Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2023

Open Access 01-12-2023 | Auranofin | Correspondence

Direct inhibition of dioxygenases TET1 by the rheumatoid arthritis drug auranofin selectively induces cancer cell death in T-ALL

Authors: Long Chen, Anqi Ren, Yuan Zhao, Hangyu Chen, Qifang Wu, Mengzhu Zheng, Zijian Zhang, Tongcun Zhang, Wu Zhong, Jian Lin, Haichuan Zhu

Published in: Journal of Hematology & Oncology | Issue 1/2023

Login to get access

Abstract

T-cell acute lymphoblastic leukemia (T-ALL) is a type of hematologic tumor with malignant proliferation of hematopoietic progenitor cells. However, traditional clinical treatment of T-ALL included chemotherapy and stem cell transplantation always lead to recurrence and poor prognosis, thus new therapeutic targets and drugs are urgently needed for T-ALL treatment. In this study, we showed that TET1 (ten-eleven translocation 1), a key participant of DNA epigenetic control, which catalyzes the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) to modulate gene expression, was highly upregulated in human T-ALL and negatively correlated with the prognosis of patients. Knockdown of TET1 suppressed T-ALL growth and progression, suggesting that TET1 inhibition maybe an effective way to fight T-ALL via DNA epigenetic modulation. Combining structure-guided virtual screening and cell-based high-throughput screening of FDA-approved drug library, we discovered that auranofin, a gold-containing compound, is a potent TET1 inhibitor. Auranofin inhibited the catalytic activity of TET1 through competitive binding to its substrates binding pocket and thus downregulated the genomic level of 5hmC marks and particularly epigenetically reprogramed the expression of oncogene c-Myc in T-ALL in TET1-dependent manner and resulted in suppression of T-ALL in vitro and in vivo. These results revealed that TET1 is a potential therapeutic target in human T-ALL and elucidated the mechanism that TET1 inhibitor auranofin suppressed T-ALL through the TET1/5hmC/c-Myc signaling pathway. Our work thus not only provided mechanism insights for T-ALL treatment, but also discovered potential small molecule therapeutics for T-ALL.
Appendix
Available only for authorised users
Literature
2.
go back to reference Bensberg, M. et al. TET2 as a tumor suppressor and therapeutic target in T-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA 118 (2021). Bensberg, M. et al. TET2 as a tumor suppressor and therapeutic target in T-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA 118 (2021).
3.
go back to reference Bamezai S, et al. TET1 promotes growth of T-cell acute lymphoblastic leukemia and can be antagonized via PARP inhibition. Leukemia. 2021;35:389–403.CrossRefPubMed Bamezai S, et al. TET1 promotes growth of T-cell acute lymphoblastic leukemia and can be antagonized via PARP inhibition. Leukemia. 2021;35:389–403.CrossRefPubMed
4.
go back to reference Pulikkottil AJ, et al. TET3 promotes AML growth and epigenetically regulates glucose metabolism and leukemic stem cell associated pathways. Leukemia. 2022;36:416–25.CrossRefPubMed Pulikkottil AJ, et al. TET3 promotes AML growth and epigenetically regulates glucose metabolism and leukemic stem cell associated pathways. Leukemia. 2022;36:416–25.CrossRefPubMed
5.
go back to reference Zhao A, Zhou H, Yang J, Li M, Niu T. Epigenetic regulation in hematopoiesis and its implications in the targeted therapy of hematologic malignancies. Signal Transduct Target Ther. 2023;8:71.CrossRefPubMedPubMedCentral Zhao A, Zhou H, Yang J, Li M, Niu T. Epigenetic regulation in hematopoiesis and its implications in the targeted therapy of hematologic malignancies. Signal Transduct Target Ther. 2023;8:71.CrossRefPubMedPubMedCentral
6.
go back to reference Kaplánek R, et al. TET protein inhibitors: potential and limitations. Biomed Pharmacother. 2023;166:115324.CrossRefPubMed Kaplánek R, et al. TET protein inhibitors: potential and limitations. Biomed Pharmacother. 2023;166:115324.CrossRefPubMed
8.
go back to reference Weirath NA, et al. Small molecule inhibitors of TET dioxygenases: bobcat339 activity is mediated by contaminating copper(II). ACS Med Chem Lett. 2022;13:792–8.CrossRefPubMedPubMedCentral Weirath NA, et al. Small molecule inhibitors of TET dioxygenases: bobcat339 activity is mediated by contaminating copper(II). ACS Med Chem Lett. 2022;13:792–8.CrossRefPubMedPubMedCentral
9.
go back to reference Freire Boullosa L, et al. Auranofin reveals therapeutic anticancer potential by triggering distinct molecular cell death mechanisms and innate immunity in mutant p53 non-small cell lung cancer. Redox Biol. 2021;42:101949.CrossRefPubMedPubMedCentral Freire Boullosa L, et al. Auranofin reveals therapeutic anticancer potential by triggering distinct molecular cell death mechanisms and innate immunity in mutant p53 non-small cell lung cancer. Redox Biol. 2021;42:101949.CrossRefPubMedPubMedCentral
10.
go back to reference Roder C, Thomson MJ. Auranofin: repurposing an old drug for a golden new age. Drugs R&D. 2015;15:13–20.CrossRef Roder C, Thomson MJ. Auranofin: repurposing an old drug for a golden new age. Drugs R&D. 2015;15:13–20.CrossRef
11.
go back to reference Sanchez-Martin M, Ferrando A. The NOTCH1-MYC highway toward T-cell acute lymphoblastic leukemia. Blood. 2017;129:1124–33.CrossRefPubMed Sanchez-Martin M, Ferrando A. The NOTCH1-MYC highway toward T-cell acute lymphoblastic leukemia. Blood. 2017;129:1124–33.CrossRefPubMed
12.
go back to reference Li Q, Pan S, Xie T, Liu H. MYC in T-cell acute lymphoblastic leukemia: functional implications and targeted strategies. Blood Sci (Baltimore, MD). 2021;3:65–70. Li Q, Pan S, Xie T, Liu H. MYC in T-cell acute lymphoblastic leukemia: functional implications and targeted strategies. Blood Sci (Baltimore, MD). 2021;3:65–70.
Metadata
Title
Direct inhibition of dioxygenases TET1 by the rheumatoid arthritis drug auranofin selectively induces cancer cell death in T-ALL
Authors
Long Chen
Anqi Ren
Yuan Zhao
Hangyu Chen
Qifang Wu
Mengzhu Zheng
Zijian Zhang
Tongcun Zhang
Wu Zhong
Jian Lin
Haichuan Zhu
Publication date
01-12-2023
Publisher
BioMed Central
Keyword
Auranofin
Published in
Journal of Hematology & Oncology / Issue 1/2023
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-023-01513-6

Other articles of this Issue 1/2023

Journal of Hematology & Oncology 1/2023 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