3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Exploiting DNA repair defects in triple negative breast cancer to improve cell killing

      research-article

      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

          Background:

          The lack of molecular targets for triple negative breast cancer (TNBC) has limited treatment options and reduced survivorship. Identifying new molecular targets may help improve patient survival and decrease recurrence and metastasis. As DNA repair defects are prevalent in breast cancer, we evaluated the expression and repair capacities of DNA repair proteins in preclinical models.

          Methods:

          DNA repair capacity was analyzed in four TNBC cell lines, MDA-MB-157 (MDA-157), MDA-MB-231 (MDA-231), MDA-MB-468 (MDA-468), and HCC1806, using fluorescence multiplex host cell reactivation (FM-HCR) assays. Expression of DNA repair genes was analyzed with RNA-seq, and protein expression was evaluated with immunoblot. Responses to the combination of DNA damage response inhibitors and primary chemotherapy drugs doxorubicin or carboplatin were evaluated in the cell lines.

          Results:

          Defects in base excision and nucleotide excision repair were observed in preclinical TNBC models. Gene expression analysis showed a limited correlation between these defects. Loss in protein expression was a better indicator of these DNA repair defects. Over-expression of PARP1, XRCC1, RPA, DDB1, and ERCC1 was observed in TNBC preclinical models, and likely contributed to altered sensitivity to chemotherapy and DNA damage response (DDR) inhibitors. Improved cell killing was achieved when primary therapy was combined with DDR inhibitors for ATM, ATR, or CHK1.

          Conclusion:

          Base excision and nucleotide excision repair pathways may offer new molecular targets for TNBC. The functional status of DNA repair pathways should be considered when evaluating new therapies and may improve the targeting for primary and combination therapies with DDR inhibitors.

          Related collections

          Most cited references54

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          A molecular portrait of microsatellite instability across multiple cancers

          Microsatellite instability (MSI) refers to the hypermutability of short repetitive sequences in the genome caused by impaired DNA mismatch repair. Although MSI has been studied for decades, large amounts of sequencing data now available allows us to examine the molecular fingerprints of MSI in greater detail. Here, we analyse ∼8,000 exomes and ∼1,000 whole genomes of cancer patients across 23 cancer types. Our analysis reveals that the frequency of MSI events is highly variable within and across tumour types. We also identify genes in DNA repair and oncogenic pathways recurrently subject to MSI and uncover non-coding loci that frequently display MSI. Finally, we propose a highly accurate exome-based predictive model for the MSI phenotype. These results advance our understanding of the genomic drivers and consequences of MSI, and our comprehensive catalogue of tumour-type-specific MSI loci will enable panel-based MSI testing to identify patients who are likely to benefit from immunotherapy.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer.

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

              Mechanisms of ATM Activation.

              The ataxia-telangiectasia mutated (ATM) protein kinase is a master regulator of the DNA damage response, and it coordinates checkpoint activation, DNA repair, and metabolic changes in eukaryotic cells in response to DNA double-strand breaks and oxidative stress. Loss of ATM activity in humans results in the pleiotropic neurodegeneration disorder ataxia-telangiectasia. ATM exists in an inactive state in resting cells but can be activated by the Mre11-Rad50-Nbs1 (MRN) complex and other factors at sites of DNA breaks. In addition, oxidation of ATM activates the kinase independently of the MRN complex. This review discusses these mechanisms of activation, as well as the posttranslational modifications that affect this process and the cellular factors that affect the efficiency and specificity of ATM activation and substrate phosphorylation. I highlight functional similarities between the activation mechanisms of ATM, phosphatidylinositol 3-kinases (PI3Ks), and the other PI3K-like kinases, as well as recent structural insights into their regulation.
                Bookmark

                Author and article information

                Contributors
                Journal
                Ther Adv Med Oncol
                Ther Adv Med Oncol
                TAM
                sptam
                Therapeutic Advances in Medical Oncology
                SAGE Publications (Sage UK: London, England )
                1758-8340
                1758-8359
                18 September 2020
                2020
                : 12
                : 1758835920958354
                Affiliations
                [1-1758835920958354]College of Medicine, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
                [2-1758835920958354]College of Medicine, University of South Alabama, Mobile, AL, USA
                [3-1758835920958354]College of Medicine, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
                [4-1758835920958354]Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
                [5-1758835920958354]Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
                [6-1758835920958354]Mitchell Cancer Institute, University of South Alabama, 1660 Springhill Avenue, Mobile, AL 36607, USA
                [7-1758835920958354]University of South Alabama College of Medicine, 307 N. University Blvd, Mobile, AL 36688, USA
                Author notes
                Article
                10.1177_1758835920958354
                10.1177/1758835920958354
                7502856
                32994807
                590b6dac-e90d-474c-a235-4550e5278500
                © The Author(s), 2020

                This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License ( https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages ( https://us.sagepub.com/en-us/nam/open-access-at-sage).

                History
                : 20 April 2020
                : 19 August 2020
                Funding
                Funded by: breast cancer research foundation of alabama, FundRef https://doi.org/10.13039/100016419;
                Award ID: Innovation Award
                Funded by: National Cancer Institute, FundRef https://doi.org/10.13039/100000054;
                Award ID: P01CA092584
                Categories
                Original Research
                Custom metadata
                January-December 2020
                ts1

                chemotherapy,dna damage,homologous recombination,nonhomologous end joining,nucleotide excision repair,small molecule inhibitor

                Comments

                Comment on this article