Low dose decitabine treatment induces CD80 expression in cancer cells and stimulates tumor specific cytotoxic T lymphocyte responses

Li-Xin Wang, Zhen-Yang Mei, Ji-Hao Zhou, Yu-Shi Yao, Yong-Hui Li, Yi-Han Xu, Jing-Xin Li, Xiao-Ning Gao, Min-Hang Zhou, Meng-Meng Jiang, Li Gao, Yi Ding, Xue-Chun Lu, Jin-Long Shi, Xu-Feng Luo, Jia Wang, Li-Li Wang, Chunfeng Qu, Xue-Feng Bai, Li Yu, Li-Xin Wang, Zhen-Yang Mei, Ji-Hao Zhou, Yu-Shi Yao, Yong-Hui Li, Yi-Han Xu, Jing-Xin Li, Xiao-Ning Gao, Min-Hang Zhou, Meng-Meng Jiang, Li Gao, Yi Ding, Xue-Chun Lu, Jin-Long Shi, Xu-Feng Luo, Jia Wang, Li-Li Wang, Chunfeng Qu, Xue-Feng Bai, Li Yu

Abstract

Lack of immunogenicity of cancer cells has been considered a major reason for their failure in induction of a tumor specific T cell response. In this paper, we present evidence that decitabine (DAC), a DNA methylation inhibitor that is currently used for the treatment of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and other malignant neoplasms, is capable of eliciting an anti-tumor cytotoxic T lymphocyte (CTL) response in mouse EL4 tumor model. C57BL/6 mice with established EL4 tumors were treated with DAC (1.0 mg/kg body weight) once daily for 5 days. We found that DAC treatment resulted in infiltration of IFN-γ producing T lymphocytes into tumors and caused tumor rejection. Depletion of CD8(+), but not CD4(+) T cells resumed tumor growth. DAC-induced CTL response appeared to be elicited by the induction of CD80 expression on tumor cells. Epigenetic evidence suggests that DAC induces CD80 expression in EL4 cells via demethylation of CpG dinucleotide sites in the promoter of CD80 gene. In addition, we also showed that a transient, low-dose DAC treatment can induce CD80 gene expression in a variety of human cancer cells. This study provides the first evidence that epigenetic modulation can induce the expression of a major T cell co-stimulatory molecule on cancer cells, which can overcome immune tolerance, and induce an efficient anti-tumor CTL response. The results have important implications in designing DAC-based cancer immunotherapy.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Impacts of DAC treatment on…
Figure 1. Impacts of DAC treatment on EL4 tumorigenicity and tumor growth.
(A) EL4 cells were treated with DAC (0.25 µM in culture medium) or Cytidine or PBS for 72 h. Cells were then injected into each mouse s.c. at a dose of 1×104 cells/mouse. Tumor size was measured in two directions every two or three days. Tumor volume was calculated using a formula: volume = (L x W2)/2 where L = length; W = width. *P<0.05 when DAC-treatment is compared to PBS or Cytidine treatment. Five to six mice were used in each group and data were pooled from two experiments. (B) EL4 cells treated with DAC were injected into C57BL/6 mice s.c. at a dose of 1×104 cells/mouse (G1), 8×104 cells/mouse (G2) or 16×104 cells/mouse (G3). PBS-treated EL4 cells (1×104 cells/mouse) served as control. Mouse survival data is shown. Five mice were included in each group and data shown are representative of three independent experiments. (C) DAC-treated or PBS-treated EL4 cells were injected into each mouse i.v. at a dose of 1×104 cells/mouse. Mouse survival was monitored up to 100 days after tumor cell injection. Ten mice per group were used, and data shown are representative of three independent experiments. (D) Mice with established EL4 tumors were treated with DAC or PBS i.p. for 5 consecutive days starting on day 10. Data shown are representative of three experiments. Asterisks indicate statistical significance of P<0.05.
Figure 2. DAC treatment induces T cell…
Figure 2. DAC treatment induces T cell infiltration into tumors.
(A–B) C57BL/6 mice with established EL4 tumors were treated with DAC (1 mg/kg body weight) or PBS once daily for 5 consecutive days. 7–10 days after treatment, mice were sacrificed, tumors were harvested, and disassociated tumor cells were stained for the expression of different cell surface markers, followed by flow cytometry quantification for CD8+, CD4+ and NK1.1+CD3− cells. (C) Flow cytometric analysis and quantification of intracellular IFN-γ production by tumor infiltrating CD8+ T cells. Bars represent mean+SD; n = 3–7 mice per group. (D) Flow cytometric analysis of intracellular IFN-γ production by tumor infiltrating CD4+ T cells. Bars represent mean+SD; n = 3–7 mice per group. Student’s t test was used for the statistical analysis. Numbers in flow cytometric figures indicate % positive cells corresponding to each gate.
Figure 3. DAC-treatment leads to CD8+ T…
Figure 3. DAC-treatment leads to CD8+ T cell dependent tumor rejection.
Four doses of anti-CD8 or anti-CD4 antibody (400 µg/per mouse, i.p.) were injected at 4 day intervals beginning on day 1 after DAC treatment. Three mice per group were used for the experiment shown. (A) CD8+ T cells in spleens and tumors were analyzed by flow cytometry after anti-CD8 antibody or an isotype-matched control antibody treatment. (B) Tumor growth in mice treated with anti-CD8 or an isotype-matched control mAb following DAC administration. Data is representative of two independent experiments with similar results. Asterisks indicate statistical significance of P<0.05. (C) CD4+ T cells in spleens and tumors were analyzed by flow cytometry after anti-CD4 antibody or an isotype-matched control antibody treatment. (D) Tumor growth of mice treated with anti-CD4 or an isotype-matched control mAb following DAC administration. Data is representative of two independent experiments with similar results. Numbers in flow cytometric figures indicate % positive cells corresponding to each gate.
Figure 4. Up-regulated gene expression in DAC…
Figure 4. Up-regulated gene expression in DAC treated EL4 cells.
The mouse SmartArray chips were hybridized with RNA derived from DAC or PBS treated EL4 cells. (A) Heatmap of some upregulated genes by DAC treatment. Columns represent microarray data obtained from 3 independent biological replicates. (B) RT-PCR was used to validate the up-regulated genes. (C) qRT-PCR was used to quantify up-regulated genes in DAC and PBS treated EL4 cells.
Figure 5. Characterization of DAC-induced CD80 gene…
Figure 5. Characterization of DAC-induced CD80 gene expression in cancer cells.
(A) EL4 cells were treated with DAC (0.25 µM) or PBS for 72 h. RT-PCR was used to detect CD80 gene expression in DAC and PBS treated EL4 cells. The GAPDH gene was simultaneously amplified as a loading control. (B) Flow cytometric analysis of CD80 expression in DAC-treated and PBS- treated EL4 cells. (C) EL4 cells were treated with DAC and PBS for 3 consecutive days and were maintained in the culture. Cells were harvested every two days and stained with anti-CD80 followed by flow cytometric analysis. (D) EL4 cells (CD45.2) were injected s.c. into C57BL/6 mice or i.v. into CD45.1 congenic C57BL/6 mice. DAC was administrated i.p. 2 weeks later for 5 consecutive days. EL4 cells from disassociated tumors (left panel) and bone marrows (BM) (right panel) were analyzed for the CD80 expression by flow cytometry. (E) Induction of CD80 expression in human cancer cell lines. Cancer cells were treated with DAC (0.25 µM) or PBS for 72 h. RT-PCR was used to determine CD80 gene expression. Numbers in flow cytometric figures indicate % positive cells corresponding to each gate.
Figure 6. DNA methylation of CD80 promoter…
Figure 6. DNA methylation of CD80 promoter region in EL4 and its variant cells.
(A) The distribution of CpG dinucleotides in a region 1 kb upstream of Exon 1 of CD80 gene. Numbers refer to position relative to the 5′ end of CD80. Two CpG enriched regions (F1 and F2) were selected for Bisulfite sequencing analysis. (B) Flow cytometric analysis of CD80 expression on two EL4 variants (EL4C3 and EL4C45). Numbers in flow cytometric figures indicate % positive cells corresponding to each gate. (C) Bisulfite sequencing of the CD80 promoter region in EL4C45 (upper panel) and EL4C3 (lower panel). Open and filled circles represent demethylated and methylated CpG sites, respectively. The methylation frequency at CpG sites of each cell line is indicated. (D) Bisulfite sequencing analysis of CD80 promoter region in DAC- and PBS-treated EL4 cells. (E) EL4 cells were first treated with DAC or PBS, CD80+ and CD80− EL4 cells were then sorted by flow cytometry-based sorting. Bisulfite sequencing analysis was performed on DAC-treated, CD80+ and CD80− EL4 cells.
Figure 7. DAC educated CD80 positive, but…
Figure 7. DAC educated CD80 positive, but not CD80 negative EL4 cells induce T cell responses.
(A) EL4 cells were treated with DAC. CD80+ and CD80− EL4 cells were separated by flow cytometry-based sorting. Flow cytometric analysis of CD80 expression in EL4 cells before and after sorting is shown. (B) Tumor growth kinetics of DAC-treated CD80+ and CD80− EL4 cells. 2×104 CD80+ or CD80− DAC-treated EL4 cells were injected s.c. into each mouse. Tumor growth in individual mouse is shown. (C) Flow cytometry analysis of T cell and NK cell infiltration in CD80+ and CD80− tumors. Single cell suspensions of tumors were stained for CD4, CD8, CD3, NK1.1 followed by flow cytometry analysis. Percentage of CD8+ (D), CD4+ (E) and NK1.1+CD3− (F) in CD80+ tumors (n = 6) and CD80− tumors (n = 6) were quantified. Data shown are mean+SEM and are representative of four independent experiments with similar results. Numbers in flow cytometric figures indicate % positive cells corresponding to each gate.
Figure 8. Role of DAC induced CD80…
Figure 8. Role of DAC induced CD80 expression in stimulation of T cells.
Six million irradiated EL4 cells were injected i.p. into each BALB/c mouse twice with a week interval between injections. Spleen and lymph node cells from EL4 immunized BALB/C mice were co-cultured with DAC-treated or PBS-treated EL4 cells for 6 days. (A) T cell proliferation was assessed using Cell Counting Kit-8 (CCK8, Dojindo, Kumomoto, Japan) on day 6. IL-2 (B) and IFN-γ (C) production in the culture supernatants were detected by ELISA. For CD80 blocking, spleen and lymph node cells from EL4-immunized mice were co-cultured with DAC-treated EL4 cells in the presence of 5 µg/ml anti-CD80 antibody or an isotype matched control antibody. (D) T cell proliferation was assessed using the CCK8 assay. Supernatants of co-culture were harvested 48 hours later for detection of IL-2 (E) and IFN-γ (F) by ELISA. Student’s t-test was used for statistical analysis. Data shown are representative of three experiments with similar results.
Figure 9. The model of DAC action…
Figure 9. The model of DAC action in T cell response.
DNA hypermethylation in the promoter and exon 1 region of CD80 gene repress the expression of CD80 in leukemic/cancer cells. Lack of co-stimulatory signal leads to T cell tolerance. Low dose decitabine treatment can restore cancer cell expression of CD80 by demethylation of the CD80 promoter region in leukemic/cancer cells. The expression of CD80 on leukemic/cancer cells can overcome T cell tolerance and lead to activation of T cells. MHC = Major histocompatibility complex; TCR = T cell receptor.

References

    1. Mapara MY, Sykes M (2004) Tolerance and cancer: mechanisms of tumor evasion and strategies for breaking tolerance. J Clin Oncol 22: 1136–1151.
    1. Zou W (2005) Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 5: 263–274.
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM (2007) Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 25: 267–296.
    1. Zheng P, Guo Y, Niu Q, Levy DE, Dyck JA, et al. (1998) Proto-oncogene PML controls genes devoted to MHC class I antigen presentation. Nature 396: 373–376.
    1. Seliger B, Maeurer MJ, Ferrone S (1997) TAP off–tumors on. Immunol Today 18: 292–299.
    1. Uyttenhove C, Maryanski J, Boon T (1983) Escape of mouse mastocytoma P815 after nearly complete rejection is due to antigen-loss variants rather than immunosuppression. J Exp Med 157: 1040–1052.
    1. Thurner B, Haendle I, Roder C, Dieckmann D, Keikavoussi P, et al. (1999) Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med 190: 1669–1678.
    1. Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, et al. (2002) Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A 99: 16168–16173.
    1. Zou W, Chen L (2008) Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol 8: 467–477.
    1. Weber J, Salgaller M, Samid D, Johnson B, Herlyn M, et al. (1994) Expression of the MAGE-1 tumor antigen is up-regulated by the demethylating agent 5-aza-2′-deoxycytidine. Cancer Res 54: 1766–1771.
    1. Guo ZS, Hong JA, Irvine KR, Chen GA, Spiess PJ, et al. (2006) De novo induction of a cancer/testis antigen by 5-aza-2′-deoxycytidine augments adoptive immunotherapy in a murine tumor model. Cancer Res 66: 1105–1113.
    1. Coral S, Sigalotti L, Altomonte M, Engelsberg A, Colizzi F, et al. (2002) 5-aza-2′-deoxycytidine-induced expression of functional cancer testis antigens in human renal cell carcinoma: immunotherapeutic implications. Clin Cancer Res 8: 2690–2695.
    1. Calabro L, Fonsatti E, Altomonte M, Pezzani L, Colizzi F, et al. (2005) Methylation-regulated expression of cancer testis antigens in primary effusion lymphoma: immunotherapeutic implications. J Cell Physiol 202: 474–477.
    1. Dubovsky JA, McNeel DG, Powers JJ, Gordon J, Sotomayor EM, et al. (2009) Treatment of chronic lymphocytic leukemia with a hypomethylating agent induces expression of NXF2, an immunogenic cancer testis antigen. Clin Cancer Res 15: 3406–3415.
    1. Konkankit VV, Kim W, Koya RC, Eskin A, Dam MA, et al. (2011) Decitabine immunosensitizes human gliomas to NY-ESO-1 specific T lymphocyte targeting through the Fas/Fas ligand pathway. J Transl Med 9: 192.
    1. Coral S, Sigalotti L, Gasparollo A, Cattarossi I, Visintin A, et al. (1999) Prolonged upregulation of the expression of HLA class I antigens and costimulatory molecules on melanoma cells treated with 5-aza-2′-deoxycytidine (5-AZA-CdR). J Immunother 22: 16–24.
    1. Serrano A, Tanzarella S, Lionello I, Mendez R, Traversari C, et al. (2001) Rexpression of HLA class I antigens and restoration of antigen-specific CTL response in melanoma cells following 5-aza-2′-deoxycytidine treatment. Int J Cancer 94: 243–251.
    1. Fonsatti E, Nicolay HJ, Sigalotti L, Calabro L, Pezzani L, et al. (2007) Functional up-regulation of human leukocyte antigen class I antigens expression by 5-aza-2′-deoxycytidine in cutaneous melanoma: immunotherapeutic implications. Clin Cancer Res 13: 3333–3338.
    1. Magner WJ, Kazim AL, Stewart C, Romano MA, Catalano G, et al. (2000) Activation of MHC class I, II, and CD40 gene expression by histone deacetylase inhibitors. J Immunol 165: 7017–7024.
    1. Maeda T, Towatari M, Kosugi H, Saito H (2000) Up-regulation of costimulatory/adhesion molecules by histone deacetylase inhibitors in acute myeloid leukemia cells. Blood 96: 3847–3856.
    1. Sigalotti L, Coral S, Fratta E, Lamaj E, Danielli R, et al. (2005) Epigenetic modulation of solid tumors as a novel approach for cancer immunotherapy. Semin Oncol 32: 473–478.
    1. Jones PA, Taylor SM (1980) Cellular differentiation, cytidine analogs and DNA methylation. Cell 20: 85–93.
    1. Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, et al. (2002) Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 20: 2429–2440.
    1. Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M, et al. (2006) Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer 106: 1794–1803.
    1. Issa JP, Garcia-Manero G, Giles FJ, Mannari R, Thomas D, et al. (2004) Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood 103: 1635–1640.
    1. Blum W, Klisovic RB, Hackanson B, Liu Z, Liu S, et al. (2007) Phase I study of decitabine alone or in combination with valproic acid in acute myeloid leukemia. J Clin Oncol 25: 3884–3891.
    1. Cashen AF, Schiller GJ, O’Donnell MR, DiPersio JF (2010) Multicenter, phase II study of decitabine for the first-line treatment of older patients with acute myeloid leukemia. J Clin Oncol 28: 556–561.
    1. Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, et al. (2011) Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov 1: 598–607.
    1. Oki Y, Jelinek J, Shen L, Kantarjian HM, Issa JP (2008) Induction of hypomethylation and molecular response after decitabine therapy in patients with chronic myelomonocytic leukemia. Blood 111: 2382–2384.
    1. Tsai HC, Li H, Van Neste L, Cai Y, Robert C, et al. (2012) Transient Low Doses of DNA-Demethylating Agents Exert Durable Antitumor Effects on Hematological and Epithelial Tumor Cells. Cancer Cell 21: 430–446.
    1. Wang LX, Talebian F, Liu JQ, Khattabi M, Yu L, et al. (2012) IL-10 contributes to the suppressive function of tumour-associated myeloid cells and enhances myeloid cell accumulation in tumours. Scand J Immunol 75: 273–281.
    1. Reiter H (1979) Effect of thymidine on the survival of mice with EL4 tumors. Cancer Res 39: 4856–4860.
    1. Gollob JA, Sciambi CJ (2007) Decitabine up-regulates S100A2 expression and synergizes with IFN-gamma to kill uveal melanoma cells. Clin Cancer Res 13: 5219–5225.
    1. Odobasic D, Leech MT, Xue JR, Holdsworth SR (2008) Distinct in vivo roles of CD80 and CD86 in the effector T-cell responses inducing antigen-induced arthritis. Immunology 124: 503–513.
    1. Chen L, Ashe S, Brady WA, Hellstrom I, Hellstrom KE, et al. (1992) Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 71: 1093–1102.
    1. Townsend SE, Allison JP (1993) Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. Science 259: 368–370.
    1. Ramarathinam L, Castle M, Wu Y, Liu Y (1994) T cell costimulation by B7/BB1 induces CD8 T cell-dependent tumor rejection: an important role of B7/BB1 in the induction, recruitment, and effector function of antitumor T cells. J Exp Med 179: 1205–1214.
    1. Wu TC, Huang AY, Jaffee EM, Levitsky HI, Pardoll DM (1995) A reassessment of the role of B7–1 expression in tumor rejection. J Exp Med 182: 1415–1421.
    1. Bai XF, Bender J, Liu J, Zhang H, Wang Y, et al. (2001) Local costimulation reinvigorates tumor-specific cytolytic T lymphocytes for experimental therapy in mice with large tumor burdens. J Immunol 167: 3936–3943.
    1. Bai XF, Gao JX, Liu J, Wen J, Zheng P, et al. (2001) On the site and mode of antigen presentation for the initiation of clonal expansion of CD8 T cells specific for a natural tumor antigen. Cancer Res 61: 6860–6867.

Source: PubMed

3
Předplatit