655
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

“Diagnostic and Prognostic Biomarkers of Luminal Breast Cancer: Where are We Now?”

, ORCID Icon, & ORCID Icon
Pages 525-540 | Received 01 Apr 2023, Accepted 12 Jul 2023, Published online: 28 Jul 2023

References

  • Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33. doi:10.3322/caac.21708
  • Perou CM, Sørlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi:10.1038/35021093
  • Yersal O, Barutca S. Biological subtypes of breast cancer: prognostic and therapeutic implications. World J Clin Oncol. 2014;5:412–424. doi:10.5306/wjco.v5.i3.412
  • Richards MA, Westcombe AM, Love SB, Littlejohns P, Ramirez AJ. Influence of delay on survival in patients with breast cancer: a systematic review. Lancet. 1999;353:1119–1126. doi:10.1016/s0140-6736(99)02143-1
  • Waks AG, Winer EP. Breast Cancer Treatment: a Review. JAMA. 2019;321:288–300. doi:10.1001/jama.2018.19323
  • Hayes DF, Isaacs C, Stearns V. Prognostic factors in breast cancer: current and new predictors of metastasis. J Mammary Gland Biol Neoplasia. 2001;6:375–392. doi:10.1023/a:1014778713034
  • Weigel MT, Dowsett M. Current and emerging biomarkers in breast cancer: prognosis and prediction. Endocr Relat Cancer. 2010;17:R245–262. doi:10.1677/ERC-10-0136
  • Denoix PF, Baclesse F. Presentation of the project for clinical classification of malignant breast tumors by the international union for cancer control. Mem Acad Chir. 1956;82:407–410.
  • Toft D, Shyamala G, Gorski J. A receptor molecule for estrogens: studies using a cell-free system. Proc Natl Acad Sci U S A. 1967;57:1740–1743. doi:10.1073/pnas.57.6.1740
  • Jensen EV, DeSombre ER. Estrogen-receptor interaction. Science. 1973;182:126–134. doi:10.1126/science.182.4108.126
  • Rakha EA, Reis-Filho JS, Ellis IO. Combinatorial biomarker expression in breast cancer. Breast Cancer Res Treat. 2010;120:293–308. doi:10.1007/s10549-010-0746-x
  • Slamon DJ, Clark GM, Wong SG, et al. Human Breast Cancer: Correlation of Relapse and Survival with Amplification of the HER-2/ neu Oncogene. Science. 1987;235:177–182. doi:10.1126/science.3798106
  • Kolata G. Breaking ranks, lab offers test to assess risk of breast cancer. N Y Times Web. 1996;A1–A15.
  • Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci US A. 2001;98:10869–10874. doi:10.1073/pnas.191367098
  • Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol. 2009;27:1160–1167. doi:10.1200/JCO.2008.18.1370
  • Sotiriou C, Pusztai L. Gene-expression signatures in breast cancer. N Engl J Med. 2009;360:790–800. doi:10.1056/NEJMra0801289
  • Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;351:2817–2826. doi:10.1056/NEJMoa041588
  • Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med. 2015;373:2005–2014. doi:10.1056/NEJMoa1510764
  • Dowsett M, Nielsen TO, A’Hern R, et al. Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. J Natl Cancer Inst. 2011;103:1656–1664. doi:10.1093/jnci/djr393
  • Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi:10.1038/nature11412
  • Cole MP, Jones CT, Todd ID. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474. Br J Cancer. 1971;25:270–275. doi:10.1038/bjc.1971.33
  • Baum M. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial. Lancet. 2002;359:2131–2139. doi:10.1016/s0140-6736(02)09088-8
  • Howell A, Robertson JFR, Quaresma Albano J, et al. Fulvestrant,formerly ICI 182,780,is as effective as anastrozole in postmenopausal women with advanced breast cancer progressing after prior endocrine treatment. J Clin Oncol. 2002;20:3396–3403. doi:10.1200/JCO.2002.10.057
  • Pegram MD, Lipton A, Hayes DF, et al. Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. J Clin Oncol. 1998;16:2659–2671. doi:10.1200/JCO.1998.16.8.2659
  • Geyer CE, Cameron D, Lindquist D. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733–2743. doi:10.1056/NEJMoa064320
  • Baselga J, Cortés J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109–119. doi:10.1056/NEJMoa1113216
  • Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783–1791. doi:10.1056/NEJMoa1209124
  • Modi S, Jacot W, Yamashita T, et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N Engl J Med. 2022;387:9–20. doi:10.1056/NEJMoa2203690
  • Robson M, Im S-A, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377:523–533. doi:10.1056/NEJMoa1706450
  • Litton JK, Rugo HS, Ettl J, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379:753–763. doi:10.1056/NEJMoa1802905
  • Piccart M, Sørlie T, Perou CM, et al. 70-gene signature as an aid for treatment decisions in early breast cancer: updated results of the phase 3 randomised MINDACT trial with an exploratory analysis by age. Lancet Oncol. 2021;22:476–488. doi:10.1016/S1470-2045(21)00007-3
  • Andre F, Ismaila N, Henry NL, et al. Use of biomarkers to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer: ASCO clinical practice guideline update-integration of results from TAILORx. J Clin Oncol. 2019;37:1956–1964. doi:10.1200/JCO.19.00945
  • Kalinsky K, Barlow WE, Gralow JR, et al. 21-gene assay to inform chemotherapy benefit in node-positive breast cancer. N Engl J Med. 2021;385:2336–2347. doi:10.1056/NEJMoa2108873
  • Andre F. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N Engl J Med. 2019;380:1929–1940. doi:10.1056/NEJMoa1813904
  • Finn RS, Martin M, Rugo HS, et al. Palbociclib and letrozole in advanced breast cancer. N Engl J Med. 2016;375(20):1925–1936. doi:10.1056/NEJMoa1607303
  • Hortobagyi GN, Stemmer SM, Burris HA, et al. Ribociclib as First-Line Therapy for HR-Positive, Advanced Breast Cancer. N Engl J Med. 2016;375:1738–1748. doi:10.1056/NEJMoa1609709
  • Sledge GW Jr, Toi M, Neven P, et al. MONARCH 2: abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol. 2017;35:2875–2884. doi:10.1200/JCO.2017.73.7585
  • Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101:1446–1452. doi:10.1093/jnci/djp335
  • Atkins D. Grading quality of evidence and strength of recommendations. BMJ. 2004;328:1490. doi:10.1136/bmj.328.7454.1490
  • Ebell MH, Siwek J, Weiss BD, et al. Strength of recommendation taxonomy (SORT): a patient-centered approach to grading evidence in the medical literature. J Am Board Fam Pract. 2004;17:59–67. doi:10.3122/jabfm.17.1.59
  • McShane LM, Chauhan SP, Ward TC, et al. Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst. 2005;97:1180–1184. doi:10.1093/jnci/dji237
  • Giuliano AE, Edge SB, Hortobagyi GN. Eighth edition of the AJCC cancer staging manual: breast cancer. Ann Surg Oncol. 2018;25:1783–1785. doi:10.1245/s10434-018-6486-6
  • Narod SA. Tumour size predicts long-term survival among women with lymph node-positive breast cancer. Curr Oncol. 2012;19:249–253. doi:10.3747/co.19.1043
  • Sopik V, Narod SA. The relationship between tumour size, nodal status and distant metastases: on the origins of breast cancer. Breast Cancer Res Treat. 2018;170:647–656. doi:10.1007/s10549-018-4796-9
  • Schijven MP, Vingerhoets AJ, Rutten HJ, et al. Comparison of morbidity between axillary lymph node dissection and sentinel node biopsy. Eur J Surg Oncol. 2003;29:341–350. doi:10.1053/ejso.2002.1385
  • Galimberti V, Cole BF, Zurrida S, et al. Axillary dissection versus no axillary dissection in patients with sentinel-node micrometastases (IBCSG 23-01): a phase 3 randomised controlled trial. Lancet Oncol. 2013;14:297–305. doi:10.1016/S1470-2045(13)70035-4
  • Hersh EH, King TA. De-escalating axillary surgery in early-stage breast cancer. Breast. 2022;62(1):S43–S49. doi:10.1016/j.breast.2021.11.018
  • Gennari A, André F, Barrios CH, et al. ESMO clinical practice guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol. 2021;32:1475–1495. doi:10.1016/j.annonc.2021.09.019
  • Groheux D, Cochet A, Humbert O, et al. 18 F-FDG PET/CT for Staging and Restaging of Breast Cancer. J Nucl Med. 2016;57(1):17S–26S. doi:10.2967/jnumed.115.157859
  • Groheux D, Hindie E. Breast cancer: initial workup and staging with FDG PET/CT. Clin Transl Imaging. 2021;9:221–231. doi:10.1007/s40336-021-00426-z
  • Arnaout A, Varela NP, Allarakhia M, et al. Baseline staging imaging for distant metastasis in women with stages I, II, and III breast cancer. Curr Oncol. 2020;27:e123–e145. doi:10.3747/co.27.6147
  • Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology. 2002;41:154–161.
  • Ellis IO, GALEA M, BROUGHTON N, et al. Pathological prognostic factors in breast cancer. II. Histological type. Relationship with survival in a large study with long-term follow-up. Histopathology. 1992;20:479–489. doi:10.1111/j.1365-2559.1992.tb01032.x
  • Galea MH, Blamey RW, Elston CE, Ellis IO. The Nottingham Prognostic Index in primary breast cancer. Breast Cancer Res Treat. 1992;22:207–219. doi:10.1007/BF01840834
  • Rakha EA, Reis-Filho JS, Baehner F, et al. Breast cancer prognostic classification in the molecular era: the role of histological grade. Breast Cancer Res. 2010;12:207. doi:10.1186/bcr2607
  • Sundquist M, Thorstenson S, Brudin L, Nordenskjold B. Applying the Nottingham Prognostic Index to a Swedish breast cancer population. South East Swedish Breast Cancer Study Group. Breast Cancer Res Treat. 1999;53:1–8. doi:10.1023/a:1006052115874
  • Rakha EA, El-Sayed ME, Lee AHS, et al. Prognostic significance of Nottingham histologic grade in invasive breast carcinoma. J Clin Oncol. 2008;26:3153–3158. doi:10.1200/JCO.2007.15.5986
  • Henson DE, Ries L, Freedman LS, Carriaga M. Relationship among outcome, stage of disease, and histologic grade for 22,616 cases of breast cancer. The basis for a prognostic index. Cancer. 1991;68:2142–2149. doi:10.1002/1097-0142(19911115)68:10<2142::aid-cncr2820681010>3.0.co;2-d
  • Duffy MJ, Harbeck N, Nap M, et al. Clinical use of biomarkers in breast cancer: updated guidelines from the European Group on Tumor Markers (EGTM). Eur J Cancer. 2017;75:284–298. doi:10.1016/j.ejca.2017.01.017
  • 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;20:691–722. doi:10.6004/jnccn.2022.0030
  • Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-updagger. Ann Oncol. 2019;30:1194–1220. doi:10.1093/annonc/mdz173
  • Early Breast Cancer Trialists’ Collaborative, G, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet. 2011;378:771–784. doi:10.1016/S0140-6736(11)60993-8
  • Elledge RM, Green S, Pugh R, et al. Estrogen receptor (ER) and progesterone receptor (PgR), by ligand-binding assay compared with ER, PgR and pS2, by immuno-histochemistry in predicting response to tamoxifen in metastatic breast cancer: a Southwest Oncology Group Study. Int J Cancer. 2000;89:111–117. doi:10.1002/(SICI)1097-0215(20000320)89:2<111::AID-IJC2>3.0.CO;2-W
  • Ruhstaller T, Giobbie-Hurder A, Colleoni M, et al. Adjuvant Letrozole and Tamoxifen Alone or Sequentially for Postmenopausal Women With Hormone Receptor-Positive Breast Cancer: Long-Term Follow-Up of the BIG 1-98 Trial. J Clin Oncol. 2019;37:105–114. doi:10.1200/JCO.18.00440
  • Howell A. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years’ adjuvant treatment for breast cancer. Lancet. 2005;365:60–62. doi:10.1016/S0140-6736(04)17666-6
  • Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors alpha (ERalpha) and beta (ERbeta): subtype-selective ligands and clinical potential. Steroids. 2014;90:13–29. doi:10.1016/j.steroids.2014.06.012
  • Kastner P, Krust A, Turcotte B, et al. Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J. 1990;9:1603–1614. doi:10.1002/j.1460-2075.1990.tb08280.x
  • Yu WC, Leung BS, Gao YL. Effects of 17 beta-estradiol on progesterone receptors and the uptake of thymidine in human breast cancer cell line CAMA-1. Cancer Res. 1981;41:5004–5009.
  • Hilton HN, Clarke CL, Graham JD. Estrogen and progesterone signalling in the normal breast and its implications for cancer development. Mol Cell Endocrinol. 2018;466:2–14. doi:10.1016/j.mce.2017.08.011
  • Hammond ME, Hayes DF, Dowsett M, et al. American society of clinical oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 2010;28:2784–2795. doi:10.1200/JCO.2009.25.6529
  • Dowsett M, Houghton J, Iden C, et al. Benefit from adjuvant tamoxifen therapy in primary breast cancer patients according oestrogen receptor, progesterone receptor, EGF receptor and HER2 status. Ann Oncol. 2006;17:818–826. doi:10.1093/annonc/mdl016
  • Ravdin PM, Green S, Dorr TM, et al. Prognostic significance of progesterone receptor levels in estrogen receptor-positive patients with metastatic breast cancer treated with tamoxifen: results of a prospective Southwest Oncology Group study. J Clin Oncol. 1992;10:1284–1291. doi:10.1200/JCO.1992.10.8.1284
  • Clark GM, Osborne CK, McGuire WL. Correlations between estrogen receptor, progesterone receptor, and patient characteristics in human breast cancer. J Clin Oncol. 1984;2:1102–1109. doi:10.1200/JCO.1984.2.10.1102
  • Reinert T, Cascelli F, Resende CAAD, et al. Clinical implication of low estrogen receptor (ER-low) expression in breast cancer. Front Endocrinol (Lausanne). 2022;13:1015388. doi:10.3389/fendo.2022.1015388
  • Allison KH, Hammond MEH, Dowsett M, et al. Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Guideline Update. J Clin Oncol. 2020;38:1346–1366. doi:10.1200/JCO.19.02309
  • Harvey JM, Clark GM, Osborne CK, Allred DC. Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol. 1999;17:1474–1481. doi:10.1200/JCO.1999.17.5.1474
  • Remmele W, Stegner HE. Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue. Pathologe. 1987;8:138–140.
  • Pecci A, Ogara MF, Sanz RT, Vicent GP. Choosing the right partner in hormone-dependent gene regulation: Glucocorticoid and progesterone receptors crosstalk in breast cancer cells. Front Endocrinol (Lausanne). 2022;13:1037177. doi:10.3389/fendo.2022.1037177
  • Belachew EB, Sewasew DT. Molecular mechanisms of endocrine resistance in estrogen-positive breast cancer. Front Endocrinol (Lausanne). 2021;12:599586. doi:10.3389/fendo.2021.599586
  • Roskoski R Jr. Cyclin-dependent protein kinase inhibitors including palbociclib as anticancer drugs. Pharmacol Res. 2016;107:249–275. doi:10.1016/j.phrs.2016.03.012
  • Andrade de Oliveira K, Sengupta S, Yadav AK, Clarke R. The complex nature of heterogeneity and its roles in breast cancer biology and therapeutic responsiveness. Front Endocrinol (Lausanne). 2023;14:1083048. doi:10.3389/fendo.2023.1083048
  • Sgroi DC, Carney E, Zarrella E, et al. Prediction of late disease recurrence and extended adjuvant letrozole benefit by the HOXB13/IL17BR biomarker. J Natl Cancer Inst. 2013;105:1036–1042. doi:10.1093/jnci/djt146
  • Sgroi DC, Sestak I, Cuzick J, et al. Prediction of late distant recurrence in patients with oestrogen-receptor-positive breast cancer: a prospective comparison of the breast-cancer index (BCI) assay, 21-gene recurrence score, and IHC4 in the TransATAC study population. Lancet Oncol. 2013;14:1067–1076. doi:10.1016/S1470-2045(13)70387-5
  • Gerdes J, Schwab U, Lemke H, Stein H. Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer. 1983;31:13–20. doi:10.1002/ijc.2910310104
  • Lopez F, Belloc F, Lacombe F, et al. Modalities of synthesis of Ki67 antigen during the stimulation of lymphocytes. Cytometry. 1991;12:42–49. doi:10.1002/cyto.990120107
  • Guarneri V, Piacentini F, Ficarra G, et al. A prognostic model based on nodal status and Ki-67 predicts the risk of recurrence and death in breast cancer patients with residual disease after preoperative chemotherapy. Ann Oncol. 2009;20:1193–1198. doi:10.1093/annonc/mdn761
  • Urruticoechea A, Smith IE, Dowsett M. Proliferation marker Ki-67 in early breast cancer. J Clin Oncol. 2005;23:7212–7220. doi:10.1200/JCO.2005.07.501
  • Goldhirsch A, Wood WC, Coates AS, et al. Strategies for subtypes--dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer 2011. Ann Oncol. 2011;22:1736–1747. doi:10.1093/annonc/mdr304
  • Goldhirsch A, Winer EP, Coates AS, et al. 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;24:2206–2223. doi:10.1093/annonc/mdt303
  • Luporsi E, André F, Spyratos F, et al. Ki-67: level of evidence and methodological considerations for its role in the clinical management of breast cancer: analytical and critical review. Breast Cancer Res Treat. 2012;132:895–915. doi:10.1007/s10549-011-1837-z
  • Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA. Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol. 2010;11:174–183. doi:10.1016/S1470-2045(09)70262-1
  • Viale G, Giobbie-Hurder A, Regan MM, et al. Prognostic and predictive value of centrally reviewed Ki-67 labeling index in postmenopausal women with endocrine-responsive breast cancer: results from Breast International Group Trial 1-98 comparing adjuvant tamoxifen with letrozole. J Clin Oncol. 2008;26:5569–5575. doi:10.1200/JCO.2008.17.0829
  • Dumontet C, Krajewska M, Treilleux I, et al. BCIRG 001 molecular analysis: prognostic factors in node-positive breast cancer patients receiving adjuvant chemotherapy. Clin Cancer Res. 2010;16:3988–3997. doi:10.1158/1078-0432.CCR-10-0079
  • Anderson RL, Balasas T, Callaghan J, et al. A framework for the development of effective anti-metastatic agents. Nat Rev Clin Oncol. 2019;16:185–204. doi:10.1038/s41571-018-0134-8
  • Polley MY, Leung SCY, Gao D, et al. An international study to increase concordance in Ki67 scoring. Mod Pathol. 2015;28:778–786. doi:10.1038/modpathol.2015.38
  • Varga Z, Diebold J, Dommann-Scherrer C, et al. How reliable is Ki-67 immunohistochemistry in grade 2 breast carcinomas? A QA study of the Swiss Working Group of Breast- and Gynecopathologists. PLoS One. 2012;7:e37379. doi:10.1371/journal.pone.0037379
  • Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/ neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–712. doi:10.1126/science.2470152
  • Wolff AC, Elizabeth Hale Hammond M, 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;36:2105–2122. doi:10.1200/JCO.2018.77.8738
  • Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol. 2002;20:719–726. doi:10.1200/JCO.2002.20.3.719
  • Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. doi:10.1056/NEJM200103153441101
  • Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107:1039–1046. doi:10.1111/cas.12966
  • Tarantino P, Hamilton E, Tolaney SM, et al. HER2-low breast cancer: pathological and clinical landscape. J Clin Oncol. 2020;38:1951–1962. doi:10.1200/JCO.19.02488
  • Zhang X, Huang AC, Chen F, et al. Novel development strategies and challenges for anti-Her2 antibody-drug conjugates. Antib Ther. 2022;5:18–29. doi:10.1093/abt/tbac001
  • Li J, Ma M, Yang X, et al. Circular HER2 RNA positive triple negative breast cancer is sensitive to Pertuzumab. Mol Cancer. 2020;19:142. doi:10.1186/s12943-020-01259-6
  • Pegram M, Jackisch C, Johnston SRD. Estrogen/HER2 receptor crosstalk in breast cancer: combination therapies to improve outcomes for patients with hormone receptor-positive/HER2-positive breast cancer. NPJ Breast Cancer. 2023;9:45. doi:10.1038/s41523-023-00533-2
  • Burstein HJ. Systemic Therapy for Estrogen Receptor-Positive, HER2-Negative Breast Cancer. N Engl J Med. 2020;383:2557–2570. doi:10.1056/NEJMra1307118
  • 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;26:5172–5177. doi:10.1158/1078-0432.CCR-20-0224
  • Hurtado A, Holmes KA, Ross-Innes CS, Schmidt D, Carroll JS. FOXA1 is a key determinant of estrogen receptor function and endocrine response. Nat Genet. 2011;43:27–33. doi:10.1038/ng.730
  • Mukherjee S, Conrad SE. c-Myc suppresses p21WAF1/CIP1 expression during estrogen signaling and antiestrogen resistance in human breast cancer cells. J Biol Chem. 2005;280:17617–17625. doi:10.1074/jbc.M502278200
  • Zwijsen RM, Wientjens E, Klompmaker R, et al. CDK-independent activation of estrogen receptor by cyclin D1. Cell. 1997;88:405–415. doi:10.1016/s0092-8674(00)81879-6
  • Sabine VS, Crozier C, Brookes CL, et al. Mutational analysis of PI3K/AKT signaling pathway in tamoxifen exemestane adjuvant multinational pathology study. J Clin Oncol. 2014;32:2951–2958. doi:10.1200/JCO.2013.53.8272
  • Bosch A, Li Z, Bergamaschi A, et al. PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer. Sci Transl Med. 2015;7:283ra251. doi:10.1126/scitranslmed.aaa4442
  • Henry NL, Somerfield MR, Dayao Z, et al. Biomarkers for systemic therapy in metastatic breast cancer: ASCO guideline update. J Clin Oncol. 2022;40:3205–3221. doi:10.1200/JCO.22.01063
  • Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366:520–529. doi:10.1056/NEJMoa1109653
  • Spring LM, Wander SA, Andre F, et al. Cyclin-dependent kinase 4 and 6 inhibitors for hormone receptor-positive breast cancer: past, present, and future. Lancet. 2020;395:817–827. doi:10.1016/S0140-6736(20)30165-3
  • Turner NC, Slamon DJ, Ro J, et al. Overall survival with palbociclib and fulvestrant in advanced breast cancer. N Engl J Med. 2018;379:1926–1936. doi:10.1056/NEJMoa1810527
  • Im SA, Lu Y-S, Bardia A, et al. Overall survival with ribociclib plus endocrine therapy in breast cancer. N Engl J Med. 2019;381:307–316. doi:10.1056/NEJMoa1903765
  • Sledge GW Jr, Toi M, Neven P, et al. The effect of abemaciclib plus fulvestrant on overall survival in hormone receptor-positive, ERBB2-negative breast cancer that progressed on endocrine therapy-MONARCH 2: a Randomized clinical trial. JAMA Oncol. 2020;6:116–124. doi:10.1001/jamaoncol.2019.4782
  • Cottu P, D’Hondt V, Dureau S, et al. Letrozole and palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer. Ann Oncol. 2018;29:2334–2340. doi:10.1093/annonc/mdy448
  • Johnston SRD, Harbeck N, Hegg R, et al. Abemaciclib combined with endocrine therapy for the adjuvant treatment of HR+, HER2-, node-positive, high-risk, early breast cancer (monarchE). J Clin Oncol. 2020;38:3987–3998. doi:10.1200/JCO.20.02514
  • Trenner A, Sartori AA. Harnessing DNA double-strand break repair for cancer treatment. Front Oncol. 2019;9:1388. doi:10.3389/fonc.2019.01388
  • Kurian AW, Gong GD, John EM, et al. Performance of prediction models for BRCA mutation carriage in three racial/ethnic groups: findings from the Northern California breast cancer family registry. Cancer Epidemiol Biomarkers Prev. 2009;18:1084–1091. doi:10.1158/1055-9965.EPI-08-1090
  • Malone KE, Daling JR, Doody DR, et al. Prevalence and Predictors of BRCA1 and BRCA2 Mutations in a Population-Based Study of Breast Cancer in White and Black American Women Ages 35 to 64 Years. Cancer Res. 2006;66:8297–8308. doi:10.1158/0008-5472.CAN-06-0503
  • Harbeck N, Schmitt M, Meisner C, et al. Ten-year analysis of the prospective multicentre Chemo-N0 trial validates American Society of Clinical Oncology (ASCO)-recommended biomarkers uPA and PAI-1 for therapy decision making in node-negative breast cancer patients. Eur J Cancer. 2013;49:1825–1835. doi:10.1016/j.ejca.2013.01.007
  • Harbeck N, Kates RE, Look MP, et al. Enhanced benefit from adjuvant chemotherapy in breast cancer patients classified high-risk according to urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor type 1 (n = 3424). Cancer Res. 2002;62(62):4617–4622.
  • Cheung KL, Graves CR, Robertson JF. Tumour marker measurements in the diagnosis and monitoring of breast cancer. Cancer Treat Rev. 2000;26:91–102. doi:10.1053/ctrv.1999.0151
  • Harris L, Fritsche H, Mennel R, et al. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol. 2007;25:5287–5312. doi:10.1200/JCO.2007.14.2364
  • Kwa M, Makris A, Esteva FJ. Clinical utility of gene-expression signatures in early stage breast cancer. Nat Rev Clin Oncol. 2017;14:595–610. doi:10.1038/nrclinonc.2017.74
  • Sparano JA, Bornschein R, Brown K, et al. adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med. 2018;379:111–121. doi:10.1056/NEJMoa1804710
  • Dubsky P, Brase JC, Jakesz R, et al. The EndoPredict score provides prognostic information on late distant metastases in ER+/HER2- breast cancer patients. Br J Cancer. 2013;109:2959–2964. doi:10.1038/bjc.2013.671
  • Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006;24:3726–3734. doi:10.1200/JCO.2005.04.7985
  • Mamounas EP, Tang G, Fisher B, et al. Association between the 21-gene recurrence score assay and risk of locoregional recurrence in node-negative, estrogen receptor-positive breast cancer: results from NSABP B-14 and NSABP B-20. J Clin Oncol. 2010;28:1677–1683. doi:10.1200/JCO.2009.23.7610
  • Albain KS, Barlow WE, Shak S, et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol. 2010;11:55–65. doi:10.1016/S1470-2045(09)70314-6
  • Dowsett M, Cuzick J, Wale C, et al. Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study. J Clin Oncol. 2010;28:1829–1834. doi:10.1200/JCO.2009.24.4798
  • Drukker CA, Bueno‐de‐Mesquita JM, Retèl VP, et al. A prospective evaluation of a breast cancer prognosis signature in the observational RASTER study. Int J Cancer. 2013;133:929–936. doi:10.1002/ijc.28082
  • van ‘t Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530–536. doi:10.1038/415530a
  • van de Vijver MJ, He YD, van ‘t Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009. doi:10.1056/NEJMoa021967
  • Cardoso F, Van’t Veer LJ, Bogaerts J, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med. 2016;375:717–729. doi:10.1056/NEJMoa1602253
  • Martin M, Brase JC, Calvo L, et al. Clinical validation of the EndoPredict test in node-positive, chemotherapy-treated ER+/HER2- breast cancer patients: results from the GEICAM 9906 trial. Breast Cancer Res. 2014;16:R38. doi:10.1186/bcr3642
  • Gnant M, Filipits M, Greil R, et al. Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 postmenopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann Oncol. 2014;25:339–345. doi:10.1093/annonc/mdt494
  • Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant recurrence after 5 years of endocrine treatment: a combined analysis of patients from the Austrian breast and colorectal cancer study group 8 and arimidex, tamoxifen alone or in combination randomized trials using the PAM50 risk of recurrence score. J Clin Oncol. 2015;33:916–922. doi:10.1200/JCO.2014.55.6894
  • Bartlett JM, Bayani J, Marshall A, et al. Comparing breast cancer multiparameter tests in the OPTIMA prelim trial: no test is more equal than the others. J Natl Cancer Inst. 2016;108:djw050. doi:10.1093/jnci/djw050
  • Buus R, Sestak I, Kronenwett R, et al. Molecular Drivers of Onco type DX, Prosigna, EndoPredict, and the Breast Cancer Index: a TransATAC Study. J Clin Oncol. 2021;39:126–135. doi:10.1200/JCO.20.00853
  • Ignatiadis M, Sledge GW, Jeffrey SS. Liquid biopsy enters the clinic - implementation issues and future challenges. Nat Rev Clin Oncol. 2021;18:297–312. doi:10.1038/s41571-020-00457-x
  • Alix-Panabieres C, Pantel K. Liquid Biopsy: From Discovery to Clinical Application. Cancer Discov. 2021;11:858–873. doi:10.1158/2159-8290.CD-20-1311
  • Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367. doi:10.1126/science.aau6977
  • Hamam R, Kassem M, Zaher W, et al. Circulating microRNAs in breast cancer: novel diagnostic and prognostic biomarkers. Cell Death Dis. 2017;8:e3045. doi:10.1038/cddis.2017.440
  • Amelio I, Bernassola F, Candi E. Emerging roles of long non-coding RNAs in breast cancer biology and management. Semin Cancer Biol. 2021;72:36–45. doi:10.1016/j.semcancer.2020.06.019
  • Ring A, Nguyen-Strauli BD, Wicki A, Aceto N. Biology, vulnerabilities and clinical applications of circulating tumour cells. Nat Rev Cancer. 2022;22:1–17. doi:10.1038/s41568-022-00536-4
  • Martinez-Jimenez F, Movasati A, Brunner SR, et al. Pan-cancer whole-genome comparison of primary and metastatic solid tumours. Nature. 2023;618:333–341. doi:10.1038/s41586-023-06054-z
  • Bidard FC, Peeters DJ, Fehm T, et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol. 2014;15:406–414. doi:10.1016/S1470-2045(14)70069-5
  • Cristofanilli M, Matera J, Miller MC, 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
  • Allard WJ, Repollet M. Connelly MC, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10:6897–6904. doi:10.1158/1078-0432.CCR-04-0378
  • Hayes DF, Miller EM, GV MJ. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 2006;12:4218–4224. doi:10.1158/1078-0432.CCR-05-2821
  • Giuliano M, Giordano A, Jackson S, et al. Circulating tumor cells as prognostic and predictive markers in metastatic breast cancer patients receiving first-line systemic treatment. Breast Cancer Res. 2011;13:R67. doi:10.1186/bcr2907
  • Rack B, Schindlbeck C, Jückstock J, et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J Natl Cancer Inst. 2014;106. doi:10.1093/jnci/dju066
  • Ignatiadis M, Litière S, Rothe F, et al. Trastuzumab versus observation for HER2 nonamplified early breast cancer with circulating tumor cells (EORTC 90091-10093, BIG 1-12, Treat CTC): a randomized phase II trial. Ann Oncol. 2018;29:1777–1783. doi:10.1093/annonc/mdy211
  • Janni WJ, Rack B, Terstappen LWMM, 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
  • Cristofanilli M, Pierga J-Y, Reuben J, et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): international expert consensus paper. Crit Rev Oncol Hematol. 2019;134:39–45. doi:10.1016/j.critrevonc.2018.12.004
  • Riethdorf S, Müller V, Zhang L, et al. Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin Cancer Res. 2010;16:2634–2645. doi:10.1158/1078-0432.CCR-09-2042
  • Pierga JY, Bidard F-C, Mathiot C, et al. Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial. Clin Cancer Res. 2008;14:7004–7010. doi:10.1158/1078-0432.CCR-08-0030
  • Georgoulias V, Bozionelou V, Agelaki S, et al. Trastuzumab decreases the incidence of clinical relapses in patients with early breast cancer presenting chemotherapy-resistant CK-19mRNA-positive circulating tumor cells: results of a randomized phase II study. Ann Oncol. 2012;23:1744–1750. doi:10.1093/annonc/mds020
  • Smerage JB, Barlow WE, Hortobagyi GN, et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol. 2014;32:3483–3489. doi:10.1200/JCO.2014.56.2561
  • Jacot W, Cottu P, Berger F, et al. Actionability of HER2-amplified circulating tumor cells in HER2-negative metastatic breast cancer: the CirCe T-DM1 trial. Breast Cancer Res. 2019;21:121. doi:10.1186/s13058-019-1215-z
  • Pestrin M, Bessi S, Puglisi F, et al. Final results of a multicenter phase II clinical trial evaluating the activity of single-agent lapatinib in patients with HER2-negative metastatic breast cancer and HER2-positive circulating tumor cells. A proof-of-concept study. Breast Cancer Res Treat. 2012;134:283–289. doi:10.1007/s10549-012-2045-1
  • Bidard FC, Jacot W, Kiavue N, et al. Efficacy of circulating tumor cell count-driven vs clinician-driven first-line therapy choice in hormone receptor-positive, ERBB2-negative metastatic breast cancer: the STIC CTC randomized clinical trial. JAMA Oncol. 2021;7:34–41. doi:10.1001/jamaoncol.2020.5660
  • Muller V, Banys-Paluchowski M, Friedl TWP, et al. Prognostic relevance of the HER2 status of circulating tumor cells in metastatic breast cancer patients screened for participation in the DETECT study program. ESMO Open. 2021;6:100299. doi:10.1016/j.esmoop.2021.100299
  • Riethdorf S, Fritsche H, Muller V, et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res. 2007;13:920–928. doi:10.1158/1078-0432.CCR-06-1695
  • Miller MC, Robinson PS, Wagner C, et al. The Parsortix Cell Separation System-A versatile liquid biopsy platform. Cytometry A. 2018;93:1234–1239. doi:10.1002/cyto.a.23571
  • Lin D, Shen L, Luo M, et al. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther. 2021;6:404. doi:10.1038/s41392-021-00817-8
  • Saucedo-Zeni N, Mewes S, Niestroj R, et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int J Oncol. 2012;41:1241–1250. doi:10.3892/ijo.2012.1557
  • Eifler RL, Lind J, Falkenhagen D, et al. Enrichment of circulating tumor cells from a large blood volume using leukapheresis and elutriation: proof of concept. Cytometry B Clin Cytom. 2011;80B:100–111. doi:10.1002/cyto.b.20560
  • Crosbie PA, Shah R, Krysiak P, et al. Circulating Tumor Cells Detected in the Tumor-Draining Pulmonary Vein Are Associated with Disease Recurrence after Surgical Resection of NSCLC. J Thorac Oncol. 2016;11:1793–1797. doi:10.1016/j.jtho.2016.06.017
  • Chemi F, Rothwell DG, McGranahan N, et al. Pulmonary venous circulating tumor cell dissemination before tumor resection and disease relapse. Nat Med. 2019;25:1534–1539. doi:10.1038/s41591-019-0593-1
  • Diamantopoulou Z, Castro-Giner F, Schwab FD, et al. The metastatic spread of breast cancer accelerates during sleep. Nature. 2022;607:156–162. doi:10.1038/s41586-022-04875-y
  • Donato C, Buczak K, Schmidt A, Aceto N. Mass spectrometry analysis of circulating breast cancer cells from a Xenograft mouse model. STAR Protoc. 2021;2:100480. doi:10.1016/j.xpro.2021.100480
  • Donato C, Kunz L, Castro-Giner F, et al. Hypoxia triggers the intravasation of clustered circulating tumor cells. Cell Rep. 2020;32:108105. doi:10.1016/j.celrep.2020.108105
  • Gkountela S, Castro-Giner F, Szczerba BM, et al. Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding. Cell. 2019;176:98–112.e114. doi:10.1016/j.cell.2018.11.046
  • Szczerba BM, Castro-Giner F, Vetter M, et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature. 2019;566:553–557. doi:10.1038/s41586-019-0915-y
  • Yu M, Bardia A, Aceto N, et al. Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science. 2014;345:216–220. doi:10.1126/science.1253533
  • 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. doi:10.1126/scitranslmed.aab0021
  • McDonald BR, Contente-Cuomo T, Sammut S-J, et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci Transl Med. 2019;11. doi:10.1126/scitranslmed.aax7392
  • Rothe F, Silva MJ, Venet D, et al. Circulating Tumor DNA in HER2-Amplified Breast Cancer: A Translational Research Substudy of the NeoALTTO Phase III Trial. Clin Cancer Res. 2019;25:3581–3588. doi:10.1158/1078-0432.CCR-18-2521
  • 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;21:1296–1308. doi:10.1016/S1470-2045(20)30444-7
  • Schiavon G, Hrebien S, Garcia-Murillas I, et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci Transl Med. 2015;7:313ra182. doi:10.1126/scitranslmed.aac7551
  • Kodahl AR, Ehmsen S, Pallisgaard N, et al. Correlation between circulating cell-free PIK 3 CA tumor DNA levels and treatment response in patients with PIK 3 CA -mutated metastatic breast cancer. Mol Oncol. 2018;12:925–935. doi:10.1002/1878-0261.12305
  • Cullinane C, Fleming C, O’Leary DP, et al. Association of circulating tumor DNA with disease-free survival in breast cancer: a systematic review and meta-analysis. JAMA Netw Open. 2020;3:e2026921. doi:10.1001/jamanetworkopen.2020.26921
  • Magbanua MJM, Swigart LB, Wu H-T, et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol. 2021;32:229–239. doi:10.1016/j.annonc.2020.11.007
  • Lipsyc-Sharf M, de Bruin EC, Santos K, et al. Circulating Tumor DNA and Late Recurrence in High-Risk Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer. J Clin Oncol. 2022;40:2408–2419. doi:10.1200/JCO.22.00908
  • Cristiano S, Leal A, Phallen J, et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature. 2019;570:385–389. doi:10.1038/s41586-019-1272-6
  • Surveillance, Epidemiology, and End Results (SEER) Program SEER*Stat Database: Mortality - All COD, Aggregated With State, Total U.S. (1969–2019) <Katrina/Rita Population Adjustment>, National Cancer Institute, DCCPS, Surveillance Research Program, released April 2021. Underlying mortality data provided by NCHS. Available from: www.cdc.gov/nchs. Accessed July 20, 2023.
  • Tevaarwerk AJ, Gray RJ, Schneider BP, et al. Survival in patients with metastatic recurrent breast cancer after adjuvant chemotherapy: little evidence of improvement over the past 30 years. Cancer. 2013;119:1140–1148. doi:10.1002/cncr.27819
  • Pedersen RN, Esen BÖ, Mellemkjær L, et al. The incidence of breast cancer recurrence 10–32 years after primary diagnosis. J Natl Cancer Inst. 2022;114:391–399. doi:10.1093/jnci/djab202
  • Hu Z, Ding J, Ma Z, et al. Quantitative evidence for early metastatic seeding in colorectal cancer. Nat Genet. 2019;51:1113–1122. doi:10.1038/s41588-019-0423-x
  • Friberg S, Nystrom A. Cancer Metastases: Early Dissemination and Late Recurrences. Cancer Growth Metastasis. 2015;8:43–49. doi:10.4137/CGM.S31244
  • Phan TG, Croucher PI. The dormant cancer cell life cycle. Nat Rev Cancer. 2020;20:398–411. doi:10.1038/s41568-020-0263-0
  • Ring A, Spataro M, Wicki A, Aceto N. Clinical and biological aspects of disseminated tumor cells and dormancy in breast cancer. Front Cell Dev Biol. 2022;10:929893. doi:10.3389/fcell.2022.929893
  • Park SY, Nam JS. The force awakens: metastatic dormant cancer cells. Exp Mol Med. 2020;52:569–581. doi:10.1038/s12276-020-0423-z