Oncotarget

Research Papers:

Osteosarcoma cells with genetic signatures of BRCAness are susceptible to the PARP inhibitor talazoparib alone or in combination with chemotherapeutics

Florian Engert, Michal Kovac, Daniel Baumhoer, Michaela Nathrath and Simone Fulda _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:48794-48806. https://doi.org/10.18632/oncotarget.10720

Metrics: PDF 4811 views  |   HTML 5175 views  |   ?  


Abstract

Florian Engert1,2,3, Michal Kovac4, Daniel Baumhoer4, Michaela Nathrath5,6,7, Simone Fulda1,2,3

1Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany

2German Cancer Consortium (DKTK), Heidelberg, Germany

3German Cancer Research Center (DKFZ), Heidelberg, Germany

4Bone Tumour Reference Centre at the Institute of Pathology, University Hospital Basel, Basel, Switzerland

5Institute of Radiation Biology, Clinical Cooperation Group Osteosarcoma, Helmholtz Zentrum München, Neuherberg, Germany

6Pediatric Oncology Center, Department of Pediatrics, Technische Universität München and Comprehensive Cancer Center, Munich, Germany

7Department of Pediatric Hematology and Oncology, Klinikum Kassel, Kassel, Germany

Correspondence to:

Simone Fulda, email: [email protected]

Keywords: PARP1, PARP inhibitor, apoptosis, cell death, osteosarcoma

Received: March 23, 2016     Accepted: May 13, 2016     Published: July 20, 2016

ABSTRACT

We recently discovered mutation signatures reminiscent of BRCA deficiency in the vast majority of a set of primary osteosarcomas (OS). In the current study, we therefore investigated the sensitivity of a panel of OS cell lines to the poly(ADP)-ribose polymerase (PARP) inhibitor talazoparib alone and in combination with several chemotherapeutic drugs (i.e. temozolomide (TMZ), SN-38, doxorubicin, cisplatin, methotrexate (MTX), etoposide/carboplatin). Here, we identified an association between homologous recombination (HR) repair deficiency and the response of OS cell lines to talazoparib. All OS cell lines with molecular features characteristic of BRCA1/2 mutant tumors (so-called “BRCAness”), such as disruptive gains in PTEN or FANCD2 and/or losses of ATM, BAP1, BARD1 or CHEK2, were susceptible to talazoparib-induced reduction of cell viability (i.e. MG63, ZK-58,, SaOS-2 and MNNG-HOS). Consistent with their high sensitivity to talazoparib, MG63 and ZK-58 cells scored positive in a DNA-based measure of genomic instability (i.e. homologous recombination deficiency (HRD)-loss of heterozygosity (LOH) score). In contrast, U2OS cells that carry a heterozygous BRCA2 mutation and therefore most likely have one intact BRCA2 allele left proved to be resistant to talazoparib. Furthermore, we identified TMZ as the most potent chemotherapeutic drug together with talazoparib to synergistically reduce cell viability, as confirmed by calculation of combination index (CI) values, and to suppress long-term clonogenic survival. Mechanistically, talazoparib and TMZ cooperated to induce apoptotic cell death, as demonstrated by activation of BAX and BAK, loss of mitochondrial membrane potential (MMP), caspase activation, DNA fragmentation and caspase-dependent cell death. Genetic silencing of BAX and BAK or pharmacological inhibition of caspases by zVAD.fmk significantly rescued OS cells from talazoparib/TMZ-induced apoptosis. These findings have important implications for the development of novel treatment strategies using PARP inhibitors alone or together with chemotherapy in a subset of OS with features of BRCAness.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 10720