33
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Polθ inhibitors elicit BRCA-gene synthetic lethality and target PARP inhibitor resistance

      research-article
      1 , 2 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 3 , 1 , 2 , 1 , 2 , 1 , 2 , 1 , 2 , 4 , 2 , 2 , 2 , 4 , 2 , 5 , 6 , 7 , 8 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 9 , 5 , 7 , 2 , 4 , 3 , 6 , 8 , 1 , 2 , , 3 , , 1 , 2 ,
      Nature Communications
      Nature Publishing Group UK
      Cancer, Drug discovery, Biomarkers

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          To identify approaches to target DNA repair vulnerabilities in cancer, we discovered nanomolar potent, selective, low molecular weight (MW), allosteric inhibitors of the polymerase function of DNA polymerase Polθ, including ART558. ART558 inhibits the major Polθ-mediated DNA repair process, Theta-Mediated End Joining, without targeting Non-Homologous End Joining. In addition, ART558 elicits DNA damage and synthetic lethality in BRCA1- or BRCA2-mutant tumour cells and enhances the effects of a PARP inhibitor. Genetic perturbation screening revealed that defects in the 53BP1/Shieldin complex, which cause PARP inhibitor resistance, result in in vitro and in vivo sensitivity to small molecule Polθ polymerase inhibitors. Mechanistically, ART558 increases biomarkers of single-stranded DNA and synthetic lethality in 53BP1-defective cells whilst the inhibition of DNA nucleases that promote end-resection reversed these effects, implicating these in the synthetic lethal mechanism-of-action. Taken together, these observations describe a drug class that elicits BRCA-gene synthetic lethality and PARP inhibitor synergy, as well as targeting a biomarker-defined mechanism of PARPi-resistance.

          Abstract

          Polθ has been recently identified as a therapeutic target in cancer but specific inhibitors are currently unavailable. Here, the authors identify small molecule inhibitors of Polθ’s polymerase activity which elicit BRCA1/2 synthetic lethality, enhance the effect of PARP inhibitors and target PARP inhibitor resistance caused by 53BP1/Shieldin pathway defects.

          Related collections

          Most cited references67

          • Record: found
          • Abstract: found
          • Article: not found

          Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy.

          BRCA1 and BRCA2 are important for DNA double-strand break repair by homologous recombination, and mutations in these genes predispose to breast and other cancers. Poly(ADP-ribose) polymerase (PARP) is an enzyme involved in base excision repair, a key pathway in the repair of DNA single-strand breaks. We show here that BRCA1 or BRCA2 dysfunction unexpectedly and profoundly sensitizes cells to the inhibition of PARP enzymatic activity, resulting in chromosomal instability, cell cycle arrest and subsequent apoptosis. This seems to be because the inhibition of PARP leads to the persistence of DNA lesions normally repaired by homologous recombination. These results illustrate how different pathways cooperate to repair damage, and suggest that the targeted inhibition of particular DNA repair pathways may allow the design of specific and less toxic therapies for cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase.

            Poly(ADP-ribose) polymerase (PARP1) facilitates DNA repair by binding to DNA breaks and attracting DNA repair proteins to the site of damage. Nevertheless, PARP1-/- mice are viable, fertile and do not develop early onset tumours. Here, we show that PARP inhibitors trigger gamma-H2AX and RAD51 foci formation. We propose that, in the absence of PARP1, spontaneous single-strand breaks collapse replication forks and trigger homologous recombination for repair. Furthermore, we show that BRCA2-deficient cells, as a result of their deficiency in homologous recombination, are acutely sensitive to PARP inhibitors, presumably because resultant collapsed replication forks are no longer repaired. Thus, PARP1 activity is essential in homologous recombination-deficient BRCA2 mutant cells. We exploit this requirement in order to kill BRCA2-deficient tumours by PARP inhibition alone. Treatment with PARP inhibitors is likely to be highly tumour specific, because only the tumours (which are BRCA2-/-) in BRCA2+/- patients are defective in homologous recombination. The use of an inhibitor of a DNA repair enzyme alone to selectively kill a tumour, in the absence of an exogenous DNA-damaging agent, represents a new concept in cancer treatment.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              PARP inhibitors: Synthetic lethality in the clinic.

              PARP inhibitors (PARPi), a cancer therapy targeting poly(ADP-ribose) polymerase, are the first clinically approved drugs designed to exploit synthetic lethality, a genetic concept proposed nearly a century ago. Tumors arising in patients who carry germline mutations in either BRCA1 or BRCA2 are sensitive to PARPi because they have a specific type of DNA repair defect. PARPi also show promising activity in more common cancers that share this repair defect. However, as with other targeted therapies, resistance to PARPi arises in advanced disease. In addition, determining the optimal use of PARPi within drug combination approaches has been challenging. Nevertheless, the preclinical discovery of PARPi synthetic lethality and the route to clinical approval provide interesting lessons for the development of other therapies. Here, we discuss current knowledge of PARP inhibitors and potential ways to maximize their clinical effectiveness.
                Bookmark

                Author and article information

                Contributors
                Stephen.Pettitt@icr.ac.uk
                Graeme.Smith@artiospharma.com
                Chris.Lord@icr.ac.uk
                Journal
                Nat Commun
                Nat Commun
                Nature Communications
                Nature Publishing Group UK (London )
                2041-1723
                17 June 2021
                17 June 2021
                2021
                : 12
                : 3636
                Affiliations
                [1 ]GRID grid.18886.3f, ISNI 0000 0001 1271 4623, CRUK Gene Function Laboratory, , The Institute of Cancer Research, ; London, UK
                [2 ]GRID grid.18886.3f, ISNI 0000 0001 1271 4623, The Breast Cancer Now Toby Robins Research Centre, , The Institute of Cancer Research, ; London, UK
                [3 ]GRID grid.418195.0, ISNI 0000 0001 0694 2777, Artios Pharma, , The Glenn Berge Building, Babraham Research Campus, ; Cambridge, UK
                [4 ]GRID grid.13097.3c, ISNI 0000 0001 2322 6764, The Breast Cancer Now Research Unit, King’s College London, ; London, UK
                [5 ]GRID grid.48336.3a, ISNI 0000 0004 1936 8075, Laboratory of Genome Integrity, , National Cancer Institute, NIH, ; Bethesda, MD USA
                [6 ]GRID grid.451388.3, ISNI 0000 0004 1795 1830, The Francis Crick Institute, ; London, UK
                [7 ]GRID grid.10419.3d, ISNI 0000000089452978, Department of Human Genetics, , Leiden University Medical Center, ; Leiden, The Netherlands
                [8 ]GRID grid.4991.5, ISNI 0000 0004 1936 8948, Medical Research Council Oxford Institute for Radiation Oncology, , University of Oxford, Old Road Campus Research Building, Roosevelt Drive, ; Oxford, UK
                [9 ]GRID grid.418195.0, ISNI 0000 0001 0694 2777, Cancer Research UK, , Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Research Campus, ; Cambridge, UK
                Author information
                http://orcid.org/0000-0001-6770-9274
                http://orcid.org/0000-0003-3316-205X
                http://orcid.org/0000-0002-5301-054X
                http://orcid.org/0000-0001-6685-5480
                http://orcid.org/0000-0001-6225-4047
                http://orcid.org/0000-0003-0070-3403
                http://orcid.org/0000-0001-6942-6067
                http://orcid.org/0000-0001-8465-9002
                http://orcid.org/0000-0001-6936-6834
                http://orcid.org/0000-0003-3313-3857
                http://orcid.org/0000-0001-8320-4057
                http://orcid.org/0000-0002-3226-0515
                Article
                23463
                10.1038/s41467-021-23463-8
                8211653
                34140467
                e1a37386-cfad-4e59-9a79-86942b6cd162
                © The Author(s) 2021

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 17 March 2021
                : 30 April 2021
                Funding
                Funded by: FundRef https://doi.org/10.13039/100009794, Breast Cancer Now (BCN);
                Award ID: CTR-Q5-Y1
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/501100010757, National Centre for the Replacement Refinement and Reduction of Animals in Research (NC3Rs);
                Award ID: NC/P001262/1
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/501100000289, Cancer Research UK (CRUK);
                Award ID: C30061/A24439
                Award Recipient :
                Categories
                Article
                Custom metadata
                © The Author(s) 2021

                Uncategorized
                cancer,drug discovery,biomarkers
                Uncategorized
                cancer, drug discovery, biomarkers

                Comments

                Comment on this article