled to an E7 polypeptide (STxB-E7) used at 20 μg. αGalCer (1 μg) was mixed with the vaccine during the prime. PD-1 expression on total CD8 or E749–57 specific CD8+ T cells was determined on dissociated cells by cytometry analysis from biopsies of mice tumors (previously vaccinated or not) at a time corresponding to 5 to 6 days after the second vaccination. B, PD-1 expression on total CD8+ T cells and E749–57–specific CD8+ T cells in mice vaccinated or not with STxB-E7. PD-1 expression on E749–57–specific CD8+ T cells from the nonvaccinated group is not shown, as we could not detect these cells in this group. This experiment is representative of 3 experiments with 4 mice per group. C, the epithelia tumor cell line, TC-1, was stained in vitro with anti–PD-L1 or isotype control antibodies. D, mice were s.c. grafted with the E7-expressing TC-1 epithelial cell line. Mice were then either vaccinated or not vaccinated at day 9 and day 18 with STxB-E7 mixed during the prime with αGalCer. The vaccine was combined with anti–PD-L1 or isotype control antibodies (200 μg/injection) that were administered 1, 4, and 9 days following the vaccine injection. A group of mice receiving the anti–PD-L1 mAb without the vaccine was also included. Tumor growth was monitored every 3 days. Results shown are representative of 3 experiments with 7 to 10 mice per group. For statistical analysis, each group was compared with the control group. *, P < 0.05; **, P < 0.01; ***, P < 0.005.

Close modal

When mice were vaccinated with a nonreplicative delivery vector targeting dendritic cells, the B subunit of Shiga toxin, coupled to E7, 44% of CD8+ T cells expressed PD-1 (Fig. 6A and B). In addition, 89% of specific anti-E749–57 CD8+ T cells, induced by the vaccine, also expressed PD-1 (Fig. 6A and B). The vaccine therefore appeared to elicit PD-1 expression in both total and E749–57 specific CD8+ T cells. The vaccine alone had a partial inhibitory effect on the growth of established TC-1 tumors (Fig. 6D). This effect was significantly enhanced by the addition of anti-PD-L1 (Fig. 6D). In contrast, the anti-PD-L1 antibody alone had no effect on tumor growth (Fig. 6D), which may be explained by the low (6%) PD-1 expression in infiltrating T cells in the absence of vaccination (Fig. 6A). Similar results were obtained, when overall survival or the percentage of tumor-free mice were selected as endpoints (Supplementary Fig. S6).

This study shows that HPV-positive head and neck tumors were heavily infiltrated by PD-1+ cells, as the number of PD-1+CD4+ infiltrating T cells and the total number of PD-1+CD4+ and PD-1+CD8+ T cells were significantly higher in HPV-positive than in HPV-negative head and neck cancers. The relevance of these results is strengthened by the matching between HPV-positive and HPV-negative tumors for various confounding parameters (tumor–node–metastasis, treatment modalities, etc).

Surprisingly, we found that high levels of PD-1+ cells and the total number of PD-1+CD4+ and PD-1+CD8+ T cells correlated with better survival compared with low levels of infiltration by these cells in primary HPV-positive head and neck cancers.

These results were unexpected because of the known inhibitory function of PD-1 on T cells and immune cells (20, 31). In addition, in renal cell carcinoma, nasopharyngeal cancer and Hodgkin lymphoma, PD-1 expression on immune cells was correlated with poor prognosis and was associated with shorter survival (32–34). However, Hsu and colleagues showed that the prognostic value of PD-1 differed between CD4+ and CD8+ T cells (33). The other studies did not conduct double immunofluorescence staining or cytometry analysis to more precisely characterize PD-1 positive cells that can be expressed by many types of infiltrating cells (NK, B cells, macrophages, etc.). In the present study, we showed that most (>95%) PD-1 positive cells corresponded to T cells (Supplementary Fig. S1) and that the prognostic value of PD-1 was associated with the total number of CD4+ and CD8+ T cells expressing this marker. It should also be mentioned that in contrast to many tumors, CD8+ T cells are associated with bad prognosis in renal cell carcinoma and hodgkin lymphoma supporting the fact that the clinical significance of molecules expressed by these cells could also vary depending on the tumor histology (35, 36).

Interestingly, recent studies have reported that follicular lymphomas infiltrated by PD-1+ T cells were associated with a lower risk of transformation, a higher progression-free survival and a better overall survival than tumors not infiltrated by PD-1+ cells (37). PD-1 was preferentially expressed by infiltrating CD4+ T cells in these follicular lymphomas, as also observed in our study. In line with our results, PD-1 mRNA expression determined by reverse transcriptase-PCR (RT-PCR) in colorectal carcinoma was also associated with a good prognosis (38). It is therefore noteworthy that the 3 tumors (lymphoma, colorectal cancer, and head and neck cancer), in which PD-1 expression was significantly associated with a favorable outcome (37, 38), were also those with an unexpected clinical benefit of Treg infiltration (17, 39, 40). The present study, conducted in an independent series of patients, confirms our previous findings that high levels of Treg in head and neck cancer patients were also positively correlated with a better prognosis (17). These results concerning the good prognostic value of intratumor regulatory T cells in head and neck cancer have subsequently been reproduced by various groups (27, 41). In addition, the frequency of regulatory Foxp3+CD4+ T cells has been shown to be higher in patients with HNSCC with no evidence of disease after oncologic therapy, than in patients with active disease (42). The plasticity of regulatory T cells, transient expression of Foxp3 by activated T cells and a possible role of regulatory T cells in the inhibition of deleterious cancer inflammation could explain the paradoxically good clinical prognosis associated with regulatory T cells in some circumstances (43, 44).

Several hypotheses can also be proposed to explain this paradoxical favorable outcome associated with PD-1 expression on T cells. PD-1 is upregulated on lymphocytes following T-cell receptor activation and remains elevated in the context of persistent antigen-specific immune stimulation (45). The absence of co-expression of Tim-3 and PD-1 in one-half of PD-1–positive cells strongly suggests that some PD-1+ T cells in the tumor microenvironment of HPV-positive head and neck cancers are not exhausted T cells (Fig. 4). We did not assess the expression of other inhibitory receptors (Lag-3, CTLA-4…) on PD-1+ T cells, but Tim-3 is the most frequent coinhibitory receptors associated with PD-1+ T cells in tumor and an hallmark of exhausted T cells (29, 46). It has been reported that high PD-1 levels on T cells more closely reflect their “exhausted status” than intermediate PD-1 levels. However, in our study, PD-1 expression was homogeneous and it was, therefore, difficult to divide T cells into 2 groups. Some of our PD-1 infiltrating T cells remained responsive to anti-PD-1 in vitro (Fig. 5). From these results, PD-1 expression also indicates, that T-cell activation has occurred during the development of HPV-associated head and neck cancers and PD-1 could, therefore, also be viewed as a witness of the antitumor immune response.

Indeed, we showed that PD-1–positive T cells expressed higher levels of the HLA-DR and CD38 activation markers, than PD-1–negative T cells (Fig. 4). Although this response was not effective to clear the tumor, it may have slowed tumor growth compared with a clinical setting associated with absence of host response to the tumor. Although the tumor specificity of PD-1 expressing T cells could not be shown because of the small number of cells present in tumor biopsies, it has been previously shown that tumor-reactive CD8+ T cells generated after in vitro culture of dissociated biopsies of melanoma were more frequently derived from PD-1 expressing T cells present at the beginning of culture (47). In preclinical tumor models, the CD137+CD8+ T cells were enriched in antitumor effector T cells (48).

Our preclinical model showed that grafting of the E7-expressing TC-1 epithelial cell line, which constitutively expresses PD-L1 led to tumor growth without therapy. Analysis of its tumor microenvironment showed the absence of PD-1 expression by CD8+T cells. In contrast, activation of an anti-HPV response by an anti-E7 vaccine, elicited PD-1 expression by CD8+T cells and E7-specific T cells, which were associated with partial regression of the tumor. In humans, it has been shown that TIL derived from spontaneously regressing melanomas also express substantial levels of PD-1 (49).

Another important result of this study is that the efficacy of anti-PD-L1 mAb depended on the presence of PD-1 on specific T cells induced after vaccination. In preclinical models, immunotherapy approaches that should lead to upregulation of PD-1 following activation of the immune system, such as recombinant cytokines or stimulatory antibodies or transfer of activated adoptive T cells, synergized with blockade of the PD-1–PD-L1 pathway (21, 25, 50).

Consistent with these results, a recent report showed that expression of PD-L1, the ligand of PD-1, which was considered to be an immunosuppressive molecule, positively correlated with better survival in melanoma (51). In melanoma, PD-L1 is mainly induced by IFNγ produced by infiltrating T cells and the distribution of T cells within the tumor was correlated with that of PD-L1 (51). In line with these results, in patients treated with anti-PD-1 mAb, tumor cell surface PD-L1 expression appeared to be correlated with the likelihood of response to treatment, possibly because of a reprogramming of infiltrating antitumor PD-1+T cells (52). PD-1 and PD-L1 expression may, therefore, represent surrogate markers of endogenous antitumor immune response, explaining their unexpected association with good prognosis in some tumors in which PD-L1 expression is not oncogene driven. Indeed, in glioma, loss of tumor suppressor PTEN function increased PD-L1 (53). In our study, PD-L1 was not associated with a good prognosis in head and neck cancer patients, regardless of HPV status, but low PTEN expression was detected in the majority of these tumors (54).

This study, therefore, revisits the significance of PD-1–positive tumor-infiltrating T cells in cancer. The inhibitory function linked to PD-1 should not mask the fact that its detection could also reflect a past antitumor immune response ready to be reprogrammed by PD-1–PD-L1 blockade.

W.H. Fridman is a consultant/advisory board member of CureTech LTD. No potential conflicts of interest were disclosed by the other authors.

Conception and design: C. Badoual, S. Hans, E. Tartour

Development of methodology: C. Badoual, N. Merillon, C. Van Ryswick, N. Benhamouda, E. Levionnois, A. Si-Mohamed, N. Besnier, A. Gey, H. Pere, C.L. Guerin, A. Chauvat, C. Alanio, D. Olive, D. Brasnu, E. Tartour

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C. Badoual, S. Hans, N. Merillon, N. Benhamouda, E. Levionnois, M. Nizard, A. Si-Mohamed, N. Besnier, A. Gey, H. Pere, A. Chauvat, E. Dransart, C. Alanio, S. Albert, B. Barry, F. Sandoval, F. Quintin-Colonna, P. Bruneval, W.H. Fridman, F.M. Lemoine, S. Oudard, L. Johannes, D. Olive, D. Brasnu, E. Tartour

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C. Badoual, S. Hans, N. Merillon, C. Van Ryswick, P. Ravel, N. Benhamouda, E. Levionnois, A. Si-Mohamed, A. Gey, R. Rotem-Yehudar, H. Pere, C.L. Guerin, A. Chauvat, C. Alanio, F. Sandoval, F. Quintin-Colonna, P. Bruneval, W.H. Fridman, F.M. Lemoine, S. Oudard, L. Johannes, E. Tartour

Writing, review, and/or revision of the manuscript: C. Badoual, S. Hans, R. Rotem-Yehudar, F. Sandoval, D. Olive, D. Brasnu, E. Tartour

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C. Badoual, S. Hans, E. Tartour

Study supervision: C. Badoual, E. Tartour

This work was supported by grants from the Ligue contre le Cancer, the Agence Nationale de la Recherche (ANR), the Association pour la Recherche contre le Cancer (ARC), Labex, Immuno-Oncology, Canceropole Region Ile de France, Fonds d'amorçage AP-HP, and Institut National du Cancer (INCA).

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.
Ramqvist
T
,
Dalianis
T
. 
Oropharyngeal cancer epidemic and human papillomavirus
.
Emerg Infect Dis
2010
;
16
:
1671
7
.
2.
Gillison
ML
,
D'Souza
G
,
Westra
W
,
Sugar
E
,
Xiao
W
,
Begum
S
, et al
Distinct risk factor profiles for human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck cancers
.
J Natl Cancer Inst
2008
;
100
:
407
20
.
3.
Ang
KK
,
Harris
J
,
Wheeler
R
,
Weber
R
,
Rosenthal
DI
,
Nguyen-Tan
PF
, et al
Human papillomavirus and survival of patients with oropharyngeal cancer
.
N Engl J Med
2010
;
363
:
24
35
.
4.
Marur
S
,
D'Souza
G
,
Westra
WH
,
Forastiere
AA
. 
HPV-associated head and neck cancer: a virus-related cancer epidemic
.
Lancet Oncol
2010
;
11
:
781
9
.
5.
Fakhry
C
,
Westra
WH
,
Li
S
,
Cmelak
A
,
Ridge
JA
,
Pinto
H
, et al
Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial
.
J Natl Cancer Inst
2008
;
100
:
261
9
.
6.
Chung
CH
,
Gillison
ML
. 
Human papillomavirus in head and neck cancer: its role in pathogenesis and clinical implications
.
Clin Cancer Res
2009
;
15
:
6758
62
.
7.
Fischer
CA
,
Zlobec
I
,
Green
E
,
Probst
S
,
Storck
C
,
Lugli
A
, et al
Is the improved prognosis of p16 positive oropharyngeal squamous cell carcinoma dependent of the treatment modality?
Int J Cancer
2010
;
126
:
1256
62
.
8.
Williams
R
,
Lee
DW
,
Elzey
BD
,
Anderson
ME
,
Hostager
BS
,
Lee
JH
. 
Preclinical models of HPV+ and HPV- HNSCC in mice: an immune clearance of HPV+ HNSCC
.
Head Neck
2009
;
31
:
911
8
.
9.
Cesson
V
,
Rivals
JP
,
Escher
A
,
Piotet
E
,
Thielemans
K
,
Posevitz
V
, et al
MAGE-A3 and MAGE-A4 specific CD4(+) T cells in head and neck cancer patients: detection of naturally acquired responses and identification of new epitopes
.
Cancer Immunol Immunother
2011
;
60
:
23
35
.
10.
Sirianni
N
,
Ha
PK
,
Oelke
M
,
Califano
J
,
Gooding
W
,
Westra
W
, et al
Effect of human papillomavirus-16 infection on CD8+ T-cell recognition of a wild-type sequence p53264-272 peptide in patients with squamous cell carcinoma of the head and neck
.
Clin Cancer Res
2004
;
10
:
6929
37
.
11.
Albers
A
,
Abe
K
,
Hunt
J
,
Wang
J
,
Lopez-Albaitero
A
,
Schaefer
C
, et al
Antitumor activity of human papillomavirus type 16 E7-specific T cells against virally infected squamous cell carcinoma of the head and neck
.
Cancer Res
2005
;
65
:
11146
55
.
12.
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
.
13.
Tanaka
K
,
Jinhua
P
,
Omura
K
,
Azuma
M
. 
Multipotency of CD11b(high)Gr-1(+) immature myeloid cells accumulating in oral squamous cell carcinoma-bearing mice
.
Oral Oncol
2007
;
43
:
586
92
.
14.
Mandapathil
M
,
Szczepanski
MJ
,
Szajnik
M
,
Ren
J
,
Lenzner
DE
,
Jackson
EK
, et al
Increased ectonucleotidase expression and activity in regulatory T cells of patients with head and neck cancer
.
Clin Cancer Res
2009
;
15
:
6348
57
.
15.
Chen
Z
,
Malhotra
PS
,
Thomas
GR
,
Ondrey
FG
,
Duffey
DC
,
Smith
CW
, et al
Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer
.
Clin Cancer Res
1999
;
5
:
1369
79
.
16.
Badoual
C
,
Bouchaud
G
,
Agueznay
Nel H
,
Mortier
E
,
Hans
S
,
Gey
A
, et al
The soluble alpha chain of interleukin-15 receptor: a proinflammatory molecule associated with tumor progression in head and neck cancer
.
Cancer Res
2008
;
68
:
3907
14
.
17.
Badoual
C
,
Hans
S
,
Rodriguez
J
,
Peyrard
S
,
Klein
C
,
Agueznay
Nel H
, et al
Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers
.
Clin Cancer Res
2006
;
12
:
465
72
.
18.
van der Burg
SH
,
Piersma
SJ
,
de Jong
A
,
van der Hulst
JM
,
Kwappenberg
KM
,
van den Hende
M
, et al
Association of cervical cancer with the presence of CD4 +regulatory T cells specific for human papillomavirus antigens
.
Proc Natl Acad Sci U S A
2007
;
104
:
12087
92
.
19.
Sharpe
AH
,
Wherry
EJ
,
Ahmed
R
,
Freeman
GJ
. 
The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection
.
Nat Immunol
2007
;
8
:
239
45
.
20.
Pardoll
DM
. 
The blockade of immune checkpoints in cancer immunotherapy
.
Nat Rev Cancer
2012
;
12
:
252
64
.
21.
Strome
SE
,
Dong
H
,
Tamura
H
,
Voss
SG
,
Flies
DB
,
Tamada
K
, et al
B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma
.
Cancer Res
2003
;
63
:
6501
5
.
22.
Katou
F
,
Ohtani
H
,
Watanabe
Y
,
Nakayama
T
,
Yoshie
O
,
Hashimoto
K
. 
Differing phenotypes between intraepithelial and stromal lymphocytes in early-stage tongue cancer
.
Cancer Res
2007
;
67
:
11195
201
.
23.
Adotevi
O
,
Vingert
B
,
Freyburger
L
,
Shrikant
P
,
Lone
YC
,
Quintin-Colonna
F
, et al
B subunit of shiga toxin-based vaccines synergize with {alpha}-galactosylceramide to break tolerance against self antigen and elicit antiviral immunity
.
J Immunol
2007
;
179
:
3371
9
.
24.
Benson
DM
 Jr
,
Bakan
CE
,
Mishra
A
,
Hofmeister
CC
,
Efebera
Y
,
Becknell
B
, et al
The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody
.
Blood
2010
;
116
:
2286
94
.
25.
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
.
26.
Xerri
L
,
Chetaille
B
,
Serriari
N
,
Attias
C
,
Guillaume
Y
,
Arnoulet
C
, et al
Programmed death 1 is a marker of angioimmunoblastic T-cell lymphoma and B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia
.
Hum Pathol
2008
;
39
:
1050
8
.
27.
Loose
D
,
Signore
A
,
Bonanno
E
,
Vermeersch
H
,
Dierckx
R
,
Deron
P
, et al
Prognostic value of CD25 expression on lymphocytes and tumor cells in squamous-cell carcinoma of the head and neck
.
Cancer Biother Radiopharm
2008
;
23
:
25
33
.
28.
Sauce
D
,
Almeida
JR
,
Larsen
M
,
Haro
L
,
Autran
B
,
Freeman
GJ
, et al
PD-1 expression on human CD8 T cells depends on both state of differentiation and activation status
.
AIDS
2007
;
21
:
2005
13
.
29.
Fourcade
J
,
Sun
Z
,
Benallaoua
M
,
Guillaume
P
,
Luescher
IF
,
Sander
C
, et al
Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients
.
J Exp Med
2010
;
207
:
2175
86
.
30.
Sakuishi
K
,
Apetoh
L
,
Sullivan
JM
,
Blazar
BR
,
Kuchroo
VK
,
Anderson
AC
. 
Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity
.
J Exp Med
2010
;
207
:
2187
94
.
31.
Terme
M
,
Ullrich
E
,
Aymeric
L
,
Meinhardt
K
,
Desbois
M
,
Delahaye
N
, et al
IL-18 induces PD-1-dependent immunosuppression in cancer
.
Cancer Res
2011
;
71
:
5393
9
.
32.
Thompson
RH
,
Dong
H
,
Lohse
CM
,
Leibovich
BC
,
Blute
ML
,
Cheville
JC
, et al
PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma
.
Clin Cancer Res
2007
;
13
:
1757
61
.
33.
Hsu
MC
,
Hsiao
JR
,
Chang
KC
,
Wu
YH
,
Su
IJ
,
Jin
YT
, et al
Increase of programmed death-1-expressing intratumoral CD8 T cells predicts a poor prognosis for nasopharyngeal carcinoma
.
Mod Pathol
2010
;
23
:
1393
403
.
34.
Muenst
S
,
Hoeller
S
,
Dirnhofer
S
,
Tzankov
A
. 
Increased programmed death-1 +tumor-infiltrating lymphocytes in classical Hodgkin lymphoma substantiate reduced overall survival
.
Hum Pathol
2009
;
40
:
1715
22
.
35.
Nakano
O
,
Sato
M
,
Naito
Y
,
Suzuki
K
,
Orikasa
S
,
Aizawa
M
, et al
Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity
.
Cancer Res
2001
;
61
:
5132
6
.
36.
Asano
N
,
Oshiro
A
,
Matsuo
K
,
Kagami
Y
,
Ishida
F
,
Suzuki
R
, et al
Prognostic significance of T-cell or cytotoxic molecules phenotype in classical Hodgkin's lymphoma: a clinicopathologic study
.
J Clin Oncol
2006
;
24
:
4626
33
.
37.
Carreras
J
,
Lopez-Guillermo
A
,
Roncador
G
,
Villamor
N
,
Colomo
L
,
Martinez
A
, et al
High numbers of tumor-infiltrating programmed cell death 1-positive regulatory lymphocytes are associated with improved overall survival in follicular lymphoma
.
J Clin Oncol
2009
;
27
:
1470
6
.
38.
Mlecnik
B
,
Tosolini
M
,
Charoentong
P
,
Kirilovsky
A
,
Bindea
G
,
Berger
A
, et al
Biomolecular network reconstruction identifies T-cell homing factors associated with survival in colorectal cancer
.
Gastroenterology
2010
;
138
:
1429
40
.
39.
Carreras
J
,
Lopez-Guillermo
A
,
Fox
BC
,
Colomo
L
,
Martinez
A
,
Roncador
G
, et al
High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma
.
Blood
2006
;
108
:
2957
64
.
40.
Salama
P
,
Phillips
M
,
Grieu
F
,
Morris
M
,
Zeps
N
,
Joseph
D
, et al
Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer
.
J Clin Oncol
2009
;
27
:
186
92
.
41.
Bron
L
,
Jandus
C
,
Andrejevic-Blant
S
,
Speiser
DE
,
Monnier
P
,
Romero
P
, et al
Prognostic value of arginase-II expression and regulatory T cell infiltration in head and neck squamous cell carcinoma
.
Int J Cancer. Epub
2012
Oct 3.
42.
Strauss
L
,
Bergmann
C
,
Szczepanski
M
,
Gooding
W
,
Johnson
JT
,
Whiteside
TL
. 
A unique subset of CD4+CD25highFoxp3+ T cells secreting interleukin-10 and transforming growth factor-beta1 mediates suppression in the tumor microenvironment
.
Clin Cancer Res
2007
;
13
:
4345
54
.
43.
Badoual
C
,
Hans
S
,
Fridman
WH
,
Brasnu
D
,
Erdman
S
,
Tartour
E
. 
Revisiting the prognostic value of regulatory T cells in patients with cancer
.
J Clin Oncol
2009
;
27
:
e5
6
;
author reply e7
.
44.
Pere
H
,
Tanchot
C
,
Bayry
J
,
Terme
M
,
Taieb
J
,
Badoual
C
, et al
Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer
.
Oncoimmunology
2012
;
1
:
326
33
.
45.
Wherry
EJ
,
Ha
SJ
,
Kaech
SM
,
Haining
WN
,
Sarkar
S
,
Kalia
V
, et al
Molecular signature of CD8+ T cell exhaustion during chronic viral infection
.
Immunity
2007
;
27
:
670
84
.
46.
Baitsch
L
,
Legat
A
,
Barba
L
,
Fuertes
Marraco SA
,
Rivals
JP
,
Baumgaertner
P
, et al
Extended co-expression of inhibitory receptors by human CD8 T-cells depending on differentiation, antigen-specificity and anatomical localization
.
PLoS One
2012
;
7
:
e30852
.
47.
Inozume
T
,
Hanada
K
,
Wang
QJ
,
Ahmadzadeh
M
,
Wunderlich
JR
,
Rosenberg
SA
, et al
Selection of CD8+PD-1+ lymphocytes in fresh human melanomas enriches for tumor-reactive T cells
.
J Immunother
2010
;
33
:
956
64
.
48.
Verbrugge
I
,
Hagekyriakou
J
,
Sharp
LL
,
Galli
M
,
West
A
,
McLaughlin
NM
, et al
Radiotherapy increases the permissiveness of established mammary tumors to rejection by immunomodulatory antibodies
.
Cancer Res
2012
;
72
:
3163
74
.
49.
Chapon
M
,
Randriamampita
C
,
Maubec
E
,
Badoual
C
,
Fouquet
S
,
Wang
SF
, et al
Progressive upregulation of PD-1 in primary and metastatic melanomas associated with blunted TCR signaling in infiltrating T lymphocytes
.
J Invest Dermatol
2011
;
131
:
1300
7
.
50.
Hirano
F
,
Kaneko
K
,
Tamura
H
,
Dong
H
,
Wang
S
,
Ichikawa
M
, et al
Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity
.
Cancer Res
2005
;
65
:
1089
96
.
51.
Taube
JM
,
Anders
RA
,
Young
GD
,
Xu
H
,
Sharma
R
,
McMiller
TL
, et al
Colocalization of inflammatory response with b7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape
.
Sci Transl Med
2012
;
4
:
127ra37
.
52.
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
.
53.
Parsa
AT
,
Waldron
JS
,
Panner
A
,
Crane
CA
,
Parney
IF
,
Barry
JJ
, et al
Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma
.
Nat Med
2007
;
13
:
84
8
.
54.
Snietura
M
,
Jaworska
M
,
Mlynarczyk-Liszka
J
,
Goraj-Zajac
A
,
Piglowski
W
,
Lange
D
, et al
PTEN as a prognostic and predictive marker in postoperative radiotherapy for squamous cell cancer of the head and neck
.
PLoS One
2012
;
7
:
e33396
.