Tumor antigen‑specific interleukin‑10‑producing T‑cell response in patients with head and neck squamous cell carcinoma
- Authors:
- Published online on: July 24, 2024 https://doi.org/10.3892/ol.2024.14589
- Article Number: 456
-
Copyright: © Horikawa et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Interleukin-10 (IL-10) is a pleiotropic cytokine that regulates immunological homeostasis through anti-inflammatory and/or immunostimulatory functions (1–3). IL-10 has been implicated in immunopathogenesis during tumor development and progression. Accumulating evidence indicates that IL-10 plays a key role in establishing and maintaining a protumor microenvironment as a potent immunosuppressive cytokine. In particular, major immunosuppressive cells, such as regulatory T cells (Tregs), myeloid-derived suppressor cells, and tumor-associated macrophages, in the tumor microenvironment utilize IL-10 as one of the multiple immunosuppressive mechanisms (4,5). Conversely, IL-10 can also exhibit immunostimulatory properties, including the induction of proliferation and cytotoxic activity of CD8+ T cells. Emmerich et al demonstrated that treatment with IL-10 could activate tumor-resident CD8+ T cells and suppress well-established large tumors in mouse tumor models (6). Guo et al also revealed that the IL-10-Fc fusion protein expands and enhances the cytotoxic function of terminally exhausted CD8+ tumor-infiltrating lymphocytes that do not respond to immune checkpoint inhibitors (7). Thus, IL-10 possesses diverse roles in tumor immunology and immunotherapy (8,9). In addition, the amount and timing of IL-10 exposure may critically impact its function in antitumor immunity.
IL-10 is produced by various cell types, including CD4+ T cells. Among CD4+ T cells, T helper (Th)1, Th2, Th17, and T regulatory type 1 (Tr1) cells are notable producers of IL-10 (10,11). Simultaneously, IL-10 can directly and/or indirectly suppress Th responses following specific antigen stimulation and potentially induce the formation of a negative feedback loop to regulate immune responses. To date, several reports have shown that tumor antigen-specific regulatory T cells that produce IL-10 exist in the peripheral circulation, as well as in the tumor microenvironment (12–14). Conversely, tumor antigen-specific Th effector cells producing IL-10 in patients with cancer have received considerably less attention. Regarding IL-10 expression in Th effector cells, IL-10 secretion from Th2 cells is stable, whereas that from Th1 and Th17 cells was found to be unstable and conditional (10). Thus, the role of tumor antigen-specific IL-10-producing T cells in antitumor responses is more complex than that of CD8+ cytotoxic T lymphocytes.
In the present study, we identified circulating tumor antigen-specific IL-10-producing T cells in patients with head and neck squamous cell carcinoma (HNSCC) and explored factors influencing the immunodynamics of IL-10-producing T cells.
Materials and methods
Patients and blood collection
During March 2019 to April 2021, blood samples were obtained at Gunma University Hospital from 18 patients with HNSCC who did not receive any anticancer drugs, radiotherapy, or surgery prior to blood collection. Patients with autoimmune diseases, severe infections, or receiving steroid treatment were excluded in this study. The median age of patients was 63.5 years (range: 48–77 years). Peripheral blood mononuclear cells (PBMCs) were isolated using density gradient centrifugation, followed by cryopreservation. This study was approved by the Ethics Committee of Gunma University Hospital (approval no. HS2017-152). Written informed consent was obtained from all patients.
Immunohistochemical expression of tumor antigens, p53 and MAGE-A4 in HNSCC
During March 2019 to April 2021, HNSCC samples were obtained by biopsy or surgical resection at Gunma University Hospital from the same patients who provided blood samples. The use of HNSCC samples was also approved by the Ethics Committee and patient consent was obtained. Immunohistochemical analysis of p53 and MAGE-A4 expression in tumor specimens was performed as described previously (15). Briefly, formalin-fixed paraffin-embedded specimens sectioned at 3 µm were deparaffinized. Antigen retrieval was achieved by boiling samples at 98°C for 30 min with 20% zinc sulfate solution and citrate buffer (pH 6.0) for p53 and MAGE-A4 staining, respectively. After blocking, slides were incubated for 2 h with primary antibodies (anti-p53 antibody, NCL-L-p53-DO7, NOVOCASTRA; anti-MAGE-A4 antibody, clone 57 B, MERCK), followed by overnight incubation at 4°C. Subsequently, slides were incubated with a secondary antibody (Histofine Simple Stain MAX-PO (MULTI), Nichirei), and the reaction products were detected with 3,3′-diaminobenzidine (DAB, DOJINDO, Kumamoto, Japan). Sections were counterstained with Mayer's hematoxylin.
The sections were evaluated by two independent, blinded researchers (H. Tada and K.C.). For p53, specific staining in >10% of tumor cells was defined as positive expression. For MAGE-A4, each specimen was considered positive if specific staining was present.
In vitro sensitization and interferon (IFN)-γ/IL-10 double-color enzyme-linked immunosorbent spot (ELISPOT) assay
Thawed PBMCs were cultured with recombinant tumor antigen protein (10 µg/ml of p53 or MAGE-A4) in a final volume of 0.5 ml AIM-V medium, supplemented with 10 IU/ml IL-2 and 5 ng/ml IL-7 in a 48-well tissue culture plate. After 4 days, AIM-V medium (0.5 ml) containing 10 IU/ml IL-2 was added to each well. After three days of culture, PBMCs were harvested as effector cells and examined for IFN-γ/IL-10 production using the ELISPOT assay. For blocking assay, mouse control IgG1κ (P3.6.2.8.1; eBioscience) or anti-lymphocyte activation gene 3 (Lag-3) antibodies (17B4; AdipoGen, Liestal, Switzerland) (10 µg/ml each) were added throughout the culture period.
ELISPOT assays were performed using the Human IFN-γ/IL-10 double-color ELISPOT kit (Cellular Technology Ltd., Cleveland, OH, USA) according to manufacturer protocol. Briefly, a 96-well plate was precoated with IFN-γ/IL-10 capture antibody and incubated at 4°C overnight. Harvested effector cells (1–5×104 cells/well) were plated into a precoated plate and co-cultured with PBMCs (1×105 cells/well) in the presence of p53 or MAGE-A4 protein (10 µg/ml each). The plates were incubated at 37°C for 24 h. After incubation, the plates were washed and developed with anti-human IFN-γ (FITC) and FITC-horseradish peroxidase and IL-10 (Biotin) and streptavidin-alkaline phosphatase, respectively. The number of spot-forming cells (SFC) in each well was counted using a CTL-ImmunoSpot Analyzer (Cellular Technology Ltd.). The mean number of spots in control wells (no protein) was subtracted from the mean number of spots in the experimental wells, and the results were expressed as SFC per 5×104 cells, as described previously (15). A T-cell response to a given tumor antigen was considered to be positive if at least 10 cells per 5×104 responder cells secreted IFN-γ or IL-10.
Flow cytometric analysis
Flow cytometry was performed using a FACSVerse flow cytometer (BD Biosciences) to analyze the proportion of T cells expressing immune checkpoint molecules in PBMCs, as reported previously (16). Briefly, cryopreserved PBMCs were thawed, blocked using BD Fc Block (BD Bioscience, San Jose, CA, USA), and stained with antibodies specific for CD3, CD4, CD8, programmed cell death-1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), T-cell immunoglobulin and mucin-domain containing-3 (Tim-3), and Lag-3. As a negative control, cells were stained with a mouse IgG isotype control (BD Biosciences). The data were analyzed using FlowJo software (TreeStar, Ashland, OR, USA). The gating strategy is illustrated in Fig. S1.
Statistical analysis
Statistical analyses were performed using GraphPad Prism software (version 8.0; GraphPad Software, San Diego, CA, USA). Unpaired two-tailed t-test was performed to determine the presence of a significant difference between the number of SFC in protein-stimulated and unstimulated wells, as described previously (17). Fisher's exact test of independence was used to determine differences in categorical variables. Kaplan-Meier curves were plotted and compared using log-rank tests to compare survival curves between patients with and without p53-specific IL-10 production. P<0.05 was considered to indicate a statistically significant difference.
Results
Patients' characteristics and p53 and MAGE-A4 expression
Table I summarizes the characteristics of the included patients. The primary tumor sites included the larynx (n=6), oropharynx (n=5), and hypopharynx (n=7). Immunohistochemical analyses were performed on 17 available tumor specimens. Fig. 1A and B show representative immunohistochemical staining results for p53 and MAGE-A4. p53 and MAGE-A4 were detected in 8 (47.1%) and 13 (76.5%) patients, respectively (Table I).
Tumor antigen-specific IFN-γ/IL-10 production
In vitro, PBMCs stimulated with p53 or MAGE-A4 protein were evaluated using IFN-γ/IL-10 double-color ELISPOT assays (Fig. 2). Of the 18 patients with HNSCC, 2 (11.1%) and 9 (50.0%) patients showed p53-specific IFN-γ and IL-10 production, respectively. Meanwhile, MAGE-A4 specific IFN-γ and IL-10 production were detected in 4 (28.6%) and 7 (50.0%) of 14 patients (Table I). Three patients (pt-8, 12, 16) exhibited both IFN-γ and IL-10 production in response to the same tumor antigen. In the p53-specific responses, IL-10-producing T cells were observed in significantly more patients than IFN-γ producing T cells (P=0.0275, Table II). There was no significant correlation between clinical factors and tumor antigen-specific IL-10 production (Table SI). To evaluate the prognostic significance of p53-specific IL-10 production, Kaplan-Meier survival analyses were performed for patients with and without p53-specific IL-10 production (Fig. S2). Although patients with p53-specific IL-10 production appeared to have a better prognosis for overall survival, the difference was non-significant (overall survival, P=0.2518; relapse-free survival, P=0.5868).
Comparison of proportions of T-cells expressing immune checkpoint molecules
To further elucidate the immunological context underlying tumor antigen-specific IL-10 production, we focused on p53-specific IL-10 production and determined the proportion of T cells expressing immune checkpoint molecules in the peripheral blood. In both CD4+ and CD8+ T cells, the proportion of T cells expressing Lag-3 was significantly lower in patients who exhibited p53-specific IL-10 production than in those who did not, as shown in Fig. 3.
Enhancement of tumor antigen-specific IL-10 production by blockade of Lag-3
Finally, we investigated whether anti-Lag-3 antibodies enhanced tumor antigen-specific IL-10 production. In some patients, Lag-3 blockade significantly enhanced tumor antigen-specific IL-10 production (Fig. 4A and B). Meanwhile, Lag-3 blockade failed to enhance p53 and MAGE-A4-specific IFN-γ production in all patients (Fig. S3).
Discussion
In the current study, we demonstrated that tumor antigen-specific IL-10-producing T cells circulate in the peripheral blood of patients with HNSCC, and their detection rate was significantly higher than that of IFN-γ-producing T cells. In patients with HNSCC, similar to other types of cancers, various immunosuppressive mechanisms are activated, and the functions of relevant effector cells are suppressed not only within the tumor sites but also at systemic sites, including the peripheral blood, bone marrow, and lymph nodes (18–20). Our results may reflect the systemic immunosuppressive status of patients with HNSCC. In some patients whose tumors did not express p53 or MAGE-A4, we detected the presence of tumor antigen-specific T-cells. Consistently, Heusinkveld et al (21) and Hoffmann et al (22,23) reported similar findings regarding p53. The authors discussed the possibility that p53-negative tumors represent immune escape variants and/or harbor p53 mutations that do not result in overexpression. There was no significant association between tumor antigen-specific IL-10-producing T cells and clinical factors, including prognosis, mirroring the high heterogeneity of IL-10-producing T cells and the dual function of IL-10 in tumor promotion and suppression. To date, high serum IL-10 levels have been associated with poor prognosis in several malignancies, including gastric cancer (24), malignant myeloma (25), and lung cancer (26). In contrast, studies have shown that serum IL-10 levels do not correlate with prognosis (27–29). More recently, a relationship between IL-10 and the clinical benefits of immune checkpoint inhibitors has been reported (30,31). The level of serum IL-10 and percentage of CD4+ IL-10+ PBMCs were associated with prognosis and treatment response in patients treated with immune checkpoint inhibitors, respectively. However, IL-10 is produced not only by tumor antigen-specific T cells but also by several immune cells, including dendritic cells, macrophages, B cells, and neutrophils (32). To clarify the clinical significance of tumor antigen-specific IL-10 production in patients with cancer, it is necessary to consider the types of tumor antigens and CD4+ T cell subsets that produce IL-10. Furthermore, it is also essential to elucidate the relationship between IL-10-producing T cells within the tumor microenvironment and tumor characteristics such as PD-L1 expression, tumor mutation burden, and microsatellite instability.
Notably, the proportion of Lag-3+ T-cells was significantly lower in patients with p53-specific IL-10-producing T cells. Lag-3 is an immune inhibitory checkpoint expressed on exhausted CD4+ and CD8+ T cells in the context of persistent tumor antigen stimulation, as well as on immune regulatory cells, including Tregs and Tr1 cells (33,34). Therefore, in addition to the exhausted status of CD4+ and CD8+ T cells, Lag-3+ regulatory cells may be involved in tumor antigen-specific IL-10-producing T-cell responses. Particularly, CD4+ Tr1 cells are induced in the periphery upon antigen stimulation, producing high amounts of IL-10 and exhibiting robust immunosuppressive effects (35). As expected, the blockade of Lag-3 could reinvigorate tumor antigen-specific IL-10 production in some patients, suggesting the existence of a mechanism through which IL-10 production from T cells may be suppressed by IL-10 from Lag-3+ regulatory cells. Conversely, tumor antigen-specific IFN-γ production was not induced. Matsuzaki et al have reported that CD8+Lag-3+PD-1+ T cells were more impaired in IFN-γ/tumor necrosis factor (TNF)-α production than Lag-3+PD-1− or Lag-3−PD-1− T cell subsets in NY-ESO-1-specific CD8+ T cells derived from patients with ovarian cancer; therefore, dual blockade of PD-1 and Lag-3 efficiently augmented cytokine production of tumor antigen-specific CD8+ T cells (36). Thus, Lag-3 blockade alone may be insufficient to induce and activate tumor antigen-specific IFN-γ production.
The present study has several limitations other than the small number of cases. As naïve CD4+ Th cells differentiate into different subsets of Th cells depending on the cytokine milieu, the Th cell balance continuously changes depending on the immune status and/or composition of the tumor microenvironment. Moreover, Bonertz et al demonstrated that the repertoires of tumor antigens recognized by Tregs and effector/memory T cells differ in patients with colorectal cancer (14). Thus, tumor antigen-specific T cells that produce IL-10 may exhibit distinct behaviors depending on whether they are effector or regulatory T cells. To identify the type of Th cells or Tr1 producing IL-10 in response to tumor antigens, further analyses, such as single-cell proteomic analysis, are required.
To the best of our knowledge, this is the first report to indicate that tumor antigen-specific IL-10-producing T cells are present in the peripheral blood of patients with HNSCC. Nevertheless, it remains unclear whether these tumor antigen-specific IL-10-producing T cells function as effectors or regulatory cells. Lag-3+ T cells play an important role in modulating IL-10-producing T cells. These findings provide new insights into the roles of IL-10 and Lag-3 in mediating antitumor immune responses.
Supplementary Material
Supporting Data
Supporting Data
Acknowledgements
Not applicable.
Funding
This work was supported in part by a Grant-in-Aid for Scientific Research (grant nos. 22K16894 to HTad, 23K08956 to YT and 20H03834 to KC) from the Ministry of Education, Culture, Sports, Science, and Technology, Japan.
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
KC conceived and designed the study. MH, KM, HTak and HTad acquired the data. KC and ST confirm the authenticity of all the raw data. KC, HTak, YT, MM, TO, ST and KC performed data analysis and interpretation. KC wrote the manuscript. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
The present study was approved by the Ethics Committee of Gunma University Hospital (approval no. HS2017-152). Written informed consent was obtained from all patients.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
IL-10 |
interleukin-10 |
PBMC |
peripheral blood mononuclear cell |
IFN-γ |
interferon-γ |
ELISPOT |
enzyme-linked immunosorbent spot |
Lag-3 |
lymphocyte activation gene-3 |
HNSCC |
head and neck squamous cell carcinoma |
Treg |
regulatory T-cell |
Th |
T helper |
Tr1 |
T regulatory type 1 |
SFC |
spot-forming-cells |
PD-1 |
programmed cell death-1 |
CTLA-4 |
cytotoxic T-lymphocyte-associated antigen-4 |
Tim-3 |
T-cell immunoglobulin and mucin-domain containing-3 |
TNF-α |
tumor necrosis factor-α |
References
Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L and Albini A: The multifaceted nature of IL-10: Regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol. 14:11610672023. View Article : Google Scholar | |
Mocellin S, Panelli MC, Wang E, Nagorsen D and Marincola FM: The dual role of IL-10. Trends Immunol. 24:36–43. 2003. View Article : Google Scholar | |
Saraiva M, Vieira P and O'Garra A: Biology and therapeutic potential of interleukin-10. J Exp Med. 217:e201904182020. View Article : Google Scholar | |
Fujimura T, Kambayashi Y and Aiba S: Crosstalk between regulatory T cells (Tregs) and myeloid derived suppressor cells (MDSCs) during melanoma growth. Oncoimmunology. 1:1433–1434. 2012. View Article : Google Scholar | |
Pan Y, Yu Y, Wang X and Zhang T: Tumor-associated macrophages in tumor immunity. Front Immunol. 11:5830842020. View Article : Google Scholar | |
Emmerich J, Mumm JB, Chan IH, LaFace D, Truong H, McClanahan T, Gorman DM and Oft M: IL-10 directly activates and expands tumor-resident CD8(+) T cells without de novo infiltration from secondary lymphoid organs. Cancer Res. 72:3570–3581. 2012. View Article : Google Scholar | |
Guo Y, Xie YQ, Gao M, Zhao Y, Franco F, Wenes M, Siddiqui I, Bevilacqua A, Wang H, Yang H, et al: Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat Immunol. 22:746–756. 2021. View Article : Google Scholar | |
Oft M: IL-10: Master switch from tumor-promoting inflammation to antitumor immunity. Cancer Immunol Res. 2:194–199. 2014. View Article : Google Scholar | |
Mannino MH, Zhu Z, Xiao H, Bai Q, Wakefield MR and Fang Y: The paradoxical role of IL-10 in immunity and cancer. Cancer Lett. 367:103–107. 2015. View Article : Google Scholar | |
Jankovic D, Kugler DG and Sher A: IL-10 production by CD4+ effector T cells: A mechanism for self-regulation. Mucosal Immunol. 3:239–246. 2010. View Article : Google Scholar | |
Roncarolo MG, Gregori S, Bacchetta R, Battaglia M and Gagliani N: The biology of T regulatory type 1 cells and their therapeutic application in immune-mediated diseases. Immunity. 49:1004–1019. 2018. View Article : Google Scholar | |
Vence L, Palucka AK, Fay JW, Ito T, Liu YJ, Banchereau J and Ueno H: Circulating tumor antigen-specific regulatory T cells in patients with metastatic melanoma. Proc Natl Acad Sci USA. 104:20884–20889. 2007. View Article : Google Scholar | |
Wang HY, Peng G, Guo Z, Shevach EM and Wang RF: Recognition of a new ARTC1 peptide ligand uniquely expressed in tumor cells by antigen-specific CD4+ regulatory T cells. J Immunol. 174:2661–2670. 2005. View Article : Google Scholar | |
Bonertz A, Weitz J, Pietsch DHK, Rahbari NN, Schlude C, Ge Y, Juenger S, Vlodavsky I, Khazaie K, Jaeger D, et al: Antigen-specific Tregs control T cell responses against a limited repertoire of tumor antigens in patients with colorectal carcinoma. J Clin Invest. 119:3311–3321. 2009. | |
Yamada K, Masuda K, Ida S, Tada H, Bando M, Abe K, Tatematsu KI, Sezutsu H, Oyama T, Chikamatsu K and Takeda S: In vitro assessment of antitumor immune responses using tumor antigen proteins produced by transgenic silkworms. J Mater Sci Mater Med. 32:582021. View Article : Google Scholar | |
Tada H, Takahashi H, Yamada K, Masuda K, Nagata Y, Uchida M, Shino M, Ida S, Mito I, Matsuyama T, et al: Dynamic alterations of circulating T lymphocytes and the clinical response in patients with head and neck squamous cell carcinoma treated with nivolumab. Cancer Immunol Immunother. 71:851–863. 2022. View Article : Google Scholar | |
Nagorsen D, Keilholz U, Rivoltini L, Schmittel A, Letsch A, Asemissen AM, Berger G, Buhr HJ, Thiel E and Scheibenbogen C: Natural T-cell response against MHC class I epitopes of epithelial cell adhesion molecule, her-2/neu, and carcinoembryonic antigen in patients with colorectal cancer. Cancer Res. 60:4850–4854. 2000. | |
Duray A, Demoulin S, Hubert P, Delvenne P and Saussez S: Immune suppression in head and neck cancers: A review. Clin Dev Immunol. 2010:7016572010. | |
Kostecki KL, Iida M, Crossman BE, Salgia R, Harari PM, Bruce JY and Wheeler DL: Immune escape strategies in head and neck cancer: Evade, resist, inhibit, recruit. Cancers (Basel). 16:3122024. View Article : Google Scholar | |
Elmusrati A, Wang J and Wang CY: Tumor microenvironment and immune evasion in head and neck squamous cell carcinoma. Int J Oral Sci. 13:242021. View Article : Google Scholar | |
Heusinkveld M, Goedemans R, Briet RJP, Gelderblom H, Nortier JWR, Gorter A, Smit VTHBM, Langeveld APM, Jansen JC and van der Burg SH: Systemic and local human papillomavirus 16-specific T-cell immunity in patients with head and neck cancer. Int J Cancer. 131:E74–E85. 2012. View Article : Google Scholar | |
Hoffmann TK, Donnenberg AD, Finkelstein SD, Donnenberg VS, Friebe-Hoffmann U, Myers EN, Appella E, DeLeo AB and Whiteside TL: Frequencies of tetramer+ T cells specific for the wild-type sequence p53(264–272) peptide in the circulation of patients with head and neck cancer. Cancer Res. 62:3521–3529. 2002. | |
Hoffmann TK, Nakano K, Elder EM, Dworacki G, Finkelstein SD, Appella E, Whiteside TL and DeLeo AB: Generation of T cells specific for the wild-type sequence p53(264–272) peptide in cancer patients: Implications for immunoselection of epitope loss variants. J Immunol. 165:5938–5944. 2000. View Article : Google Scholar | |
Ikeguchi M, Hatada T, Yamamoto M, Miyake T, Matsunaga T, Fukumoto Y, Yamada Y, Fukuda K, Saito H and Tatebe S: Serum interleukin-6 and −10 levels in patients with gastric cancer. Gastric Cancer. 12:95–100. 2009. View Article : Google Scholar | |
Wang H, Wang L, Chi PD, Wang WD, Chen XQ, Geng QR, Xia ZJ and Lu Y: High level of interleukin-10 in serum predicts poor prognosis in multiple myeloma. Br J Cancer. 114:463–468. 2016. View Article : Google Scholar | |
De Vita F, Orditura M, Galizia G, Romano C, Roscigno A, Lieto E and Catalano G: Serum interleukin-10 levels as a prognostic factor in advanced non-small cell lung cancer patients. Chest. 117:365–373. 2000. View Article : Google Scholar | |
Cortes JE, Talpaz M, Cabanillas F, Seymour JF and Kurzrock R: Serum levels of interleukin-10 in patients with diffuse large cell lymphoma: Lack of correlation with prognosis. Blood. 85:2516–2520. 1995. View Article : Google Scholar | |
Evans C, Morrison I, Heriot AG, Bartlett JB, Finlayson C, Dalgleish AG and Kumar D: The correlation between colorectal cancer rates of proliferation and apoptosis and systemic cytokine levels; plus their influence upon survival. Br J Cancer. 94:1412–1419. 2006. View Article : Google Scholar | |
Green VL, Irune E, Prasai A, Alhamarneh O, Greenman J and Stafford ND: Serum IL10, IL12 and circulating CD4+CD25high T regulatory cells in relation to long-term clinical outcome in head and neck squamous cell carcinoma patients. Int J Oncol. 40:833–839. 2012. | |
Kim Y, Yang H, Lee WS, Cheon J, Sang YB, Kang B, Chon HJ and Kim C: High levels of baseline serum IL-10 are associated with reduced clinical benefit from first-line immune checkpoint inhibitor therapy in advanced renal cell carcinoma. J Cancer. 14:935–942. 2023. View Article : Google Scholar | |
Giunta EF, Barra G, De Falco V, Argenziano G, Napolitano S, Vitale P, Zanaletti N, Terminiello M, Martinelli E, Morgillo F, et al: Baseline IFN-γ and IL-10 expression in PBMCs could predict response to PD-1 checkpoint inhibitors in advanced melanoma patients. Sci Rep. 10:176262020. View Article : Google Scholar | |
Rutz S and Ouyang W: Regulation of interleukin-10 expression. Adv Exp Med Biol. 941:89–116. 2016. View Article : Google Scholar | |
Huo JL, Wang YT, Fu WJ, Lu N and Liu ZS: The promising immune checkpoint LAG-3 in cancer immunotherapy: From basic research to clinical application. Front Immunol. 13:9560902022. View Article : Google Scholar | |
Joller N and Kuchroo VK: Tim-3, Lag-3, and TIGIT. Curr Top Microbiol Immunol. 410:127–156. 2017. | |
Gagliani N, Magnani CF, Huber S, Gianolini ME, Pala M, Licona-Limon P, Guo B, Herbert DR, Bulfone A, Trentini F, et al: Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med. 19:739–746. 2013. View Article : Google Scholar | |
Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, Eppolito C, Qian F, Lele S, Shrikant P, et al: Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA. 107:7875–7880. 2010. View Article : Google Scholar |