Monitoring of Early Changes of Circulating Tumor DNA in the Plasma of Rectal Cancer Patients Receiving Neoadjuvant Concomitant Chemoradiotherapy: Evaluation for Prognosis and Prediction of Therapeutic Response

Filip Pazdirek, Marek Minarik, Lucie Benesova, Tereza Halkova, Barbora Belsanova, Milan Macek, Lubomír Stepanek, Jiri Hoch, Filip Pazdirek, Marek Minarik, Lucie Benesova, Tereza Halkova, Barbora Belsanova, Milan Macek, Lubomír Stepanek, Jiri Hoch

Abstract

Introduction: Patients with locally advanced rectal cancer (LARC) are undergoing neoadjuvant chemoradiotherapy (NCRT) prior to surgery. Although in some patients the NCRT is known to prevent local recurrence, it is also accompanied by side effects. Accordingly, there is an unmet need to identify predictive markers allowing to identify non-responders to avoid its adverse effects. We monitored circulating tumor DNA (ctDNA) as a potential liquid biopsy-based biomarker. We have investigated ctDNA changes plasma during the early days of NCRT and its relationship to the overall therapy outcome. Methods and Patients: The studied cohort included 36 LARC patients (stage II or III) undergoing NCRT with subsequent surgical treatment. We have detected somatic mutations in tissue biopsies taken during endoscopic examination prior to the therapy. CtDNA was extracted from patient plasma samples prior to therapy and at the end of the first week. In order to optimize the analytical costs of liquid-biopsy testing, we have utilized a two-level approach in which first a low-cost detection method of denaturing capillary electrophoresis was used followed by examination of initially negative samples by a high-sensitivity BEAMING assay. The ctDNA was related to clinical parameters including tumor regression grade (TRG) and TNM tumor staging. Results: We have detected a somatic mutation in 33 out of 36 patients (91.7%). Seven patients (7/33, 21.2%) had ctDNA present prior to therapy. The ctDNA positivity before treatment reduced post-operative disease-free survival and overall survival by an average of 1.47 and 1.41 years, respectively (p = 0.015, and p = 0.010). In all patients, ctDNA was strongly reduced or completely eliminated from plasma by the end of the first week of NCRT, with no correlation to any of the parameters analyzed. Conclusions: The baseline ctDNA presence represented a statistically significant negative prognostic biomarker for the overall patient survival. As ctDNA was reduced indiscriminately from circulation of all patients, dynamics during the first week of NCRT is not suited for predicting the outcome of LARC. However, the general effect of rapid ctDNA disappearance apparently occurring during the initial days of NCRT is noteworthy and should further be studied.

Keywords: biomarker; circulating tumor ctDNA; neoadjuvant chemoradiotherapy; prediction; prognosis; rectal cancer; response.

Copyright © 2020 Pazdirek, Minarik, Benesova, Halkova, Belsanova, Macek, Stepanek and Hoch.

Figures

Figure 1
Figure 1
A multilevel cascade testing algorithm for evaluation of circulating tumor DNA (ctDNA) involving standard [denaturing capillary electrophoresis (DCE)] and high-sensitivity (BEAMING*) approaches. *The BEAMING technology was performed using an experimental setting on a set of archived ctDNA samples. This research setting is different from that of protocols applied in regular testing under the IVD-CE certification.
Figure 2
Figure 2
MRI of the rectal tumor (initially T3, N1) before (A) and after (B) treatment. Arrows show tumor and enlarged lymph node. After treatment, no tumor and lymph nodes are presented (complete clinical tumor response).
Figure 3
Figure 3
Illustration of a complete therapy response in a patient. Pretreatment endoscopic examination of the tumor (A) and surgical specimen (B).
Figure 4
Figure 4
Impact of pretreatment circulating tumor DNA (ctDNA) positivity on disease-free survival (A) and overall survival (B) of locally advanced rectal cancer (LARC) patients.
Figure 5
Figure 5
Probability of disease-free survival (A) and overall survival (B) in locally advanced rectal cancer (LARC) patients according to circulating tumor DNA (ctDNA) status prior to the therapy onset.
Figure 6
Figure 6
Dynamics of circulating tumor DNA (ctDNA) during the first week of neoadjuvant chemoradiotherapy (NCRT). The ctDNA quantity is presented as a percentage of DNA fragments bearing tumor-specific mutation detected in plasma [denoted as minor allele fraction (MAF)].

References

    1. Glynne-Jones R, Wyrwicz L, Tiret E, Brown G, Rödel C, Cervantes A, et al. Rectal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. (2017) 28(Suppl. 4):iv22–40. 10.1093/annonc/mdx224
    1. Pahlman L, Bohe M, Cedermark B, Dahlberg M, Lindmark G, Sjodahl R, et al. The Swedish rectal cancer registry. Br J Surg. (2007) 94:1285–92. 10.1002/bjs.5679
    1. Fokas E, Liersch T, Fietkau R, Hohenberger W, Beissbarth T, Hess C, et al. Tumor regression grading after preoperative chemoradiotherapy for locally advanced rectal carcinoma revisited: updated results of the CAO/ARO/AIO-94 trial. J Clin Oncol. (2014) 32:1554–62. 10.1200/JCO.2013.54.3769
    1. Song C, Chung J-H, Kang S-B, Kim D-W, Oh H-K, Lee HS, et al. . Impact of tumor regression grade as a major prognostic factor in locally advanced rectal cancer after neoadjuvant chemoradiotherapy: a proposal for a modified staging system. Cancers. (2018) 10:319. 10.3390/cancers10090319
    1. Dworak O, Keilholz L, Hoffmann A. Pathological features of rectal cancer after preoperative radiochemotherapy. Int J Colorectal Dis. (1997) 12:19–23. 10.1007/s003840050072
    1. Contin P, Kulu Y, Bruckner T, Sturm M, Welsch T, Müller-Stich BP, et al. Comparative analysis of late functional outcome following preoperative radiation therapy or chemoradiotherapy and surgery or surgery alone in rectal cancer. Int J Colorect Dis. (2014) 29:165–75. 10.1007/s00384-013-1780-z
    1. Birgisson H, Pahlman L, Gunnarsson U, Glimelius B. Adverse effects of preoperative radiation therapy for rectal cancer: long-term follow-up of the Swedish rectal cancer trial. J Clin Oncol. (2005) 23:8697–705. 10.1200/JCO.2005.02.9017
    1. Dayde D, Tanaka I, Jain R, Tai MC, Taguchi A. Predictive and prognostic molecular biomarkers for response to neoadjuvant chemoradiation in rectal cancer. Int J Mol Sci. (2017) 18:573. 10.3390/ijms18030573
    1. Vaupel P, Thews O, Hoeckel M. Treatment resistance of solid tumors: role of hypoxia and anemia. Med Oncol. (2001) 18:243–59. 10.1385/MO:18:4:243
    1. DeVries AF, Kremser C, Hein PA, Griebel J, Krezcy A, Ofner D, et al. . Tumor microcirculation and diffusion predict therapy outcome for primary rectal carcinoma. Int J Radiat Oncol Biol Phys. (2003) 56:958–65. 10.1016/S0360-3016(03)00208-6
    1. Devries AF, Griebel J, Kremser C, Judmaier W, Gneiting T, Kreczy A, et al. . Tumor microcirculation evaluated by dynamic magnetic resonance imaging predicts therapy outcome for primary rectal carcinoma. Cancer Res. (2001) 61:2513–6. 10.1016/s0360-3016(02)03106-1
    1. Kremser C, Trieb T, Rudisch A, Judmaier W, de Vries A. Dynamic T(1) mapping predicts outcome of chemoradiation therapy in primary rectal carcinoma: sequence implementation and data analysis. J Magn Reson Imaging. (2007) 26:662–71. 10.1002/jmri.21034
    1. Lambrecht M, Vandecaveye V, De Keyzer F, Roels S, Penninckx F, Van Cutsem E, et al. . Value of diffusion-weighted magnetic resonance imaging for prediction and early assessment of response to neoadjuvant radiochemotherapy in rectal cancer: preliminary results. Int J Radiat Oncol Biol Phys. (2012) 82:863–70. 10.1016/j.ijrobp.2010.12.063
    1. Barbaro B, Vitale R, Valentini V, Illuminati S, Vecchio FM, Rizzo G, et al. . Diffusion-weighted magnetic resonance imaging in monitoring rectal cancer response to neoadjuvant chemoradiotherapy. Int J Radiat Oncol Biol Phys. (2012) 83:594–9. 10.1016/j.ijrobp.2011.07.017
    1. Elmi A, Hedgire SS, Covarrubias D, Abtahi SM, Hahn PF, Harisinghani M. Apparent diffusion coefficient as a non-invasive predictor of treatment response and recurrence in locally advanced rectal cancer. Clin Radiol. (2013) 68:e524–31. 10.1016/j.crad.2013.05.094
    1. Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, et al. . COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res. (2019) 47:D941–7. 10.1093/nar/gky1015
    1. Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, et al. . KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. (2006) 66:3992–5. 10.1158/0008-5472.CAN-06-0191
    1. Luna-Perez P, Segura J, Alvarado I, Labastida S, Santiago-Payan H, Quintero A. Specific c-K-ras gene mutations as a tumor-response marker in locally advanced rectal cancer treated with preoperative chemoradiotherapy. Ann Surg Oncol. (2000) 7:727–31. 10.1007/s10434-000-0727-0
    1. Carpinetti P, Donnard E, Bettoni F, Asprino P, Koyama F, Rozanski A, et al. . The use of personalized biomarkers and liquid biopsies to monitor treatment response and disease recurrence in locally advanced rectal cancer after neoadjuvant chemoradiation. Oncotarget. (2015) 6:38360–71. 10.18632/oncotarget.5256
    1. Grimminger PP, Danenberg P, Dellas K, Arnold D, Rodel C, Machiels JP, et al. . Biomarkers for cetuximab-based neoadjuvant radiochemotherapy in locally advanced rectal cancer. Clin Cancer Res. (2011) 17:3469–77. 10.1158/1078-0432.CCR-10-2273
    1. Garcia-Aguilar J, Chen Z, Smith DD, Li W, Madoff RD, Cataldo P, et al. . Identification of a biomarker profile associated with resistance to neoadjuvant chemoradiation therapy in rectal cancer. Ann Surg. (2011) 254:486–92. 10.1097/SLA.0b013e31822b8cfa
    1. Spitz FR, Giacco GG, Hess K, Larry L, Rich TA, Janjan N, et al. . p53 immunohistochemical staining predicts residual disease after chemoradiation in patients with high-risk rectal cancer. Clin Cancer Res. (1997) 3:1685–90.
    1. Rebischung C, Gerard JP, Gayet J, Thomas G, Hamelin R, Laurent-Puig P. Prognostic value of P53 mutations in rectal carcinoma. Int J Cancer. (2002) 100:131–5. 10.1002/ijc.10480
    1. Chen MB, Wu XY, Yu R, Li C, Wang LQ, Shen W, et al. . P53 status as a predictive biomarker for patients receiving neoadjuvant radiation-based treatment: a meta-analysis in rectal cancer. PLoS ONE. (2012) 7:e45388. 10.1371/journal.pone.0045388
    1. Huh JW, Lee JH, Kim HR. Pretreatment expression of 13 molecular markers as a predictor of tumor responses after neoadjuvant chemoradiation in rectal cancer. Ann Surg. (2014) 259:508–15. 10.1097/SLA.0b013e31829b3916
    1. Kudrimoti M, Lee EY, Kang Y, Ahmed M, Mohiuddin M. Genetic markers predictive of response to induction chemoradiotherapy for locally advanced rectal cancers. J Ky Med Assoc. (2007) 105:18–22.
    1. Osumi H, Shinozaki E, Yamaguchi K, Zembutsu H. Clinical utility of circulating tumor DNA for colorectal cancer. Cancer Sci. (2019) 110:1148–55. 10.1111/cas.13972
    1. Meddeb R, Dache ZAA, Thezenas S, Otandault A, Tanos R, Pastor B, et al. . Quantifying circulating cell-free DNA in humans. Sci Rep. (2019) 9:5220. 10.1038/s41598-019-41593-4
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. . Circulating mutant DNA to assess tumor dynamics. Nat Med. (2008) 14:985–90. 10.1038/nm.1789
    1. Ondrouskova E, Hrstka R. [Circulating tumor DNA in blood and its utilization as a potential biomarker for cancer]. Klin Onkol. (2015) 28(Suppl. 2):2s69–74. 10.14735/amko20152S69
    1. Sefrioui D, Sarafan-Vasseur N, Beaussire L, Baretti M, Gangloff A, Blanchard F, et al. . Clinical value of chip-based digital-PCR platform for the detection of circulating DNA in metastatic colorectal cancer. Dig Liver Dis. (2015) 47:884–90. 10.1016/j.dld.2015.05.023
    1. Spindler KL, Pallisgaard N, Andersen RF, Jakobsen A. Changes in mutational status during third-line treatment for metastatic colorectal cancer–results of consecutive measurement of cell free DNA, KRAS and BRAF in the plasma. Int J Cancer. (2014) 135:2215–22. 10.1002/ijc.28863
    1. Tie J, Kinde I, Wang Y, Wong HL, Roebert J, Christie M, et al. . Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann Oncol. (2015) 26:1715–22. 10.1093/annonc/mdv177
    1. Khier S, Lohan L. Kinetics of circulating cell-free DNA for biomedical applications: critical appraisal of the literature. Future Sci OA. (2018) 4:Fso295. 10.4155/fsoa-2017-0140
    1. Minarik M, Minarikova L, Hrabikova M, Minarikova P, Hrabal P, Zavoral M. Application of cycling gradient capillary electrophoresis to detection of APC, K-ras, and DCC point mutations in patients with sporadic colorectal tumors. Electrophoresis. (2004) 25:1016–21. 10.1002/elps.200305770
    1. Prochazkova K, Pavlikova K, Minarik M, Sumerauer D, Kodet R, Sedlacek Z. Somatic TP53 mutation mosaicism in a patient with Li-Fraumeni syndrome. Am J Med Genet A. (2009) 149a:206–11. 10.1002/ajmg.a.32574
    1. Hinselwood DC, Abrahamsen TW, Ekstrom PO. BRAF mutation detection and identification by cycling temperature capillary electrophoresis. Electrophoresis. (2005) 26:2553–61. 10.1002/elps.200410427
    1. Fiala O, Pesek M, Finek J, Benesova L, Bortlicek Z, Minarik M. Gene mutations in squamous cell NSCLC: insignificance of EGFR, KRAS and PIK3CA mutations in prediction of EGFR-TKI treatment efficacy. Anticancer Res. (2013) 33:1705–11. 10.1016/S0169-5002(13)70293-9
    1. Levy M, Benesova L, Lipska L, Belsanova B, Minarikova P, Veprekova G, et al. . Utility of cell-free tumour DNA for post-surgical follow-up of colorectal cancer patients. Anticancer Res. (2012) 32:1621–6.
    1. Diehl F, Li M, He Y, Kinzler KW, Vogelstein B, Dressman D. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods. (2006) 3:551–9. 10.1038/nmeth898
    1. Team. RDC A Language and Environment For Statistical Computing. Vienna: R Foundation for Statistical Computing; (2008).
    1. Zitt M, Muller HM, Rochel M, Schwendinger V, Goebel G, Devries A, et al. . Circulating cell-free DNA in plasma of locally advanced rectal cancer patients undergoing preoperative chemoradiation: a potential diagnostic tool for therapy monitoring. Dis Markers. (2008) 25:159–65. 10.1155/2008/598071
    1. Agostini M, Pucciarelli S, Enzo MV, Del Bianco P, Briarava M, Bedin C, et al. . Circulating cell-free DNA: a promising marker of pathologic tumor response in rectal cancer patients receiving preoperative chemoradiotherapy. Ann Surg Oncol. (2011) 18:2461–8. 10.1245/s10434-011-1638-y
    1. Sun W, Li G, Wan J, Zhu J, Shen W, Zhang Z. Circulating tumor cells: a promising marker of predicting tumor response in rectal cancer patients receiving neoadjuvant chemo-radiation therapy. Oncotarget. (2016) 7:69507–17. 10.18632/oncotarget.10875
    1. Li M, Xiao W, Zhao GQ, Guo Z, Yang X, Lin S, et al. Predictive value of circulating tumor DNA in locally advanced rectal cancer patients receiving neoadjuvant radiochemotherapy. J Clin Oncol. (2017) 35(Suppl. 15):e15125 10.1200/JCO.2017.35.15_suppl.e15125
    1. Yang L, Wang Y, Shen L, Wan J, Deng W, Zhu J, et al. Predicting treatment outcome of rectal cancer patients underwent neoadjuvant chemoradiotherapy by ctDNA: The potential use of ctDNA monitoring as organ-sparing approach. J Clin Oncol. (2018) 36(Suppl. 15):3608 10.1200/JCO.2018.36.15_suppl.3608
    1. Benesova L, Belsanova B, Suchanek S, Kopeckova M, Minarikova P, Lipska L, et al. . Mutation-based detection and monitoring of cell-free tumor DNA in peripheral blood of cancer patients. Anal Biochem. (2013) 433:227–34. 10.1016/j.ab.2012.06.018
    1. Tie J, Cohen JD, Wang Y, Li L, Christie M, Simons K, et al. . Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: a prospective biomarker study. Gut. (2018) 68:663–71. 10.1136/gutjnl-2017-315852
    1. Hrebien S, Citi V, Garcia-Murillas I, Cutts R, Fenwick K, Kozarewa I, et al. . Early ctDNA dynamics as a surrogate for progression-free survival in advanced breast cancer in the BEECH trial. Ann Oncol. (2019) 30:945–52. 10.1093/annonc/mdz085
    1. Osumi H, Shinozaki E, Yamaguchi K, Zembutsu H. Early change in circulating tumor DNA as a potential predictor of response to chemotherapy in patients with metastatic colorectal cancer. Sci Rep. (2019) 9:17358. 10.1038/s41598-019-53711-3
    1. Reece M, Saluja H, Hollington P, Karapetis CS, Vatandoust S, Young GP, et al. . The use of circulating tumor DNA to monitor and predict response to treatment in colorectal cancer. Front Genet. (2019) 10:1118. 10.3389/fgene.2019.01118
    1. Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther. (2019) 20:1057–67. 10.1080/15384047.2019.1598759
    1. Vanpouille-Box C, Alard A, Aryankalayil MJ, Sarfraz Y, Diamond JM, Schneider RJ, et al. . DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun. (2017) 8:15618. 10.1038/ncomms15618
    1. Lyskjaer I, Kronborg CS, Rasmussen MH, Sorensen BS, Demuth C, Rosenkilde M, et al. . Correlation between early dynamics in circulating tumour DNA and outcome from FOLFIRI treatment in metastatic colorectal cancer. Sci Rep. (2019) 9:11542. 10.1038/s41598-019-47708-1
    1. Vymetalkova V, Cervena K, Bartu L, Vodicka P. Circulating cell-free DNA and colorectal cancer: a systematic review. Int J Mol Sci. (2018) 19:3356. 10.3390/ijms19113356
    1. Raja R, Kuziora M, Brohawn PZ, Higgs BW, Gupta A, Dennis PA, et al. . Early reduction in ctDNA predicts survival in patients with lung and bladder cancer treated with durvalumab. Clin Cancer Res. (2018) 24:6212–6222. 10.1158/1078-0432.CCR-18-0386
    1. Husain H, Melnikova VO, Kosco K, Woodward B, More S, Pingle SC, et al. . Monitoring daily dynamics of early tumor response to targeted therapy by detecting circulating tumor DNA in urine. Clin Cancer Res. (2017) 23:4716–4723. 10.1158/1078-0432.CCR-17-0454
    1. Khakoo S, Carter PD, Brown G, Valeri N, Picchia S, Bali MA, et al. . MRI tumor regression grade and circulating tumor DNA as complementary tools to assess response and guide therapy adaptation in rectal cancer. Clin Cancer Res. (2020) 26:183–92. 10.1158/1078-0432.CCR-19-1996

Source: PubMed

3
Předplatit