Cell-free tumor DNA analysis in advanced or metastatic breast cancer patients: mutation frequencies, testing intention, and clinical impact

Hanna Huebner , Pauline Wimberger , Elena Laakmann , Eugen Ruckhäberle , Matthias Ruebner , Sarah Lehle , Sabrina Uhrig , Philipp Ziegler , Theresa Link , Carolin C. Hack , Erik Belleville , Iris Faull , Marcus Hausch , Diethelm Wallwiener , Andreas Schneeweiss , Hans Tesch , Sara Y. Brucker , Matthias W. Beckmann , Peter A. Fasching , Volkmar Müller , Tanja N. Fehm

Precision Clinical Medicine ›› 2025, Vol. 8 ›› Issue (1) : pbae034

PDF (1186KB)
Precision Clinical Medicine ›› 2025, Vol. 8 ›› Issue (1) :pbae034 DOI: 10.1093/pcmedi/pbae034
Research Article
research-article

Cell-free tumor DNA analysis in advanced or metastatic breast cancer patients: mutation frequencies, testing intention, and clinical impact

Author information +
History +
PDF (1186KB)

Abstract

Background: Circulating cell-free tumor DNA (ctDNA) provides a non-invasive approach for assessing somatic alterations. The German PRAEGNANT registry study aims to explore molecular biomarkers and investigate their integration into clinical practice. In this context, ctDNA testing was included to understand the motivations of clinicians to initiate testing, to identify somatic alterations, and to assess the clinical impact of the results obtained.

Methods: Patients with advanced/metastatic breast cancer were prospectively enrolled in the Prospective Academic Translational Research Network for the Optimization of Oncological Health Care Quality in the Adjuvant and Advanced/Metastatic Setting (PRAEGNANT study; NCT02338167). The FDA-approved and CE-marked GUARDANT360 CDx test was used to assess somatic alterations. A ctDNA-analysis report was provided to the treating physician along with a questionnaire about the intent for testing and the clinical implications of test results.

Results: ctDNA from 49 patients was analyzed prospectively: 37 (76%) had at least one somatic alteration in the analyzed geneset; 14 patients (29%) harbored alterations in TP53, 12 (24%) in PIK3CA, and 6 (12%) in ESR1. Somatic mutations in BRCA1 or BRCA2 were detected in 3 (6%) and 4 (8%) patients, respectively, and 59% of patients had hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer. Questionnaires regarding test intentions and clinical impact were completed for 48 (98%) patients. These showed that ctDNA testing influenced treatment decisions for 35% of patients.

Discussion: The high prevalence of somatic alterations in TP53, PIK3CA, ESR1, and BRCA1/2 genes, identified by ctDNA genotyping, highlights their potential as biomarkers for targeted therapies. Detection of specific mutations affected treatment decisions, such as eligibility for alpelisib, and might further facilitate treatment with e.g. elacestrant or capiversatib in future treatment lines.

Keywords

ctDNA / cfDNA / cell-free DNA / breast cancer / genetic testing / Clinical trial registration No.: NCT02338167

Cite this article

Download citation ▾
Hanna Huebner, Pauline Wimberger, Elena Laakmann, Eugen Ruckhäberle, Matthias Ruebner, Sarah Lehle, Sabrina Uhrig, Philipp Ziegler, Theresa Link, Carolin C. Hack, Erik Belleville, Iris Faull, Marcus Hausch, Diethelm Wallwiener, Andreas Schneeweiss, Hans Tesch, Sara Y. Brucker, Matthias W. Beckmann, Peter A. Fasching, Volkmar Müller, Tanja N. Fehm. Cell-free tumor DNA analysis in advanced or metastatic breast cancer patients: mutation frequencies, testing intention, and clinical impact. Precision Clinical Medicine, 2025, 8(1): pbae034 DOI:10.1093/pcmedi/pbae034

登录浏览全文

4963

注册一个新账户 忘记密码

Acknowledgments

Guardant Health, Inc provided test kits and performed the sequencing analysis. The PRAEGNANT network is supported by grants from Pfizer, Hexal, Celgene, Daiichi-Sankyo, Roche, Merrimack, Eisai, AstraZeneca, and Novartis. These companies did not have any involvement in the study design, in the collection, analysis, or interpretation of the data, in the writing of the report, or in the decision to submit this article for publication.

Author contributions

Hanna Huebner (Formal analysis, Methodology, Visualization, Writing - original draft, Writing - review & editing), Pauline Wimberger (Investigation, Writing - review & editing), Elena Laakmann (Investigation, Writing - review & editing), Eugen Ruckhäberle (Investigation, Writing - review & editing), Matthias Ruebner (Resources, Supervision, Writing - review & editing), Sarah Lehle (Formal analysis, Writing - review & editing), Sabrina Uhrig (Data curation, Writing - review & editing), Philipp Ziegler (Data curation, Writing - review & editing), Theresa Link (Investigation, Writing - review & editing), Carolin C. Hack (Investigation, Writing - review & editing), Erik Belleville (Project administration, Resources, Writing - review & editing), Iris Faull (Resources, Writing - review & editing), Marcus Hausch (Resources, Writing - review & editing), Diethelm Wallwiener (Investigation, Writing - review & editing), Andreas Schneeweiss (Investigation, Writing - review & editing), Hans Tesch (Investigation, Writing - review & editing), Sara Y. Brucker (Investigation, Writing - review & editing), Matthias W. Beckmann (Investigation, Resources, Writing - review & editing), Peter A. Fasching (Conceptualization, Resources, Supervision, Writing - review & editing), Volkmar Müller (Investigation, Writing - review & editing) and Tanja N. Fehm (Funding acquisition, Investigation, Resources, Writing - review & editing).

Conflict of interest

P.W. has received honoraria from Roche, Novartis, Amgen, AstraZeneca, Pfizer, MSD, Clovis, Tesaro, Celgene, Teva, Eisai, Daiichi Sankyo, Seagen, and Eli Lilly. E.B. has received honoraria from Novartis, Celgene, Eisai, Daiichi Sankyo, Merrimack, AstraZeneca, Riemser, Pfizer, Hexal, Amgen, and onkowissen.de for consulting, clinical research management, or medical education activities. A.S. has received honoraria from Roche, Celgene, AstraZeneca, Novartis, Pfizer, Zuckschwerdt Verlag GmbH, Georg Thieme Verlag, Aurikamed GmbH, MCI Deutschland GmbH, bsh medical communications GmbH, and promedicis GmbH. H.T. has received honoraria from Novartis, Roche, Celgene, Teva, and Pfizer, and travel support from Roche, Celgene, and Pfizer. S.Y.B. received honoraria from Roche, Novartis, Pfizer, MSD, Teva, and AstraZeneca. V.M. has received speaker honoraria from Astra Zeneca, Daiichi-Sankyo, Eisai, Pfizer, MSD, Medac, Novartis, Roche, Seagen, Onkowissen, high5 Oncology, Lilly, Medscape, Gilead, Pierre Fabre, and iMED Institut, consultancy honoraria from Roche, Pierre Fabre, PINK, ClinSol, Novartis, MSD, Daiichi-Sankyo, Eisai, Lilly, Seagen, Gilead, Stemline, institutional research support from Novartis, Roche, Seagen, Genentech, and Astra Zeneca, and travel grants from Astra Zeneca, Roche, Pfizer, Daiichi Sankyo, and Gilead. T.N.F. received honoraria from Novartis, Roche, Pfizer, TEVA, Daiichi Sankyo, AstraZeneca, MSD, Onkowisseng GmBH, and Medconcept. H.H. received speaker honoraria from Lilly and funding from Novartis and Sysmex. P.A.F. has received honoraria from Agendia, AstraZeneca, ClinSol GmbH, Daiichi Sankyo, Eisai, Gilead, Lilly, MSD, Novartis, Pfizer, Seagen, Stemline Therapeutics, TRIO Oncology, and Veracyte for consulting, participation in advisory boards and steering committees and/or lectures. His institution conducts research for Novartis. T.L. received honoraria for lectures or presentations from Amgen, Roche, MSD, Novartis, Pfizer, Lilly, GSK, Gilead, Astra Zeneca, Daiichi Sankyo, Stemline, and Seagen, advisory board/advise honoraria from MSD, Roche, Pfizer, Lilly, Myriad, Esai, GSK, Gilead, Daiichi Sankyo, Roche, and Astra Zeneca, and support for attending meetings and/or travel from Pfizer, Astra Zeneca, Gilead, Daiichi Sankyo, and Stemline. I.F. and M.H. are employees and stockholders at Guardant Health, Inc. E.L. reports travel expenses from Pierre Fabre, honoraria for educational events from Astra Zeneca, and Seagen, and consultancy honoraria from Novartis, Astra Zeneca, and Daiichi Sankyo. E.R. has received Speaker honoraria from Astra Zeneca, Daiichi-Sankyo, Eisai, Pfizer, MSD, Novartis, Roche, Seagen, Onkowissen, Lilly, Gilead, and Pierre Fabre, consultancy honoraria from Roche, Pierre Fabre, ClinSol, Novartis, MSD, Daiichi-Sankyo, Eisai, Lilly, Seagen, Gilead, and Stemline, research research support from Roche, and travel grants from Astra Zeneca, Roche, Pfizer, and Pierre Fabre.

Ethics statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Ethics Committee of the Medical Faculty, University of Tübingen, Tübingen, Germany (ethics approval number: 234/2014BO1, first approval on 17 June 2014, approval of Amendment 1 on 11 June 2015, approval of Amendment 2 on 18 March 2019). Informed consent was obtained from all subjects involved in the study.

Data availability statement

The datasets generated during and/or analyzed during the current study are not publicly available due to GDPR regulations but are available from the corresponding author on reasonable request.

References

[1]

Tutt ANJ, Garber JE, Kaufman B et al. Adjuvant Olaparib for patients with BRCA1-or BRCA2-mutated breast cancer. N Engl J Med 2021;384:2394-405. https://doi.org/10.1056/NEJMoa2105215.

[2]

Bertucci F, Ng CKY, Patsouris A et al. Genomic characterization of metastatic breast cancers. Nature 2019;569:560-64. https://doi.org/10.1038/s41586-019-1056-z.

[3]

Razavi P, Chang MT, Xu G et al. The genomic landscape of endocrine-resistant advanced breast cancers. Cancer Cell 2018;34:427-38 e6. https://doi.org/10.1016/j.ccell.2018.08.008.

[4]

Toy W, Shen Y, Won H et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet 2013;45:1439-45. https://doi.org/10.1038/ng.2822.

[5]

Pearson A, Proszek P, Pascual J et al. Inactivating NF1 mutations are enriched in advanced breast cancer and contribute to endocrine therapy resistance. Clin Cancer Res 2020;26:608-22. https://doi.org/10.1158/1078-0432.CCR-18-4044.

[6]

Xu G, Chhangawala S, Cocco E et al. ARID1A determines luminal identity and therapeutic response in estrogen-receptor-positive breast cancer. Nat Genet 2020;52:198-207. https://doi.org/10.1038/s41588-019-0554-0.

[7]

Andre F, Ciruelos EM, Juric D et al. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1. Ann Oncol 2021;32:208-17. https://doi.org/10.1016/j.annonc.2020.11.011.

[8]

Bidard FC, Kaklamani VG, Neven P et al. Elacestrant (oral selective estrogen receptor degrader) versus standard endocrine therapy for estrogen receptor-positive, Human epidermal growth factor receptor 2-negative advanced breast cancer: results from the randomized phase III EMERALD trial. J Clin Oncol 2022;40:3246-56. https://doi.org/10.1200/JCO.22.00338.

[9]

Turner NC, Oliveira M, Howell SJ et al. C. APItello-291 Study Group. Capivasertib in hormone receptor-positive advanced breast cancer. N Engl J Med 2023;388:2058-70. https://doi.org/10.1056/NEJMoa2214131.

[10]

Kingston B, Cutts RJ, Bye H et al. Genomic profile of advanced breast cancer in circulating tumour DNA. Nat Commun 2021;12:2423. https://doi.org/10.1038/s41467-021-22605-2.

[11]

Lehle S, Emons J, Hack CC et al. Evaluation of automated techniques for extraction of circulating cell-free DNA for implementation in standardized high-throughput workflows. Sci Rep 2023;13:373. https://doi.org/10.1038/s41598-022-27216-5.

[12]

Zannini G, Facchini G, De Sio M et al. Implementation of BRCA mutations testing in formalin-fixed paraffin-embedded (FFPE) samples of different cancer types. Pathol Res Pract 2023;243:154336. https://doi.org/10.1016/j.prp.2023.154336.

[13]

Pixberg C, Zapatka M, Hlevnjak M et al. COGNITION: a prospective precision oncology trial for patients with early breast cancer at high risk following neoadjuvant chemotherapy. ESMO Open 2022;7:100637. https://doi.org/10.1016/j.esmoop.2022.100637.

[14]

Turner NC, Kingston B, Kilburn LS et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): a multicentre, multicohort, phase 2a, platform trial. Lancet Oncol 2020;21:1296-308. https://doi.org/10.1016/S1470-2045(20)30444-7.

[15]

Jhaveri KLIS, Im S-A, Saura C et al. Inavolisib or placebo in combination with palbociclib and fulvestrant in patients with PIK3CAmutated, hormone receptor-positive, HER2-negative locally advanced or metastatic breast cancer:phase III, INAVO120 primary analysis. In Proceedings of the 2023 San Antonio Breast Cancer Symposium, San Antonio, TX, USA, 2023;5-9.

[16]

Venetis K, Pepe F, Pescia C et al. ESR1 mutations in HR+/HER2metastatic breast cancer: enhancing the accuracy of ctDNA testing. Cancer Treat Rev 2023;121:102642. https://doi.org/10.1016/j.ctrv.2023.102642.

[17]

Pascual J, Attard G, Bidard FC et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2022;33:750-68. https://doi.org/10.1016/j.annonc.2022.05.520.

[18]

Huebner H, Kurbacher CM, Kuesters G et al. Heregulin (HRG) assessment for clinical trial eligibility testing in a molecular registry (PRAEGNANT) in Germany. BMC Cancer 2020;20:1091. https://doi.org/10.1186/s12885-020-07546-1.

[19]

Huebner H, Ruebner M, Kurbacher C et al. Return of individual genomic research results within the PRAEGNANT multicenter registry study. Breast Cancer Res Treat 2023;197:355-68. https://doi.org/10.1007/s10549-022-06795-x.

[20]

Fasching PA, Brucker SY, Fehm TN et al. Biomarkers in patients with metastatic breast cancer and the PRAEGNANT Study network. Geburtshilfe Frauenheilkd 2015;75:41-50.

[21]

Engler T, Fasching PA, Luftner D et al. Implementation of CDK4/6 inhibitors and its influence on the treatment landscape of advanced breast cancer patients-data from the realworld registry PRAEGNANT. Geburtshilfe Frauenheilkd 2022;82:1055-67.

[22]

Fasching PA, Yadav S, Hu C et al. Mutations in BRCA1/2 and other panel genes in patients with metastatic breast cancer association with patient and disease characteristics and effect on prognosis. J Clin Oncol 2021;39:1619-30. https://doi.org/10.1200/JCO.20.01200.

[23]

Stout LA, Kassem N, Hunter C et al. Identification of germline cancer predisposition variants during clinical ctDNA testing. Sci Rep 2021;11:13624. https://doi.org/10.1038/s41598-021-93084-0.

[24]

Wang JF, Pu X, Zhang X et al. Variants with a low allele frequency detected in genomic DNA affect the accuracy of mutation detection in cell-free DNA by next-generation sequencing. Cancer 2018;124:1061-69. https://doi.org/10.1002/cncr.31152.

[25]

Martinez-Saez O, Pascual T, Braso-Maristany F et al. Circulating tumor DNA dynamics in advanced breast cancer treated with CDK4/6 inhibition and endocrine therapy. NPJ Breast Cancer 2021;7:8. https://doi.org/10.1038/s41523-021-00218-8.

[26]

Odegaard JI, Vincent JJ, Mortimer S et al. Validation of a plasmabased comprehensive cancer genotyping assay utilizing orthogonal tissue-and plasma-based methodologies. Clin Cancer Res 2018;24:3539-49. https://doi.org/10.1158/1078-0432.CCR-17-3831.

[27]

Fuentes-Antras J, Martinez-Rodriguez A, Guevara-Hoyer K et al. Real-world use of highly sensitive liquid biopsy monitoring in metastatic breast cancer patients treated with endocrine agents after exposure to aromatase inhibitors. Int J Mol Sci 2023;24:11419. https://doi.org/10.3390/ijms241411419.

[28]

Muendlein A, Geiger K, Gaenger S et al. Significant impact of circulating tumour DNA mutations on survival in metastatic breast cancer patients. Sci Rep 2021;11:6761. https://doi.org/10.1038/s41598-021-86238-7.

[29]

Liu B, Hu Z, Ran J et al. The circulating tumor DNA (ctDNA) alteration level predicts therapeutic response in metastatic breast cancer: novel prognostic indexes based on ctDNA. Breast 2022;65:116-23. https://doi.org/10.1016/j.breast.2022.07.010.

[30]

Jeselsohn R, Buchwalter G,De Angelis C et al. ESR1 mutations-a mechanism for acquired endocrine resistance in breast cancer. Nat Rev Clin Oncol 2015;12:573-83. https://doi.org/10.1038/nrclinonc.2015.117.

[31]

Najim O, Papadimitriou K, Broeckx G et al. Validation of liquid biopsy for ESR1-mutation analysis in hormone-sensitive breast cancer: a pooled meta-analysis. Front Oncol 2023;13:1221773. https://doi.org/10.3389/fonc.2023.1221773.

[32]

Chandarlapaty S, Chen D, He W et al. Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: A secondary analysis of the BOLERO-2 clinical trial. JAMA Oncol 2016;2:1310-15. https://doi.org/10.1001/jamaoncol.2016.1279.

[33]

Goetz MP, Hamilton EP, Campone M et al. Landscape of baseline and acquired genomic alterations in circulating tumor DNA with abemaciclib alone or with endocrine therapy in advanced breast cancer. Clin Cancer Res 2023;30:2233-2244. https://doi.org/10.1158/1078-0432.CCR-22-3573.

[34]

Martin M, Zielinski C, Ruiz-Borrego M et al. Palbociclib in combination with endocrine therapy versus capecitabine in hormonal receptor-positive, human epidermal growth factor 2-negative, aromatase inhibitor-resistant metastatic breast cancer: a phase III randomised controlled trial-PEARL. Ann Oncol 2021;32:488-99. https://doi.org/10.1016/j.annonc.2020.12.013.

[35]

O'Leary B, Hrebien S, Morden JP et al. Early circulating tumor DNA dynamics and clonal selection with palbociclib and fulvestrant for breast cancer. Nat Commun 2018;9:896. https://doi.org/10.1038/s41467-018-03215-x.

[36]

Turner NC, Swift C, Kilburn L et al. ESR1 Mutations and overall survival on Fulvestrant versus Exemestane in advanced hormone receptor-positive breast cancer: A combined analysis of the phase III SoFEA and EFECT trials. Clin Cancer Res 2020;26:5172-77. https://doi.org/10.1158/1078-0432.CCR-20-0224.

[37]

Tung NM, Robson ME, Ventz S et al. TBCRC 048: Phase II study of Olaparib for metastatic breast cancer and mutations in homologous recombination-related genes. J Clin Oncol 2020;38:4274-82. https://doi.org/10.1200/JCO.20.02151.

[38]

Vidula N, Damodaran S, Blouch EL et al. Phase II study of talazoparib, a PARP inhibitor, in somatic BRCA1/2 mutant metastatic breast cancer identified by cell-free DNA or tumor tissue genotyping. Cancer Res 2023;83:OT1-11-01-. https://doi.org/10.1158/1538-7445.SABCS22-OT1-11-01.

[39]

Smyth LM, Tamura K, Oliveira M et al. Capivasertib, an AKT kinase inhibitor, as monotherapy or in combination with Fulvestrant in patients with AKT1 (E17K)-mutant, ER-positive metastatic breast cancer. Clin Cancer Res 2020;26:3947-57. https://doi.org/10.1158/1078-0432.CCR-19-3953.

[40]

Andre F, Filleron T, Kamal M et al. Genomics to select treatment for patients with metastatic breast cancer. Nature 2022;610:34348. https://doi.org/10.1038/s41586-022-05068-3.

[41]

Garcia-Murillas I, Schiavon G, Weigelt B et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med 2015;7:302ra133. https://doi.org/10.1126/scitranslmed.aab0021.

[42]

Magbanua MJM, Swigart LB, Wu HT et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and sur-vival. Ann Oncol 2021;32:229-39. https://doi.org/10.1016/j.annonc.2020.11.007.

[43]

Zhou Q, Gampenrieder SP, Frantal S et al. Persistence of ctDNA in patients with breast cancer during neoadjuvant treatment is a significant predictor of poor tumor response. Clin Cancer Res 2022;28:697-707. https://doi.org/10.1158/1078-0432.CCR-21-3231.

[44]

Hlevnjak M, Schulze M, Elgaafary S et al. CATCH: A prospective precision oncology trial in metastatic breast cancer. JCO Precis Oncol 2021;5:676-686. https://doi.org/10.1200/PO.20.0024.

[45]

Colomer R, Miranda J, Romero-Laorden N et al. Usefulness and real-world outcomes of next generation sequencing testing in patients with cancer: an observational study on the impact of selection based on clinical judgement. E Clinical Medicine 2023;60:102029. https://doi.org/10.1016/j.eclinm.2023.102029.

[46]

Schneider BP, Jiang G, Ballinger TJ et al. BRE12-158: A postneoadjuvant, randomized phase II trial of personalized therapy versus treatment of physician's choice for patients with residual triple-negative breast cancer. J Clin Oncol 2022;40:345-55. https://doi.org/10.1200/JCO.21.01657.

[47]

Jahangiri L, Hurst T. Assessing the concordance of genomic alterations between circulating-free DNA and tumour tissue in cancer patients. Cancers (Basel) 2019;11:1938. https://doi.org/10.3390/cancers11121938.

[48]

Rosenberg S, Ben Cohen G, Kato S et al. Concordance between cancer gene alterations in tumor and circulating tumor DNA correlates with poor survival in a real-world precision-medicine population. Mol Oncol 2023;17:1844-56. https://doi.org/10.1002/1878-0261.13383.

[49]

Urso L, Vernaci G, Carlet J et al. ESR1 Gene mutation in hormone receptor-positive HER2-negative metastatic breast cancer patients: concordance between tumor tissue and circulating tumor DNA analysis. Front Oncol 2021;11:625636. https://doi.org/10.3389/fonc.2021.625636.

[50]

Brown D, Smeets D, Szekely B et al. Phylogenetic analysis of metastatic progression in breast cancer using somatic mutations and copy number aberrations. Nat Commun 2017;8:14944. https://doi.org/10.1038/ncomms14944.

[51]

Yates LR, Knappskog S, Wedge D et al. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell 2017;32:169-84 e7. https://doi.org/10.1016/j.ccell.2017.07.005.

[52]

Zelinova K, Jagelkova M, Laucekova Z et al. Molecular analysis of circulating tumor DNA from breast cancer patients before and after surgery and following adjuvant chemotherapy. Mol Clin Oncol 2020;13:26.

AI Summary AI Mindmap
PDF (1186KB)

0

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/