ATM in DNA repair in cancer

https://doi.org/10.1016/j.pharmthera.2019.07.002Get rights and content

Abstract

Alterations in DNA damage response (DDR) pathways are hallmarks of cancer. Incorrect repair of DNA lesions often leads to genomic instability. Ataxia telangiectasia mutated (ATM), a core component of the DNA repair system, is activated to enhance the homologous recombination (HR) repair pathway upon DNA double-strand breaks. Although ATM signaling has been widely studied in different types of cancer, its research is still lacking compared with other DDR-involved molecules such as PARP and ATR. There is still a vast research opportunity for the development of ATM inhibitors as anticancer agents. Here, we focus on the recent findings of ATM signaling in DNA repair of cancer. Previous studies have identified several partners of ATM, some of which promote ATM signaling, while others have the opposite effect. ATM inhibitors, including KU-55933, KU-60019, KU-59403, CP-466722, AZ31, AZ32, AZD0156, and AZD1390, have been evaluated for their antitumor effects. It has been revealed that ATM inhibition increases a cancer cell's sensitivity to radiotherapy. Moreover, the combination with PARP or ATR inhibitors has synergistic lethality in some cancers. Of note, among these ATM inhibitors, AZD0156 and AZD1390 achieve potent and highly selective ATM kinase inhibition and have an excellent ability to penetrate the blood-brain barrier. Currently, AZD0156 and AZD1390 are under investigation in phase I clinical trials. Taken together, targeting ATM may be a promising strategy for cancer treatment. Hence, further development of ATM inhibitors is urgently needed in cancer research.

Section snippets

Introduction: an overview of the DNA repair signaling network

Numerous DNA damage events occur each day in the human body by exposure to various conditions (Jackson & Bartek, 2009). These DNA lesions include simple base modifications, base mismatches, bulky DNA adducts, inter-strand and intra-strand crosslinks, protein-DNA crosslinks, DNA single-strand break (SSB) and double-strand break (DSB) (Roos, Thomas, & Kaina, 2016). When normal cells are subjected to stress that induces DNA damage, they can repair the damage through intact DNA repair pathways

ATM signaling pathways in DNA repair

Ataxia telangiectasia is an inherited recessive disease. Patients with ataxia telangiectasia exhibit multiple clinical manifestations such as cerebellar degeneration, immunodeficiency, susceptibility to malignancies, increased radiosensitivity, and metabolic diseases (Amirifar, Ranjouri, Yazdani, Abolhassani, & Aghamohammadi, 2019). Ataxia telangiectasia mutated, named ATM, belongs to the phosphatidylinositol 3-kinase-related kinase family. It has multiple functions in cancer development such

ATM alterations in cancers

Cytotoxic chemotherapeutic agents induce DNA damage in cancer cells that engage numerous factors to repair the lesions for survival (Hosoya & Miyagawa, 2014; Matt & Hofmann, 2016). Activation of ATM signaling is one of the barriers to chemotherapeutic activity and radiation resistance (Matt & Hofmann, 2016). Currently, several transcription factors have been identified to facilitate ATM activation in response to chemotherapeutic agents (Palmieri et al., 2011). To improve a patients' response to

ATM inhibitors

The antitumor effects of various kinds of ATM inhibitors have been studied in both human cancer cells and mouse models (Weber & Ryan, 2015). In this review, we outline the recent research advances in the effects of these ATM inhibitors on DNA repair pathways, including KU-55933, KU-60019, KU-59403, CP-466722, AZ31, AZ32, AZD0156, and AZD1390 (Table 1).

Clinical trials of ATM inhibitors

Among the diverse DDR-targeting agents, research on PARP inhibitors is the most advanced (Pilie et al., 2019; Sonnenblick, de Azambuja, Azim Jr., & Piccart, 2015). Olaparib and rucaparib are approved by the Food and Drug Administration for the treatment of advanced stage ovarian and breast cancers with BRCA1/2 mutation after chemotherapy (Pilie et al., 2019). In addition, there are hundreds of ongoing clinical trials evaluating PARP inhibitors.

Compared with PARP inhibitors, clinical development

Conclusions and future directions

In whole DDR networks, ATM not only plays a critical role in DNA damage repair, but also controls cell cycle progression, apoptosis, chromatin remodeling, and transcriptional regulation (O'Connor, 2015; Weber & Ryan, 2015). Because PARP inhibitors have been approved for patients with breast and ovarian cancers, hundreds of preclinical and clinical trials are being performed to evaluate DDR-targeted agents for various cancer types. Several ATM cofactors have been identified, including the MRN

Declarations of Competing Interest

D-Y Oh is a consultant and advisory board member of AstraZeneca, Novartis, Genentech, Roche, Merck Serono, Bayer, Taiho, ASLAN, Halozyme, and Zymework. D-Y Oh has received research grants from AstraZeneca, Novartis, Array, Eli Lilly, and Green Cross. There is no conflict of interest to declare by MH Jin.

Acknowledgments

This study was supported by a grant from The SNUH Research Fund (03-2017-0100).

References (118)

  • N.S. Gavande et al.

    DNA repair targeted therapy: The past or future of cancer treatment?

    Pharmacology & Therapeutics

    (2016)
  • A. Guleria et al.

    ATM kinase: Much more than a DNA damage responsive protein

    DNA Repair (Amst)

    (2016)
  • T.E. Johnson et al.

    Homeodomain proteins directly regulate ATM kinase activity

    Cell Reports

    (2018)
  • H. Katoh et al.

    CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis

    Cancer Cell

    (2013)
  • A. Konishi et al.

    Involvement of histone H1.2 in apoptosis induced by DNA double-strand breaks

    Cell

    (2003)
  • S.V. Kozlov et al.

    Autophosphorylation and ATM activation: Additional sites add to the complexity

    The Journal of Biological Chemistry

    (2011)
  • K. Li et al.

    ATM inhibition induces synthetic lethality and enhances sensitivity of PTEN-deficient breast cancer cells to cisplatin

    Experimental Cell Research

    (2018)
  • R. Liu et al.

    ASCIZ/ATMIN is dispensable for ATM signaling in response to replication stress

    DNA Repair (Amst)

    (2017)
  • R. Liu et al.

    The depletion of ATM inhibits colon cancer proliferation and migration via B56gamma2-mediated Chk1/p53/CD44 cascades

    Cancer Letters

    (2017)
  • A. Malkova et al.

    Break-induced replication: Functions and molecular mechanism

    Current Opinion in Genetics & Development

    (2013)
  • M.J. O'Connor

    Targeting the DNA damage response in cancer

    Molecular Cell

    (2015)
  • P. Ruff et al.

    RPA stabilization of single-stranded DNA is critical for break-induced replication

    Cell Reports

    (2016)
  • P. Samadder et al.

    Cancer TARGETases: DSB repair as a pharmacological target

    Pharmacology & Therapeutics

    (2016)
  • L. Schmidt et al.

    ATMIN is required for the ATM-mediated signaling and recruitment of 53BP1 to DNA damage sites upon replication stress

    DNA Repair (Amst)

    (2014)
  • P. Amirifar et al.

    Ataxia-telangiectasia: A review of clinical features and molecular pathology

    Pediatric Allergy and Immunology

    (2019)
  • H.K. Angell et al.

    PD-L1 and immune infiltrates are differentially expressed in distinct subgroups of gastric cancer

    Oncoimmunology

    (2019)
  • P. Awasthi et al.

    ATM and ATR signaling at a glance

    Journal of Cell Science

    (2015)
  • M. Ayars et al.

    Susceptibility of ATM-deficient pancreatic cancer cells to radiation

    Cell Cycle

    (2017)
  • D. Baretic et al.

    Structures of closed and open conformations of dimeric human ATM

    Science Advances

    (2017)
  • S.P. Barry et al.

    STAT3 modulates the DNA damage response pathway

    International Journal of Experimental Pathology

    (2010)
  • M.A. Batey et al.

    Preclinical evaluation of a novel ATM inhibitor, KU59403, in vitro and in vivo in p53 functional and dysfunctional models of human cancer

    Molecular Cancer Therapeutics

    (2013)
  • J.J. Bednarski et al.

    At the intersection of DNA damage and immune responses

    Nature Reviews. Immunology

    (2019)
  • N. Begam et al.

    Promoter Hypermethylation of the ATM gene as a novel biomarker for breast cancer

    Asian Pacific Journal of Cancer Prevention

    (2017)
  • K.P. Bhat et al.

    RPA and RAD51: Fork reversal, fork protection, and genome stability

    Nature Structural & Molecular Biology

    (2018)
  • L. Biddlestone-Thorpe et al.

    ATM kinase inhibition preferentially sensitizes p53-mutant glioma to ionizing radiation

    Clinical Cancer Research

    (2013)
  • D. Branzei et al.

    Regulation of DNA repair throughout the cell cycle

    Nature Reviews Molecular Cell Biology

    (2008)
  • S. Buglioni et al.

    The clinical significance of PD-L1 in advanced gastric cancer is dependent on ARID1A mutations and ATM expression

    Oncoimmunology

    (2018)
  • H.H.Y. Chang et al.

    Non-homologous DNA end joining and alternative pathways to double-strand break repair

    Nature Reviews Molecular Cell Biology

    (2017)
  • C.C. Chen et al.

    Homology-directed repair and the role of BRCA1, BRCA2, and related proteins in genome integrity and Cancer

    Annual Review Cancer Biology

    (2018)
  • H. Chen et al.

    INPP4B overexpression suppresses migration, invasion and angiogenesis of human prostate cancer cells

    Clinical and Experimental Pharmacology & Physiology

    (2017)
  • W.T. Chen et al.

    ATM regulation of IL-8 links oxidative stress to cancer cell migration and invasion

    Elife

    (2015)
  • M. Choi et al.

    ATM mutations in Cancer: Therapeutic implications

    Molecular Cancer Therapeutics

    (2016)
  • C.A. Cremona et al.

    ATM signalling and cancer

    Oncogene

    (2013)
  • N.J. Curtin

    DNA repair dysregulation from cancer driver to therapeutic target

    Nature Reviews Cancer

    (2012)
  • S.T. Durant et al.

    The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models

    Science Advances

    (2018)
  • M.S. Gilardini Montani et al.

    ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition

    Journal of Experimental & Clinical Cancer Research

    (2013)
  • S.E. Golding et al.

    Improved ATM kinase inhibitor KU-60019 radiosensitizes glioma cells, compromises insulin, AKT and ERK prosurvival signaling, and inhibits migration and invasion

    Molecular Cancer Therapeutics

    (2009)
  • S.T. Guo et al.

    INPP4B is an oncogenic regulator in human colon cancer

    Oncogene

    (2016)
  • G.H. Ha et al.

    Pellino1 regulates reversible ATM activation via NBS1 ubiquitination at DNA double-strand breaks

    Nature Communications

    (2019)
  • T. Helleday et al.

    DNA repair pathways as targets for cancer therapy

    Nature Reviews Cancer

    (2008)
  • Cited by (138)

    View all citing articles on Scopus
    View full text