Circulating tumor DNA and magnetic resonance imaging to predict neoadjuvant chemotherapy response and recurrence risk

Mark Jesus M Magbanua, Wen Li, Denise M Wolf, Christina Yau, Gillian L Hirst, Lamorna Brown Swigart, David C Newitt, Jessica Gibbs, Amy L Delson, Ekaterina Kalashnikova, Alexey Aleshin, Bernhard Zimmermann, A Jo Chien, Debu Tripathy, Laura Esserman, Nola Hylton, Laura van 't Veer, Mark Jesus M Magbanua, Wen Li, Denise M Wolf, Christina Yau, Gillian L Hirst, Lamorna Brown Swigart, David C Newitt, Jessica Gibbs, Amy L Delson, Ekaterina Kalashnikova, Alexey Aleshin, Bernhard Zimmermann, A Jo Chien, Debu Tripathy, Laura Esserman, Nola Hylton, Laura van 't Veer

Abstract

We investigated whether serial measurements of circulating tumor DNA (ctDNA) and functional tumor volume (FTV) by magnetic resonance imaging (MRI) can be combined to improve prediction of pathologic complete response (pCR) and estimation of recurrence risk in early breast cancer patients treated with neoadjuvant chemotherapy (NAC). We examined correlations between ctDNA and FTV, evaluated the additive value of ctDNA to FTV-based predictors of pCR using area under the curve (AUC) analysis, and analyzed the impact of FTV and ctDNA on distant recurrence-free survival (DRFS) using Cox regressions. The levels of ctDNA (mean tumor molecules/mL plasma) were significantly correlated with FTV at all time points (p < 0.05). Median FTV in ctDNA-positive patients was significantly higher compared to those who were ctDNA-negative (p < 0.05). FTV and ctDNA trajectories in individual patients showed a general decrease during NAC. Exploratory analysis showed that adding ctDNA information early during treatment to FTV-based predictors resulted in numerical but not statistically significant improvements in performance for pCR prediction (e.g., AUC 0.59 vs. 0.69, p = 0.25). In contrast, ctDNA-positivity after NAC provided significant additive value to FTV in identifying patients with increased risk of metastatic recurrence and death (p = 0.004). In this pilot study, we demonstrate that ctDNA and FTV were correlated measures of tumor burden. Our preliminary findings based on a limited cohort suggest that ctDNA at surgery improves FTV as a predictor of metastatic recurrence and death. Validation in larger studies is warranted.

Conflict of interest statement

The following authors are employees of Natera, Inc. (E.K., A.A. and B.Z.). L.V.V. is co-founder, stockholder and part-time employee of Agendia NV. The rest of the authors declare no potential conflicts of interest.

Figures

Fig. 1. Pretreatment functional tumor volume (FTV)…
Fig. 1. Pretreatment functional tumor volume (FTV) and association with clinicopathologic characteristics.
P values were calculated using Mann–Whitney (2 groups) or Kruskal–Wallis test (3 groups). For each box plot, the center line, the boundaries of the box, the ends of the whiskers and points beyond the whiskers represent the median value, the interquartile range, the minimum and maximum values, and the outliers, respectively.
Fig. 2. Relationship between ctDNA and FTV…
Fig. 2. Relationship between ctDNA and FTV at different time points.
a Scatterplot for all patients with paired data at different time points to show correlation between ctDNA and MRI-based FTV as measures of tumor burden. b ctDNA-positivity and tumor burden by MRI-based FTV. Box plots showing distribution of FTV according to ctDNA status (ctDNA-positive or ctDNA-negative) at different time points. T0: pretreatment; T1: 3 weeks after initiation of paclitaxel-based treatment; T2: between paclitaxel and anthracycline regimens; T3: after NAC prior to surgery. For each box plot, the center line, the boundaries of the box, the ends of the whiskers and points beyond the whiskers represent the median value, the interquartile range, the minimum and maximum values, and the outliers, respectively.
Fig. 3. Correlation between ctDNA and MRI-based…
Fig. 3. Correlation between ctDNA and MRI-based FTV dynamics during NAC.
a Mean levels of ctDNA (mean tumor molecules per mL plasma, MTM) and FTV (cm3) across time points. The error bars represent standard deviation. The vertical lines represent standard deviation. b Schematic for calculating the mean Pearson correlation for actual data and means for simulated datasets using Monte Carlo method. The right panel is histogram showing the distribution of simulated means of Fisher z-transformed Pearson’s correlation coefficient. The red line corresponds to the mean Fisher z-transformed Pearson’s correlation coefficient calculated based on the actual data. c Plots showing ctDNA (mean tumor molecules per mL plasma, MTM) and FTV (cm3) levels across time points in 4 representative cases. Unfilled dot at T3 in patient 4 indicates ctDNA below the limit of detection. T0: pretreatment; T1: 3 weeks after initiation of paclitaxel-based treatment; T2: between paclitaxel and anthracycline regimens; T3: after NAC prior to surgery.
Fig. 4. Additive value of ctDNA to…
Fig. 4. Additive value of ctDNA to FTV-based predictors of pCR at T1.
a Area under the curve (AUC) for prediction of pCR early during NAC (T1: 3 weeks after initiation treatment) for FTV alone and FTV + ctDNA treated as a continuous variable. b Positive predictive value (PPV), negative predictive value (NPV) and accuracy for prediction of pCR of FTV alone and FTV + ctDNA treated as a dichotomous variable.
Fig. 5. Study schema showing time points…
Fig. 5. Study schema showing time points for collection of MRI-based functional tumor volume (FTV) and circulating tumor DNA (ctDNA) data during neoadjuvant chemotherapy (NAC).
Patients received paclitaxel-based treatment followed by anthracycline-based chemotherapy. Data and biospecimens were collected at pretreatment (T0), 3 weeks after initiation of paclitaxel-based treatment (T1), between paclitaxel and anthracycline regimens (T2), and after NAC prior to surgery (T3). Pathologic complete response (pCR), the primary endpoint of this study, was assessed at surgical time point.

References

    1. Wang M, et al. Neoadjuvant chemotherapy creates surgery opportunities for inoperable locally advanced breast cancer. Sci. Rep. 2017;7:44673. doi: 10.1038/srep44673.
    1. Tromberg BJ, et al. Predicting responses to neoadjuvant chemotherapy in breast cancer: ACRIN 6691 trial of diffuse optical spectroscopic imaging. Cancer Res. 2016;76:5933–5944. doi: 10.1158/0008-5472.CAN-16-0346.
    1. DeMichele A, et al. The neoadjuvant model is still the future for drug development in breast cancer. Clin. Cancer Res. 2015;21:2911–2915. doi: 10.1158/1078-0432.CCR-14-1760.
    1. Berry DA, Hudis CA. Neoadjuvant therapy in breast cancer as a basis for drug approval. JAMA Oncol. 2015;1:875–876. doi: 10.1001/jamaoncol.2015.1293.
    1. Cortazar P, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:164–172. doi: 10.1016/S0140-6736(13)62422-8.
    1. Spring LM, et al. Pathologic complete response after neoadjuvant chemotherapy and impact on breast cancer recurrence and survival: a comprehensive meta-analysis. Clin. Cancer Res. 2020;26:2838–2848. doi: 10.1158/1078-0432.CCR-19-3492.
    1. Consortium, I. S. T. et al. Association of event-free and distant recurrence-free survival with individual-level pathologic complete response in neoadjuvant treatment of stages 2 and 3 breast cancer: three-year follow-up analysis for the I-SPY2 adaptively randomized clinical trial. JAMA Oncol., 10.1001/jamaoncol.2020.2535 (2020).
    1. Caparica R, et al. Post-neoadjuvant treatment and the management of residual disease in breast cancer: state of the art and perspectives. Ther. Adv. Med. Oncol. 2019;11:1758835919827714. doi: 10.1177/1758835919827714.
    1. Reig, B., Heacock, L., Lewin, A., Cho, N. & Moy, L. Role of MRI to assess response to neoadjuvant therapy for breast cancer. J. Magn. Reson. Imaging, 10.1002/jmri.27145 (2020).
    1. Hylton N. MR imaging for the prediction of breast cancer response to neoadjuvant chemotherapy. Radiology. 2013;266:367.
    1. Hylton NM, et al. Neoadjuvant chemotherapy for breast cancer: functional tumor volume by mr imaging predicts recurrence-free survival-results from the ACRIN 6657/CALGB 150007 I-SPY 1 trial. Radiology. 2016;279:44–55. doi: 10.1148/radiol.2015150013.
    1. Li W, et al. Predicting breast cancer response to neoadjuvant treatment using multi-feature MRI: results from the I-SPY 2 TRIAL. npj Breast Cancer. 2020;6:63. doi: 10.1038/s41523-020-00203-7.
    1. Esserman, L. et al. Platform trial designs in drug development: umbrella trials and basket trials (eds Zoran Antonijevic & Robert A. Beckman) 3-22 (Chapman and Hall/CRC, 2018).
    1. Wolf DM, et al. DNA repair deficiency biomarkers and the 70-gene ultra-high risk signature as predictors of veliparib/carboplatin response in the I-SPY 2 breast cancer trial. NPJ Breast Cancer. 2017;3:31. doi: 10.1038/s41523-017-0025-7.
    1. Wolf DM, et al. Mechanism of action biomarkers predicting response to AKT inhibition in the I-SPY 2 breast cancer trial. NPJ Breast Cancer. 2020;6:48. doi: 10.1038/s41523-020-00189-2.
    1. Magbanua MJM, et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann. Oncol. 2021;32:229–239. doi: 10.1016/j.annonc.2020.11.007.
    1. Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017;14:531–548. doi: 10.1038/nrclinonc.2017.14.
    1. Donaldson J, Park BH. Circulating tumor DNA: measurement and clinical utility. Annu Rev. Med. 2018;69:223–234. doi: 10.1146/annurev-med-041316-085721.
    1. McDonald, B. R. et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci. Transl. Med.11, 10.1126/scitranslmed.aax7392 (2019).
    1. Rothe, F. et al. Circulating tumor DNA in HER2 amplified breast cancer: a translational research substudy of the NeoALTTO phase 3 trial. Clin. Cancer Res., 10.1158/1078-0432.CCR-18-2521 (2019).
    1. Riva F, et al. Patient-specific circulating tumor DNA detection during neoadjuvant chemotherapy in triple-negative breast cancer. Clin. Chem. 2017;63:691–699. doi: 10.1373/clinchem.2016.262337.
    1. Hylton NM, et al. Locally advanced breast cancer: MR imaging for prediction of response to neoadjuvant chemotherapy–results from ACRIN 6657/I-SPY trial. Radiology. 2012;263:663–672. doi: 10.1148/radiol.12110748.
    1. Li W, et al. Additive value of diffusion-weighted MRI in the I-SPY 2 TRIAL. J. Magn. Reson. Imaging. 2019;50:1742–1753. doi: 10.1002/jmri.26770.
    1. Abbosh C, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature. 2017;545:446–451. doi: 10.1038/nature22364.
    1. Christensen, E. et al. Early detection of metastatic relapse and monitoring of therapeutic efficacy by ultra-deep sequencing of plasma cell-free DNA in patients with urothelial bladder carcinoma. J. Clin. Oncol., JCO1802052, 10.1200/JCO.18.02052 (2019).
    1. Reinert, T. et al. Analysis of plasma cell-free DNA by Ultradeep Sequencing in Patients With Stages I to III Colorectal Cancer. JAMA Oncol., 10.1001/jamaoncol.2019.0528 (2019).
    1. Coombes, R. C. et al. Personalized detection of circulating tumor DNA antedates breast cancer metastatic recurrence. Clin Cancer Res., 10.1158/1078-0432.CCR-18-3663 (2019).
    1. Phallen, J. et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med.9, 10.1126/scitranslmed.aan2415 (2017).
    1. Rodriguez, B. J. et al. Detection of TP53 and PIK3CA mutations in circulating tumor DNA using next-generation sequencing in the screening process for early breast cancer diagnosis. J. Clin. Med.8, 10.3390/jcm8081183 (2019).
    1. Rossi G, et al. Cell-free DNA and circulating tumor cells: comprehensive liquid biopsy analysis in advanced breast cancer. Clin. Cancer Res. 2018;24:560–568. doi: 10.1158/1078-0432.CCR-17-2092.
    1. Newman AM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 2016;34:547–555. doi: 10.1038/nbt.3520.
    1. Partridge SC, et al. Diffusion-weighted MRI findings predict pathologic response in neoadjuvant treatment of breast cancer: the ACRIN 6698 multicenter trial. Radiology. 2018;289:618–627. doi: 10.1148/radiol.2018180273.
    1. Chien AJ, et al. MK-2206 and standard neoadjuvant chemotherapy improves response in patients with human epidermal growth factor receptor 2-positive and/or hormone receptor-negative breast cancers in the I-SPY 2 trial. J. Clin. Oncol. 2020;38:1059–1069. doi: 10.1200/JCO.19.01027.
    1. Sauerbrei W, Taube SE, McShane LM, Cavenagh MM, Altman DG. Reporting recommendations for tumor marker prognostic studies (REMARK): an abridged explanation and elaboration. J. Natl Cancer Inst. 2018;110:803–811. doi: 10.1093/jnci/djy088.
    1. Gorsuch RL, Lehmann CS. Correlation coefficients: mean bias and confidence interval distortions. J. Methods Meas. Soc. Sci. 2010;1:52–65. doi: 10.2458/jmm.v1i2.114.
    1. Grambsch P, Therneau T. Proportional hazards tests and diagnostics based on weighted residuals. Biometrika. 1994;81:515–526. doi: 10.1093/biomet/81.3.515.
    1. Akaike, H. in Selected papers of Hirotugu Akaike (eds Emanuel Parzen, Kunio Tanabe, & Genshiro Kitagawa) 199-213 (Springer New York, 1998).
    1. Magbanua, M. J. M. et al. Metadata record for the manuscript: Circulating tumor DNA and magnetic resonance imaging to predict neoadjuvant chemotherapy response and recurrence risk. figshare10.6084/m9.figshare.14075876 (2021).
    1. Wolf, D. M. et al. Gene expression omnibus. figshare (2020).
    1. Li, W. et al. Metadata record for the manuscript: predicting breast cancer response to neoadjuvant treatment using multi-feature MRI: results from the I-SPY 2 TRIAL. figshare10.6084/m9.figshare.12912191.v1 (2020).

Source: PubMed

3
Předplatit