Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 1/2021

Open Access 01-01-2021 | Colon Cancer | Original Article

Romidepsin (FK228) regulates the expression of the immune checkpoint ligand PD-L1 and suppresses cellular immune functions in colon cancer

Authors: Yehui Shi, Ying Fu, Xin Zhang, Gang Zhao, Yuan Yao, Yan Guo, Gang Ma, Shuai Bai, Hui Li

Published in: Cancer Immunology, Immunotherapy | Issue 1/2021

Login to get access

Abstract

Romidepsin (FK228), a histone deacetylase inhibitor (HDACi), has anti-tumor effects against several types of solid tumors. Studies have suggested that HDACi could upregulate PD-L1 expression in tumor cells and change the state of anti-tumor immune responses in vivo. However, the influence of enhanced PD-L1 expression in tumor cells induced by romidepsin on anti-tumor immune responses is still under debate. So, the purpose of this study was to explore the anti-tumor effects and influence on immune responses of romidepsin in colon cancer. The results indicated that romidepsin inhibited proliferation, induced G0/G1 cell cycle arrest and increased apoptosis in CT26 and MC38 cells. Romidepsin treatment increased PD-L1 expression in vivo and in vitro via increasing the acetylation levels of histones H3 and H4 and regulating the transcription factor BRD4. In subcutaneous transplant tumor mice and colitis-associated cancer (CAC) mice, romidepsin increased the percentage of FOXP3+ regulatory T cells (Tregs), decreased the ratio of Th1/Th2 cells and the percentage of IFN-γ+ CD8+ T cells in the peripheral blood and the tumor microenvironment. Upon combination with an anti-PD-1 antibody, the anti-tumor effects of romidepsin were enhanced and the influence on CD4+ and CD8+ T cells was partially reversed. Therefore, the combination of romidepsin and anti-PD-1 immunotherapy provides a more potential treatment for colon cancer.
Literature
3.
go back to reference Briere D, Sudhakar N, Woods DM, Hallin J, Engstrom LD, Aranda R, Chiang H, Sodre AL, Olson P, Weber JS, Christensen JG (2018) The class I/IV HDAC inhibitor mocetinostat increases tumor antigen presentation, decreases immune suppressive cell types and augments checkpoint inhibitor therapy. Cancer Immunol Immunother 67(3):381–392. https://doi.org/10.1007/s00262-017-2091-yCrossRefPubMed Briere D, Sudhakar N, Woods DM, Hallin J, Engstrom LD, Aranda R, Chiang H, Sodre AL, Olson P, Weber JS, Christensen JG (2018) The class I/IV HDAC inhibitor mocetinostat increases tumor antigen presentation, decreases immune suppressive cell types and augments checkpoint inhibitor therapy. Cancer Immunol Immunother 67(3):381–392. https://​doi.​org/​10.​1007/​s00262-017-2091-yCrossRefPubMed
4.
go back to reference Knox T, Sahakian E, Banik D, Hadley M, Palmer E, Noonepalle S, Kim J, Powers J, Gracia-Hernandez M, Oliveira V, Cheng F, Chen J, Barinka C, Pinilla-Ibarz J, Lee NH, Kozikowski A, Villagra A (2019) Selective HDAC6 inhibitors improve anti-PD-1 immune checkpoint blockade therapy by decreasing the anti-inflammatory phenotype of macrophages and down-regulation of immunosuppressive proteins in tumor cells. Sci Rep 9(1):6136. https://doi.org/10.1038/s41598-019-42237-3CrossRefPubMedPubMedCentral Knox T, Sahakian E, Banik D, Hadley M, Palmer E, Noonepalle S, Kim J, Powers J, Gracia-Hernandez M, Oliveira V, Cheng F, Chen J, Barinka C, Pinilla-Ibarz J, Lee NH, Kozikowski A, Villagra A (2019) Selective HDAC6 inhibitors improve anti-PD-1 immune checkpoint blockade therapy by decreasing the anti-inflammatory phenotype of macrophages and down-regulation of immunosuppressive proteins in tumor cells. Sci Rep 9(1):6136. https://​doi.​org/​10.​1038/​s41598-019-42237-3CrossRefPubMedPubMedCentral
5.
go back to reference Fournel M, Bonfils C, Hou Y, Yan PT, Trachy-Bourget MC, Kalita A, Liu J, Lu AH, Zhou NZ, Robert MF, Gillespie J, Wang JJ, Ste-Croix H, Rahil J, Lefebvre S, Moradei O, Delorme D, Macleod AR, Besterman JM, Li Z (2008) MGCD0103, a novel isotype-selective histone deacetylase inhibitor, has broad spectrum antitumor activity in vitro and in vivo. Mol Cancer Ther 7(4):759–768. https://doi.org/10.1158/1535-7163.Mct-07-2026CrossRefPubMed Fournel M, Bonfils C, Hou Y, Yan PT, Trachy-Bourget MC, Kalita A, Liu J, Lu AH, Zhou NZ, Robert MF, Gillespie J, Wang JJ, Ste-Croix H, Rahil J, Lefebvre S, Moradei O, Delorme D, Macleod AR, Besterman JM, Li Z (2008) MGCD0103, a novel isotype-selective histone deacetylase inhibitor, has broad spectrum antitumor activity in vitro and in vivo. Mol Cancer Ther 7(4):759–768. https://​doi.​org/​10.​1158/​1535-7163.​Mct-07-2026CrossRefPubMed
10.
11.
go back to reference Ciardiello C, Roca MS, Noto A, Bruzzese F, Moccia T, Vitagliano C, Di Gennaro E, Ciliberto G, Roscilli G, Aurisicchio L, Marra E, Mancini R, Budillon A, Leone A (2016) Synergistic antitumor activity of histone deacetylase inhibitors and anti-ErbB3 antibody in NSCLC primary cultures via modulation of ErbB receptors expression. Oncotarget 7(15):19559–19574. https://doi.org/10.18632/oncotarget.7195CrossRefPubMedPubMedCentral Ciardiello C, Roca MS, Noto A, Bruzzese F, Moccia T, Vitagliano C, Di Gennaro E, Ciliberto G, Roscilli G, Aurisicchio L, Marra E, Mancini R, Budillon A, Leone A (2016) Synergistic antitumor activity of histone deacetylase inhibitors and anti-ErbB3 antibody in NSCLC primary cultures via modulation of ErbB receptors expression. Oncotarget 7(15):19559–19574. https://​doi.​org/​10.​18632/​oncotarget.​7195CrossRefPubMedPubMedCentral
15.
17.
23.
go back to reference Hu X, Lu X, Liu R, Ai N, Cao Z, Li Y, Liu J, Yu B, Liu K, Wang H, Zhou C, Wang Y, Han A, Ding F, Chen R (2014) Histone cross-talk connects protein phosphatase 1α (PP1α) and histone deacetylase (HDAC) pathways to regulate the functional transition of bromodomain-containing 4 (BRD4) for inducible gene expression. J Biol Chem 289(33):23154–23167. https://doi.org/10.1074/jbc.M114.570812CrossRefPubMedPubMedCentral Hu X, Lu X, Liu R, Ai N, Cao Z, Li Y, Liu J, Yu B, Liu K, Wang H, Zhou C, Wang Y, Han A, Ding F, Chen R (2014) Histone cross-talk connects protein phosphatase 1α (PP1α) and histone deacetylase (HDAC) pathways to regulate the functional transition of bromodomain-containing 4 (BRD4) for inducible gene expression. J Biol Chem 289(33):23154–23167. https://​doi.​org/​10.​1074/​jbc.​M114.​570812CrossRefPubMedPubMedCentral
26.
go back to reference Riganti C, Lingua MF, Salaroglio IC, Falcomata C, Righi L, Morena D, Picca F, Oddo D, Kopecka J, Pradotto M, Libener R, Orecchia S, Bironzo P, Comunanza V, Bussolino F, Novello S, Scagliotti GV, Di Nicolantonio F, Taulli R (2018) Bromodomain inhibition exerts its therapeutic potential in malignant pleural mesothelioma by promoting immunogenic cell death and changing the tumor immune-environment. Oncoimmunology 7(3):e1398874. https://doi.org/10.1080/2162402X.2017.1398874CrossRefPubMed Riganti C, Lingua MF, Salaroglio IC, Falcomata C, Righi L, Morena D, Picca F, Oddo D, Kopecka J, Pradotto M, Libener R, Orecchia S, Bironzo P, Comunanza V, Bussolino F, Novello S, Scagliotti GV, Di Nicolantonio F, Taulli R (2018) Bromodomain inhibition exerts its therapeutic potential in malignant pleural mesothelioma by promoting immunogenic cell death and changing the tumor immune-environment. Oncoimmunology 7(3):e1398874. https://​doi.​org/​10.​1080/​2162402X.​2017.​1398874CrossRefPubMed
Metadata
Title
Romidepsin (FK228) regulates the expression of the immune checkpoint ligand PD-L1 and suppresses cellular immune functions in colon cancer
Authors
Yehui Shi
Ying Fu
Xin Zhang
Gang Zhao
Yuan Yao
Yan Guo
Gang Ma
Shuai Bai
Hui Li
Publication date
01-01-2021
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 1/2021
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-020-02653-1

Other articles of this Issue 1/2021

Cancer Immunology, Immunotherapy 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine