Artemis inhibition as a therapeutic strategy for acute lymphoblastic leukemia

As effective therapies for relapse and refractory B-cell acute lymphoblastic leukemia (B-ALL) remain problematic, novel therapeutic strategies are needed. Artemis is a key endonuclease in V(D)J recombination and nonhomologous end joining (NHEJ) of DNA double-strand break (DSB) repair. Inhibition of Artemis would cause chromosome breaks during maturation of RAG-expressing T- and B-cells. Though this would block generation of new B- and T-cells temporarily, it could be oncologically beneficial for reducing the proliferation of B-ALL and T-ALL cells by causing chromosome breaks in these RAG-expressing tumor cells. Currently, pharmacological inhibition is not available for Artemis. According to gene expression analyses from 207 children with high-risk pre-B acute lymphoblastic leukemias high Artemis expression is correlated with poor outcome. Therefore, we evaluated four compounds (827171, 827032, 826941, and 825226), previously generated from a large Artemis targeted drug screen. A biochemical assay using a purified Artemis:DNA-PKcs complex shows that the Artemis inhibitors 827171, 827032, 826941, 825226 have nanomolar IC50 values for Artemis inhibition. We compared these 4 compounds to a DNA-PK inhibitor (AZD7648) in three patient-derived B-ALL cell lines (LAX56, BLQ5 and LAX7R) and in two mature B-cell lines (3301015 and 5680001) as controls. We found that pharmacological Artemis inhibition substantially decreases proliferation of B-ALL cell lines while normal mature B-cell lines are not markedly affected. Inhibition of DNA-PKcs (which regulates Artemis) using the DNA-PK inhibitor AZD7648 had minor effects on these same primary patient-derived ALL lines, indicating that inhibition of V(D)J hairpin opening requires direct inhibition of Artemis, rather than indirect suppression of the kinase that regulates Artemis. Our data provides a basis for further evaluation of pharmacological Artemis inhibition of proliferation of B- and T-ALL.

[1]  M. Lieber,et al.  Partial deletions of the autoregulatory C-terminal domain of Artemis and their effect on its nuclease activity. , 2022, DNA repair.

[2]  M. Lieber,et al.  Dynamics of the Artemis and DNA-PKcs Complex in the Repair of Double-Strand Breaks. , 2022, Journal of molecular biology.

[3]  M. Lieber,et al.  Structural analysis of the basal state of the Artemis:DNA-PKcs complex , 2022, Nucleic acids research.

[4]  N. Mahlaoui,et al.  Inborn errors of immunity caused by defects in the DNA damage response pathways: Importance of minimizing treatment‐related genotoxicity , 2022, Pediatric allergy and immunology : official publication of the European Society of Pediatric Allergy and Immunology.

[5]  Y. Matsumoto Development and Evolution of DNA-Dependent Protein Kinase Inhibitors toward Cancer Therapy , 2022, International journal of molecular sciences.

[6]  A. Shibata,et al.  Roles for 53BP1 in the repair of radiation-induced DNA double strand breaks. , 2020, DNA repair.

[7]  T. Graeber,et al.  Integrin α6 mediates drug resistance of acute lymphoblastic B-cell leukemia. , 2020, Blood.

[8]  M. Bétermier,et al.  Coupling DNA Damage and Repair: an Essential Safeguard during Programmed DNA Double-Strand Breaks? , 2019, Trends in cell biology.

[9]  M. Lieber,et al.  Effects of DNA end configuration on XRCC4-DNA ligase IV and its stimulation of Artemis activity , 2017, The Journal of Biological Chemistry.

[10]  M. van der Burg,et al.  Functional analysis of naturally occurring DCLRE1C mutations and correlation with the clinical phenotype of ARTEMIS deficiency. , 2015, The Journal of allergy and clinical immunology.

[11]  J. de Villartay Congenital defects in V(D)J recombination. , 2015, British medical bulletin.

[12]  M. Stratton,et al.  RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia , 2014, Nature Genetics.

[13]  R. Legerski,et al.  Artemis interacts with the Cul4A-DDB1DDB2 ubiquitin E3 ligase and regulates degradation of the CDK inhibitor p27 , 2011, Cell cycle.

[14]  D. Schatz,et al.  V(D)J recombination: mechanisms of initiation. , 2011, Annual review of genetics.

[15]  M. Lieber,et al.  Mechanistic Basis for RAG Discrimination between Recombination Sites and the Off-Target Sites of Human Lymphomas , 2011, Molecular and Cellular Biology.

[16]  N. Adachi,et al.  Functions and regulation of Artemis: a goddess in the maintenance of genome integrity. , 2010, Journal of radiation research.

[17]  M. Lieber,et al.  The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. , 2010, Annual review of biochemistry.

[18]  J. Downing,et al.  Gene expression classifiers for relapse-free survival and minimal residual disease improve risk classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia. , 2010, Blood.

[19]  R. Legerski,et al.  Artemis Regulates Cell Cycle Recovery from the S Phase Checkpoint by Promoting Degradation of Cyclin E* , 2009, The Journal of Biological Chemistry.

[20]  K. Schwarz,et al.  A Biochemically Defined System for Coding Joint Formation in V(D)J Recombination , 2008, Molecular cell.

[21]  Yunmei Ma,et al.  A biochemically defined system for mammalian nonhomologous DNA end joining. , 2004, Molecular cell.

[22]  Yunmei Ma,et al.  Hairpin Opening and Overhang Processing by an Artemis/DNA-Dependent Protein Kinase Complex in Nonhomologous End Joining and V(D)J Recombination , 2002, Cell.

[23]  A. Fischer,et al.  Artemis, a Novel DNA Double-Strand Break Repair/V(D)J Recombination Protein, Is Mutated in Human Severe Combined Immune Deficiency , 2001, Cell.

[24]  F. Sigaux,et al.  Expression of human recombination activating genes (RAG1 and RAG2) in neoplastic lymphoid cells: correlation with cell differentiation and antigen receptor expression. , 1991, Blood.