DNA demethylating agent decitabine broadens the peripheral T cell receptor repertoire

Jing Nie, Yan Zhang, Xiang Li, Meixia Chen, Chuanjie Liu, Weidong Han, Jing Nie, Yan Zhang, Xiang Li, Meixia Chen, Chuanjie Liu, Weidong Han

Abstract

Purpose: Decitabine, a promising epi-immunotherapeutic agent has shown clinical responses in solid tumor patients, while the anti-tumor mechanisms were unclear. We aimed to investigate the immunomodulatory effect of decitabine in peripheral T cells.

Experimental design: We applied next-generation sequencing to investigate the complementarity-determining region 3 (CDR3) of the TCRβ gene, the diversity of which acts as the prerequisite for the host immune system to recognize the universal foreign antigens. We collected the peripheral blood mononuclear cells (PBMCs) from 4 patients, at baseline and after 2 cycles of low-dose decitabine therapy.

Results: An increase of the unique productive sequences of the CDR3 of TCRβ was observed in all of the 4 patients after decitabine treatment, which was characterized by a lower abundance of expanded clones and increased TCR diversity compared with before decitabine treatment. Further analysis showed a tendency for CD4 T cells with an increased CD4/CD8 ratio in response to decitabine therapy. In addition, the genome-wide expression alterations confirmed the effects of decitabine on immune reconstitution, and the increase of TCR excision circles (TRECs) was validated.

Conclusions: The low-dose DNMT inhibitor decitabine broadens the peripheral T cell repertoire, providing a novel role for the epigenetic modifying agent in anti-tumor immune enhancement.

Keywords: T cell receptor repertoire; decitabine; epigenetic therapy; pharmacometabonomics; solid tumor.

Conflict of interest statement

None.

Figures

Figure 1. Global analysis of V β…
Figure 1. Global analysis of Vβ-Jβ gene segment combinations
(A, B) The frequencies of TCRβ V (A) and J (B) gene segments in peripheral blood samples at baseline (pre) and following 2 cycles of decitabine therapy (post) from UPN1 and UPN2 are shown. (CF) Frequencies of specific Vβ-Jβ gene segment combinations in TCRβ CDR3 sequences are expressed in UPN1 (C), UPN2 (D), UPN3 (E), and UPN4 (F) before and after decitabine therapy.
Figure 2. Analysis of CDR3 spectrums
Figure 2. Analysis of CDR3 spectrums
(AD) A comparison of CDR3 region length and distribution between baseline and after 2 cycles of decitabine therapy in UPN1 (A), UPN2 (B), UPN3 (C), and UPN4 (D).
Figure 3. Analysis of high clonal V-J…
Figure 3. Analysis of high clonal V-J combinations
(AD) The 10 most frequent V-J combinations of PBMCs from UPN1 (A), UPN2 (B), UPN3 (C) and UPN4 (D) at baseline and after 2 cycles of decitabine therapy are indicated in color; the remaining V-J combinations are grouped in white.
Figure 4. Analysis of changes in relative…
Figure 4. Analysis of changes in relative TCRβ V and TCRβ J
(A) PBMCs from the 4 patients were collected as detected by flow cytometry using antibodies against CD3, CD4, and CD8. The average ratios of CD3+CD4+: CD3+CD8+ from these patients are shown. (BE) The frequencies of TCRβ V5-4 (B), TCRβ V7-2 (C), TCRβ V18 (D) and TCRβ V7-9 (E) in PBMCs from each patient before and after decitabine therapy are shown. (F) The top 10 V-J combinations in PBMCs from the 4 patients following decitabine therapy are listed.
Figure 5. Low-dose decitabine increased TCR diversity…
Figure 5. Low-dose decitabine increased TCR diversity in PBMCs
(AD) Frequency distribution of clonotypes in plots of UPN1 (A), UPN2 (B), UPN3 (C), and UPN4 (D) at baseline (in blue) and after 2 cycles of decitabine therapy (in red). Each dot represents a distinct TCRβ clonotype. Clonotypes are presented at frequencies > 1% and the top 10 clonotypes with their cumulative frequency (percentage of reads). Values in the lower-left corner depict TCR diversity. (E) The 10 most frequent clonotypes in the 4 patients at baseline (pre) and after 2 cycles of decitabine therapy (post) are indicated in color and listed at the right; the remaining clonotypes are grouped in black. (F) TCR Simpson diversity of the 4 patients after 2 cycles of decitabine therapy compared to that of the baseline. (G) The average T cell repertoire Shannon diversity of the 4 patients before and after decitabine therapy.
Figure 6. Genome-wide expression alteration and PRKDC…
Figure 6. Genome-wide expression alteration and PRKDC induction
(A) The peripheral T cells were sorted by flow cytometry using anti-CD3 antibody from blood samples in these 4 patients (UPN1 to UPN4). The percentages of 5-mC in T cells from the 4 patients before and after decitabine therapy were detected relative to the total cytosine content. (B) Beta value changes in CpG islands in patients UPN1 and UPN2 before and after decitabine therapy. (C) Heat map and sample clustering analysis of the differentially expressed genes before and after decitabine therapy in the same patients as (B). (D) Significantly enriched up-regulated and down-regulated GO terms in biological process (BP) clusters were analyzed. The data shown are the negative log2 P-values within each category. (E) Central pathways and differentially expressed genes are shown according to the chip data. Genes in red type indicate the up-regulated genes, and genes in black type indicate the down-regulated genes. (F) The relative mRNA expression level of PRKDC was detected by quantitative RT-PCR assay. (G) TRECs and GAPDH expression levels were detected in the 4 patients before and after decitabine therapy as described in the “Method” section. UPN# (line 7 and 8) was shown as negative control, who was not received the DAC treatment.

References

    1. Arstila TP, Casrouge A, Baron V, Even J, Kanellopoulos J, Kourilsky P. A direct estimate of the human alphabeta T cell receptor diversity. Science. 1999;286:958–961.
    1. Freeman JD, Warren RL, Webb JR, Nelson BH, Holt RA. Profiling the T-cell receptor beta-chain repertoire by massively parallel sequencing. Genome Res. 2009;19:1817–1824.
    1. van Heijst JW, Ceberio I, Lipuma LB, Samilo DW, Wasilewski GD, Gonzales AM, Nieves JL, van den Brink MR, Perales MA, Pamer EG. Quantitative assessment of T cell repertoire recovery after hematopoietic stem cell transplantation. Nat Med. 2013;19:372–377.
    1. Yager EJ, Ahmed M, Lanzer K, Randall TD, Woodland DL, Blackman MA. Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus. J Exp Med. 2008;205:711–723.
    1. Cicin-Sain L, Smyk-Pearson S, Currier N, Byrd L, Koudelka C, Robinson T, Swarbrick G, Tackitt S, Legasse A, Fischer M, Nikolich-Zugich D, Park B, Hobbs T, et al. Loss of naive T cells and repertoire constriction predict poor response to vaccination in old primates. J Immunol. 2010;184:6739–6745.
    1. Luo W, Liao WJ, Huang YT, Shi M, Zhang Y, Wen Q, Zhou MQ, Ma L. Normalization of T cell receptor repertoire diversity in patients with advanced colorectal cancer who responded to chemotherapy. Cancer Sci. 2011;102:706–712.
    1. Muraro PA, Robins H, Malhotra S, Howell M, Phippard D, Desmarais C, de Paula Alves Sousa A, Griffith LM, Lim N, Nash RA, Turka LA. T cell repertoire following autologous stem cell transplantation for multiple sclerosis. J Clin Invest. 2014;124:1168–1172.
    1. Robins H. Immunosequencing: applications of immune repertoire deep sequencing. Curr Opin Immunol. 2013;25:646–652.
    1. Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, Fong L. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med. 2014;6:238ra270.
    1. Britanova OV, Putintseva EV, Shugay M, Merzlyak EM, Turchaninova MA, Staroverov DB, Bolotin DA, Lukyanov S, Bogdanova EA, Mamedov IZ, Lebedev YB, Chudakov DM. Age-related decrease in TCR repertoire diversity measured with deep and normalized sequence profiling. J Immunol. 2014;192:2689–2698.
    1. Fan H, Lu X, Wang X, Liu Y, Guo B, Zhang Y, Zhang W, Nie J, Feng K, Chen M, Zhang Y, Wang Y, Shi F, et al. Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a phase I/II report. J Immunol Res. 2014;2014:371087.
    1. Nie J, Liu L, Li X, Han W. Decitabine, a new star in epigenetic therapy: the clinical application and biological mechanism in solid tumors. Cancer Lett. 2014;354:12–20.
    1. Appleton K, Mackay HJ, Judson I, Plumb JA, McCormick C, Strathdee G, Lee C, Barrett S, Reade S, Jadayel D, Tang A, Bellenger K, Mackay L, et al. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol. 2007;25:4603–4609.
    1. Stathis A, Hotte SJ, Chen EX, Hirte HW, Oza AM, Moretto P, Webster S, Laughlin A, Stayner LA, McGill S, Wang L, Zhang WJ, Espinoza-Delgado I, et al. Phase I study of decitabine in combination with vorinostat in patients with advanced solid tumors and non-Hodgkin's lymphomas. Clin Cancer Res. 2011;17:1582–1590.
    1. Mei Q, Chen M, Lu X, Li X, Duan F, Wang M, Luo G, Han W. An open-label, single-arm, phase I/II study of lower-dose decitabine based therapy in patients with advanced hepatocellular carcinoma. Oncotarget. 2015;6:16698–16711. doi: 10.18632/oncotarget.3677.
    1. Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, Berry WA, Huang T, Nephew KP. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res. 2012;72:2197–2205.
    1. Heninger E, Krueger TE, Lang JM. Augmenting antitumor immune responses with epigenetic modifying agents. Front Immunol. 2015;6:29.
    1. Roulois D, Loo Yau H, Singhania R, Wang Y, Danesh A, Shen SY, Han H, Liang G, Jones PA, Pugh TJ, O'Brien C, De Carvalho DD. DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell. 2015;162:961–973.
    1. Farace F, Angevin E, Poullion I, Leboullaire C, Ferir G, Elias D, Escudier B, Triebel F. T-cell receptor CDR3 size distribution analysis to evaluate specific T-cell response to cancer vaccines. Int J Cancer. 1997;71:972–977.
    1. Emerson R, Sherwood A, Desmarais C, Malhotra S, Phippard D, Robins H. Estimating the ratio of CD4+ to CD8+ T cells using high-throughput sequence data. J Immunol Methods. 2013;391:14–21.
    1. Vrisekoop N, Monteiro JP, Mandl JN, Germain RN. Revisiting thymic positive selection and the mature T cell repertoire for antigen. Immunity. 2014;41:181–190.
    1. Robert L, Tsoi J, Wang X, Emerson R, Homet B, Chodon T, Mok S, Huang RR, Cochran AJ, Comin-Anduix B, Koya RC, Graeber TG, Robins H, et al. CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res. 2014;20:2424–2432.
    1. Machnes-Maayan D, Lev A, Katz U, Mishali D, Vardi A, Simon AJ, Somech R. Insight into normal thymic activity by assessment of peripheral blood samples. Immunol Res. 2015;61:198–205.
    1. Greiff V, Miho E, Menzel U, Reddy ST. Bioinformatic and Statistical Analysis of Adaptive Immune Repertoires. Trends Immunol. 2015;36:738–749.
    1. Busch DH, Pamer EG. T cell affinity maturation by selective expansion during infection. J Exp Med. 1999;189:701–710.
    1. Mason D. A very high level of crossreactivity is an essential feature of the T-cell receptor. Immunol. 1998;19:395–404.
    1. Messaoudi I, Guevara Patino JA, Dyall R, LeMaoult J, Nikolich-Zugich J. Direct link between mhc polymorphism, T cell avidity, and diversity in immune defense. Science. 2002;298:1797–1800.
    1. Meyer-Olson D, Shoukry NH, Brady KW, Kim H, Olson DP, Hartman K, Shintani AK, Walker CM, Kalams SA. Limited T cell receptor diversity of HCV-specific T cell responses is associated with CTL escape. J Exp Med. 2004;200:307–319.
    1. Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM, Herati RS, Mansfield KD, Patsch D, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520:373–377.
    1. Sportes C, Hakim FT, Memon SA, Zhang H, Chua KS, Brown MR, Fleisher TA, Krumlauf MC, Babb RR, Chow CK, Fry TJ, Engels J, Buffet R, et al. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J Exp Med. 2008;205:1701–1714.
    1. Al-Chami E, Tormo A, Pasquin S, Kanjarawi R, Ziouani S, Rafei M. Interleukin-21 administration to aged mice rejuvenates their peripheral T-cell pool by triggering de novo thymopoiesis. Aging cell. 2016;15:349–60. doi: 10.1111/acel.12440.
    1. Li X, Mei Q, Nie J, Fu X, Han W. Decitabine: a promising epi-immunotherapeutic agent in solid tumors. Expert Rev Clin Immunol. 2015;11:363–375.
    1. Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC, Lim SG, Bertoletti A. Profile of tumor antigen-specific CD8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology. 2009;137:682–690.
    1. Peng D, Kryczek I, Nagarsheth N, Zhao L, Wei S, Wang W, Sun Y, Zhao E, Vatan L, Szeliga W, Kotarski J, Tarkowski R, Dou Y, et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature. 2015;527:249–253.
    1. Wu J, Liu D, Tu W, Song W, Zhao X. T-cell receptor diversity is selectively skewed in T-cell populations of patients with Wiskott-Aldrich syndrome. J Allergy Clin Immunol. 2015;135:209–216.
    1. Langerak AW, van Den Beemd R, Wolvers-Tettero IL, Boor PP, van Lochem EG, Hooijkaas H, van Dongen JJ. Molecular and flow cytometric analysis of the Vbeta repertoire for clonality assessment in mature TCRalphabeta T-cell proliferations. Blood. 2001;98:165–173.
    1. Yousfi Monod M, Giudicelli V, Chaume D, Lefranc MP. IMGT/JunctionAnalysis: the first tool for the analysis of the immunoglobulin and T cell receptor complex V-J and V-D-J JUNCTIONs. Bioinformatics. 2004;20:i379–385.
    1. Li Y, Geng S, Yin Q, Chen S, Yang L, Wu X, Li B, Du X, Schmidt CA, Przybylski GK. Decreased level of recent thymic emigrants in CD4+ and CD8+T cells from CML patients. J Transl Med. 2010;8:47.

Source: PubMed

3
Sottoscrivi