Inhibitors of histone deacetylase 1 reverse the immune evasion phenotype to enhance T-cell mediated lysis of prostate and breast carcinoma cells

Sofia R Gameiro, Anthony S Malamas, Kwong Y Tsang, Soldano Ferrone, James W Hodge, Sofia R Gameiro, Anthony S Malamas, Kwong Y Tsang, Soldano Ferrone, James W Hodge

Abstract

The clinical promise of cancer immunotherapy relies on the premise that the immune system can recognize and eliminate tumor cells identified as non-self. However, tumors can evade host immune surveillance through multiple mechanisms, including epigenetic silencing of genes involved in antigen processing and immune recognition. Hence, there is an unmet clinical need to develop effective therapeutic strategies that can restore tumor immune recognition when combined with immunotherapy, such as immune checkpoint blockade and therapeutic cancer vaccines. We sought to examine the potential of clinically relevant exposure of prostate and breast human carcinoma cells to histone deacetylase (HDAC) inhibitors to reverse tumor immune escape to T-cell mediated lysis. Here we demonstrate that prostate (LNCAP) and breast (MDA-MB-231) carcinoma cells are more sensitive to T-cell mediated lysis in vitro after clinically relevant exposure to epigenetic therapy with either the pan-HDAC inhibitor vorinostat or the class I HDAC inhibitor entinostat. This pattern of immunogenic modulation was observed against a broad range of tumor-associated antigens, such as CEA, MUC1, PSA, and brachyury, and associated with augmented expression of multiple proteins involved in antigen processing and tumor immune recognition. Genetic and pharmacological inhibition studies identified HDAC1 as a key determinant in the reversal of carcinoma immune escape. Further, our findings suggest that the observed reversal of tumor immune evasion is driven by a response to cellular stress through activation of the unfolded protein response. This offers the rationale for combining HDAC inhibitors with immunotherapy, including therapeutic cancer vaccines.

Keywords: antigen-processing machinery; entinostat; histone deacetylases; immunogenic modulation; vorinostat.

Conflict of interest statement

CONFLICTS OF INTEREST

Authors have no other potential conflicts of interest.

Figures

Figure 1. Vorinostat decreases pan-HDAC activity and…
Figure 1. Vorinostat decreases pan-HDAC activity and proliferation of human carcinoma cells in an exposure-dependent manner
Human prostate (LNCaP) and breast (MDA-MB-231) carcinoma cells were exposed to vorinostat (1 μM, grey circles and bars; 3 μM, black circles and bars), or vehicle (DMSO, open squares and bars). A. and B. HDAC activity determined at 96h. Results are presented as mean ± S.E.M. from replicate wells. C. and D. Cell number at the indicated time points. Insets denote viability at 96 h. Results are presented as mean ± S.D. from 6 replicate wells. Asterisks denote statistical significance relative to control cells exposed to vehicle (DMSO, P < 0.001). This experiment was repeated 2-3 times with similar results.
Figure 2. Carcinoma cells exposed to vorinostat…
Figure 2. Carcinoma cells exposed to vorinostat are significantly more sensitive to CTL-mediated killing
Human prostate (LNCaP) and breast (MDA-MB-231) carcinoma cells were exposed to vorinostat (3 μM, black bars) or to vehicle (DMSO, open bars) as described in Materials and Methods, prior to being used as targets for antigen-specific CTL lysis using CEA-, brachyury-, MUC1-, or PSA-specific CD8+ T cells as effector cells (E:T = 30:1). To verify that effector T cells were HLA-restricted, CTLs were incubated with HLA-A2 negative AsPC-1 pancreatic carcinoma cells exposed to vehicle (DMSO) or vorinostat. Results are presented as mean ± S.E.M. from 3-6 replicate wells, and are representative of 1-4 independent experiments. Asterisks denote statistical significance relative to controls.
Figure 3. Vorinostat-induced immunogenic modulation of MDA-MB-231…
Figure 3. Vorinostat-induced immunogenic modulation of MDA-MB-231 carcinoma cells is mediated by HDAC1
MDA-MB-231 cells were exposed to siRNA control or targeting HDAC1 for 24 h prior to being exposed to vehicle (DMSO) or vorinostat (3 μM) as described in Materials and Methods. A. At the end of treatment, total cell lysates were examined by Western blotting to determine expression of HDAC1. GAPDH was used as internal control for total protein levels. Silencing ratio denotes HDAC1 protein expression relative to GAPDH, further normalized to protein levels after treatment in the presence of silencing RNA control. B. At the end of treatment, MDA-MB-231 cells were used as targets in a CTL-lysis assay where effector brachyury-specific CD8+ T cells were used at an E:T ratio of 30:1. Results are presented as mean ± S.E.M. from 4-6 replicate wells. Asterisks denote statistical significance relative to controls (*P = 0.002). Data is representative of two independent experiments.
Figure 4. HDAC inhibition activates the ER…
Figure 4. HDAC inhibition activates the ER stress responsive element in LNCaP carcinoma cells in a dose-dependent manner
A. Single-cell clones of LNCaP cells stably transduced with an ER stress responsive element driving firefly luciferase expression were exposed to vorinostat or entinostat, at the designated concentrations, or DMSO controls, as described in Materials and Methods. At the end of treatment, firefly and renilla luciferase activities were determined. Data are shown as the ratio of firefly luciferase activity relative to that of control renilla luciferase within each well, further normalized to DMSO control. Results are presented as mean ± S.E.M. from 4-6 replicate wells, and are representative of two independent experiments. B. Parental LNCaP prostate carcinoma cells were exposed to vorinostat (3 μM), entinostat (500 nM) or to vehicle (DMSO) controls as described in Materials and Methods, prior to being used as targets for antigen-specific CTL lysis using PSA-specific CD8+ T cells as effector cells (E:T = 30:1). Results are presented as mean ± S.E.M. from 6 replicate wells. Asterisks denote statistical significance relative to controls (P < 0.05). C. Schematic representation of immunogenic modulation induced by HDAC inhibition in human carcinoma cells.
Figure 5. Vorinostat-induced immunogenic modulation is mediated…
Figure 5. Vorinostat-induced immunogenic modulation is mediated by the unfolded protein response
MDA-MB-231 cells were exposed to siRNA control or targeting ERN1 or PERK for 24 h prior to being exposed to vehicle (DMSO) or vorinostat (3 μM) as described in Materials and Methods. A. and B. At the end of treatment, total cell lysates were examined by Western blotting to determine expression of ERN1 (A) or PERK (B). GAPDH was used as internal control for total protein levels. Silencing ratio denotes target protein expression relative to GAPDH, further normalized to protein levels after treatment in the presence of silencing RNA control. C. and D. At the end of treatment, MDA-MB-231 cells were used as targets in a CTL lysis assay using CEA-specific CD8+ T cells as effectors (E:T = 30:1). Results are presented as mean ± S.E.M. from 6 replicate wells, and are representative of 2-3 independent experiments. Asterisks denote statistical significance relative to controls (P < 0.0001).

References

    1. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.
    1. Hicklin DJ, Marincola FM, Ferrone S. HLA class I antigen downregulation in human cancers: T-cell immunotherapy revives an old story. Mol Med Today. 1999;5:178–186.
    1. Johnsen AK, Templeton DJ, Sy M, Harding CV. Deficiency of transporter for antigen presentation (TAP) in tumor cells allows evasion of immune surveillance and increases tumorigenesis. J Immunol. 1999;163:4224–4231.
    1. Azad N, Zahnow CA, Rudin CM, Baylin SB. The future of epigenetic therapy in solid tumours—lessons from the past. Nat Rev Clin Oncol. 2013;10:256–266.
    1. Wrangle J, Wang W, Koch A, Easwaran H, Mohammad HP, Vendetti F, Vancriekinge W, Demeyer T, Du Z, Parsana P, Rodgers K, Yen RW, Zahnow CA, et al. Alterations of immune response of Non-Small Cell Lung Cancer with Azacytidine. Oncotarget. 2013;4:2067–2079. doi: 10.18632/oncotarget.1542.
    1. Ardiani A, Gameiro SR, Kwilas AR, Donahue RN, Hodge JW. Androgen deprivation therapy sensitizes prostate cancer cells to T-cell killing through androgen receptor dependent modulation of the apoptotic pathway. Oncotarget. 2014;5:9335–9348. doi: 10.18632/oncotarget.2429.
    1. Del Campo AB, Carretero J, Munoz JA, Zinchenko S, Ruiz-Cabello F, Gonzalez-Aseguinolaza G, Garrido F, Aptsiauri N. Adenovirus expressing beta2-microglobulin recovers HLA class I expression and antitumor immunity by increasing T-cell recognition. Cancer Gene Ther. 2014;21:317–332.
    1. Hodge JW, Garnett CT, Farsaci B, Palena C, Tsang KY, Ferrone S, Gameiro SR. Chemotherapy-induced immunogenic modulation of tumor cells enhances killing by cytotoxic T lymphocytes and is distinct from immunogenic cell death. Int J Cancer. 2013;133:624–636.
    1. Gameiro SR, Ardiani A, Kwilas A, Hodge JW. Radiation-induced survival responses promote immunogenic modulation to enhance immunotherapy in combinatorial regimens. Oncoimmunology. 2014;3:e28643.
    1. Gameiro SR, Jammeh ML, Wattenberg MM, Tsang KY, Ferrone S, Hodge JW. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. 2014;5:403–416. doi: 10.18632/oncotarget.1719.
    1. Hellebrekers DM, Castermans K, Vire E, Dings RP, Hoebers NT, Mayo KH, Oude Egbrink MG, Molema G, Fuks F, van Engeland M, Griffioen AW. Epigenetic regulation of tumor endothelial cell anergy: silencing of intercellular adhesion molecule-1 by histone modifications. Cancer Res. 2006;66:10770–10777.
    1. Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, Olsen JV, Mann M. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science. 2009;325:834–840.
    1. West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Invest. 2014;124:30–39.
    1. Burdelski C, Ruge OM, Melling N, Koop C, Simon R, Steurer S, Sauter G, Kluth M, Hube-Magg C, Minner S, Wittmer C, Wilczak W, Hinsch A, et al. HDAC1 overexpression independently predicts biochemical recurrence and is associated with rapid tumor cell proliferation and genomic instability in prostate cancer. Exp Mol Pathol. 2015;98:419–426.
    1. Muller BM, Jana L, Kasajima A, Lehmann A, Prinzler J, Budczies J, Winzer KJ, Dietel M, Weichert W, Denkert C. Differential expression of histone deacetylases HDAC1, 2 and 3 in human breast cancer—overexpression of HDAC2 and HDAC3 is associated with clinicopathological indicators of disease progression. BMC Cancer. 2013;13:215.
    1. Campoli M, Ferrone S. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. 2008;27:5869–5885.
    1. Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, Sebree R, Rodgers K, Hooker CM, Franco N, Lee B, Tsai S, Delgado IE, et al. Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer Discov. 2011;1:598–607.
    1. Ahuja N, Easwaran H, Baylin SB. Harnessing the potential of epigenetic therapy to target solid tumors. J Clin Invest. 2014;124:56–63.
    1. Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, Diaz LA, Jr, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A. 2014;111:11774–11779.
    1. Christiansen AJ, West A, Banks KM, Haynes NM, Teng MW, Smyth MJ, Johnstone RW. Eradication of solid tumors using histone deacetylase inhibitors combined with immune-stimulating antibodies. Proc Natl Acad Sci U S A. 2011;108:4141–4146.
    1. Schech A, Kazi A, Yu S, Shah P, Sabnis G. Histone Deacetylase Inhibitor Entinostat Inhibits Tumor-Initiating Cells in Triple-Negative Breast Cancer Cells. Mol Cancer Ther. 2015;14:1848–1857.
    1. Sigalotti L, Fratta E, Coral S, Maio M. Epigenetic drugs as immunomodulators for combination therapies in solid tumors. Pharmacol Ther. 2014;142:339–350.
    1. Pili R, Salumbides B, Zhao M, Altiok S, Qian D, Zwiebel J, Carducci MA, Rudek MA. Phase I study of the histone deacetylase inhibitor entinostat in combination with 13-cis retinoic acid in patients with solid tumours. Br J Cancer. 2012;106:77–84.
    1. Setiadi AF, Omilusik K, David MD, Seipp RP, Hartikainen J, Gopaul R, Choi KB, Jefferies WA. Epigenetic enhancement of antigen processing and presentation promotes immune recognition of tumors. Cancer Res. 2008;68:9601–9607.
    1. Woan KV, Sahakian E, Sotomayor EM, Seto E, Villagra A. Modulation of antigen-presenting cells by HDAC inhibitors: implications in autoimmunity and cancer. Immunol Cell Biol. 2012;90:55–65.
    1. Pellicciotta I, Cortez-Gonzalez X, Sasik R, Reiter Y, Hardiman G, Langlade-Demoyen P, Zanetti M. Presentation of telomerase reverse transcriptase, a self-tumor antigen, is down-regulated by histone deacetylase inhibition. Cancer Res. 2008;68:8085–8093.
    1. Fiegler N, Textor S, Arnold A, Rolle A, Oehme I, Breuhahn K, Moldenhauer G, Witzens-Harig M, Cerwenka A. Downregulation of the activating NKp30 ligand B7-H6 by HDAC inhibitors impairs tumor cell recognition by NK cells. Blood. 2013;122:684–693.
    1. Butler LM, Agus DB, Scher HI, Higgins B, Rose A, Cordon-Cardo C, Thaler HT, Rifkind RA, Marks PA, Richon VM. Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res. 2000;60:5165–5170.
    1. Richon VM, Sandhoff TW, Rifkind RA, Marks PA. Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci U S A. 2000;97:10014–10019.
    1. Iwamoto M, Friedman EJ, Sandhu P, Agrawal NG, Rubin EH, Wagner JA. Clinical pharmacology profile of vorinostat, a histone deacetylase inhibitor. Cancer Chemother Pharmacol. 2013;72:493–508.
    1. Yang PM, Lin PJ, Chen CC. CD1d induction in solid tumor cells by histone deacetylase inhibitors through inhibition of HDAC1/2 and activation of Sp1. Epigenetics. 2012;7:390–399.
    1. Nebbioso A, Clarke N, Voltz E, Germain E, Ambrosino C, Bontempo P, Alvarez R, Schiavone EM, Ferrara F, Bresciani F, Weisz A, de Lera AR, Gronemeyer H, et al. Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat Med. 2005;11:77–84.
    1. Baumeister P, Luo S, Skarnes WC, Sui G, Seto E, Shi Y, Lee AS. Endoplasmic reticulum stress induction of the Grp78/BiP promoter: activating mechanisms mediated by YY1 and its interactive chromatin modifiers. Mol Cell Biol. 2005;25:4529–4540.
    1. Hetz C, Chevet E, Oakes SA. Proteostasis control by the unfolded protein response. Nat Cell Biol. 2015;17:829–838.
    1. Kortenhorst MS, Wissing MD, Rodriguez R, Kachhap SK, Jans JJ, Van der Groep P, Verheul HM, Gupta A, Aiyetan PO, van der Wall E, Carducci MA, Van Diest PJ, Marchionni L. Analysis of the genomic response of human prostate cancer cells to histone deacetylase inhibitors. Epigenetics. 2013;8:907–920.
    1. Seliger B. Different regulation of MHC class I antigen processing components in human tumors. J Immunotoxicol. 2008;5:361–367.
    1. Sabbatino F, Schwab JH, Ferrone S, Ferrone CR. Evolution of studies of HLA class I antigen processing machinery (APM) components in malignant cells. Clin Transpl. 2013:453–463.
    1. Kelly RD, Cowley SM. The physiological roles of histone deacetylase (HDAC) 1 and 2: complex co-stars with multiple leading parts. Biochem Soc Trans. 2013;41:741–749.
    1. Telles E, Seto E. Modulation of cell cycle regulators by HDACs. Front Biosci (Schol Ed) 2012;4:831–839.
    1. Stearns V, Jacobs LK, Fackler M, Tsangaris TN, Rudek MA, Higgins M, Lange J, Cheng Z, Slater SA, Jeter SC, Powers P, Briest S, Chao C, et al. Biomarker modulation following short-term vorinostat in women with newly diagnosed primary breast cancer. Clin Cancer Res. 2013;19:4008–4016.
    1. Stein WD, Gulley JL, Schlom J, Madan RA, Dahut W, Figg WD, Ning YM, Arlen PM, Price D, Bates SE, Fojo T. Tumor regression and growth rates determined in five intramural NCI prostate cancer trials: the growth rate constant as an indicator of therapeutic efficacy. Clin Cancer Res. 2011;17:907–917.
    1. Schlom J, Hodge JW, Palena C, Tsang KY, Jochems C, Greiner JW, Farsaci B, Madan RA, Heery CR, Gulley JL. Therapeutic cancer vaccines. Adv Cancer Res. 2014;121:67–124.
    1. Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton JM, Schlom J. Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine. J Natl Cancer Inst. 1995;87:982–990.
    1. Correale P, Walmsley K, Nieroda C, Zaremba S, Zhu M, Schlom J, Tsang KY. In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen. J Natl Cancer Inst. 1997;89:293–300.
    1. Tsang KY, Palena C, Gulley J, Arlen P, Schlom J. A human cytotoxic T-lymphocyte epitope and its agonist epitope from the nonvariable number of tandem repeat sequence of MUC1. Clin Cancer Res. 2004;10:2139–2149.
    1. Tucker JA, Jochems C, Boyerinas B, Fallon J, Greiner JW, Palena C, Rodell TC, Schlom J, Tsang KY. Identification and characterization of a cytotoxic T-lymphocyte agonist epitope of brachyury, a transcription factor involved in epithelial to mesenchymal transition and metastasis. Cancer Immunol Immunother. 2014;63:1307–1317.
    1. Ogino T, Wang X, Ferrone S. Modified flow cytometry and cell-ELISA methodology to detect HLA class I antigen processing machinery components in cytoplasm and endoplasmic reticulum. J Immunol Methods. 2003;278:33–44.
    1. Bandoh N, Ogino T, Cho HS, Hur SY, Shen J, Wang X, Kato S, Miyokawa N, Harabuchi Y, Ferrone S. Development and characterization of human constitutive proteasome and immunoproteasome subunit-specific monoclonal antibodies. Tissue Antigens. 2005;66:185–194.
    1. Ogino T, Wang X, Kato S, Miyokawa N, Harabuchi Y, Ferrone S. Endoplasmic reticulum chaperone-specific monoclonal antibodies for flow cytometry and immunohistochemical staining. Tissue Antigens. 2003;62:385–393.
    1. Wang X, Campoli M, Cho HS, Ogino T, Bandoh N, Shen J, Hur SY, Kageshita T, Ferrone S. A method to generate antigen-specific mAb capable of staining formalin-fixed, paraffin-embedded tissue sections. J Immunol Methods. 2005;299:139–151.

Source: PubMed

3
Abonnieren