Persistence of ctDNA in Patients with Breast Cancer During Neoadjuvant Treatment Is a Significant Predictor of Poor Tumor Response

Qing Zhou, Simon P Gampenrieder, Sophie Frantal, Gabriel Rinnerthaler, Christian F Singer, Daniel Egle, Georg Pfeiler, Rupert Bartsch, Viktor Wette, Angelika Pichler, Edgar Petru, Peter C Dubsky, Zsuzsanna Bago-Horvath, Christian Fesl, Margaretha Rudas, Anders Ståhlberg, Ricarda Graf, Sabrina Weber, Nadia Dandachi, Martin Filipits, Michael Gnant, Marija Balic, Ellen Heitzer, Qing Zhou, Simon P Gampenrieder, Sophie Frantal, Gabriel Rinnerthaler, Christian F Singer, Daniel Egle, Georg Pfeiler, Rupert Bartsch, Viktor Wette, Angelika Pichler, Edgar Petru, Peter C Dubsky, Zsuzsanna Bago-Horvath, Christian Fesl, Margaretha Rudas, Anders Ståhlberg, Ricarda Graf, Sabrina Weber, Nadia Dandachi, Martin Filipits, Michael Gnant, Marija Balic, Ellen Heitzer

Abstract

Purpose: Accurate response assessment during neoadjuvant systemic treatment (NST) poses a clinical challenge. Therefore, a minimally invasive assessment of tumor response based on cell-free circulating tumor DNA (ctDNA) may be beneficial to guide treatment decisions.

Experimental design: We profiled 93 genes in tissue from 193 patients with early breast cancer. Patient-specific assays were designed for 145 patients to track ctDNA during NST in plasma. ctDNA presence and levels were correlated with complete pathological response (pCR) and residual cancer burden (RCB) as well as clinicopathologic characteristics of the tumor to identify potential proxies for ctDNA release.

Results: At baseline, ctDNA could be detected in 63/145 (43.4%) patients and persisted in 25/63 (39.7%) patients at mid-therapy (MT) and 15/63 (23.8%) patients at the end of treatment. ctDNA detection at MT was significantly associated with higher RCB (OR = 0.062; 95% CI, 0.01-0.48; P = 0.0077). Of 31 patients with detectable ctDNA at MT, 30 patients (96.8%) were nonresponders (RCB II, n = 8; RCB III, n = 22) and only one patient responded to the treatment (RCB I). Considering all 145 patients with baseline (BL) plasma, none of the patients with RCB 0 and only 6.7% of patients with RCB I had ctDNA detectable at MT, whereas 30.6% and 29.6% of patients with RCB II/III, respectively, had a positive ctDNA result.

Conclusions: Overall, our results demonstrate that the detection and persistence of ctDNA at MT may have the potential to negatively predict response to neoadjuvant treatment and identify patients who will not achieve pCR or be classified with RCB II/III.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Study design and flow chart and molecular profiling results. A, Shown is an overview of the study design. Patients with early breast cancer were treated with standard of care (SoC) NCT or NET. After mutation analysis of the primary tumors using a 93-gene panel, patient-specific high-resolution assays were designed to track ctDNA during treatment. B, Flow chart showing patients and samples evaluated in the study. NA, not available; QC, quality control; UV, variants with unknown clinical significance. Created with BioRender.com.
Figure 2.
Figure 2.
Somatic driver alterations in tumor and plasma. A, Heat map representing the landscape of somatic driver alterations, including mutations and CNAs in tumor samples. B, Heat map representing number of mutations detected by SiMSen-seq in all plasma samples (All), at BL, MT, and EOT. C, Time courses of exemplary patients with various numbers of screened mutations in plasma. Shown are the VAF in plasma at BL, MT, and EOT (left y-axis) and the VAFs of the mutations detected in matched tissue samples (right y-axis). LOD, limit of detection.
Figure 3.
Figure 3.
Predictive value of ctDNA. A, Flow chart depicting patients with (ctDNA+) and without (ctDNA−) detectable ctDNA at BL, MT, and EOT. NA, no plasma available. B, OR with 95% confidence intervals (CI) and P values for ctDNA detection at BL, MT, EOT, or all time points to predict tumor response (RCB 0/I vs. RCB II/III) calculated from univariate logistic regression models. C, Plotted are the fractions of patients stratified by response (RCB 0–III) with (ctDNA+) and without (ctDNA−) detectable ctDNA at MT. D, Detection rates of ctDNA at BL, MT, and EOT stratified by RCB scores (E) same as in B but for pCR. NA, not analyzable.
Figure 4.
Figure 4.
ctDNA levels and dynamics over the course of neoadjuvant therapy. A, Plotted are VAF distributions for three time points stratified by RCB (left plot). At BL, ctDNA levels were significantly elevated in patients with poor tumor responses (RCB II/III) compared with responders (RCB 0/I). For MT and the EOT, no association could be established due to a small number of positive samples for responders (right plot). B, Shown are patients with ctDNA data at BL and at least one additional time point stratified by tumor response. Top plot denotes responders, lower plot represents nonresponders. C, Waterfall plot of ctDNA changes (in percent) from BL to MT. *, Clearance of ctDNA at MT was considered as a 100% decrease; #, ctDNA detection at MT but not at BL was considered as a 100% increase.
Figure 5.
Figure 5.
Association between circulating tumor DNA and clinicopathologic characteristics. A, Association between ctDNA presence (ctDNA+) and absence (ctDNA−) at baseline and clinicopathologic characteristics. P values were calculated using Fisher exact test. B, Distribution of tumor size, Ki-67 scores, EndoPredict scores (MS, 12-gene molecular score; EPclin, combined clinic-molecular score), and stromal and intratumoral TILs in patients with (ctDNA+) and without (ctDNA−) detectable ctDNA at baseline.

References

    1. Early Breast Cancer Trialists' Collaborative G. Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol 2018;19:27–39.
    1. Heil J, Kuerer HM, Pfob A, Rauch G, Sinn HP, Golatta M, et al. . Eliminating the breast cancer surgery paradigm after neoadjuvant systemic therapy: current evidence and future challenges. Ann Oncol 2020;31:61–71.
    1. Burstein HJ, Curigliano G, Loibl S, Dubsky P, Gnant M, Poortmans P, et al. . Estimating the benefits of therapy for early-stage breast cancer: the St. Gallen International Consensus Guidelines for the primary therapy of early breast cancer 2019. Ann Oncol 2019;30:1541–57.
    1. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. . Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 2014;384:164–72.
    1. Saura C, Hlauschek D, Oliveira M, Zardavas D, Jallitsch-Halper A, de la Peña L, et al. . Neoadjuvant letrozole plus taselisib versus letrozole plus placebo in postmenopausal women with oestrogen receptor-positive, HER2-negative, early-stage breast cancer (LORELEI): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2019;20:1226–38.
    1. Marinovich ML, Houssami N, Macaskill P, von Minckwitz G, Blohmer J-U, Irwig L. Accuracy of ultrasound for predicting pathologic response during neoadjuvant therapy for breast cancer. Int J Cancer 2015;136:2730–7.
    1. Gampenrieder SP, Peer A, Weismann C, Meissnitzer M, Rinnerthaler G, Webhofer J, et al. . Radiologic complete response (rCR) in contrast-enhanced magnetic resonance imaging (CE-MRI) after neoadjuvant chemotherapy for early breast cancer predicts recurrence-free survival but not pathologic complete response (pCR). Breast Cancer Res 2019;21:19.
    1. van Loevezijn AA, van der Noordaa MEM, van Werkhoven ED, Loo CE, Winter-Warnars GAO, Wiersma T, et al. . Minimally invasive complete response assessment of the breast after neoadjuvant systemic therapy for early breast cancer (MICRA trial): Interim analysis of a multicenter observational cohort study. Ann Surg Oncol 2021;28:3243–53.
    1. Tasoulis MK, Lee H-B, Yang W, Pope R, Krishnamurthy S, Kim S-Y, et al. . Accuracy of post-neoadjuvant chemotherapy image-guided breast biopsy to predict residual cancer. JAMA Surg 2020;155:e204103.
    1. Heil J, Pfob A, Sinn HP, Rauch G, Bach P, Thomas B, et al. . Diagnosing pathologic complete response in the breast after neoadjuvant systemic treatment of breast cancer patients by minimal invasive biopsy: oral presentation at the San Antonio Breast Cancer Symposium on Friday, December 13, 2019, Program Number GS5–03. Ann Surg 2020.
    1. Alix-Panabieres C, Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov 2021;11:858–73,
    1. Ignatiadis M, Sledge GW, Jeffrey SS. Liquid biopsy enters the clinic: implementation issues and future challenges. Nat Rev Clin Oncol 2021;18:297–312.
    1. Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem 2015;61:112–23,
    1. Heitzer E, Haque IS, Roberts CES, Speicher MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet 2019;20:71–88.
    1. Garcia-Murillas I, Chopra N, Comino-Méndez I, Beaney M, Tovey H, Cutts RJ, et al. . Assessment of molecular relapse detection in early-stage breast cancer. JAMA Oncol 2019;5:1473–8.
    1. Butler TM, Boniface CT, Johnson-Camacho K, Tabatabaei S, Melendez D, Kelley T, et al. . Circulating tumor DNA dynamics using patient-customized assays are associated with outcome in neoadjuvantly treated breast cancer. Cold Spring Harb Mol Case Stud 2019;5:a003772.
    1. Riva F, Bidard F-C, Houy A, Saliou A, Madic J, Rampanou A, et al. . Patient-specific circulating tumor DNA detection during neoadjuvant chemotherapy in triple-negative breast cancer. Clin Chem 2017;63:691–9.
    1. Moss J, Zick A, Grinshpun A, Carmon E, Maoz M, Ochana BL, et al. . Circulating breast-derived DNA allows universal detection and monitoring of localized breast cancer. Ann Oncol 2020;31:395–403.
    1. Rothé F, Silva MJ, Venet D, Campbell C, Bradburry I, Rouas G, et al. . Circulating tumor DNA in HER2-amplified breast cancer: a translational research substudy of the NeoALTTO phase III trial. Clin Cancer Res 2019;25:3581–8.
    1. Magbanua MJM, Swigart LB, Wu H-T, Hirst GL, Yau C, Wolf DM, et al. . Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol 2021;32:229–39.
    1. Singer CF, Pfeiler G, Hubalek M, Bartsch R, Stöger H, Pichler A, et al. . Efficacy and safety of the therapeutic cancer vaccine tecemotide (L-BLP25) in early breast cancer: Results from a prospective, randomised, neoadjuvant phase II study (ABCSG 34). Eur J Cancer 2020;132:43–52,
    1. Bartsch R, Singer CF, Pfeiler G, Hubalek M, Stoeger H, Pichler A, et al. . Conventional versus reverse sequence of neoadjuvant epirubicin/cyclophosphamide and docetaxel: sequencing results from ABCSG-34. Br J Cancer 2021;124:1795–802,
    1. Yau C, van der Noordaa M, Wei J, et al. . Residual cancer burden after neoadjuvant therapy and long-term survival outcomes in breast cancer: a multi-center pooled analysis. Cancer Res 2020;80.
    1. Symmans WF, Wei C, Gould R, Yu X, Zhang Y, Liu M, et al. . Long-term prognostic risk after neoadjuvant chemotherapy associated with residual cancer burden and breast cancer subtype. J Clin Oncol 2017;35:1049–60.
    1. Dubsky PC, Singer CF, Egle D, Wette V, Petru E, Balic M, et al. . The EndoPredict score predicts response to neoadjuvant chemotherapy and neoendocrine therapy in hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer patients from the ABCSG-34 trial. Eur J Cancer 2020;134:99–106.
    1. Denkert C, von Minckwitz G, Darb-Esfahani S, Lederer B, Heppner BI, Weber KE, et al. . Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 2018;19:40–50.
    1. Denkert C, Wienert S, Poterie A, Loibl S, Budczies J, Badve S, et al. . Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: results of the ring studies of the international immuno-oncology biomarker working group. Mod Pathol 2016;29:1155–64.
    1. Denkert C, Kronenwett R, Schlake W, Bohmann K, Penzel R, Weber KE, et al. . Decentral gene expression analysis for ER+/Her2- breast cancer: results of a proficiency testing program for the EndoPredict assay. Virchows Arch 2012;460:251–9.
    1. Kronenwett R, Bohmann K, Prinzler J, Sinn BV, Haufe F, Roth C, et al. . Decentral gene expression analysis: analytical validation of the Endopredict genomic multianalyte breast cancer prognosis test. BMC Cancer 2012;12:456.
    1. Filipits M, Rudas M, Jakesz R, Dubsky P, Fitzal F, Singer CF, et al. . A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res 2011;17:6012–20.
    1. Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. . Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol 2007;25:4414–22.
    1. Heitzer E, Ulz P, Belic J, Gutschi S, Quehenberger F, Fischereder K, et al. . Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med 2013;5:30.
    1. Suppan C, Brcic I, Tiran V, Mueller HD, Posch F, Auer M, et al. . Untargeted assessment of tumor fractions in plasma for monitoring and prognostication from metastatic breast cancer patients undergoing systemic treatment. Cancers (Basel) 2019;11:1171.
    1. Xu C, Ranjbar MRN, Wu Z, DiCarlo J, Wang Y. Detecting very low allele fraction variants using targeted DNA sequencing and a novel molecular barcode-aware variant caller. BMC Genomics 2017;18:5.
    1. Ulz P, Belic J, Graf R, Auer M, Lafer I, Fischereder K, et al. . Whole-genome plasma sequencing reveals focal amplifications as a driving force in metastatic prostate cancer. Nat Commun 2016;7:12008.
    1. Zhou Q, Perakis SO, Ulz P, Mohan S, Riedl JM, Talakic E, et al. . Cell-free DNA analysis reveals POLR1D-mediated resistance to bevacizumab in colorectal cancer. Genome Med 2020;12:20.
    1. Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. . Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 2017;8:1324.
    1. Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol 2011;12:R41.
    1. Mohan S, Heitzer E, Ulz P, Lafer I, Lax S, Auer M, et al. . Changes in colorectal carcinoma genomes under anti-EGFR therapy identified by whole-genome plasma DNA sequencing. PLoS Genet 2014;10:e1004271.
    1. Unseld M, Belic J, Pierer K, Zhou Q, Moser T, Bauer R, et al. . A higher ctDNA fraction decreases survival in regorafenib-treated metastatic colorectal cancer patients. Results from the regorafenib's liquid biopsy translational biomarker phase II pilot study. Int J Cancer 2021;148:1452–61.
    1. Ståhlberg A, Krzyzanowski PM, Egyud M, Filges S, Stein L, Godfrey TE. Simple multiplexed PCR-based barcoding of DNA for ultrasensitive mutation detection by next-generation sequencing. Nat Protoc 2017;12:664–82.
    1. Ståhlberg A, Krzyzanowski PM, Jackson JB, Egyud M, Stein L, Godfrey TE. Simple, multiplexed, PCR-based barcoding of DNA enables sensitive mutation detection in liquid biopsies using sequencing. Nucleic Acids Res 2016;44:e105.
    1. Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61–70.
    1. Aftimos P, Oliveira M, Irrthum A, Fumagalli D, Sotiriou C, Gal-Yam EN, et al. . Genomic and transcriptomic analyses of breast cancer primaries and matched metastases in AURORA, the Breast International Group (BIG) molecular screening initiative. Cancer Discov 2021;11:2796–811.
    1. Luen SJ, Asher R, Lee CK, Savas P, Kammler R, Dell'Orto P, et al. . Association of somatic driver alterations with prognosis in postmenopausal, hormone receptor-positive, HER2-negative early breast cancer: a secondary analysis of the BIG 1–98 randomized clinical trial. JAMA Oncol 2018;4:1335–43.
    1. Masuda N, Lee S-J, Ohtani S, Im Y-H, Lee E-S, Yokota I, et al. . Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med 2017;376:2147–59.
    1. von Minckwitz G, Huang C-S, Mano MS, Loibl S, Mamounas EP, Untch M, et al. . Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med 2019;380:617–28.
    1. Prowell TM, Pazdur R. Pathological complete response and accelerated drug approval in early breast cancer. N Engl J Med 2012;366:2438–41,
    1. Suppan C, Posch F, Mueller HD, Mischitz N, Steiner D, Klocker EV, et al. . Patterns of recurrence after neoadjuvant therapy in early breast cancer, according to the residual cancer burden index and reductions in neoadjuvant treatment intensity. Cancers (Basel) 2021;13:2492.
    1. Müller HD, Posch F, Suppan C, Bargfrieder U, Gumpoldsberger M, Hammer R, et al. . Validation of residual cancer burden as prognostic factor for breast cancer patients after neoadjuvant therapy. Ann Surg Oncol 2019;26:4274–83.
    1. Li X, Wang M, Wang M, Yu X, Guo J, Sun T, et al. . Predictive and prognostic roles of pathological indicators for patients with breast cancer on neoadjuvant chemotherapy. J Breast Cancer 2019;22:497–521.
    1. Asano Y, Kashiwagi S, Goto W, Takada K, Takahashi K, Hatano T, et al. . Prediction of survival after neoadjuvant chemotherapy for breast cancer by evaluation of tumor-infiltrating lymphocytes and residual cancer burden. BMC Cancer 2017;17:888.
    1. Luen SJ, Salgado R, Dieci MV, Vingiani A, Curigliano G, Gould RE, et al. . Prognostic implications of residual disease tumor-infiltrating lymphocytes and residual cancer burden in triple-negative breast cancer patients after neoadjuvant chemotherapy. Ann Oncol 2019;30:236–42.
    1. Sheri A, Smith IE, Johnston SR, A'Hern R, Nerurkar A, Jones RL, et al. . Residual proliferative cancer burden to predict long-term outcome following neoadjuvant chemotherapy. Ann Oncol 2015;26:75–80,
    1. McDonald BR, Contente-Cuomo T, Sammut S-J, Odenheimer-Bergman A, Ernst B, Perdigones N, et al. . Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci Transl Med 2019;11:eaax7392,
    1. Zviran A, Schulman RC, Shah M, Hill STK, Deochand S, Khamnei CC, et al. . Genome-wide cell-free DNA mutational integration enables ultra-sensitive cancer monitoring. Nat Med 2020;26:1114–24,
    1. Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, et al. . Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446–51,
    1. Chen M-B, Zhu Y-Q, Xu J-Y, Wang L-Q, Liu C-Y, Ji Z-Y, et al. . Value of TP53 status for predicting response to neoadjuvant chemotherapy in breast cancer: a meta-analysis. PLoS One 2012;7:e39655,
    1. Loibl S, Majewski I, Guarneri V, Nekljudova V, Holmes E, Bria E, et al. . PIK3CA mutations are associated with reduced pathological complete response rates in primary HER2-positive breast cancer: pooled analysis of 967 patients from five prospective trials investigating lapatinib and trastuzumab. Ann Oncol 2019;30:1180,

Source: PubMed

3
Subscribe