The DNA damage response network in the treatment of head and neck squamous cell carcinoma

A Psyrri, M Gkotzamanidou, G Papaxoinis, L Krikoni, P Economopoulou, I Kotsantis, M Anastasiou, V L Souliotis, A Psyrri, M Gkotzamanidou, G Papaxoinis, L Krikoni, P Economopoulou, I Kotsantis, M Anastasiou, V L Souliotis

Abstract

Background: We sought to determine whether DNA damage response (DDR)-related aberrations predict therapeutic benefit in cisplatin-treated head and neck squamous cell carcinoma (HNSCC) patients and how DDR pathways are modulated after treatment with olaparib alone or in combination with cisplatin or durvalumab.

Patients and methods: Oxidative stress, abasic sites and DDR-related parameters, including endogenous DNA damage, DNA repair mechanisms and apoptosis rates, were evaluated in HNSCC cell lines and peripheral blood mononuclear cells from 46 healthy controls (HC) and 70 HNSCC patients at baseline and following treatment with cisplatin-containing chemoradiation or nivolumab or enrolled in the OPHELIA phase II trial (NCT02882308; olaparib alone, olaparib plus cisplatin, olaparib plus durvalumab).

Results: HNSCC patients at diagnosis exhibited deregulated DDR-related parameters and higher levels of oxidative stress and abasic sites compared with HC (all P < 0.05). Accordingly, nucleotide excision repair (NER; ERCC1, ERCC2/XPD, XPA, XPC) and base excision repair (APEX1, XRCC1) genes were downregulated in patients versus HC whereas double-strand breaks repair (MRE11A, RAD50, RAD51, XRCC2) and mismatch repair (MLH1, MSH2, MSH3) genes were overexpressed. Corresponding results were obtained in cell lines (all P < 0.001). Excellent correlations were observed between individual ex vivo and in vivo/therapeutic results, with cisplatin non-responders showing higher levels of endogenous DNA damage, augmented oxidative stress and abasic sites, increased NER capacities and reduced apoptosis than responders (all P < 0.05). Also, longer progression-free survival correlated with lower NER capacity (P = 0.037) and increased apoptosis (P = 0.029). Interestingly, treatment with olaparib-containing regimens results in the accumulation of cytotoxic DNA damage and exerts an extra antitumor effect by elevating oxidative stress (all P < 0.05). Nivolumab induced no significant changes in the DDR parameters examined.

Conclusions: Aberrations in DDR signals are implicated in the response to HNSCC chemotherapy and can be exploited as novel therapeutic targets, sensitive/effective non-invasive biomarkers as well as for the design of novel clinical trials.

Keywords: DNA damage response; cisplatin-containing chemoradiation; endogenous DNA damage; head and neck squamous cell carcinoma; olaparib-containing regimens; oxidative stress.

Conflict of interest statement

Disclosure The authors have declared no conflicts of interest. Data sharing Data are available upon reasonable request.

Copyright © 2021 The Author(s). Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
DNA damage response in HNSCC cell lines. (A) Alkaline comet assay images of normal and HNSCC cell lines. (B) Bar charts showing distribution of the endogenous DNA damage measured by comet assay. (C) Typical images showing γH2AX staining using confocal microscopy. Upper images, γH2AX staining; middle, cell nuclei labeled with DAPI; bottom, merged. (D) Bar charts showing distribution of the endogenous DSBs measured by γH2AX staining. Bar charts showing distribution of (E) the oxidative stress and (F) AP-sites. (G) Southern blot of the cisplatin-induced N-ras-specific monoadducts in normal and HNSCC cell lines. Bar charts showing (H) the kinetics of monoadducts repair and (I) the accumulation of monoadducts following cisplatin treatment. N CL, normal cell line; HNSCC CL, HNSCC cell line. Bar charts showing (J) the kinetics of γH2AX foci formation/removal and (K) the accumulation of γH2AX foci after cisplatin treatment. (L) Bar charts showing distribution of the lowest concentrations of cisplatin required for the induction of apoptosis 24 h, 48 h and 72 h after cisplatin treatment. Error bars represent SD; ∗∗∗P < 0.001 by Mann–Whitney U test. The experiments shown were based on a minimum of three independent repeats. AP, abasic sites; AUC, area under the curve; CL, cell line; DAPI, 4′,6-diamidino-2-phenylindole; DSBs, double-strand breaks; HNSCC, head and neck squamous cell cancer; IS, internal standard; N CL, normal cell line; SD, standard deviation.
Figure 2
Figure 2
Endogenous DNA damage in PBMCs from HNSCC patients. (A) Alkaline comet assay images of two healthy controls (HC1, HC2) and two HNSCC patients at baseline. (B) Box plots showing statistical distribution of the endogenous DNA damage measured by comet assay in HC and HNSCC patients at baseline. (C) The immunofluorescence γH2AX staining of two HC and two HNSCC patients. (D) Box plots showing statistical distribution of the endogenous DNA damage in HC and patients at baseline. Box plots showing statistical distribution of the oxidative stress (E) and the AP-sites (F) in HC and patients at baseline. Error bars represent SD; ∗∗∗P < 0.001 by Mann–Whitney U test. The experiments shown were based on a minimum of three independent repeats. AP, abasic sites; DAPI, 4′,6-diamidino-2-phenylindole; HC, healthy control; HNSCC, head and neck squamous cell cancer; PBMCs, peripheral blood mononuclear cells; NR, non-responder; R, responder; SD, standard deviation.
Figure 3
Figure 3
DDR signals in PBMCs following ex vivo cisplatin treatment. (A) Southern blot of the ex vivo cisplatin-induced monoadducts in a HC and two HNSCC patients (R, responder; NR, non-responder). (B) The kinetics of monoadducts repair and (C) the statistical distribution of the accumulation of the ex vivo cisplatin-induced monoadducts, in HC and HNSCC patients at baseline. (D) The kinetics of γH2AX foci and (E) the statistical distribution of the ex vivo cisplatin-induced γH2AX foci accumulation in HC and HNSCC patients. (F) Box plots showing statistical distribution of the lowest concentrations of cisplatin required for the induction of apoptosis 24 h, 48 h and 72 h after ex vivo treatment with cisplatin. Error bars represent SD; ∗P < 0.05, ∗∗P < 0.01 and ∗∗∗P < 0.001 by Mann–Whitney U test. The experiments shown were based on a minimum of three independent repeats. AUC, area under the curve; DDR, DNA damage response; HC, healthy control; HNSCC, head and neck squamous cell cancer; IS, internal standard. NR, non-responder; PBMCs, peripheral blood mononuclear cells; R, responder.
Figure 4
Figure 4
Kaplan–Meier PFS curves and expression of DDR-associated genes. (A) Kaplan–Meier PFS curves according to response to cisplatin-containing chemoradiation. Kaplan–Meier curves demonstrating that longer PFS is associated with (B) lower NER resulting in higher DNA damage burden, (C) increased apoptosis, (D) lower endogenous DNA damage, and (E) decreased oxidative stress at baseline. (F) Hierarchical clustergram of 84 DDR-associated genes in eight HNSCC patients at baseline versus eight HC. (G, H) Genes demonstrating at least two-fold difference in the transcription activity between patients and HC. AUC, area under the curve; CI, confidence interval; DDR, DNA damage response; GSH, glutathione; GSSG, glutathione disulfide; HC, healthy control; HNSCC, head and neck squamous cell cancer; HR, hazard ratio; NER, nucleotide excision repair; PFS, progression-free survival.
Figure 5
Figure 5
DDR signals following various in vivo/therapeutic treatments. (A) Box plots showing the kinetics of monoadducts repair in patients following various therapeutic treatments. (B) Box plots showing statistical distribution of the accumulation of monoadducts after cisplatin-only therapy. (C) Correlation between monoadducts burden following therapeutic and ex vivo cisplatin treatment in the same patients. (D) Box plots showing the kinetics of γH2AX foci formation/removal following various therapeutic treatments. (E) Box plots showing the statistical distribution of the accumulation of γH2AX foci after cisplatin-only therapy. (F) Correlation between DSB burden following therapeutic and ex vivo cisplatin treatment in the same patients. Box plots showing statistical distribution of the oxidative stress (G) and the abasic sites (H) after various therapeutic treatments. Error bars represent SD; ∗P < 0.05, ∗∗P < 0.01 and ∗∗∗P < 0.001 by Mann–Whitney U test. The experiments shown were based on a minimum of three independent repeats. AUC, area under the curve; DDR, DNA damage response; DSB, double-strand break; GSH, glutathione; GSSG, glutathione disulfide; NR, non-responder; NS, not significant; PBMCs, peripheral blood mononuclear cells; R, responder; SD, standard deviation.

References

    1. Du E., Mazul A.L., Farquhar D. Long term survival in head and neck cancer: impact of site, stage, smoking, and human papillomavirus status. Laryngoscope. 2019;129:2506–2513.
    1. Economopoulou P., de Bree R., Kotsantis I. Diagnostic tumor markers in head and neck squamous cell carcinoma (HNSCC) in the clinical setting. Front Oncol. 2019;9:827.
    1. Tubbs A., Nussenzweig A. Endogenous DNA damage as a source of genomic instability in cancer. Cell. 2017;168:644–656.
    1. De Bont R., van Larebeke N. Endogenous DNA damage in humans: a review of quantitative data. Mutagenesis. 2004;19:169–185.
    1. Pateras I.S., Havaki S., Nikitopoulou X. The DNA damage response and immune signaling alliance: is it good or bad? Nature decides when and where. Pharmacol Ther. 2015;154:36–56.
    1. Burrell R.A., McGranahan N., Bartek J. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501:338–345.
    1. Jeggo P.A., Pearl L.H., Carr A.M. DNA repair, genome stability and cancer: a historical perspective. Nat Rev Cancer. 2016;16:35–42.
    1. Srinivas U.S., Tan B.W.Q., Vellayappan B.A. ROS and the DNA damage response in cancer. Redox Biol. 2019;25:101084.
    1. Visconti R., Della Monica R., Grieco D. Cell cycle checkpoint in cancer: a therapeutically targetable double-edged sword. J Exp Clin Cancer Res. 2016;35:153.
    1. Ceccaldi R., Rondinelli B., D'Andrea A.D. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 2016;26:52–64.
    1. Chakravarthi B.V., Nepal S., Varambally S. Genomic and epigenomic alterations in cancer. Am J Pathol. 2016;186:1724–1735.
    1. Mouw K.W., Goldberg M.S., Konstantinopoulos P.A. DNA damage and repair biomarkers of immunotherapy response. Cancer Discov. 2017;7:675–693.
    1. Cooper J.S., Pajak T.F., Forastiere A.A. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med. 2004;350:1937–1944.
    1. Kartalou M., Essigmann J.M. Mechanisms of resistance to cisplatin. Mutat Res. 2001;478:23–43.
    1. Mogi S., Oh D.H. Gamma-H2AX formation in response to interstrand crosslinks requires 7XPF in human cells. DNA Repair (Amst) 2006;5:731–740.
    1. Helleday T. Homologous recombination in cancer development, treatment and development of drug resistance. Carcinogenesis. 2010;31:955–960.
    1. Chakraborty A., Tapryal N., Venkova T. Classical non-homologous end-joining pathway utilizes nascent RNA for error-free double-strand break repair of transcribed genes. Nat Commun. 2016;7:13049.
    1. Ray Chaudhuri A., Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol. 2017;18:610–621.
    1. Ledermann J.A., Pujade-Lauraine E. Olaparib as maintenance treatment for patients with platinum-sensitive relapsed ovarian cancer. Ther Adv Med Oncol. 2019;11 1758835919849753.
    1. Qin S., Xu L., Yi M. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18:155.
    1. Yarchoan M., Johnson B.A., 3rd, Lutz E.R. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer. 2017;17:209–222.
    1. Li T., Chen Z.J. The cGAS–cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med. 2018;215:1287–1299.
    1. Chen Q., Sun L., Chen Z.J. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–1149.
    1. Motwani M., Pesiridis S., Fitzgerald K.A. DNA sensing by the cGAS–STING pathway in health and disease. Nat Rev Genet. 2019;20:657–674.
    1. Kwon J., Bakhoum S.F. The cytosolic DNA-sensing cGAS–STING pathway in cancer. Cancer Discov. 2020;10:26–39.
    1. Zhang Q., Green M.D., Lang X. Inhibition of ATM increases interferon signaling and sensitizes pancreatic cancer to immune checkpoint blockade therapy. Cancer Res. 2019;79:3940–3951.
    1. Mateo J., Lord C.J., Serra V. A decade of clinical development of PARP inhibitors in perspective. Ann Oncol. 2019;30:1437–1447.
    1. Yu E.Y., Massard C., Retz M. Keynote-365 cohort a: pembrolizumab (pembro) plus olaparib in docetaxel-pretreated patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC) J Clin Oncol. 2019;37(suppl 7):145.
    1. Karzai F., VanderWeele D., Madan R.A. Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J Immunother Cancer. 2018;6:141.
    1. Lee J.M., Cimino-Mathews A., Peer C.J. Safety and clinical activity of the programmed death-ligand 1 inhibitor durvalumab in combination with poly (ADP-Ribose) polymerase inhibitor olaparib or vascular endothelial growth factor receptor 1-3 inhibitor cediranib in women's cancers: a dose-escalation, phase I study. J Clin Oncol. 2017;35:2193–2202.
    1. Drew Y., de Jonge M., Hong S.H. An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): results in germline BRCA-mutated (gBRCA m) platinum-sensitive relapsed (PSR) ovarian cancer (OC) Gynecol Oncol. 2018;149:246–247.
    1. Vinayak S., Tolaney S.M., Schwartzberg L.S. TOPACIO/Keynote-162: niraparib + pembrolizumab in patients (pts) with metastatic triple-negative breast cancer (TNBC), a Phase II trial. J Clin Oncol. 2018;36(suppl 15):1011.
    1. Friedlander M., Meniawy T., Markman B. A Phase Ib study of the anti-PD-1 monoclonal antibody BGB-A317 (A317) in combination with the PARP inhibitor BGB-290 (290) in advanced solid tumors. J Clin Oncol. 2018;36(suppl 5):48.
    1. Patel M.R., Falchook G.S., Wang J.S.-Z. Open-label, multicenter, phase I study to assess safety and tolerability of adavosertib plus durvalumab in patients with advanced solid tumors. J Clin Oncol. 2019;37(suppl 15):2562.
    1. Amin M.B., Greene F.L., Edge S.B. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin. 2017;67:93–99.
    1. Schwartz L.H., Litière S., de Vries E. RECIST 1.1-Update and clarification: from the RECIST committee. Eur J Cancer. 2016;62:132–137.
    1. Gkotzamanidou M., Sfikakis P.P., Kyrtopoulos S.A. Chromatin structure, transcriptional activity and DNA repair efficiency affect the outcome of chemotherapy in multiple myeloma. Br J Cancer. 2014;111:1293–1304.
    1. Theron T., Fousteri M.I., Volker M. Transcription-associated breaks in xerodermapigmentosum group D cells from patients with combined features of xeroderma pigmentosum and Cockayne syndrome. Mol Cell Biol. 2005;25:8368–8378.
    1. Baker S.R. An in vivo model for squamous cell carcinoma of the head and neck. Laryngoscope. 1985;95:43–56.
    1. Gioanni J., Courdi A., Lalanne C.M. Establishment, characterization, chemosensitivity, and radiosensitivity of two different cell lines derived from a human breast cancer biopsy. Cancer Res. 1985;45:1246–1258.
    1. Souliotis V.L., Vlachogiannis N.I., Pappa M. DNA damage accumulation, defective chromatin organization and deficient DNA repair capacity in patients with rheumatoid arthritis. Clin Immunol. 2019;203:28–36.
    1. Gkotzamanidou M., Terpos E., Bamia C. DNA repair of myeloma plasma cells correlates with clinical outcome: the effect of the nonhomologous end-joining inhibitor SCR7. Blood. 2016;128:1214–1225.
    1. Vichai V., Kirtikara K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc. 2006;1:1112–1116.
    1. Souliotis V.L., Vougas K., Gorgoulis V.G. Defective DNA repair and chromatin organization in patients with quiescent systemic lupus erythematosus. Arthritis Res Ther. 2016;18:182.
    1. Souliotis V.L., Dimopoulos M.A., Episkopou H.G. Preferential in vivo DNA repair of melphalan-induced damage in human genes is greatly affected by the local chromatin structure. DNA Repair (Amst) 2006;5:972–985.
    1. Souliotis V.L., Vlachogiannis N.I., Pappa M. DNA damage response and oxidative stress in systemic autoimmunity. Int J Mol Sci. 2019;21:E55.
    1. Vitale I., Kroemer G. Spontaneous DNA damage propels tumorigenicity. Cell Res. 2017;27:720–721.
    1. Singh A.K., Pandey P., Tewari M. Free radicals hasten head and neck cancer risk: a study of total oxidant, total antioxidant, DNA damage, and histological grade. J Postgrad Med. 2016;62:96–101.
    1. Hegde M.L., Izumi T., Mitra S. Oxidized base damage and single-strand break repair in mammalian genomes: role of disordered regions and posttranslational modifications in early enzymes. Prog Mol Biol Transl Sci. 2012;110:123–153.
    1. Dequanter D., Dok R., Nuyts S. Basal oxidative stress ratio of head and neck squamous cell carcinomas correlates with nodal metastatic spread in patients under therapy. Onco Targets Ther. 2017;10:259–263.
    1. Schärer O.D. Nucleotide excision repair in eukaryotes. Cold Spring Harb Perspect Biol. 2013;5:a012609.
    1. Stefanou D.T., Bamias A., Episkopou H. Aberrant DNA damage response pathways may predict the outcome of platinum chemotherapy in ovarian cancer. PLoS One. 2015;10:e0117654.
    1. Sliwinski T., Markiewicz L., Rusin P. Impaired nucleotide excision repair pathway as a possible factor in pathogenesis of head and neck cancer. Mutat Res. 2011;716:51–58.
    1. Dylawerska A., Barczak W., Wegner A. Association of DNA repair genes polymorphisms and mutations with increased risk of head and neck cancer: a review. Med Oncol. 2017;34:197.
    1. Schena M., Guarrera S., Buffoni L. DNA repair gene expression level in peripheral blood and tumour tissue from non-small cell lung cancer and head and neck squamous cell cancer patients. DNA Repair (Amst) 2012;11:374–380.
    1. Dimopoulos M.A., Souliotis V.L., Anagnostopoulos A. Extent of damage and repair in the p53 tumor-suppressor gene after treatment of myeloma patients with high-dose melphalan and autologous blood stem-cell transplantation is individualized and may predict clinical outcome. J Clin Oncol. 2005;23:4381–4389.
    1. Shammas M.A., Shmookler Reis R.J., Koley H. Dysfunctional homologous recombination mediates genomic instability and progression in myeloma. Blood. 2009;113:2290–2297.
    1. Sliwinski T., Walczak A., Przybylowska K. Polymorphisms of the XRCC3 C722T and the RAD51 G135C genes and the risk of head and neck cancer in a Polish population. Exp Mol Pathol. 2010;89:358–366.
    1. Choudhury J.H., Choudhury B., Kundu S. Combined effect of tobacco and DNA repair genes polymorphisms of XRCC1 and XRCC2 influence high risk of head and neck squamous cell carcinoma in northeast Indian population. Med Oncol. 2014;31:67.
    1. Senghore T., Wang W.C., Chien H.T. Polymorphisms of mismatch repair pathway genes predict clinical outcomes in oral squamous cell carcinoma patients receiving adjuvant concurrent chemoradiotherapy. Cancers (Basel) 2019;11:598.
    1. Mahjabeen I., Ali K., Zhou X. Deregulation of base excision repair gene expression and enhanced proliferation in head and neck squamous cell carcinoma. Tumour Biol. 2014;35:5971–5983.
    1. Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
    1. Hassan M., Watari H., Abu-Almaaty A. Apoptosis and molecular targeting therapy in cancer. Biomed Res Int. 2014;2014:150845.
    1. Bochum S., Berger S., Martens U.M. Olaparib. Recent Results Cancer Res. 2018;211:217–233.
    1. Hou H., Liu Z., Xu X. Increased oxidative stress mediates the antitumor effect of PARP inhibition in ovarian cancer. Redox Biol. 2018;17:99–111.
    1. Jiao S., Xia W., Yamaguchi H. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res. 2017;23:3711–3720.
    1. Fikrova P., Stetina R., Hrnciarik M. DNA crosslinks, DNA damage and repair in peripheral blood lymphocytes of non-small cell lung cancer patients treated with platinum derivatives. Oncol Rep. 2014;31:391–396.
    1. Burgaz S., Coskun E., Demircigil G.C. Micronucleus frequencies in lymphocytes and buccal epithelial cells from patients having head and neck cancer and their first-degree relatives. Mutagenesis. 2011;26:351–356.
    1. Vasavi M., Vedicherala B., Vattam K.K. Assessment of genetic damage in inflammatory, precancerous, and cancerous pathologies of the esophagus using the comet assay. Genet Test Mol Biomarkers. 2010;14:477–482.
    1. Santos R.A., Teixeira A.C., Mayorano M.B. Basal levels of DNA damage detected by micronuclei and comet assays in untreated breast cancer patients and healthy women. Clin Exp Med. 2010;10:87–92.
    1. Synowiec E., Stefanska J., Morawiec Z. Association between DNA damage, DNA repair genes variability and clinical characteristics in breast cancer patients. Mutat Res. 2008;648:65–72.
    1. Schabath M.B., Spitz M.R., Grossman H.B. Genetic instability in bladder cancer assessed by the comet assay. J Natl Cancer Inst. 2003;95:540–547.
    1. Allione A., Pardini B., Viberti C. The prognostic value of basal DNA damage level in peripheral blood lymphocytes of patients affected by bladder cancer. Urol Oncol. 2018;36:241.e15–241.e23.
    1. Buchynska L., Brieieva O., Glushchenko N. DNA repair deficiency in peripheral blood lymphocytes of endometrial cancer patients with a family history of cancer. BMC Cancer. 2014;14:765.
    1. Liu Z., Liu H., Han P. Apoptotic capacity and risk of squamous cell carcinoma of the head and neck. Eur J Cancer. 2017;72:166–176.
    1. Raturi V.P., Wu C.T., Mohammad S. Could excision repair cross-complementing group-1 mRNA expression from peripheral blood lymphocytes predict locoregional failure with cisplatin chemoradiation for locally advanced laryngeal cancer? Asia Pac J Clin Oncol. 2020;16:e19–e26.
    1. Romanet T., Ré J.L., De Méo M. Detection of hypoxia by measurement of DNA damage and repair in human lymphocytes (comet assay): a predictive variable for tumor response during chemotherapy in patients with head and neck squamous cell carcinoma. In Vivo. 1999;13:343–348.
    1. Quintela-Fandino M., Hitt R., Medina P.P. DNA-repair gene polymorphisms predict favorable clinical outcome among patients with advanced squamous cell carcinoma of the head and neck treated with cisplatin-based induction chemotherapy. J Clin Oncol. 2006;24:4333–4339.
    1. Borgmann K., Röper B., El-Awady R. Indicators of late normal tissue response after radiotherapy for head and neck cancer: fibroblasts, lymphocytes, genetics, DNA repair, and chromosome aberrations. Radiother Oncol. 2002;64:141–152.
    1. Fikrova P., Stetina R., Hrnciarik M. Detection of DNA crosslinks in peripheral lymphocytes isolated from patients treated with platinum derivates using modified comet assay. Neoplasma. 2013;60:413–418.
    1. Lobachevsky P.N., Bucknell N.W., Mason J. Monitoring DNA damage and repair in peripheral blood mononuclear cells of lung cancer radiotherapy patients. Cancers (Basel) 2020;12:2517.
    1. Suna X., Li F., Suna N. Polymorphisms in XRCC1 and XPG and response to platinum-based chemotherapy in advanced non-small cell lung cancer patients. Lung Cancer. 2009;65:230–236.
    1. Zhou C., Ren S., Zhou S. Predictive effects of ERCC1 and XRCC3 SNP on efficacy of platinum-based chemotherapy in advanced NSCLC patients. Jpn J Clin Oncol. 2010;40:954–960.
    1. Reed E., Yuspa S.H., Zwelling L.A. Quantitation of cis-diamminedichloroplatinum II (cisplatin)-DNA-intrastrand adducts in testicular and ovarian cancer patients receiving cisplatin chemotherapy. J Clin Invest. 1986;77:545–550.
    1. Zimmermann M., Li T., Semrad T.J. Oxaliplatin-DNA adducts as predictive biomarkers of FOLFOX response in colorectal cancer: a potential treatment optimization strategy. Mol Cancer Ther. 2020;19:1070–1079.

Source: PubMed

3
Abonnieren