Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2018 Nov 1;128(11):4912-4923.
doi: 10.1172/JCI120612. Epub 2018 Sep 24.

Self-associated molecular patterns mediate cancer immune evasion by engaging Siglecs on T cells

Affiliations

Self-associated molecular patterns mediate cancer immune evasion by engaging Siglecs on T cells

Michal A Stanczak et al. J Clin Invest. .

Abstract

First-generation immune checkpoint inhibitors, including anti-CTLA-4 and anti-programmed death 1 (anti-PD-1) antibodies, have led to major clinical progress, yet resistance frequently leads to treatment failure. Thus, new targets acting on T cells are needed. CD33-related sialic acid-binding immunoglobulin-like lectins (Siglecs) are pattern-recognition immune receptors binding to a range of sialoglycan ligands, which appear to function as self-associated molecular patterns (SAMPs) that suppress autoimmune responses. Siglecs are expressed at very low levels on normal T cells, and these receptors were not until recently considered as interesting targets on T cells for cancer immunotherapy. Here, we show an upregulation of Siglecs, including Siglec-9, on tumor-infiltrating T cells from non-small cell lung cancer (NSCLC), colorectal, and ovarian cancer patients. Siglec-9-expressing T cells coexpressed several inhibitory receptors, including PD-1. Targeting of the sialoglycan-SAMP/Siglec pathway in vitro and in vivo resulted in increased anticancer immunity. T cell expression of Siglec-9 in NSCLC patients correlated with reduced survival, and Siglec-9 polymorphisms showed association with the risk of developing lung and colorectal cancer. Our data identify the sialoglycan-SAMP/Siglec pathway as a potential target for improving T cell activation for immunotherapy.

Keywords: Cancer immunotherapy; Immunology; Oncology; T cells.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1
Figure 1. Siglec-9 is upregulated on CD8+ TILs.
(A) Representative flow cytometry analysis of Siglec-9 expression on CD8+ T cells in PBMCs from healthy donors (left panel), PBMCs from NSCLC patients (middle panel), and TILs from a matched NSCLC patient (right panel). (B and C) Quantification of Siglec-9 expression on CD4+ (B) and CD8+ (C) TILs from NSCLC patients (PBMCs, n = 36; NSCLC, n = 41; mean ± SD). Statistical analysis by unpaired Student’s t test. (D) Paired analysis of CD8+ T cells from the peripheral blood and tumors of NSCLC patients (n = 20). Statistical analysis by paired Student’s t test. (E) Immunohistochemical staining of Siglec-9–positive cells in NSCLC sections. Scale bars: 50 μm. (F) Representative immunofluorescence analysis of CD3 and Siglec-9 double-positive cells and Siglec-9 staining or SNA staining and Siglec-9 staining (right panel). Scale bars: 30 μm (left panel); 50 μm (right panel).(G) Heatmap of expression analysis of different Siglecs in NSCLC samples on CD4+ and CD8+ TILs. (H and I) Immunofluorescence study in paraffin-embedded tissue microarrays using recombinant Siglec-9–Fc (human IgG1) fusion protein coupled to secondary PE-conjugated (Fab′)2 goat anti-human Fc antibody. Representative images (H) and Siglec ligands quantification summary (I) are shown. Original magnification, ×400. Scale bars: 50 μm. Fluorescence values were normalized against an IgG1 isotype control. Lung tissue, n = 5; adjacent lung tissue, n = 9; squamous cell carcinoma (SCC), n = 20; adenocarcinoma, n = 20; small cell lung cancer (SCLC), n = 10; broncho-alveolar carcinoma (BAC), n = 10; atypical carcinoid, n = 5. Data are shown as mean ± SEM. Statistical analysis performed by 1-way ANOVA. *P < 0.05; **P < 0.01; ***P < 0.001.
Figure 2
Figure 2. Sig9+CD8+ TILs coexpress inhibitory receptors.
(A and B) Expression of PD-1 in primary NSCLC samples on Sig9CD8+ (Sig9) or Sig9+CD8+ (Sig9+) TILs (A, n = 44) and representative FACS analysis (B). Statistical analysis by paired Student’s t test. (C and D) Expression of TIM-3 (C, n = 71) and LAG-3 (D, n = 18) on Sig9CD8+ or Sig9+CD8+ TILs from NSCLC samples. Statistical analysis by paired Student’s t test. (E) Analysis of the number of coexpressed inhibitory receptors on Sig9CD8+ or Sig9+CD8+ TILs. (F) Volcano plot of RNA-seq on sorted TILs according to their Siglec-9 expression. The 3 significantly differentially expressed genes were MIK67 (Ki67), KLF4, and SPP1. *P < 0.05; ***P < 0.001. Data are presented as mean ± SD.
Figure 3
Figure 3. Sig9+CD8+ TILs are a distinct subset within intratumoral CD8+ T cells.
(A and B) Upregulation of the activation markers CD25 (A) and CD69 (B) on Sig9CD8+ or Sig9+ TILs sorted from primary NSCLC samples and activated with anti-CD3/28 antibodies for 48 hours (n = 9). Statistical analysis by paired Student’s t test. (C) ELISA analysis of IFN-γ in the supernatant of sorted Sig9CD8+ T cells or Sig9+CD8+ T cells (n = 3–7, independent patients). Cells were sorted from PBMCs of healthy donors or primary NSCLC samples (TILs). Act, activated. Supernatants from 50,000 cells were analyzed. (D) Analysis of TNF-α in the supernatant of sorted Sig9CD8+ or Sig9+CD8+ cells from healthy donors or NSCLC patient samples (n = 3–7, independent donors/patients). Statistical analysis performed by 1-way ANOVA. (E) Expression level of CD5 in the CD8+PD-1hi population on Sig9 TILs and Sig9+ TILs (n = 9). (F) Percentage of Sig9CD8+ TILs or Sig9+CD8+ TILs in primary NSCLC samples that express Ki67 within the PD-1hi population (n = 9). (G) Frequency of CD38hiCD101hi cells on Sig9 and Sig9+CD8+PD-1hi TILs determined by flow cytometric analysis (n = 13). Statistical analysis by paired Student’s t test. *P < 0.05; **P < 0.01; ***P < 0.001. Data are presented as mean ± SD.
Figure 4
Figure 4. Sia-SAMPs inhibit T cell–mediated tumor cell killing in vitro.
(A) Inhibition of T cell activation by LGALS3BP in a dose-dependent manner measured by intracellular IFN-γ by flow cytometry. CD8+ T cells from healthy donors were activated with anti-CD3 and anti-CD28 antibodies in the presence of increasing amounts of LGALS3BP (μg/ml, n = 3). (B) Representative histograms of binding of Sig9-Fc to A549 WT cells, enzymatically desialylated A549 cells (desia), GNE-deficient A549 cells (GNE-KO), and GNE-KO A549 cells refed with 10 mM Neu5Ac. (C) Percentage of cleaved caspase-3–positive (clCasp3+) WT A549 cells, desialylated A549 cells, GNE-KO A549 cells, or GNE-KO A549 cells fed with Neu5Ac (refed) after incubation with CD8+ T cells and catumaxomab (n = 10). (D) Apoptosis of WT, desialylated, GNE-KO, and refed GNE-KO HT-29 cells measured by upregulation of cleaved caspase-3 in tumor cells (n = 6). (E and F) clCaps3+ A549 (E, n = 11) or HT-29 (F, n = 11) tumor cells after coincubation with TILs from NSCLC or CRC samples. (G) CD8+ T cells were sorted according to their Siglec-9 expression and incubated with either WT or GNE-KO A549 cells (n = 4). (H) CD19+ RAMOS cells were incubated with CD8+ T cells from healthy donors in the presence of CD3 and CD19 bispecific antibody blinatumomab (n = 7). (I) GNE-KO RAMOS cells incubated with CD8+ T cells from patients with chronic lymphocytic leukemia (n = 3). (J) Activation measured by CD25 on CD8+ T cells treated with anti-CD3 and anti-CD28 antibodies in the presence of anti–Siglec-9 antibody (clone 191240, g/ml, n = 4). (K) Relative IL-2 production of NSCLC primary tumor samples incubated with SEB and Siglec-9–blocking antibody and the Fab fragments (clone 191240, n = 5). (L) Measurement of CD69 upregulation on CD8+ TILs from NSCLC patients upon incubation with SEB in the presence of antibodies or Fab fragments (n = 5). Statistical analyses in this figure were performed by 1-way ANOVA. Data are presented as mean ± SD. *P < 0.05; **P < 0.01; ***P < 0.001.
Figure 5
Figure 5. Sialylated SAMPs enhance immune escape and tumor growth in vivo.
(A) Siglec-E expression was determined by flow cytometry on control splenocytes, splenocytes from tumor-bearing mice, and CD8+ TILs from subcutaneous MC38 tumors (n = 25–28). Statistical analysis performed by 1-way ANOVA. (B) Expression of intracellular Ki67 was examined by flow cytometry on SigECD8+ and SigE+CD8+ TILs (n = 18). Statistical analysis by paired Student’s t test. (CE) Frequencies of inhibitory immune receptor expression on SigECD8+ and SigE+CD8+ TILs from MC38 tumors, as studied by flow cytometry. PD-1 (C, n = 16), TIM-3 (D, n = 18), and LAG-3 (E, n = 7) were analyzed. Statistical analysis by paired Student’s t test. (F) Number of coexpressed inhibitory receptors on SigECD8+ or SigE+CD8+ TILs. (G) Upregulation of CD25+CD69+ upon restimulation of sorted SigECD8+ and SigE+CD8+ TILs. Statistical analysis by paired Student’s t test. (H) Growth curves of subcutaneous WT or GNE-KO MC38 tumors (n = 8–9). (I) Growth curves of subcutaneous WT and GNE-KO EMT6 tumors (n = 13-14). Experiments were replicated 2 to 3 times. Statistical analysis by 2-way ANOVA. (J and K) Frequencies of CD3+ and CD8+ cells in the tumor (n = 7). Statistical analysis by unpaired Student’s t test. **P < 0.01; ***P < 0.001. Data are presented as mean ± SD.
Figure 6
Figure 6. Engagement of inhibitory Siglecs on T cells mediates immune escape.
(A) Subcutaneous growth curves of MC38 tumors in littermate Siglec-9 transgenic control mice (HS9) or Siglec-9 transgenic mice crossed to CD4-Cre mice (HS9, CD4Cre) (n = 8–10). The experiment was repeated 3 times. Statistical analysis by 2-way ANOVA. (B) Tumor volumes after 21 days of subcutaneous MC38 tumors in littermate control mice (E/E) or homozygous (E16/E16) mice that express the chimeric receptor SigE16 (n = 7). Statistical analysis performed by 1-way ANOVA. (C) Subcutaneous growth curves of MC38 tumors in E16 mice or littermate control mice after CD4 and CD8 cell depletion by antibodies (n = 7–8). The experiment was repeated 2 times. Statistical analysis by 2-way ANOVA. (D) MC38 tumor volumes after 21 days in E16 mice and littermate control mice and independent depletion of CD4+ and CD8+ T cells. (E) Tumor volume of MC38-OVA tumors after adoptive transfer of OVA-specific OT-I CD8+ T cells from WT or SigE16 (E16) mice. Statistical analysis performed by 1-way ANOVA. *P < 0.05; **P < 0.01. Data are presented as mean ± SD. (F) Survival analysis of NSCLC patients with low (n = 18) and high percentage (above 30%, n = 11) of Siglec-9 expression on their CD8+ T cells. Differences were analyzed by Wilcoxon’s test. A multivariate analysis of the 2 groups for age and stage showed slightly reduced significance, with a P value of 0.0669 (multivariate, univariate analysis P = 0.0097) and a hazard ratio of 14.6.

References

    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–461. doi: 10.1016/j.ccell.2015.03.001. - DOI - PMC - PubMed
    1. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–330. doi: 10.1038/nature21349. - DOI - PubMed
    1. Cao X. Self-regulation and cross-regulation of pattern-recognition receptor signalling in health and disease. Nat Rev Immunol. 2016;16(1):35–50. doi: 10.1038/nri.2015.8. - DOI - PubMed
    1. Schaefer L. Complexity of danger: the diverse nature of damage-associated molecular patterns. J Biol Chem. 2014;289(51):35237–35245. - PMC - PubMed
    1. Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev. 2009;22(2):240–273. doi: 10.1128/CMR.00046-08. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances