DNA damage response and repair genes in advanced bone and soft tissue sarcomas: An 8-gene signature as a candidate predictive biomarker of response to trabectedin and olaparib combination

Alessandra Merlini, Maria Laura Centomo, Giulio Ferrero, Giulia Chiabotto, Umberto Miglio, Enrico Berrino, Giorgia Giordano, Silvia Brusco, Alberto Pisacane, Elena Maldi, Ivana Sarotto, Federica Capozzi, Cristina Lano, Claudio Isella, Giovanni Crisafulli, Massimo Aglietta, Angelo Paolo Dei Tos, Marta Sbaraglia, Dario Sangiolo, Lorenzo D'Ambrosio, Alberto Bardelli, Ymera Pignochino, Giovanni Grignani, Alessandra Merlini, Maria Laura Centomo, Giulio Ferrero, Giulia Chiabotto, Umberto Miglio, Enrico Berrino, Giorgia Giordano, Silvia Brusco, Alberto Pisacane, Elena Maldi, Ivana Sarotto, Federica Capozzi, Cristina Lano, Claudio Isella, Giovanni Crisafulli, Massimo Aglietta, Angelo Paolo Dei Tos, Marta Sbaraglia, Dario Sangiolo, Lorenzo D'Ambrosio, Alberto Bardelli, Ymera Pignochino, Giovanni Grignani

Abstract

Background: Advanced and unresectable bone and soft tissue sarcomas (BSTS) still represent an unmet medical need. We demonstrated that the alkylating agent trabectedin and the PARP1-inhibitor olaparib display antitumor activity in BSTS preclinical models. Moreover, in a phase Ib clinical trial (NCT02398058), feasibility, tolerability and encouraging results have been observed and the treatment combination is currently under study in a phase II trial (NCT03838744).

Methods: Differential expression of genes involved in DNA Damage Response and Repair was evaluated by Nanostring® technology, extracting RNA from pre-treatment tumor samples of 16 responder (≥6-month progression free survival) and 16 non-responder patients. Data validation was performed by quantitative real-time PCR, RNA in situ hybridization, and immunohistochemistry. The correlation between the identified candidate genes and both progression-free survival and overall survival was investigated in the publicly available dataset "Sarcoma (TCGA, The Cancer Genome Atlas)".

Results: Differential RNA expression analysis revealed an 8-gene signature (CDKN2A, PIK3R1, SLFN11, ATM, APEX2, BLM, XRCC2, MAD2L2) defining patients with better outcome upon trabectedin+olaparib treatment. In responder vs. non-responder patients, a significant differential expression of these genes was further confirmed by RNA in situ hybridization and by qRT-PCR and immunohistochemistry in selected experiments. Correlation between survival outcomes and genetic alterations in the identified genes was shown in the TCGA sarcoma dataset.

Conclusions: This work identified an 8-gene expression signature to improve prediction of response to trabectedin+olaparib combination in BSTS. The predictive role of these potential biomarkers warrants further investigation.

Keywords: DNA damage response and repair genes; bone and soft tissue sarcomas; olaparib; predictive biomarkers; trabectedin.

Conflict of interest statement

GGr has received fees for consulting/advisory roles from PharmaMar, Lilly, Novartis, Bayer, and Eisai. LDA received travel grant from PharmaMar and Lilly. MA has received fees for consulting/advisory roles from Bristol- Myers Squibb, Merck, and Roche. AB served in a consulting/advisory role for Illumina and Inivata. AB is cofounder and shareholder of NeoPhore. AB is a member of the NeoPhore scientific advisory board. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Merlini, Centomo, Ferrero, Chiabotto, Miglio, Berrino, Giordano, Brusco, Pisacane, Maldi, Sarotto, Capozzi, Lano, Isella, Crisafulli, Aglietta, Dei Tos, Sbaraglia, Sangiolo, D’Ambrosio, Bardelli, Pignochino and Grignani.

Figures

Figure 1
Figure 1
(A) Volcano plot showing differential expression of DDRR genes in responder vs. non-responder patients. (B) Boxplot showing differential expression of DDRR genes in responder vs. non-responder patients, with normalized expression. P-value by Wilcoxon Rank-Sum test. *p<0.05; **p<0.01.
Figure 2
Figure 2
RNA ISH of selected genes in responder vs. non-responder patients. (A) Higher expressed genes in responder patients (B) higher expressed genes in non-responders (C). Heatmap showing differential RNA ISH staining between responder and non-responder patients. ISH score was assigned by an expert pathologist on the basis of staining intensity and percentage of positive cells. P-value was calculated by Chi-square test. *p<0.05; **p<0.01.
Figure 3
Figure 3
Validation Assays (A) Expression levels of representative genes (CDKN2A, left; and APEX2, right) among responder and non-responder patients. Statistically significant differential expression was shown between the two groups (Wilcoxon rank-sum test). (B) Representative IHC staining of CDKN2A/p16 in tumor samples from responder and non-responder patients. (C) Box plot distribution of CDKN2a/p16 expression level (percentage of IHC positive cells) in responders and non-responders patients.
Figure 4
Figure 4
Oncoprint and heatmap of candidate biomarkers in the TCGA sarcoma cohort.
Figure 5
Figure 5
Kaplan-Meier curves showing Overall Sirvival in the sarcoma TCGA cohort, according to selected candidate biomarker genes (BLM, A, and MAD2L2, B).

References

    1. Gronchi A, et al. . Soft tissue and visceral sarcomas: ESMO–EURACAN–GENTURIS clinical practice guidelines for diagnosis, treatment and follow-up☆. Ann Oncol (2021) 32(11):1348–65. doi: 10.1016/j.annonc.2021.07.006
    1. Demetri GD, et al. . Efficacy and safety of trabectedin or dacarbazine for metastatic liposarcoma or leiomyosarcoma after failure of conventional chemotherapy: Results of a phase III randomized multicenter clinical trial. J Clin Oncol (2016) 34(8):786–93. doi: 10.1200/JCO.2015.62.4734
    1. Schöffski P, et al. . Eribulin versus dacarbazine in previously treated patients with advanced liposarcoma or leiomyosarcoma: A randomised, open-label, multicentre, phase 3 trial. Lancet (2016) 387(10028):1629–37. doi: 10.1016/S0140-6736(15)01283-0
    1. Judson I, et al. . Randomised phase II trial of pegylated liposomal doxorubicin (DOXIL/CAELYX) versus doxorubicin in the treatment of advanced or metastatic soft tissue sarcoma: a study by the EORTC soft tissue and bone sarcoma group. Eur J Cancer (2001) 37(7):870–7. doi: 10.1016/S0959-8049(01)00050-8
    1. van der Graaf WT, et al. . Pazopanib for metastatic soft-tissue sarcoma (PALETTE): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet (2012) 379(9829):1879–86. doi: 10.1016/S0140-6736(12)60651-5
    1. Farag S, Smith MJ, Fotiadis N, Constantinidou A, Jones RL. Revolutions in treatment options in gastrointestinal stromal tumours (GISTs): The latest updates: Revolutions in treatment options in GIST. Curr Treat Options Oncol (2020) 21:1–11. doi: 10.1007/s11864-020-00754-8
    1. Hao Z, Wang P. Lenvatinib in management of solid tumors. Oncologist (2020) 25(2):e302–10. doi: 10.1634/theoncologist.2019-0407
    1. D'Ambrosio L, et al. . Doxorubicin plus dacarbazine, doxorubicin plus ifosfamide, or doxorubicin alone as a first-line treatment for advanced leiomyosarcoma: A propensity score matching analysis from the European organization for research and treatment of cancer soft tissue and bone sarcoma group. Cancer (2020) 126(11):2637–47. doi: 10.1002/cncr.32795
    1. Pautier P, et al. . Trabectedin in combination with doxorubicin for first-line treatment of advanced uterine or soft-tissue leiomyosarcoma (LMS-02): a non-randomised, multicentre, phase 2 trial. Lancet Oncol (2015) 16(4):457–64. doi: 10.1016/S1470-2045(15)70070-7
    1. Müller D, et al. . 1669TiP a non-randomized, open-label phase II trial evaluating efficacy and feasibility of combined treatment with trabectedin and nivolumab in patients with metastatic or inoperable soft tissue sarcomas (STS) after failure of an anthracycline-containing regimen. Ann Oncol (2020) 31:S991.doi: 10.1016/annonc/annonc288
    1. Chae YK, et al. . Path toward precision oncology: Review of targeted therapy studies and tools to aid in defining "Actionability" of a molecular lesion and patient management support. Mol Cancer Ther (2017) 16(12):2645–55. doi: 10.1158/1535-7163.MCT-17-0597
    1. Pignochino Y, et al. . PARP1 expression drives the synergistic antitumor activity of trabectedin and PARP1 inhibitors in sarcoma preclinical models. Mol Cancer (2017) 16(1):1–15. doi: 10.1186/s12943-017-0652-5
    1. Grignani G, et al. . Trabectedin and olaparib in patients with advanced and non-resectable bone and soft-tissue sarcomas (TOMAS): an open-label, phase 1b study from the Italian sarcoma group. Lancet Oncol (2018) 19(10):1360–71. doi: 10.1016/S1470-2045(18)30438-8
    1. Moura DS, et al. . A DNA damage repair gene-associated signature predicts responses of patients with advanced soft-tissue sarcoma to treatment with trabectedin. Mol Oncol (2021) 15(12):3691–705. doi: 10.1002/1878-0261.12996
    1. Italiano A, et al. . ERCC5/XPG, ERCC1, and BRCA1 gene status and clinical benefit of trabectedin in patients with soft tissue sarcoma. Cancer (2011) 117(15):3445–56. doi: 10.1002/cncr.25925
    1. Massuti B, et al. . Trabectedin in patients with advanced non-small-cell lung cancer (NSCLC) with XPG and/or ERCC1 overexpression and BRCA1 underexpression and pretreated with platinum. Lung Cancer (2012) 76(3):354–61. doi: 10.1016/j.lungcan.2011.12.002
    1. Schöffski P, et al. . Predictive impact of DNA repair functionality on clinical outcome of advanced sarcoma patients treated with trabectedin: A retrospective multicentric study. Eur J Cancer (2011) 47(7):1006–12. doi: 10.1016/j.ejca.2011.01.016
    1. Moura DS, et al. . CUL4A, ERCC5, and ERCC1 as predictive factors for trabectedin efficacy in advanced soft tissue sarcomas (STS): A Spanish group for sarcoma research (GEIS) study. Cancers (2020) 12(5):1128. doi: 10.3390/cancers12051128
    1. Pilié PG, Gay CM, Byers LA, O'Connor MJ, Yap TA. PARP inhibitors: extending benefit beyond BRCA-mutant cancers. Clin Cancer Res (2019) 25(13):3759–71. doi: 10.1158/1078-0432.CCR-18-0968
    1. Cardnell RJ, et al. . Proteomic markers of DNA repair and PI3K pathway activation predict response to the PARP inhibitor BMN 673 in small cell lung cancer. Clin Cancer Res (2013) 19(22):6322–8. doi: 10.1158/1078-0432.CCR-13-1975
    1. Michels J, Vitale I, Saparbaev M, Castedo M, Kroemer G. Predictive biomarkers for cancer therapy with PARP inhibitors. Oncogene (2014) 33(30):3894–907. doi: 10.1038/onc.2013.352
    1. Mateo J, et al. . DNA-Repair defects and olaparib in metastatic prostate cancer. N Engl J Med (2015) 373(18):1697–708. doi: 10.1056/NEJMoa1506859
    1. Thomas A, Murai J, Pommier Y. The evolving landscape of predictive biomarkers of response to PARP inhibitors. J Clin Invest (2018) 128(5):1727–30. doi: 10.1172/JCI120388
    1. Miller RE, et al. . ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann Oncol (2020) 31(12):1606–22. doi: 10.1016/j.annonc.2020.08.2102
    1. Larsen AK, Galmarini CM, D'Incalci M. Unique features of trabectedin mechanism of action. Cancer Chemother Pharmacol (2016) 77(4):663–71. doi: 10.1007/s00280-015-2918-1
    1. D'Incalci M, Galmarini CM. A review of trabectedin (ET-743): a unique mechanism of action. Mol Cancer Ther (2010) 9(8):2157–63. doi: 10.1158/1535-7163.MCT-10-0263
    1. Aune GJ, et al. . Von Hippel-Lindau–coupled and transcription-coupled nucleotide excision repair–dependent degradation of RNA polymerase II in response to trabectedin. Clin Cancer Res (2008) 14(20):6449–55. doi: 10.1158/1078-0432.CCR-08-0730
    1. Tavecchio M, et al. . Role of homologous recombination in trabectedin-induced DNA damage. Eur J Cancer (2008) 44(4):609–18. doi: 10.1016/j.ejca.2008.01.003
    1. Grignani G, Merlini A, Sangiolo D, D'Ambrosio L, Pignochino Y. Delving into PARP inhibition from bench to bedside and back. Pharmacol Ther (2020) 206:107446. doi: 10.1016/j.pharmthera.2019.107446
    1. Kamel D, Gray C, Walia JS, Kumar V. PARP inhibitor drugs in the treatment of breast, ovarian, prostate and pancreatic cancers: an update of clinical trials. Curr Drug Targets (2018) 19(1):21–37. doi: 10.2174/1389450118666170711151518
    1. Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives poly (ADP-ribose) polymerase (PARP) inhibitor lethality in homologous recombination-deficient cells. Proc Natl Acad Sci (2011) 108(8):3406–11. doi: 10.1073/pnas.1013715108
    1. Corti G, et al. . A genomic analysis workflow for colorectal cancer precision oncology. Clin colorectal Cancer (2019) 18(2):91–101. e103. doi: 10.1016/j.clcc.2019.02.008
    1. Crisafulli G, et al. . Whole exome sequencing analysis of urine trans-renal tumour DNA in metastatic colorectal cancer patients. ESMO Open (2019) 4(6):e000572. doi: 10.1136/esmoopen-2019-000572
    1. Adzhubei IA, et al. . A method and server for predicting damaging missense mutations. Nat Methods (2010) 7(4):248–9. doi: 10.1038/nmeth0410-248
    1. Van Emburgh BO, et al. . Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer. Nat Commun (2016) 7:13665–5. doi: 10.1038/ncomms13665
    1. Wang F, et al. . RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol diagnostics (2012) 14(1):22–9. doi: 10.1016/j.jmoldx.2011.08.002
    1. Gao J, et al. . Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal (2013) 6(269):pl1. doi: 10.1126/scisignal.2004088
    1. Gounder MM, et al. . Impact of next-generation sequencing (NGS) on diagnostic and therapeutic options in soft-tissue and bone sarcoma. J Clin Oncol (2017) 35(15):11001–1100139. doi: 10.1200/JCO.2017.35.15_suppl.11001
    1. Zhao R, Choi BY, Lee M-H, Bode AM, Dong Z. Implications of genetic and epigenetic alterations of CDKN2A (p16INK4a) in cancer. EBioMedicine (2016) 8:30–9. doi: 10.1016/j.ebiom.2016.04.017
    1. Monzon J, et al. . CDKN2A mutations in multiple primary melanomas. New Engl J Med (1998) 338(13):879–87. doi: 10.1056/NEJM199803263381305
    1. Lu VM, et al. . The prognostic significance of CDKN2A homozygous deletion in IDH-mutant lower-grade glioma and glioblastoma: a systematic review of the contemporary literature. J Neurooncol (2020) 148(2):221–9. doi: 10.1007/s11060-020-03528-2
    1. Foulkes WD, Flanders TY, Pollock PM, Hayward NK. The CDKN2A (p16) gene and human cancer. Mol Med (1997) 3(1):5–20. doi: 10.1007/BF03401664
    1. Bui NQ, et al. . A clinico-genomic analysis of soft tissue sarcoma patients reveals CDKN2A deletion as a biomarker for poor prognosis. Clin Sarcoma Res (2019) 9:12. doi: 10.1186/s13569-019-0122-5
    1. Cizkova M, et al. . PIK3R1 underexpression is an independent prognostic marker in breast cancer. BMC Cancer (2013) 13(1):1–15. doi: 10.1186/1471-2407-13-545
    1. Lok BH, et al. . PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin Cancer Res (2017) 23(2):523–35. doi: 10.1158/1078-0432.CCR-16-1040
    1. Zoppoli G, et al. . Putative DNA/RNA helicase schlafen-11 (SLFN11) sensitizes cancer cells to DNA-damaging agents. Proc Natl Acad Sci (2012) 109(37):15030–5. doi: 10.1073/pnas.1205943109
    1. Li M, et al. . DNA Damage-induced cell death relies on SLFN11-dependent cleavage of distinct type II tRNAs. Nat Struct Mol Biol (2018) 25(11):1047–58. doi: 10.1038/s41594-018-0142-5
    1. Jensen KA, Shi X, Yan S. Genomic alterations and abnormal expression of APE2 in multiple cancers. Sci Rep (2020) 10(1):1–11. doi: 10.1038/s41598-020-60656-5
    1. Kumar S, et al. . Role of apurinic/apyrimidinic nucleases in the regulation of homologous recombination in myeloma: mechanisms and translational significance. Blood Cancer J (2018) 8(10):1–10. doi: 10.1038/s41408-018-0129-9
    1. Mengwasser KE, et al. . Genetic screens reveal FEN1 and APEX2 as BRCA2 synthetic lethal targets. Mol Cell (2019) 73(5):885–99.e886. doi: 10.1016/j.molcel.2018.12.008
    1. LaRocque JR, et al. . Interhomolog recombination and loss of heterozygosity in wild-type and bloom syndrome helicase (BLM)-deficient mammalian cells. Proc Natl Acad Sci (2011) 108(29):11971–6. doi: 10.1073/pnas.1104421108
    1. Johnson RD, Liu N, Jasin M. Mammalian XRCC2 promotes the repair of DNA double-strand breaks by homologous recombination. Nature (1999) 401(6751):397–9. doi: 10.1038/43932
    1. Boersma V, et al. . MAD2L2 controls DNA repair at telomeres and DNA breaks by inhibiting 5′ end resection. Nature (2015) 521(7553):537–40. doi: 10.1038/nature14216
    1. Mendes-Pereira AM, et al. . Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol Med (2009) 1(6-7):315–22. doi: 10.1002/emmm.200900041
    1. Becerikli M, et al. . Numerical and structural chromosomal anomalies in undifferentiated pleomorphic sarcoma. Anticancer Res (2014) 34(12):7119–27.
    1. Holtkamp N, et al. . EGFR and erbB2 in malignant peripheral nerve sheath tumors and implications for targeted therapy. Neuro-oncology (2008) 10(6):946–57. doi: 10.1215/15228517-2008-053
    1. Pahuja KB, et al. . Actionable activating oncogenic ERBB2/HER2 transmembrane and juxtamembrane domain mutations. Cancer Cell (2018) 34(5):792–806. e795. doi: 10.1016/j.ccell.2018.09.010
    1. Boone JJ, Bhosle J, Tilby MJ, Hartley JA, Hochhauser D. Involvement of the HER2 pathway in repair of DNA damage produced by chemotherapeutic agents. Mol Cancer Ther (2009) 8(11):3015–23. doi: 10.1158/1535-7163.MCT-09-0219

Source: PubMed

3
订阅