Detection of circulating tumor cells and circulating tumor DNA before and after mammographic breast compression in a cohort of breast cancer patients scheduled for neoadjuvant treatment

Daniel Förnvik, Kristina E Aaltonen, Yilun Chen, Anthony M George, Christian Brueffer, Robert Rigo, Niklas Loman, Lao H Saal, Lisa Rydén, Daniel Förnvik, Kristina E Aaltonen, Yilun Chen, Anthony M George, Christian Brueffer, Robert Rigo, Niklas Loman, Lao H Saal, Lisa Rydén

Abstract

Purpose: It is not known if mammographic breast compression of a primary tumor causes shedding of tumor cells into the circulatory system. Little is known about how the detection of circulating biomarkers such as circulating tumor cells (CTCs) or circulating tumor DNA (ctDNA) is affected by breast compression intervention.

Methods: CTCs and ctDNA were analyzed in blood samples collected before and after breast compression in 31 patients with primary breast cancer scheduled for neoadjuvant therapy. All patients had a central venous access to allow administration of intravenous neoadjuvant chemotherapy, which enabled blood collection from superior vena cava, draining the breasts, in addition to sampling from a peripheral vein.

Results: CTC and ctDNA positivity was seen in 26% and 65% of the patients, respectively. There was a significant increase of ctDNA after breast compression in central blood (p = 0.01), not observed in peripheral testing. No increase related with breast compression was observed for CTC. ctDNA positivity was associated with older age (p = 0.05), and ctDNA increase after breast compression was associated with high Ki67 proliferating tumors (p = 0.04). CTCs were more abundant in central compared to peripheral blood samples (p = 0.04).

Conclusions: There was no significant release of CTCs after mammographic breast compression but more CTCs were present in central compared to peripheral blood. No significant difference between central and peripheral levels of ctDNA was observed. The small average increase in ctDNA after breast compression is unlikely to be clinically relevant. The results give support for mammography as a safe procedure from the point of view of CTC and ctDNA shedding to the blood circulation. The results may have implications for the standardization of sampling procedures for circulating tumor markers.

Trial registration: ClinicalTrials.gov NCT02306096.

Keywords: Breast cancer; Breast compression; Circulating tumor DNA; Circulating tumor cells; Mammography; Neoadjuvant.

Conflict of interest statement

YC, AMG, CB, RR, and LHS are shareholders and employees of SAGA Diagnostics AB. LHS has received honorarium from Novartis AG. All remaining authors have declared no conflicts of interest.

Figures

Fig. 1
Fig. 1
Examples of CTCs detected with the CellSearch system from a patient in the study
Fig. 2
Fig. 2
The number of CTCs found before and after mammographic breast compression in central venous access (a), where two patients are represented by a line going from 0 to 1 CTC and from 1 to 0 CTCs, respectively. The corresponding number of CTCs before and after compression in peripheral blood (b). Figures for mutant allele frequency before and after breast compression in central (c), where two patients are represented by a line from 0 to approximately 0.35, and peripheral (d) plasma, where two patients are represented by a line going from 0 to approximately 0.04 and from approximately 0.03 to 0, respectively. Patients with an increasing value are plotted with red lines, decreasing values in blue, and constant values in green. All patients that did not have any circulating tumor markers are summarized in one line at number/frequency = 0
Fig. 3
Fig. 3
CTC (a) and ctDNA (b) detection in central and peripheral sample pairs. In 8/10 CTC-positive pairwise samples, a higher number of CTCs was detected in the central compared to the peripheral blood sample (p = 0.04). In pairwise samples 1–6, no CTCs were found in the peripheral blood sample. In 12/20 ctDNA pairwise samples, a higher mutant allele fraction was found in the peripheral plasma sample (p = 0.50). In pairwise samples 2, 4–7, no ctDNA was detected centrally, and in sample 1, no ctDNA was detected peripherally

References

    1. Janni WJ, Rack B, Terstappen LW, et al. Pooled analysis of the prognostic relevance of circulating tumor cells in primary breast cancer. Clin Cancer Res. 2016;22:2583–2593. doi: 10.1158/1078-0432.CCR-15-1603.
    1. Bidard FC, Michiels S, Riethdorf S, et al. Circulating tumor cells in breast cancer patients treated by neoadjuvant chemotherapy: a meta-analysis. J Natl Cancer Inst. 2018;110:560–567. doi: 10.1093/jnci/djy018.
    1. Bardelli A, Pantel K. Liquid biopsies, what we do not know (yet) Cancer Cell. 2017;31:172–179. doi: 10.1016/j.ccell.2017.01.002.
    1. Yan WT, Cui X, Chen Q, et al. Circulating tumor cell status monitors the treatment responses in breast cancer patients: a meta-analysis. Sci Rep. 2017;7:43464. doi: 10.1038/srep43464.
    1. Tibbe AG, Miller MC, Terstappen LW. Statistical considerations for enumeration of circulating tumor cells. Cytometry A. 2007;71:154–162. doi: 10.1002/cyto.a.20369.
    1. Wan JC, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–238. doi: 10.1038/nrc.2017.7.
    1. Beddowes E, Sammut SJ, Gao M, Caldas C. Predicting treatment resistance and relapse through circulating DNA. Breast. 2017;34:S31–S35. doi: 10.1016/j.breast.2017.06.024.
    1. Olsson E, Winter C, George A, et al. Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med. 2015;7:1034–1047. doi: 10.15252/emmm.201404913.
    1. Murtaza M, Dawson SJ, Tsui DW, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112. doi: 10.1038/nature12065.
    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;135:2215–2222. doi: 10.1002/ijc.28863.
    1. Scholer LV, Reinert T, Orntoft MW, et al. Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin Cancer Res. 2017;23:5437–5445. doi: 10.1158/1078-0432.CCR-17-0510.
    1. Loman N, Saal LH. The state of the art in prediction of breast cancer relapse using cell-free circulating tumor DNA liquid biopsies. Ann Transl Med. 2016;4:S68. doi: 10.21037/atm.2016.10.58.
    1. Sandri MT, Zorzino L, Cassatella MC, et al. Changes in circulating tumor cell detection in patients with localized breast cancer before and after surgery. Ann Surg Oncol. 2010;17:1539–1545. doi: 10.1245/s10434-010-0918-2.
    1. Papavasiliou P, Fisher T, Kuhn J, et al. Circulating tumor cells in patients undergoing surgery for hepatic metastases from colorectal cancer. Proc (Bayl Univ Med Cent) 2010;23:11–14. doi: 10.1080/08998280.2010.11928572.
    1. Koch M, Kienle P, Hinz U, et al. Detection of hematogenous tumor cell dissemination predicts tumor relapse in patients undergoing surgical resection of colorectal liver metastases. Ann Surg. 2005;241:199–205. doi: 10.1097/01.sla.0000151795.15068.27.
    1. van Dalum G, van der Stam GJ, Tibbe AG, et al. Circulating tumor cells before and during follow-up after breast cancer surgery. Int J Oncol. 2015;46:407–413. doi: 10.3892/ijo.2014.2694.
    1. Hashimoto M, Tanaka F, Yoneda K, et al. Significant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer. Interact Cardiovasc Thorac Surg. 2014;18:775–783. doi: 10.1093/icvts/ivu048.
    1. Martin OA, Anderson RL, Narayan K, MacManus MP. Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Nat Rev Clin Oncol. 2017;14:32–44. doi: 10.1038/nrclinonc.2016.128.
    1. Juratli MA, Sarimollaoglu M, Siegel ER, et al. Real-time monitoring of circulating tumor cell release during tumor manipulation using in vivo photoacoustic and fluorescent flow cytometry. Head Neck. 2014;36:1207–1215. doi: 10.1002/hed.23439.
    1. Juratli MA, Siegel ER, Nedosekin DA, et al. In vivo long-term monitoring of circulating tumor cells fluctuation during medical interventions. PLoS ONE. 2015;10:e0137613. doi: 10.1371/journal.pone.0137613.
    1. Nishizaki T, Matsumata T, Kanematsu T, et al. Surgical manipulation of VX2 carcinoma in the rabbit liver evokes enhancement of metastasis. J Surg Res. 1990;49:92–97. doi: 10.1016/0022-4804(90)90116-J.
    1. Förnvik D, Andersson I, Dustler M, et al. No evidence for shedding of circulating tumor cells to the peripheral venous blood as a result of mammographic breast compression. Breast Cancer Res Treat. 2013;141:187–195. doi: 10.1007/s10549-013-2674-z.
    1. Peeters DJE, Van den Eynden GG, van Dam PJ, et al. Circulating tumour cells in the central and the peripheral venous compartment in patients with metastatic breast cancer. Br J Cancer. 2011;104:1472–1477. doi: 10.1038/bjc.2011.122.
    1. Jiao LR, Apostolopoulos C, Jacob J, et al. Unique localization of circulating tumor cells in patients with hepatic metastases. J Clin Oncol. 2009;27:6160–6165. doi: 10.1200/JCO.2009.24.5837.
    1. Deneve E, Riethdorf S, Ramos J, et al. Capture of viable circulating tumor cells in the liver of colorectal cancer patients. Clin Chem. 2013;59:1384–1392. doi: 10.1373/clinchem.2013.202846.
    1. Reddy RM, Murlidhar V, Zhao L, et al. Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J Thorac Cardiovasc Surg. 2016;151:852–857. doi: 10.1016/j.jtcvs.2015.09.126.
    1. Mizuno N, Kato Y, Izumi Y, et al. Importance of hepatic first-pass removal in metastasis of colon carcinoma cells. J Hepatol. 1998;28:865–877. doi: 10.1016/S0168-8278(98)80238-9.
    1. Peeters DJ, Brouwer A, Van den Eynden GG, et al. Circulating tumour cells and lung microvascular tumour cell retention in patients with metastatic breast and cervical cancer. Cancer Lett. 2015;356:872–879. doi: 10.1016/j.canlet.2014.10.039.
    1. Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199–1209. doi: 10.1056/NEJMoa1213261.
    1. Shaw JA, Guttery DS, Hills A, et al. Mutation analysis of cell-free DNA and single circulating tumor cells in metastatic breast cancer patients with high CTC counts. Clin Cancer Res. 2016;23:88–96. doi: 10.1158/1078-0432.CCR-16-0825.
    1. Saal LH, Vallon-Christersson J, Hakkinen J, et al. The Sweden cancerome analysis network-breast (SCAN-B) initiative: a large-scale multicenter infrastructure towards implementation of breast cancer genomic analyses in the clinical routine. Genome Med. 2015;7:20. doi: 10.1186/s13073-015-0131-9.
    1. Rydén L, Loman N, Larsson C, et al. Minimizing inequality in access to precision medicine in breast cancer by real-time population-based molecular analysis in the SCAN-B initiative. Br J Surg. 2018;105:e158–e168. doi: 10.1002/bjs.10741.
    1. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–791. doi: 10.1056/NEJMoa040766.
    1. Brueffer C, Vallon-Christersson J, Grabau D, et al. Clinical value of rna sequencing-based classifiers for prediction of the five conventional breast cancer biomarkers: a report from the population-based multicenter Sweden cancerome analysis network—breast initiative. JCO Precis Oncol. 2018;2:1–18. doi: 10.1200/PO.18.00183.
    1. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12:357–360. doi: 10.1038/nmeth.3317.
    1. Lai Z, Markovets A, Ahdesmaki M, et al. VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research. Nucleic Acids Res. 2016;44:e108. doi: 10.1093/nar/gkw227.
    1. Pedersen BS, Layer RM, Quinlan AR. Vcfanno: fast, flexible annotation of genetic variants. Genome Biol. 2016;17:118. doi: 10.1186/s13059-016-0973-5.

Source: PubMed

3
Abonnieren