A novel liquid biopsy assay for detection of ERBB2 (HER2) amplification in circulating tumor cells (CTCs)

Authors

DOI:

https://doi.org/10.33393/jcb.2024.3046

Keywords:

Analytical validation, Breast cancer, Circulating tumor cells (CTCs), Epic CTC Platform, HER2, Liquid biopsy

Abstract

Purpose: Circulating tumor cell (CTC)-based ERBB2 (HER2) assay is a laboratory test developed by Epic Sciences using single-cell genomics to detect ERBB2 (HER2) amplification in CTCs found in the peripheral blood of metastatic breast cancer (MBC) patients.

Patients and methods: Peripheral blood was collected in Streck tubes and centrifugation was used to remove plasma and red blood cells. The remaining nucleated cells were deposited on glass slides, immunofluorescent-stained with proprietary antibodies, scanned by a high-definition digital scanner, and analyzed by a proprietary algorithm. In addition, single-cell genomics was performed on selected CTC. Analytical validation was performed using white blood cells from healthy donors and breast cancer cell lines with known levels of ERBB2 amplification. Clinical concordance was assessed on MBC patients whose blood was tested by the CTC ERBB2 (HER2) assay and those results are compared to results of matched metastatic tissue biopsy (immunohistochemistry [IHC] 3+ or IHC2+/in situ hybridization [ISH+]).

Results: Epic’s ERBB2 (HER2) assay detected 2-fold ERBB2 amplification with 85% sensitivity and 94% specificity. In the clinical concordance study, among the 50% of the cases that had ERBB2 status results from CTCs found to be chromosomally-unstable, the CTC ERBB2 (HER2) assay showed sensitivity of 69% and specificity of 78% when compared to HER2 status by metastatic tissue biopsy.

Conclusions: The CTC ERBB2 (HER2) assay can consistently detect ERBB2 status in MBC cell lines and in the population of patients with MBC with detectable chromosomally unstable CTCs for whom tissue biopsy is not available or is infeasible.

Downloads

Download data is not yet available.

References

Carey LA, Perou CM, Livasy CA, et al. Race, breast cancer subtypes, and survival in the Carolina breast cancer study. JAMA. 2006;295(21):2492-2502. doi:10.1001/jama.295.21.2492

Martínez-Sáez O, Prat A. Current and future management of HER2-positive metastatic breast cancer. JCO Oncol Pract. 2021;17(10):594-604. doi: 10.1200/OP.21.00172

Wolff AC, Hammond MEH, Hicks DG, et al; American Society of Clinical Oncology; College of American Pathologists. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013;31(31):3997-4013. https://doi.org/10.1200/JCO.2013.50.9984 PMID:24101045

Gradishar WJ, Moran MS, Abraham J, et al. Breast Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022;20(6):691-722. https://doi.org/10.6004/jnccn.2022.0030 PMID:35714673

Gilson P, Merlin JL, Harlé A. Deciphering tumour heterogeneity: from tissue to liquid biopsy. Cancers (Basel). 2022;14(6):1384. https://doi.org/10.3390/cancers14061384 PMID:35326534

Hiley C, de Bruin EC, McGranahan N, Swanton C. Deciphering intratumor heterogeneity and temporal acquisition of driver events to refine precision medicine. Genome Biol. 2014 Aug 27;15(8):453. doi: 10.1186/s13059-014-0453-8. PMID: 25222836

Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12(7):381-394. https://doi.org/10.1038/nrclinonc.2015.73 PMID:25895611

Pasha N, Turner NC. Understanding and overcoming tumor heterogeneity in metastatic breast cancer treatment. Nat Cancer. 2021;2(7):680-692. https://doi.org/10.1038/s43018-021-00229-1 PMID:35121946

Hapach LA, Carey SP, Schwager SC, et al. Phenotypic heterogeneity and metastasis of breast cancer cells. Cancer Res. 2021;81(13):3649-3663. https://doi.org/10.1158/0008-5472.CAN-20-1799 PMID:33975882

Lee JK, Choi YL, Kwon M, Park PJ. Mechanisms and consequences of cancer genome instability: lessons from genome sequencing studies. Annu Rev Pathol. 2016;11(1):283-312. https://doi.org/10.1146/annurev-pathol-012615-044446 PMID:26907526

Tellez-Gabriel M, Ory B, Lamoureux F, Heymann MF, Heymann D. Tumour heterogeneity: the key advantages of single-cell analysis. Int J Mol Sci. 2016;17(12):2142. https://doi.org/10.3390/ijms17122142 PMID:27999407

McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168(4):613-628. https://doi.org/10.1016/j.cell.2017.01.018 PMID:28187284

Kaufman PA, Bloom KJ, Burris H, et al. Assessing the discordance rate between local and central HER2 testing in women with locally determined HER2-negative breast cancer. Cancer. 2014 Sep 1;120(17):2657-2664. doi: 10.1002/cncr.28710. Epub 2014 Jun 13. PMID: 24930388

Paik S, Bryant J, Tan-Chiu E, et al. Real-world performance of HER2 testing – National Surgical Adjuvant Breast and Bowel Project experience. J Natl Cancer Inst. 2002 Jun 5;94(11):852-854. doi: 10.1093/jnci/94.11.852. PMID: 12048273

Schrijver WAME, Suijkerbuijk KPM, van Gils CH, van der Wall E, Moelans CB, van Diest PJ. Receptor conversion in distant breast cancer metastases: a systematic review and meta-analysis. J Natl Cancer Inst. 2018 Jun 1;110(6):568-580. doi: 10.1093/jnci/djx273. PMID: 29315431

Aktas B, Kasimir-Bauer S, Müller V, et al; DETECT Study Group. Comparison of the HER2, estrogen and progesterone receptor expression profile of primary tumor, metastases and circulating tumor cells in metastatic breast cancer patients. BMC Cancer. 2016;16(1):522. https://doi.org/10.1186/s12885-016-2587-4 PMID:27456970

Schrijver WAME, van der Groep P, Hoefnagel LD, et al. Influence of decalcification procedures on immunohistochemistry and molecular pathology in breast cancer. Mod Pathol. 2016;29(12):1460-1470. https://doi.org/10.1038/modpathol.2016.116 PMID:27562496

Van Poznak C, Somerfield MR, Bast RC, et al. Use of biomarkers to guide decisions on systemic therapy for women with metastatic breast cancer: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2015;33(24):2695-2704. https://doi.org/10.1200/JCO.2015.61.1459 PMID:26195705

Micalizzi DS, Maheswaran S, Haber DA. A conduit to metastasis: circulating tumor cell biology. Genes Dev. 2017 Sep 15;31(18):1827-1840. doi: 10.1101/gad.305805.117. PMID: 29051388

Alix-Panabières C, Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov. 2021;11(4):858-873. https://doi.org/10.1158/2159-8290.CD-20-1311 PMID:33811121

Alix-Panabières C, Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov. 2021 Apr;11(4):858-873. doi: 10.1158/2159-8290.CD-20-1311. PMID: 33811121

Joosse SA, Gorges TM, Pantel K. Biology, detection, and clinical implications of circulating tumor cells. EMBO Mol Med. 2015;7(1):1-11. https://doi.org/10.15252/emmm.201303698 PMID:25398926

Keomanee-Dizon K, Shishido SN, Kuhn P. Circulating tumor cells: high-throughput imaging of CTCs and bioinformatic analysis. In: Recent results in cancer research. Vol 215. Springer: New York LLC; 2020:89-104. https://doi.org/10.1007/978-3-030-26439-0_5

Werner SL, Graf RP, Landers M, et al. Analytical validation and capabilities of the epic CTC platform: enrichment-free circulating tumour cell detection and characterization. J Circ Biomark. 2015;4:3. https://doi.org/10.5772/60725 PMID:28936239

Scher HI, Lu D, Schreiber NA, et al. Association of AR-V7 on circulating tumor cells as a treatment-specific biomarker with outcomes and survival in castration-resistant prostate cancer. JAMA Oncol. 2016;2(11):1441-1449. https://doi.org/10.1001/jamaoncol.2016.1828 PMID:27262168

Armstrong AJ, Luo J, Nanus DM, et al. Prospective multicenter study of circulating tumor cell AR-V7 and taxane versus hormonal treatment outcomes in metastatic castration-resistant prostate cancer. JCO Precis Oncol. 2020;4(4):1285-1301. https://doi.org/10.1200/PO.20.00200 PMID:33154984

Graf RP, Hullings M, Barnett ES, Carbone E, Dittamore R, Scher HI. Clinical utility of the nuclear-localized AR-V7 biomarker in circulating tumor cells in improving physician treatment choice in castration-resistant prostate cancer. Eur Urol. 2020;77(2):170-177. https://doi.org/10.1016/j.eururo.2019.08.020 PMID:31648903

Lu D, Krupa R, Harvey M, et al. Development of an immunofluorescent AR-V7 circulating tumor cell assay – a blood-based test for men with metastatic prostate cancer. J Circ Biomark. 2020;9(1):13-19. https://doi.org/10.33393/jcb.2020.2163 PMID:33717359

Scher HI, Graf RP, Schreiber NA, et al. Nuclear-specific AR-V7 protein localization is necessary to guide treatment selection in metastatic castration-resistant prostate cancer. Eur Urol. 2017;71(6):874-882. https://doi.org/10.1016/j.eururo.2016.11.024 PMID:27979426

Scher HI, Graf RP, Schreiber NA, et al. Phenotypic heterogeneity of circulating tumor cells informs clinical decisions between AR signaling inhibitors and taxanes in metastatic prostate cancer. Cancer Res. 2017;77(20):5687-5698. https://doi.org/10.1158/0008-5472.CAN-17-1353 PMID:28819021

Beltran H, Jendrisak A, Landers M, et al. The initial detection and partial characterization of circulating tumor cells in neuroendocrine prostate cancer. Clin Cancer Res. 2016;22(6):1510-1519. https://doi.org/10.1158/1078-0432.CCR-15-0137 PMID:26671992

Bjartell AS. Re: The initial detection and partial characterization of circulating tumor cells in neuroendocrine prostate cancer. Eur Urol. 2016;70(4):700. https://doi.org/10.1016/j.eururo.2016.07.037 PMID:27481179

Fujii T, Reuben JM, Huo L, et al. Androgen receptor expression on circulating tumor cells in metastatic breast cancer. PLoS One. 2017;12(9):e0185231. https://doi.org/10.1371/journal.pone.0185231 PMID:28957377

Dago AE, Stepansky A, Carlsson A, et al. Rapid phenotypic and genomic change in response to therapeutic pressure in prostate cancer inferred by high content analysis of single circulating tumor cells. PLoS One. 2014;9(8):e101777. https://doi.org/10.1371/journal.pone.0101777 PMID:25084170

Greene SB, Dago AE, Leitz LJ, et al. Chromosomal instability estimation based on next generation sequencing and single cell genome wide copy number variation analysis. PLoS One. 2016;11(11):e0165089. https://doi.org/10.1371/journal.pone.0165089 PMID:27851748

Efron B. Mathematics. Bayes' theorem in the 21st century. Science. 2013 Jun 7;340(6137):1177-1178. doi: 10.1126/science.1236536. PMID: 23744934.

Bours MJ. Bayes’ rule in diagnosis. J Clin Epidemiol. 2021;131:158-160. https://doi.org/10.1016/j.jclinepi.2020.12.021 PMID:33741123

Zhang L, Beasley S, Prigozhina NL, et al. Detection and characterization of circulating tumour cells in multiple myeloma. J Circ Biomark. 2016;5:10. https://doi.org/10.5772/64124 PMID:28936258

Subik K, Lee JF, Baxter L, et al. The expression patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer (Auckl). 2010 May 20;4:35-41. Erratum in: Breast Cancer (Auckl). 2018 Oct 16;12:1178223418806626. PMID: 20697531

McCabe A, Dolled-Filhart M, Camp RL, Rimm DL. Automated quantitative analysis (AQUA) of in situ protein expression, antibody concentration, and prognosis. J Natl Cancer Inst. 2005;97(24):1808-1815. https://doi.org/10.1093/jnci/dji427 PMID:16368942

Gertych A, Mohan S, Maclary S, et al. Effects of tissue decalcification on the quantification of breast cancer biomarkers by digital image analysis. Diagn Pathol. 2014;9(1):213. https://doi.org/10.1186/s13000-014-0213-9 PMID:25421113

Brown LC, Halabi S, Schonhoft JD, et al. Circulating tumor cell chromosomal instability and neuroendocrine phenotype by immunomorphology and poor outcomes in men with mCRPC treated with abiraterone or enzalutamide. Clin Cancer Res. 2021;27(14):4077-4088. https://doi.org/10.1158/1078-0432.CCR-20-3471 PMID:33820782

Manié E, Popova T, Battistella A, et al. Genomic hallmarks of homologous recombination deficiency in invasive breast carcinomas. Int J Cancer. 2016;138(4):891-900. https://doi.org/10.1002/ijc.29829 PMID:26317927

Sansregret L, Swanton C. The role of aneuploidy in cancer evolution. Cold Spring Harb Perspect Med. 2017;7(1):a028373. https://doi.org/10.1101/cshperspect.a028373 PMID:28049655

Malihi PD, Graf RP, Rodriguez A, et al. Single-cell circulating tumor cell analysis reveals genomic instability as a distinctive feature of aggressive prostate cancer. Clin Cancer Res. 2020;26(15):4143-4153. https://doi.org/10.1158/1078-0432.CCR-19-4100 PMID:32341031

Parkes A, Clifton K, Al-Awadhi A, et al. Characterization of bone only metastasis patients with respect to tumor subtypes. NPJ Breast Cancer. 2018;4(1):2. https://doi.org/10.1038/s41523-018-0054-x PMID:29387785

Mosele F, Deluche E, Lusque A, et al. Trastuzumab deruxtecan in metastatic breast cancer with variable HER2 expression: the phase 2 DAISY trial. Nat Med. 2023;29(8):2110-2120. https://doi.org/10.1038/s41591-023-02478-2 PMID:37488289

Published

2024-10-04

How to Cite

Di Caro, G., Lam, E., Bourdon, D., Blankfard, M., Dharajiya, N., Slade, M., Williams, E., Zhang, D., Wenstrup, R., & Schwartzberg, L. (2024). A novel liquid biopsy assay for detection of ERBB2 (HER2) amplification in circulating tumor cells (CTCs). Journal of Circulating Biomarkers, 13(1), 27–35. https://doi.org/10.33393/jcb.2024.3046

Issue

Section

Original research article
Received 2024-02-15
Accepted 2024-08-19
Published 2024-10-04

Metrics

Most read articles by the same author(s)