open access

Ahead of print
Review paper
Published online: 2023-08-17
Get Citation

The importance of selected biomarkers in the clinical practice of breast cancer patients

Agata Makówka1, Beata Kotowicz1
Affiliations
  1. Cancer Biomarker and Cytokines Laboratory Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

open access

Ahead of print
Review article
Published online: 2023-08-17

Abstract

Breast cancer is considered the most commonly diagnosed tumors. Biomarkers used for the diagnosis and treatment of breast cancer are: tissue biomarkers (PR, ER, HER2, Ki-67) and serum biomarkers (CA15-3, CA125, CA27.29, CEA, cytokeratins). ECD HER2, metalloproteinases and leptin are emerging promising biomarkers for breast cancer. There is a growing need for personalized diagnostics based on tumor genome characterization, relying on liquid biopsy containing components such as CTC and ctDNA, cell-free RNA. Biomarkers can also be used use as a target for anti breast cancer treatment (PGRN and sortilin, AR, PD-1/PD-L1). Another potential fields of application of breast cancer biomarkers is monitoring of treatment side effects, such us inflammatory biomarkers causing cardiotoxicity, thyroiditis biomarkers (TSH, FT4, TPOab TgAb) in IrAE, NF-L and MCP-1 in ICI-associated neurotoxity. It is expected to develop new prognostic and predictive biomarkers that would provide accurate and reliable information for clinical application. Through the recognition of emerging biomarkers, it is possible to identify subgroups of patients who benefit from targeted therapies and managing treatment by monitoring side effects. However, these new biomarkers need to be validated and tested for their suitability before entering clinical use.

Abstract

Breast cancer is considered the most commonly diagnosed tumors. Biomarkers used for the diagnosis and treatment of breast cancer are: tissue biomarkers (PR, ER, HER2, Ki-67) and serum biomarkers (CA15-3, CA125, CA27.29, CEA, cytokeratins). ECD HER2, metalloproteinases and leptin are emerging promising biomarkers for breast cancer. There is a growing need for personalized diagnostics based on tumor genome characterization, relying on liquid biopsy containing components such as CTC and ctDNA, cell-free RNA. Biomarkers can also be used use as a target for anti breast cancer treatment (PGRN and sortilin, AR, PD-1/PD-L1). Another potential fields of application of breast cancer biomarkers is monitoring of treatment side effects, such us inflammatory biomarkers causing cardiotoxicity, thyroiditis biomarkers (TSH, FT4, TPOab TgAb) in IrAE, NF-L and MCP-1 in ICI-associated neurotoxity. It is expected to develop new prognostic and predictive biomarkers that would provide accurate and reliable information for clinical application. Through the recognition of emerging biomarkers, it is possible to identify subgroups of patients who benefit from targeted therapies and managing treatment by monitoring side effects. However, these new biomarkers need to be validated and tested for their suitability before entering clinical use.

Get Citation

Keywords

breast cancer; biomarkers; personalized diagnostics; anti-cancer therapy; adverse events

About this article
Title

The importance of selected biomarkers in the clinical practice of breast cancer patients

Journal

Nowotwory. Journal of Oncology

Issue

Ahead of print

Article type

Review paper

Published online

2023-08-17

Page views

51

Article views/downloads

38

DOI

10.5603/njo.95605

Keywords

breast cancer
biomarkers
personalized diagnostics
anti-cancer therapy
adverse events

Authors

Agata Makówka
Beata Kotowicz

References (86)
  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021; 71(3): 209–249.
  2. www.wcrf.org/cancer-trends/breast-cancer-statistics.
  3. Wojciechowska U, Barańska K, Michałek I, et al. Cancer in Poland in 2020. National Cancer Registry. Ministry of Health, Warsaw 2022.
  4. Youn HJo, Han W. A Review of the Epidemiology of Breast Cancer in Asia: Focus on Risk Factors. Asian Pac J Cancer Prev. 2020; 21(4): 867–880.
  5. Sinn HP, Kreipe H. A Brief Overview of the WHO Classification of Breast Tumors, 4th Edition, Focusing on Issues and Updates from the 3rd Edition. Breast Care (Basel). 2013; 8(2): 149–154.
  6. Goldhirsch A, Winer EP, Coates AS, et al. Panel members. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013; 24(9): 2206–2223.
  7. Denkert C, Liedtke C, Tutt A, et al. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet. 2017; 389(10087): 2430–2442.
  8. Duffy MJ, Crown J. Precision treatment for cancer: role of prognostic and predictive markers. Crit Rev Clin Lab Sci. 2014; 51(1): 30–45.
  9. BEST (Biomarkers, EdnpointS and other Tools), FDA-NIH Biomarker Working Group. Last Updated: November 16, 2020.
  10. Wolff AC, Hammond ME, Allison KH, et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol. 2018; 36(20): 2105–2122.
  11. Nicolini A, Ferrari P, Duffy MJ. Prognostic and predictive biomarkers in breast cancer: Past, present and future. Semin Cancer Biol. 2018; 52(Pt 1): 56–73.
  12. Leclercq G. Molecular forms of the estrogen receptor in breast cancer. J Steroid Biochem Mol Biol. 2002; 80(3): 259–272.
  13. Niwińska A, Litwiniuk M. Resistance to hormonotherapy in patients with breast cancer – molecular mechanisms and clinical implications. Contemporary Oncology/Współczesna Onkologia. 2007; 11(10): 469–474.
  14. Shen K, Yu H, Xie B, et al. Anticancer or carcinogenic? The role of estrogen receptor β in breast cancer progression. Pharmacol Ther. 2023; 242: 108350.
  15. Cui X, Schiff R, Arpino G, et al. Biology of progesterone receptor loss in breast cancer and its implications for endocrine therapy. J Clin Oncol. 2005; 23(30): 7721–7735.
  16. Arpino G, Weiss H, Lee AV, et al. Estrogen receptor-positive, progesterone receptor-negative breast cancer: association with growth factor receptor expression and tamoxifen resistance. J Natl Cancer Inst. 2005; 97(17): 1254–1261.
  17. Li Z, Wei H, Li S, et al. The Role of Progesterone Receptors in Breast Cancer. Drug Des Devel Ther. 2022; 16: 305–314.
  18. Keen JC, Davidson NE. The biology of breast carcinoma. Cancer. 2003; 97(3 Suppl): 825–833.
  19. Osborne CK, Schiff R, Arpino G, et al. Endocrine responsiveness: understanding how progesterone receptor can be used to select endocrine therapy. Breast. 2005; 14(6): 458–465.
  20. Perrier A, Gligorov J, Lefèvre G, et al. The extracellular domain of Her2 in serum as a biomarker of breast cancer. Lab Invest. 2018; 98(6): 696–707.
  21. Walaszczyk A, Gabryś D. Molecular markers used in breast cancer diagnosis — cur-rent practice and future perspectives. Nowotwory. Journal of Oncology. 2018; 68(5-6): 259–267.
  22. Mass RD, Press MF, Anderson S, et al. Evaluation of clinical outcomes according to HER2 detection by fluorescence in situ hybridization in women with metastatic breast cancer treated with trastuzumab. Clin Breast Cancer. 2005; 6(3): 240–246.
  23. Vici P, Pizzuti L, Natoli C, et al. Outcomes of HER2-positive early breast cancer patients in the pre-trastuzumab and trastuzumab eras: a real-world multicenter observational analysis. The RETROHER study. Breast Cancer Res Treat. 2014; 147(3): 599–607.
  24. Coates AS, Winer EP, Goldhirsch A, et al. Panel Members. Tailoring therapies--improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol. 2015; 26(8): 1533–1546.
  25. Li J, Guan X, Fan Z, et al. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers (Basel). 2020; 12(10).
  26. Ghosh SK, Pantazopoulos P, Medarova Z, et al. Expression of underglycosylated MUC1 antigen in cancerous and adjacent normal breast tissues. Clin Breast Cancer. 2013; 13(2): 109–118.
  27. Duffy MJ, Evoy D, McDermott EW. CA 15-3: uses and limitation as a biomarker for breast cancer. Clin Chim Acta. 2010; 411(23-24): 1869–1874.
  28. Lin DC, Genzen JR. Concordance analysis of paired cancer antigen (CA) 15-3 and 27.29 testing. Breast Cancer Res Treat. 2018; 167(1): 269–276.
  29. Rack B, Jückstock J, Trapp E, et al. SUCCESS Study Group. CA27.29 as a tumour marker for risk evaluation and therapy monitoring in primary breast cancer patients. Tumour Biol. 2016; 37(10): 13769–13775.
  30. Jeong S, Park MJ, Song W, et al. Current immunoassay methods and their applications to clinically used biomarkers of breast cancer. Clin Biochem. 2020; 78: 43–57.
  31. Baskić D, Ristić P, Matić S, et al. Clinical evaluation of the simultaneous determination of CA 15-3, CA 125 and sHER2 in breast cancer. Biomarkers. 2007; 12(6): 657–667.
  32. Uygur MM, Gümüş M. The utility of serum tumor markers CEA and CA 15-3 for breast cancer prognosis and their association with clinicopathological parameters. Cancer Treat Res Commun. 2021; 28: 100402.
  33. Shao Y, Sun X, He Y, et al. Elevated Levels of Serum Tumor Markers CEA and CA15-3 Are Prognostic Parameters for Different Molecular Subtypes of Breast Cancer. PLoS One. 2015; 10(7): e0133830.
  34. Li J, Liu L, Feng Z, et al. Tumor markers CA15-3, CA125, CEA and breast cancer survival by molecular subtype: a cohort study. Breast Cancer. 2020; 27(4): 621–630.
  35. Li H, Chen K, Su F, et al. Preoperative CA 15-3 levels predict the prognosis of nonmetastatic luminal A breast cancer. J Surg Res. 2014; 189(1): 48–56.
  36. Spiliotaki M, Mavroudis D, Kapranou K, et al. Evaluation of proliferation and apoptosis markers in circulating tumor cells of women with early breast cancer who are candidates for tumor dormancy. Breast Cancer Res. 2014; 16(6): 485.
  37. Xie S, Ding X, Mo W, et al. Serum tissue polypeptide-specific antigen is an independent predictor in breast cancer. Acta Histochem. 2014; 116(2): 372–376.
  38. Barak V, Goike H, Panaretakis KW, et al. Clinical utility of cytokeratins as tumor markers. Clin Biochem. 2004; 37(7): 529–540.
  39. Seale KN, Tkaczuk KHR. Circulating Biomarkers in Breast Cancer. Clin Breast Cancer. 2022; 22(3): e319–e331.
  40. Perrier A, Gligorov J, Lefèvre G, et al. The extracellular domain of Her2 in serum as a biomarker of breast cancer. Lab Invest. 2018; 98(6): 696–707.
  41. Di Gioia D, Dresse M, Mayr D, et al. Serum HER2 in combination with CA 15-3 as a parameter for prognosis in patients with early breast cancer. Clin Chim Acta. 2015; 440: 16–22.
  42. Darlix A, Lamy PJ, Lopez-Crapez E, et al. Serum NSE, MMP-9 and HER2 extracellular domain are associated with brain metastases in metastatic breast cancer patients: predictive biomarkers for brain metastases? Int J Cancer. 2016; 139(10): 2299–2311.
  43. Gonzalez-Avila G, Sommer B, Mendoza-Posada DA, et al. Matrix metalloproteinases participation in the metastatic process and their diagnostic and therapeutic applications in cancer. Crit Rev Oncol Hematol. 2019; 137: 57–83.
  44. Habanjar O, Bingula R, Decombat C, et al. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci. 2023; 24(4).
  45. Atoum MF, Alzoughool F, Al-Hourani H. Linkage Between Obesity Leptin and Breast Cancer. Breast Cancer (Auckl). 2020; 14: 1178223419898458.
  46. 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(302): 302ra133.
  47. Larsson AM, Jansson S, Bendahl PO, et al. Longitudinal enumeration and cluster evaluation of circulating tumor cells improve prognostication for patients with newly diagnosed metastatic breast cancer in a prospective observational trial. Breast Cancer Res. 2018; 20(1): 48.
  48. Liu MC, Shields PG, Warren RD, et al. Circulating tumor cells: a useful predictor of treatment efficacy in metastatic breast cancer. J Clin Oncol. 2009; 27(31): 5153–5159.
  49. Alimirzaie S, Bagherzadeh M, Akbari MR. Liquid biopsy in breast cancer: A comprehensive review. Clin Genet. 2019; 95(6): 643–660.
  50. Aktas B, Müller V, Tewes M, et al. Comparison of estrogen and progesterone receptor status of circulating tumor cells and the primary tumor in metastatic breast cancer patients. Gynecol Oncol. 2011; 122(2): 356–360.
  51. Sant M, Bernat-Peguera A, Felip E, et al. Role of ctDNA in Breast Cancer. Cancers (Basel). 2022; 14(2).
  52. Phallen J, Sausen M, Adleff V, et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 2017; 9(403).
  53. Davis AA, Zhang Q, Gerratana L, et al. Association of a novel circulating tumor DNA next-generating sequencing platform with circulating tumor cells (CTCs) and CTC clusters in metastatic breast cancer. Breast Cancer Res. 2019; 21(1): 137.
  54. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008; 14(9): 985–990.
  55. Hrebien S, Citi V, Garcia-Murillas I, et al. Early ctDNA dynamics as a surrogate for progression-free survival in advanced breast cancer in the BEECH trial. Ann Oncol. 2019; 30(6): 945–952.
  56. Chen M, Zhao H. Next-generation sequencing in liquid biopsy: cancer screening and early detection. Hum Genomics. 2019; 13(1): 34.
  57. Li H, Liu J, Chen J, et al. A serum microRNA signature predicts trastuzumab benefit in HER2-positive metastatic breast cancer patients. Nat Commun. 2018; 9(1): 1614.
  58. Park HS, Han HJu, Lee S, et al. Detection of Circulating Tumor Cells in Breast Cancer Patients Using Cytokeratin-19 Real-Time RT-PCR. Yonsei Med J. 2017; 58(1): 19–26.
  59. He Z, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med (Berl). 2003; 81(10): 600–612.
  60. Koo DH, Park CY, Lee ES, et al. Progranulin as a prognostic biomarker for breast cancer recurrence in patients who had hormone receptor-positive tumors: a cohort study. PLoS One. 2012; 7(6): e39880.
  61. Li Lq, Huang Hl, Ping Jl, et al. Clinicopathologic and prognostic implications of progranulin in breast carcinoma. Chin Med J (Engl). 2011; 124(13): 2045–2050.
  62. Roselli S, Pundavela J, Demont Y, et al. Sortilin is associated with breast cancer aggressiveness and contributes to tumor cell adhesion and invasion. Oncotarget. 2015; 6(12): 10473–10486.
  63. Berger K, Rhost S, Rafnsdóttir S, et al. Tumor co-expression of progranulin and sortilin as a prognostic biomarker in breast cancer. BMC Cancer. 2021; 21(1): 185.
  64. Purrahman D, Mahmoudian-Sani MR, Saki N, et al. Involvement of progranulin (PGRN) in the pathogenesis and prognosis of breast cancer. Cytokine. 2022; 151: 155803.
  65. Guha R, Yue B, Dong J, et al. Anti-progranulin/GP88 antibody AG01 inhibits triple negative breast cancer cell proliferation and migration. Breast Cancer Res Treat. 2021; 186(3): 637–653.
  66. Rhost S, Hughes É, Harrison H, et al. Sortilin inhibition limits secretion-induced progranulin-dependent breast cancer progression and cancer stem cell expansion. Breast Cancer Res. 2018; 20(1): 137.
  67. Iacopetta D, Rechoum Y, Fuqua SAw. The Role of Androgen Receptor in Breast Cancer. Drug Discov Today Dis Mech. 2012; 9(1-2): e19–e27.
  68. Kono M, Fujii T, Lim B, et al. Androgen Receptor Function and Androgen Receptor-Targeted Therapies in Breast Cancer: A Review. JAMA Oncol. 2017; 3(9): 1266–1273.
  69. Zatloukalová P, Pjechová M, Babčanová S, et al. [The Role of PD-1/PD-L1 Signaling Pathway in Antitumor Immune Response]. Klin Onkol. 2016; 29 Suppl 4(Suppl 4): 72–77.
  70. Schütz F, Stefanovic S, Mayer L, et al. PD-1/PD-L1 Pathway in Breast Cancer. Oncol Res Treat. 2017; 40(5): 294–297.
  71. Bou Zerdan M, Ghorayeb T, Saliba F, et al. Triple Negative Breast Cancer: Updates on Classification and Treatment in 2021. Cancers (Basel). 2022; 14(5).
  72. Schick J, Ritchie RP, Restini C. Breast Cancer Therapeutics and Biomarkers: Past, Present, and Future Approaches. Breast Cancer (Auckl). 2021; 15: 1178223421995854.
  73. Emens LA. Breast Cancer Immunotherapy: Facts and Hopes. Clin Cancer Res. 2018; 24(3): 511–520.
  74. Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative mean? J Exp Med. 2016; 213(13): 2835–2840.
  75. Tavares DF, Chaves Ribeiro V, Andrade MA, et al. Immunotherapy using PD-1/PDL-1 inhibitors in metastatic triple-negative breast cancer: A systematic review. Oncol Rev. 2021; 15(2): 497.
  76. Adams S, Gray RJ, Demaria S, et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol. 2014; 32(27): 2959–2966.
  77. Loi S, Michiels S, Salgado R, et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol. 2014; 25(8): 1544–1550.
  78. Lenneman CG, Sawyer DB. Cardio-Oncology: An Update on Cardiotoxicity of Cancer-Related Treatment. Circ Res. 2016; 118(6): 1008–1020.
  79. Kufel-Grabowska J, Katarzyński S, Szmit S, et al. Cardiotoxicity in patients with early breast cancer treated with adjuvant trastuzumab. Nowotwory. Journal of Oncology. 2022; 72(5): 288–293.
  80. Chaikijurajai T, Tang WH. Reappraisal of Inflammatory Biomarkers in Heart Failure. Curr Heart Fail Rep. 2020; 17(1): 9–19.
  81. Cardinale D, Sandri MT. Role of biomarkers in chemotherapy-induced cardiotoxicity. Prog Cardiovasc Dis. 2010; 53(2): 121–129.
  82. Muir CA, Clifton-Bligh RJ, Long GV, et al. Thyroid Immune-related Adverse Events Following Immune Checkpoint Inhibitor Treatment. J Clin Endocrinol Metab. 2021; 106(9): e3704–e3713.
  83. Gumusay O, Callan J, Rugo HS. Immunotherapy toxicity: identification and management. Breast Cancer Res Treat. 2022; 192(1): 1–17.
  84. Huehnchen P, Schinke C, Bangemann N, et al. Neurofilament proteins as a potential biomarker in chemotherapy-induced polyneuropathy. JCI Insight. 2022; 7(6).
  85. Burgess BL, Cho E, Honigberg L. Neurofilament light as a predictive biomarker of unresolved chemotherapy-induced peripheral neuropathy in subjects receiving paclitaxel and carboplatin. Sci Rep. 2022; 12(1): 15593.
  86. Möhn N, Mahjoub S, Duzzi L, et al. ICOG-CCCH (Immune Cooperative Oncology Group; Comprehensive Cancer Center Hannover). Monocyte chemoattractant protein 1 as a potential biomarker for immune checkpoint inhibitor-associated neurotoxicity. Cancer Med. 2023; 12(8): 9373–9383.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Wydawcą serwisu jest VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail: viamedica@viamedica.pl