Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation

Andreas D Ørskov, Marianne B Treppendahl, Anni Skovbo, Mette S Holm, Lone S Friis, Marianne Hokland, Kirsten Grønbæk, Andreas D Ørskov, Marianne B Treppendahl, Anni Skovbo, Mette S Holm, Lone S Friis, Marianne Hokland, Kirsten Grønbæk

Abstract

The hypomethylating agents (HMAs) are standard therapy for patients with higher-risk myelodysplastic syndrome (MDS); however, the majority of the patients will lose their response to HMAs over time due to unknown mechanisms. It has recently been shown that T cell expression of the immunoinhibitory receptor PD-1 is regulated by DNA methylation. In 12 of 27 patients (44%) PD-1 promoter demethylation was observed in sorted peripheral blood T cells isolated over consecutive cycles of treatment with 5-azacytidine (5-aza). The PD-1 promoter demethylation correlated with an increase in PD-1 expression. Moreover, demethylation of the PD-1 promoter correlated with a significantly worse overall response rate (8% vs. 60%, p = 0.014), and a trend towards a shorter overall survival (p = 0.11) was observed. A significantly higher baseline methylation level of the PD-1 promoter was observed in T cells of non-responding patients compared to healthy controls (p = 0.023). Accordingly, in addition to their beneficial function, HMAs induce PD-1 expression on T cells in the MDS microenvironment, thereby likely hampering the immune response against the MDS blasts. Thus, we suggest that activation of the PD-1 checkpoint during HMA treatment can be a possible resistance mechanism, which may be overcome by combination therapy with a PD-1 pathway inhibitor.

Keywords: DNA methylation; T cells; hypomethylating agents; myelodysplastic syndromes; programmed death-1.

Conflict of interest statement

DISCLOSURE OF CONFLICTS OF INTEREST

The authors declare no competing financial interests.

Figures

Figure 1. Mean PD-1 promoter methylation in…
Figure 1. Mean PD-1 promoter methylation in six distinct cell populations from 5 healthy donors
All cells are from peripheral blood. The graph shows the means. PBMNC = peripheral blood mononuclear cells. CD3 = CD3+ T cells. CD4 = CD4+ T cells. CD8 = CD8+ T cells. Gran = granulocytes. B cells = CD19+ B cells.
Figure 2. Dynamics of PD-1 promoter methylation…
Figure 2. Dynamics of PD-1 promoter methylation in peripheral blood mononuclear cells of 15 patients during treatment with 5-azacytidine
(A)PD-1 promoter methylation in the 9 patients in whom we observed a classifiable demethylation. (B)PD-1 promoter methylation in the 6 patients in whom we did not observe a classifiable demethylation. C = course of 5-aza treatment. D = day in treatment course. PBMNC = peripheral blood mononuclear cells.
Figure 3. Dynamics of PD-1 promoter methylation…
Figure 3. Dynamics of PD-1 promoter methylation in peripheral blood CD4+ and CD8+ T cells from the patients with PD-1 promoter demethylation during treatment with 5-azacytidine
(A)PD-1 promoter methylation in CD4+ T cells of eight patients. (B)PD-1 promoter methylation in CD8+ T cells of eight patients. In patient no. 123 we only observed demethylation in the CD4+ T cells (CD8+ T cells from patient no. 123 are included in Figure 4B) and in patient no. 60 we only observed demethylation in the CD8+ T cells (CD4+ T cells from patient no. 60 are included in Figure 4A). C = course of 5-aza treatment. D = day in treatment course.
Figure 4. Dynamics of PD-1 promoter methylation…
Figure 4. Dynamics of PD-1 promoter methylation in peripheral blood CD4+ and CD8+ T cells from patients without PD-1 promoter demethylation during treatment with 5-azacytidine
(A)PD-1 promoter methylation in CD4+ T cells of 14 patients. (B)PD-1 promoter methylation in CD8+ T cells of 14 patients. In patient no. 123 we observed demethylation in the CD4+ T cells (CD4+ T cells from patient no. 123 are included in Figure 3A) and in patient no. 60 we observed demethylation in the CD8+ T cells (CD8+ T cells from patient no. 60 are included in Figure 3A). C = course of 5-aza treatment. D = day in treatment course.
Figure 5. Baseline PD-1 promoter methylation in…
Figure 5. Baseline PD-1 promoter methylation in peripheral blood CD4+ and CD8+ T cells from 5-azacytidine treated patients and five healthy donors
The patients are grouped according to whether the PD-1 promoter demethylates or not during treatment. The values for CD4+ and CD8− T cells are pooled.
Figure 6. PD-1 protein surface expression on…
Figure 6. PD-1 protein surface expression on CD8+ T cells from 5-aza treated patients
PD-1 protein surface expression, as expressed by the median fluorescence intensity (MFI), on CD8+/CD45RO+ T cells from PBMNCs of four patients at baseline before 5-aza treatment (1) and following the first treatment cycle (2) (the first available sample for every patient after the first treatment cycle. Patient no. 94, 123, 141 and 146: C4D5, C3D1, C2D1 and C3D1, respectively (C = course of 5-aza treatment. D = day in treatment course)). In patient no. 94 and 141 an increase in PD-1 protein expression is observed (MFI: 770 to 853 (MFIR: 1.11), and MFI: 220 to 358 (MFIR: 1.63), respectively), whereas in patient no. 123 and 146 no increase is observed (MFI: 336 to 291 (MFIR: 0.89), and MFI: 426 to 373 (MFIR: 0.88), respectively). Correspondingly, we observed a significant demethylation of the PD-1 promoter during treatment in CD8+ T cells in patient no. 94 and 141 (Figure 3B) and no demethylation in patient no. 123 and 146 (Figure 4B). Moreover, we saw a higher baseline MFI of PD-1 in patient 94 compared to patient 141, which is in line with the variation in baseline methylation level.
Figure 7. Correlation between PD-1 promoter methylation…
Figure 7. Correlation between PD-1 promoter methylation and PD-1 gene expression in peripheral blood CD4+ and CD8+ T cells from 5-azacytidine treated patients
Ten patients and their corresponding values of methylation and relative expression before and during 5-aza treatment (altogether 117 pairs of observations). Methylation level is plotted as co-variate and the relative expression as outcome on a log-log scale with the base 2. The linear regression reveals a statistically significant inverse relationship between the two log-transformed variables (p < .0001) with the regression coefficient −2.0936 (SE 0.2521).
Figure 8. Baseline PD-1 promoter methylation in…
Figure 8. Baseline PD-1 promoter methylation in peripheral blood CD4+ and CD8+ T cells from 5-azacytidine treated patients and five healthy donors
The patients are grouped according to whether they responded or not responded to the treatment. The values for CD4+ and CD8− T cells are pooled.

References

    1. Tefferi A, Vardiman JW. Myelodysplastic syndromes. N. Engl. J. Med. 2009;361:1872–85.
    1. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, Schoch R, Gattermann N, Sanz G, List A, Gore SD, Seymour JF, Bennett JM, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10:223–32.
    1. Kantarjian H, Issa J-PJ, Rosenfeld CS, Bennett JM, Albitar M, DiPersio J, Klimek V, Slack J, de Castro C, Ravandi F, Helmer R, Shen L, Nimer SD, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006;106:1794–803.
    1. Lübbert M, Suciu S, Baila L, Rüter BH, Platzbecker U, Giagounidis A, Selleslag D, Labar B, Germing U, Salih HR, Beeldens F, Muus P, Pflüger K-H, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: final results of the randomized phase III study of the European Organisation for Rese. J. Clin. Oncol. 2011;29:1987–96.
    1. Fenaux P, Mufti GJ, Hellström-Lindberg E, Santini V, Gattermann N, Germing U, Sanz G, List AF, Gore S, Seymour JF, Dombret H, Backstrom J, Zimmerman L, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J. Clin. Oncol. 2010;28:562–9.
    1. Kantarjian HM, Thomas XG, Dmoszynska A, Wierzbowska A, Mazur G, Mayer J, Gau J-P, Chou W-C, Buckstein R, Cermak J, Kuo C-Y, Oriol A, Ravandi F, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J. Clin. Oncol. 2012;30:2670–7.
    1. Prébet T, Gore SD, Esterni B, Gardin C, Itzykson R, Thepot S, Dreyfus F, Rauzy OB, Recher C, Adès L, Quesnel B, Beach CL, Fenaux P, et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. J. Clin. Oncol. 2011;29:3322–7.
    1. Gore SD, Baylin S, Sugar E, Carraway H, Miller CB, Carducci M, Grever M, Galm O, Dauses T, Karp JE, Rudek M a, Zhao M, Smith BD, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res. 2006;66:6361–9.
    1. Qiu X, Hother C, Ralfkiær UM, Søgaard A, Lu Q, Workman CT, Liang G, Jones P a, Grønbæk K. Equitoxic doses of 5-azacytidine and 5-aza-2′deoxycytidine induce diverse immediate and overlapping heritable changes in the transcriptome. PLoS One. 2010;5:pii: e12994.
    1. Almstedt M, Blagitko-Dorfs N, Duque-Afonso J, Karbach J, Pfeifer D, Jäger E, Lübbert M. The DNA demethylating agent 5-aza-2′-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cells. Leuk. Res. 2010;34:899–905.
    1. Karpf AR. A potential role for epigenetic modulatory drugs in the enhancement of cancer/germ-line antigen vaccine efficacy. Epigenetics. 2007;1:116–20.
    1. Goodyear O, Agathanggelou A, Novitzky-Basso I, Siddique S, McSkeane T, Ryan G, Vyas P, Cavenagh J, Stankovic T, Moss P, Craddock C. Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. 2010;116:1908–18.
    1. Dougan M, Dranoff G. Immune therapy for cancer. Annu. Rev. Immunol. 2009;27:83–117.
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008;26:677–704.
    1. Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin. Cancer Res. 2013;19:1021–34.
    1. Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int. Immunol. 2007;19:813–24.
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon V a, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat. Med. 2002;8:793–800.
    1. Youngblood B, Oestreich KJ, Ha S-J, Duraiswamy J, Akondy RS, West EE, Wei Z, Lu P, Austin JW, Riley JL, Boss JM, Ahmed R. Chronic virus infection enforces demethylation of the locus that encodes PD-1 in antigen-specific CD8(+) T cells. Immunity. 2011;35:400–12.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer. 2012;12:252–64.
    1. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat. Rev. Immunol. 2008;8:467–77.
    1. Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, Chen L, Pardoll DM, Topalian SL, Anders R a. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014;20:5064–74.
    1. Kondo A, Yamashita T, Tamura H, Zhao W, Tsuji T, Shimizu M, Shinya E, Takahashi H, Tamada K, Chen L, Dan K, Ogata K. Interferon-gamma and tumor necrosis factor-alpha induce an immunoinhibitory molecule, B7-H1, via nuclear factor-kappaB activation in blasts in myelodysplastic syndromes. Blood. 2010;116:1124–31.
    1. Yang H, Bueso-Ramos C, Dinardo C, Estecio MR, Davanlou M, Geng Q-R, Fang Z, Nguyen M, Pierce S, Wei Y, Parmar S, Cortes J, Kantarjian H, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2013:1–32.
    1. Brahmer JR, Tykodi SS, Chow LQM, Hwu W-J, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012;366:2455–65.
    1. Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, Dronca R, Gangadhar TC, Patnaik A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N. Engl. J. Med. 2013;369:134–44.
    1. Youngblood B, Noto A, Porichis F, Akondy RS, Ndhlovu ZM, Austin JW, Bordi R, Procopio F a, Miura T, Allen TM, Sidney J, Sette A, Walker BD, et al. Cutting edge: Prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells. J. Immunol. 2013;191:540–4.
    1. Perez-Gracia JL, Labiano S, Rodriguez-Ruiz ME, Sanmamed MF, Melero I. Orchestrating immune check-point blockade for cancer immunotherapy in combinations. Curr. Opin. Immunol. 2014;27:89–97.
    1. Crespo J, Sun H, Welling TH, Tian Z, Zou W. T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment. Curr. Opin. Immunol. 2013;25:214–21.
    1. Wrangle J, Wang W, Koch A, Easwaran H, Mohammad HP, Vendetti F, Vancriekinge W, Demeyer T, Du Z, Parsana P, Rodgers K, Yen R-W, Zahnow CA, et al. Alterations of immune response of Non-Small Cell Lung Cancer with Azacytidine. Oncotarget. 2013;4:2067–79.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012;366:2443–54.
    1. Brahmer JR, Tykodi SS, Chow LQM, Hwu W-J, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012;366:2455–65.
    1. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–7.
    1. Vardiman JW, Thiele J, Arber D a, Brunning RD, Borowitz MJ, Porwit A, Harris NL, Le Beau MM, Hellström-Lindberg E, Tefferi A, Bloomfield CD. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114:937–51.
    1. Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, Ohyashiki K, Toyama K, Aul C, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079–88.
    1. Cheson BD, Greenberg PL, Bennett JM, Lowenberg B, Wijermans PW, Nimer SD, Pinto A, Beran M, de Witte TM, Stone RM, Mittelman M, Sanz GF, Gore SD, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108:419–25.
    1. Arya M, Shergill IS, Williamson M, Gommersall L, Arya N, Patel HRH. Basic principles of real-time quantitative PCR. Expert Rev. Mol. Diagn. 2005;5:209–19.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C (T)) Method. Methods. 2001;25:402–8.

Source: PubMed

3
Subskrybuj