Evaluation of the Clinical Outcome of Metastatic Breast Cancer Patients with Negative and Positive Circulating Tumor DNA: A Systematic Review and Meta-analysis

Document Type : Review Article

Authors

1 Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran

2 Department of Internal Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran

Abstract

Background and aim: The present study was conducted with the aim of evaluating the clinical outcome of metastatic breast cancer patients with negative and positive circulating tumor DNA.
Material and methods: Searching international databases PubMed, Web of Science, Scopus, Science Direct, Web of Knowledge, EBSCO, Wiley, ISI, Elsevier, Embase databases, and Google Scholar search engine based on PRISMA 2020-27-item checklist and keywords Related to the objectives of the study, it was carried out from 2019 to February 2024. a model with fixed effect and inverse–variance method was used. All statistical analyses are done using STATA/MP software. v17 was done considering the significance of less than 0.05.
Results: Six studies were selected according to the inclusion criteria. Compared with patients with negative ctDNA, those with positive ctDNA had a higher risk for progression-free survival and overall survival (HR 2.02, 95% CI 0.71-3.33; P-value < 0.001) and (HR 2.78, 95% CI 1.47-4.10; P-value < 0.001), respectively.
Conclusions: In metastatic breast cancer patients, it was individually and jointly associated with progression-free survival and overall survival-positive circulating tumor DNA.

Keywords

Main Subjects


[1] Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, et al. Current and future burden of breast cancer: Global statistics for 2020 and 2040. The Breast. 2022;66:15-23. https://doi.org/10.1016/j.breast.2022.08.010.
[2] Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. Ca Cancer J Clin. 2023;73(1):17-48. https://doi.org/10.3322/caac.21763.
[3] Lei S, Zheng R, Zhang S, Wang S, Chen R, Sun K, et al. Global patterns of breast cancer incidence and mortality: A population‐based cancer registry data analysis from 2000 to 2020. Cancer Communications. 2021;41(11):1183-94. https://doi.org/10.1002/cac2.12207.
[4] Wang L, Zhang S, Wang X. The metabolic mechanisms of breast cancer metastasis. Frontiers in Oncology. 2021;10:602416. https://doi.org/10.3389/fonc.2020.602416.
[5] Li J, Guan X, Fan Z, Ching LM, Li Y, Wang X, et al. Non-invasive biomarkers for early detection of breast cancer. Cancers. 2020;12(10):2767. https://doi.org/10.3390/cancers12102767.
[6] Botlagunta M, Botlagunta MD, Myneni MB, Lakshmi D, Nayyar A, Gullapalli JS, et al. Classification and diagnostic prediction of breast cancer metastasis on clinical data using machine learning algorithms. Scientific Reports. 2023;13(1):485. https://doi.org/10.1038/s41598-023-27548-w.
[7] Pai T, Cornell L, Seneviratne D, Niazi S, Mussallem D, Vallow L. Pre-diagnosis major life stressors and breast cancer outcomes. Breast Cancer Research and Treatment. 2021;188:459-64. https://doi.org/10.1007/s10549-021-06218-3.
[8] Łukasiewicz S, Czeczelewski M, Forma A, Baj J, Sitarz R, Stanisławek A. Breast cancer—epidemiology, risk factors, classification, prognostic markers, and current treatment strategies—an updated review. Cancers. 2021;13(17):4287. https://doi.org/10.3390/cancers13174287.
[9] Benson KR, Aggarwal S, Carter JN, Von Eyben R, Pradhan P, Prionas ND, et al. Predicting survival for patients with metastatic disease. International Journal of Radiation Oncology* Biology* Physics. 2020;106(1):52-60. https://doi.org/10.1016/j.ijrobp.2019.10.032.
[10] Bhushan A, Gonsalves A, Menon JU. Current state of breast cancer diagnosis, treatment, and theranostics. Pharmaceutics. 2021;13(5):723. https://doi.org/10.3390/pharmaceutics13050723.
[11] Peng Y, Mei W, Ma K, Zeng C. Circulating tumor DNA and minimal residual disease (MRD) in solid tumors: current horizons and future perspectives. Frontiers in oncology. 2021;11:763790. https://doi.org/10.3389/fonc.2021.763790.
[12] Alba-Bernal A, Lavado-Valenzuela R, Domínguez-Recio ME, Jiménez-Rodriguez B, Queipo-Ortuño MI, Alba E, et al. Challenges and achievements of liquid biopsy technologies employed in early breast cancer. EBioMedicine. 2020;62. https://doi.org/10.1016/j.ebiom.2020.103100.
[13] Papakonstantinou A, Gonzalez NS, Pimentel I, Suñol A, Zamora E, Ortiz C, et al. Prognostic value of ctDNA detection in patients with early breast cancer undergoing neoadjuvant therapy: A systematic review and meta-analysis. Cancer Treatment Reviews. 2022;104:102362. https://doi.org/10.1016/j.ctrv.2022.102362.
[14] Cullinane C, Fleming C, O’Leary DP, Hassan F, Kelly L, O’Sullivan MJ, et al. Association of circulating tumor DNA with disease-free survival in breast cancer: a systematic review and meta-analysis. JAMA network open. 2020;3(11):e2026921. https://doi.org/10.1001/jamanetworkopen.2020.26921.
[15] Gögenur M, Hadi NA, Qvortrup C, Andersen CL, Gögenur I. ctDNA for risk of recurrence assessment in patients treated with neoadjuvant treatment: A systematic review and meta-analysis. Annals of Surgical Oncology. 2022;29(13):8666-74. https://doi.org/10.1245/s10434-022-12366-7.
[16] Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Bmj. 2021;372. https://doi.org/10.1136/bmj.n71.
[17] Gierisch JM, Beadles C, Shapiro A, McDuffie J, Cunningham N, Bradford D. Newcastle-Ottawa scale coding manual for cohort studies. Health disparities in quality indicators of healthcare among adults with mental illness. Department of Veterans Affairs (US). 2014.
[18] Minozzi S, Cinquini M, Gianola S, Gonzalez-Lorenzo M, Banzi R. The revised Cochrane risk of bias tool for randomized trials (RoB 2) showed low interrater reliability and challenges in its application. Journal of clinical epidemiology. 2020;126:37-44. https://doi.org/10.1016/j.jclinepi.2020.06.015.
[19] Sokouti M, Shafiee-Kandjani AR, Sokouti M, Sokouti B. A meta-analysis of systematic reviews and meta-analyses to evaluate the psychological consequences of COVID-19. BMC psychology. 2023;11(1):279. https://doi.org/10.1186/s40359-023-01313-0.
[20] Park J, Chang ES, Kim JY, Chelakkot C, Sung M, Song JY,et al. c-MET-positive circulating tumor cells and cell-free DNA as independent prognostic factors in hormone receptor-positive/HER2-negative metastatic breast cancer. Breast Cancer Research. 2024;26(1):13. https://doi.org/10.1186/s13058-024-01768-y.
[21] Keup C, Storbeck M, Hauch S, Hahn P, Sprenger-Haussels M, Hoffmann O, et al. Multimodal targeted deep sequencing of circulating tumor cells and matched cell-free DNA provides a more comprehensive tool to identify therapeutic targets in metastatic breast cancer patients. Cancers. 2020;12(5):1084. https://doi.org/10.3390/cancers12051084.
[22] Clatot F, Perdrix A, Beaussire L, Lequesne J, Lévy C, Emile G, et al. Risk of early progression according to circulating ESR1 mutation, CA-15.3 and cfDNA increases under first-line anti-aromatase treatment in metastatic breast cancer. Breast Cancer Research. 2020;22:1-2. https://doi.org/10.1186/s13058-020-01290-x.
[23] Li X, Lu J, Zhang L, Luo Y, Zhao Z, Li M. Clinical implications of monitoring ESR1 mutations by circulating tumor DNA in estrogen receptor positive metastatic breast cancer: a pilot study. Translational Oncology. 2020;13(2):321-8. https://doi.org/10.1016/j.tranon.2019.11.007.
[24] Ye Z, Wang C, Wan S, Mu Z, Zhang Z, Abu-Khalaf MM, et al. Association of clinical outcomes in metastatic breast cancer patients with circulating tumour cell and circulating cell-free DNA. European Journal of Cancer. 2019;106:133-43. https://doi.org/10.1016/j.ejca.2018.10.012.
 [25] Fernandez-Garcia D, Hills A, Page K, Hastings RK, Toghill B, Goddard KS, et al. Plasma cell-free DNA (cfDNA) as a predictive and prognostic marker in patients with metastatic breast cancer. Breast Cancer Research. 2019;21:1-3. https://doi.org/10.1186/s13058-019-1235-8.
 [26] Panagopoulou M, Karaglani M, Balgkouranidou I, Biziota E, Koukaki T, Karamitrousis E, et al. Circulating cell-free DNA in breast cancer: size profiling, levels, and methylation patterns lead to prognostic and predictive classifiers. Oncogene. 2019;38(18):3387-401. https://doi.org/10.1038/s41388-018-0660-y.
[27] Qin Z, Ljubimov VA, Zhou C, Tong Y, Liang J. Cell-free circulating tumor DNA in cancer. Chinese journal of cancer. 2016;35:1-9. https://doi.org/10.1186/s40880-016-0092-4.
[28] Kidess E, Jeffrey SS. Circulating tumor cells versus tumor-derived cell-free DNA: rivals or partners in cancer care in the era of single-cell analysis?. Genome medicine. 2013;5:1-4. https://doi.org/10.1186/gm474.
[29] Khurram I, Khan MU, Ibrahim S, Saleem A, Khan Z, Mubeen M, et al. Efficacy of cell-free DNA as a diagnostic biomarker in breast cancer patients. Scientific Reports. 2023;13(1):15347. https://doi.org/10.1038/s41598-023-42726-6.
[30] Bidard FC, Peeters DJ, Fehm T, Nolé F, Gisbert-Criado R, Mavroudis D, et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. The lancet oncology. 2014;15(4):406-14. https://doi.org/10.1016/S1470-2045(14)70069-5.
[31] Ocana A, Díez-González L, García-Olmo DC, Templeton AJ, Vera-Badillo F, José Escribano M, et al. Circulating DNA and survival in solid tumors. Cancer Epidemiology, Biomarkers & Prevention. 2016;25(2):399-406. https://doi.org/10.1158/1055-9965.EPI-15-0893.
[32] Shaw JA, Guttery DS, Hills A, Fernandez-Garcia D, Page K, Rosales BM, et al. Mutation analysis of cell-free DNA and single circulating tumor cells in metastatic breast cancer patients with high circulating tumor cell counts. Clinical Cancer Research. 2017;23(1):88-96. https://doi.org/10.1158/1078-0432.CCR-16-0825.