Review Article

Prediction of Late Recurrence and Distant Metastasis in Early-stage Breast Cancer: Overview of Current and Emerging Biomarkers

Author(s): A. Gouri*, B. Benarba, A. Dekaken, H. Aoures and S. Benharkat

Volume 21, Issue 10, 2020

Page: [1008 - 1025] Pages: 18

DOI: 10.2174/1389450121666200312105908

Price: $65

Abstract

Recently, a significant number of breast cancer (BC) patients have been diagnosed at an early stage. It is therefore critical to accurately predict the risk of recurrence and distant metastasis for better management of BC in this setting. Clinicopathologic patterns, particularly lymph node status, tumor size, and hormonal receptor status are routinely used to identify women at increased risk of recurrence. However, these factors have limitations regarding their predictive ability for late metastasis risk in patients with early BC. Emerging molecular signatures using gene expression-based approaches have improved the prognostic and predictive accuracy for this indication. However, the use of their based-scores for risk assessment has provided contradictory findings. Therefore, developing and using newly emerged alternative predictive and prognostic biomarkers for identifying patients at high- and low-risk is of great importance. The present review discusses some serum biomarkers and multigene profiling scores for predicting late recurrence and distant metastasis in early-stage BC based on recently published studies and clinical trials.

Keywords: Early breast cancer, late recurrence, distant metastasis, predictive biomarkers.

Next »
Graphical Abstract
[1]
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394-424.
[http://dx.doi.org/10.3322/caac.21492] [PMID: 30207593]
[2]
Bleyer A, Welch HG. Effect of three decades of screening mammography on breast-cancer incidence. N Engl J Med 2012; 367(21): 1998-2005.
[http://dx.doi.org/10.1056/NEJMoa1206809] [PMID: 23171096]
[3]
Taneja P, Maglic D, Kai F, et al. Classical and novel prognostic markers for breast cancer and their clinical significance. Clin Med Insights Oncol 2010; 4: 15-34.
[http://dx.doi.org/10.4137/CMO.S4773] [PMID: 20567632]
[4]
Carter CL, Allen C, Henson DE. Relation of tumor size, lymph node status, and survival in 24,740 breast cancer cases. Cancer 1989; 63(1): 181-7.
[http://dx.doi.org/10.1002/1097-0142(19890101)63:1<181:AID-CNCR2820630129>3.0.CO;2-H] [PMID: 2910416]
[5]
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(3): 647-56.
[http://dx.doi.org/10.1007/s10549-018-4796-9] [PMID: 29693227]
[6]
Cianfrocca M, Goldstein LJ. Prognostic and predictive factors in early-stage breast cancer. Oncologist 2004; 9(6): 606-16.
[http://dx.doi.org/10.1634/theoncologist.9-6-606] [PMID: 15561805]
[7]
Fung F, Cornacchi SD, Vanniyasingam T, et al. Predictors of 5-year local, regional, and distant recurrent events in a population-based cohort of breast cancer patients. Am J Surg 2017; 213(2): 418-25.
[http://dx.doi.org/10.1016/j.amjsurg.2016.03.016] [PMID: 27424042]
[8]
Paoletti C, Hayes DF. Molecular testing in breast cancer. Annu Rev Med 2014; 65: 95-110.
[http://dx.doi.org/10.1146/annurev-med-070912-143853] [PMID: 24422569]
[9]
Shigematsu H, Nishina M, Yasui D, Hirata T, Ozaki S. Minimal prognostic significance of sentinel lymph node metastasis in patients with cT1-2 and cN0 breast cancer. World J Surg Oncol 2019; 17(1): 41.
[http://dx.doi.org/10.1186/s12957-019-1585-9] [PMID: 30797231]
[10]
Dings PJM, Elferink MAG, Strobbe LJA, de Wilt JH. The prognostic value of lymph node ratio in node-positive breast cancer: a Dutch nationwide population-based study. Ann Surg Oncol 2013; 20(8): 2607-14.
[http://dx.doi.org/10.1245/s10434-013-2932-7] [PMID: 23536053]
[11]
Kim J, Kim JH, Kim OB, Oh YK, Park SG. Clinical significance of the lymph node ratio in N1 breast cancer. Radiat Oncol J 2017; 35(3): 227-32.
[http://dx.doi.org/10.3857/roj.2017.00101] [PMID: 28893060]
[12]
He M, Zhang JX, Jiang YZ, et al. The lymph node ratio as an independent prognostic factor for node-positive triple-negative breast cancer. Oncotarget 2017; 8(27): 44870-80.
[http://dx.doi.org/10.18632/oncotarget.17413] [PMID: 28496004]
[13]
Liu C, Li H, Zhuo R, et al. Grade-lymph node ratio predicts the survival of breast cancer in different molecular types: A surveillance, epidemiology, and end results population-based analysis. Medicine (Baltimore) 2019; 98(28) e16436
[http://dx.doi.org/10.1097/MD.0000000000016436] [PMID: 31305469]
[14]
Dalton LW, Page DL, Dupont WD. Histologic grading of breast carcinoma. A reproducibility study. Cancer 1994; 73(11): 2765-70.
[http://dx.doi.org/10.1002/1097-0142(19940601)73:11<2765:AID-CNCR2820731119>3.0.CO;2-K] [PMID: 8194018]
[15]
Frierson HF Jr, Wolber RA, Berean KW, et al. Interobserver reproducibility of the Nottingham modification of the Bloom and Richardson histologic grading scheme for infiltrating ductal carcinoma. Am J Clin Pathol 1995; 103(2): 195-8.
[http://dx.doi.org/10.1093/ajcp/103.2.195] [PMID: 7856562]
[16]
Robbins P, Pinder S, de Klerk N, et al. Histological grading of breast carcinomas: a study of interobserver agreement. Hum Pathol 1995; 26(8): 873-9.
[http://dx.doi.org/10.1016/0046-8177(95)90010-1] [PMID: 7635449]
[17]
Metzger Filho O, Ignatiadis M, Sotiriou C. Genomic Grade Index: An important tool for assessing breast cancer tumor grade and prognosis. Crit Rev Oncol Hematol 2011; 77(1): 20-9.
[http://dx.doi.org/10.1016/j.critrevonc.2010.01.011] [PMID: 20138540]
[18]
Petrelli F, Viale G, Cabiddu M, Barni S. Prognostic value of different cut-off levels of Ki-67 in breast cancer: a systematic review and meta-analysis of 64,196 patients. Breast Cancer Res Treat 2015; 153(3): 477-91.
[http://dx.doi.org/10.1007/s10549-015-3559-0] [PMID: 26341751]
[19]
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(22): 1656-64.
[http://dx.doi.org/10.1093/jnci/djr393] [PMID: 21960707]
[20]
Polley MY, Leung SC, McShane LM, et al. International ki67 in breast cancer working group of the breast international group and north american breast cancer group. J Natl Cancer Inst 2013; 105(24): 1897-906.
[http://dx.doi.org/10.1093/jnci/djt306] [PMID: 24203987]
[21]
Colomer R, Aranda-López I, Albanell J, García-Caballero T, Ciruelos E, López-García M Á. Biomarkers in breast cancer: a consensus statement by the spanish society of medical oncology and the spanish society of pathology. clinical & translational oncology : official publication of the federation of spanish oncology societies and of the national cancer Institute of Mexico 2018; 20(7): 815-26.
[22]
Coates AS, Winer EP, Goldhirsch A, et al. 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): 1533-46.
[http://dx.doi.org/10.1093/annonc/mdv221] [PMID: 25939896]
[23]
de Azambuja E, Cardoso F, de Castro G Jr, et al. Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer 2007; 96(10): 1504-13.
[http://dx.doi.org/10.1038/sj.bjc.6603756] [PMID: 17453008]
[24]
Kermani T, Kermani I, Faham Z, Dolatkhah R. Ki-67 status in patients with primary breast cancer and its relationship with other prognostic factors. Biomed Res Ther 2019; 6(2): 2986-91.
[http://dx.doi.org/10.15419/bmrat.v6i2.520]
[25]
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: 284-98.
[http://dx.doi.org/10.1016/j.ejca.2017.01.017] [PMID: 28259011]
[26]
Harris LN, Ismaila N, McShane LM, et al. American Society of Clinical Oncology. Use of biomarkers to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34(10): 1134-50.
[http://dx.doi.org/10.1200/JCO.2015.65.2289] [PMID: 26858339]
[27]
Goetz MP, Gradishar WJ, Anderson BO, et al. NCCN guidelines insights: breast cancer version, 3.2018. J Natl Compr Canc Netw 2019; 17(2): 118-26.
[http://dx.doi.org/10.6004/jnccn.2019.0009] [PMID: 30787125]
[28]
Fasching PA, Gass P, Häberle L, et al. Prognostic effect of Ki-67 in common clinical subgroups of patients with HER2-negative, hormone receptor-positive early breast cancer. Breast Cancer Res Treat 2019; 175(3): 617-25.
[http://dx.doi.org/10.1007/s10549-019-05198-9] [PMID: 30868391]
[29]
Ellis MJ, Suman VJ, Hoog J, et al. Ki67 proliferation index as a tool for chemotherapy decisions during and after neoadjuvant aromatase inhibitor treatment of breast cancer: results from the american college of surgeons oncology group z1031 trial (alliance). J Clin Oncol 2017; 35(10): 1061-9.
[http://dx.doi.org/10.1200/JCO.2016.69.4406] [PMID: 28045625]
[30]
Ács B, Zámbó V, Vízkeleti L, et al. Ki-67 as a controversial predictive and prognostic marker in breast cancer patients treated with neoadjuvant chemotherapy. Diagn Pathol 2017; 12(1): 20.
[http://dx.doi.org/10.1186/s13000-017-0608-5] [PMID: 28222768]
[31]
Chen X, He C, Han D, et al. The predictive value of Ki-67 before neoadjuvant chemotherapy for breast cancer: a systematic review and meta-analysis. Future Oncol 2017; 13(9): 843-57.
[http://dx.doi.org/10.2217/fon-2016-0420] [PMID: 28075166]
[32]
Horimoto Y, Arakawa A, Tanabe M, et al. Ki67 expression and the effect of neo-adjuvant chemotherapy on luminal HER2-negative breast cancer. BMC Cancer 2014; 14: 550.
[http://dx.doi.org/10.1186/1471-2407-14-550] [PMID: 25073969]
[33]
Denkert C, Loibl S, Müller BM, et al. Ki67 levels as predictive and prognostic parameters in pretherapeutic breast cancer core biopsies: a translational investigation in the neoadjuvant GeparTrio trial. Ann Oncol 2013; 24(11): 2786-93.
[http://dx.doi.org/10.1093/annonc/mdt350] [PMID: 23970015]
[34]
Yip CH, Rhodes A. Estrogen and progesterone receptors in breast cancer. Future Oncol 2014; 10(14): 2293-301.
[http://dx.doi.org/10.2217/fon.14.110] [PMID: 25471040]
[35]
Davies C, Godwin J, Gray R, 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(9793): 771-84.
[http://dx.doi.org/10.1016/S0140-6736(11)60993-8] [PMID: 21802721]
[36]
Noordhoek I, de Groot AF, Cohen D, Liefers GJ, Portielje JEA, Kroep JR. Higher ER load is not associated with better outcome in stage 1-3 breast cancer: a descriptive overview of quantitative HR analysis in operable breast cancer. Breast Cancer Res Treat 2019; 176(1): 27-36.
[http://dx.doi.org/10.1007/s10549-019-05233-9] [PMID: 30997625]
[37]
Rhodes A, Jasani B, Balaton AJ, Barnes DM, Miller KD. Frequency of oestrogen and progesterone receptor positivity by immunohistochemical analysis in 7016 breast carcinomas: correlation with patient age, assay sensitivity, threshold value, and mammographic screening. J Clin Pathol 2000; 53(9): 688-96.
[http://dx.doi.org/10.1136/jcp.53.9.688] [PMID: 11041059]
[38]
Purdie CA, Quinlan P, Jordan LB, et al. Progesterone receptor expression is an independent prognostic variable in early breast cancer: a population-based study. Br J Cancer 2014; 110(3): 565-72.
[http://dx.doi.org/10.1038/bjc.2013.756] [PMID: 24300977]
[39]
Yang LH, Tseng HS, Lin C, et al. Survival benefit of tamoxifen in estrogen receptor-negative and progesterone receptor-positive low grade breast cancer patients. J Breast Cancer 2012; 15(3): 288-95.
[http://dx.doi.org/10.4048/jbc.2012.15.3.288] [PMID: 23091541]
[40]
Campbell EJ, Tesson M, Doogan F, Mohammed ZMA, Mallon E, Edwards J. The combined endocrine receptor in breast cancer, a novel approach to traditional hormone receptor interpretation and a better discriminator of outcome than ER and PR alone. Br J Cancer 2016; 115(8): 967-73.
[http://dx.doi.org/10.1038/bjc.2016.206] [PMID: 27657341]
[41]
Bardou VJ, Arpino G, Elledge RM, Osborne CK, Clark GM. Progesterone receptor status significantly improves outcome prediction over estrogen receptor status alone for adjuvant endocrine therapy in two large breast cancer databases. J Clin Oncol 2003; 21(10): 1973-9.
[http://dx.doi.org/10.1200/JCO.2003.09.099] [PMID: 12743151]
[42]
Stendahl M, Ryden L, Nordenskjold B, et al. High progesterone receptor expression correlates to the effect of adjuvant tamoxifen in premenopausal breast cancer patients. Clin Cancer Res 2006; 12(15): 4614-8. 139.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-0248]
[43]
Fernö M, Stål O, Baldetorp B, et al. Results of two or five years of adjuvant tamoxifen correlated to steroid receptor and S-phase levels. South Sweden Breast Cancer Group, and South-East Sweden Breast Cancer Group. Breast Cancer Res Treat 2000; 59(1): 69-76.
[http://dx.doi.org/10.1023/A:1006332423620] [PMID: 10752681]
[44]
Liu S, Chia SK, Mehl E, et al. Progesterone receptor is a significant factor associated with clinical outcomes and effect of adjuvant tamoxifen therapy in breast cancer patients. Breast Cancer Res Treat 2010; 119(1): 53-61.
[http://dx.doi.org/10.1007/s10549-009-0318-0] [PMID: 19205877]
[45]
Nordenskjöld A, Fohlin H, Fornander T, Löfdahl B, Skoog L, Stål O. Progesterone receptor positivity is a predictor of long-term benefit from adjuvant tamoxifen treatment of estrogen receptor positive breast cancer. Breast Cancer Res Treat 2016; 160(2): 313-22.
[http://dx.doi.org/10.1007/s10549-016-4007-5] [PMID: 27722840]
[46]
Iqbal N, Iqbal N. Human epidermal growth factor receptor 2 (her2) in cancers: overexpression and therapeutic implications. Mol Biol Int 2014; 2014852748
[http://dx.doi.org/10.1155/2014/852748] [PMID: 25276427]
[47]
Seidman AD, Berry D, Cirrincione C, et al. Randomized phase III trial of weekly compared with every-3-weeks paclitaxel for metastatic breast cancer, with trastuzumab for all HER-2 overexpressors and random assignment to trastuzumab or not in HER-2 nonoverexpressors: final results of Cancer and Leukemia Group B protocol 9840. J Clin Oncol 2008; 26(10): 1642-9.
[http://dx.doi.org/10.1200/JCO.2007.11.6699] [PMID: 18375893]
[48]
Martin M, López-Tarruella S. Emerging therapeutic options for HER2-positive breast cancer. emerging therapeutic options for HER2-positive breast cancer. Am Soc Clin Oncol Educ Book 2016; 35: e64-70.
[http://dx.doi.org/10.14694/EDBK_159167] [PMID: 27249772]
[49]
Dent S, Oyan B, Honig A, Mano M, Howell S. HER2-targeted therapy in breast cancer: a systematic review of neoadjuvant trials. Cancer Treat Rev 2013; 39(6): 622-31.
[http://dx.doi.org/10.1016/j.ctrv.2013.01.002] [PMID: 23434074]
[50]
Wolff AC, Hammond ME, Schwartz JN, et al. American society of clinical oncology/college of american pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J Clin Oncol 2007; 25(1): 118-45.
[http://dx.doi.org/10.1200/JCO.2006.09.2775] [PMID: 17159189]
[51]
Middleton LP, Price KM, Puig P, et al. Implementation of american society of clinical oncology/college of american pathologists her2 guideline recommendations in a tertiary care facility increases her2 immunohistochemistry and fluorescence in situ hybridization concordance and decreases the number of inconclusive cases. Arch Pathol Lab Med 2009; 133(5): 775-80.
[PMID: 19415952]
[52]
Todd JH, Dowle C, Williams MR, et al. Confirmation of a prognostic index in primary breast cancer. Br J Cancer 1987; 56(4): 489-92.
[http://dx.doi.org/10.1038/bjc.1987.230] [PMID: 3689666]
[53]
Quintyne KI, Woulfe B, Coffey JC, Gupta RK. Correlation between nottingham prognostic index and adjuvant! online prognostic tools in patients with early-stage breast cancer in mid-western ireland. Clin Breast Cancer 2013; 13(4): 233-8.
[http://dx.doi.org/10.1016/j.clbc.2013.02.011] [PMID: 23829889]
[54]
Kurshumliu F, Gashi-Luci L, Kadare S, Alimehmeti M, Gozalan U. Classification of patients with breast cancer according to Nottingham prognostic index highlights significant differences in immunohistochemical marker expression. World J Surg Oncol 2014; 12: 243.
[http://dx.doi.org/10.1186/1477-7819-12-243] [PMID: 25082024]
[55]
Rejali M, Tazhibi M, Mokarian F, Gharanjik N, Mokarian R. The performance of the nottingham prognosis index and the adjuvant online decision making tool for prognosis in early-stage breast cancer patients. Int J Prev Med 2015; 6: 93.
[http://dx.doi.org/10.4103/2008-7802.166503] [PMID: 26605014]
[56]
Al jarroudi O, Zaimi A, Brahmi SA, Afqir S. Nottingham Prognostic Index is an Applicable Prognostic Tool in Non-Metastatic Triple-Negative Breast Cancer. Asian Pac J Cancer Prev 2019; 20(1): 59-63.
[http://dx.doi.org/10.31557/APJCP.2019.20.1.59] [PMID: 30678381]
[57]
Albergaria A, Ricardo S, Milanezi F, et al. Nottingham Prognostic Index in triple-negative breast cancer: a reliable prognostic tool? BMC Cancer 2011; 11: 299.
[http://dx.doi.org/10.1186/1471-2407-11-299] [PMID: 21762477]
[58]
Hearne BJ, Teare MD, Butt M, Donaldson L. Comparison of Nottingham Prognostic Index and Adjuvant Online prognostic tools in young women with breast cancer: review of a single-institution experience. BMJ Open 2015; 5(1) e005576
[http://dx.doi.org/10.1136/bmjopen-2014-005576] [PMID: 25628047]
[59]
Rakha EA, Soria D, Green AR, et al. Nottingham Prognostic Index Plus (NPI+): a modern clinical decision making tool in breast cancer. Br J Cancer 2014; 110(7): 1688-97.
[http://dx.doi.org/10.1038/bjc.2014.120] [PMID: 24619074]
[60]
Green AR, Soria D, Powe DG, et al. Nottingham prognostic index plus (NPI+) predicts risk of distant metastases in primary breast cancer. Breast Cancer Res Treat 2016; 157(1): 65-75. [published correction appears in Breast Cancer Res Treat. 2016 Aug;159(1):199].
[http://dx.doi.org/10.1007/s10549-016-3804-1] [PMID: 27116185]
[61]
Vieira AF, Schmitt F. An update on breast cancer multigene prognostic tests-emergent clinical biomarkers. Front Med (Lausanne) 2018; 5: 248.
[http://dx.doi.org/10.3389/fmed.2018.00248] [PMID: 30234119]
[62]
Meirson T, Gil-Henn H. Targeting invadopodia for blocking breast cancer metastasis. Drug Resist Updat 2018; 39: 1-17.
[http://dx.doi.org/10.1016/j.drup.2018.05.002] [PMID: 30075834]
[63]
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(25): 1999-2009.
[http://dx.doi.org/10.1056/NEJMoa021967] [PMID: 12490681]
[64]
Fayanju OM, Park KU, Lucci A. Molecular genomic testing for breast cancer: utility for surgeons. Ann Surg Oncol 2018; 25(2): 512-9.
[http://dx.doi.org/10.1245/s10434-017-6254-z] [PMID: 29159748]
[65]
Cuadros M, Llanos A. [Validation and clinical application of MammaPrint® in patients with breast cancer]. Med Clin (Barc) 2011; 136(14): 627-32.
[http://dx.doi.org/10.1016/j.medcli.2010.02.009] [PMID: 20416899]
[66]
Xin L, Liu YH, Martin TA, Jiang WG. The era of multigene panels comes? the clinical utility of oncotype dx and mammaprint. World J Oncol 2017; 8(2): 34-40.
[http://dx.doi.org/10.14740/wjon1019w] [PMID: 29147432]
[67]
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(8): 717-29.
[http://dx.doi.org/10.1056/NEJMoa1602253] [PMID: 27557300]
[68]
Sánchez-Forgach ER, Carpinteyro-Espín U, Alemán-Áviles JA, Sánchez-Basurto C. [Validation and clinical application of MammaPrint® in patients with breast cancer]. Cir Cir 2017; 85(4): 320-4.
[http://dx.doi.org/10.1016/j.circen.2016.10.008] [PMID: 27979362]
[69]
Groenendijk FH, Jager A, Cardoso F, van Deurzen CHM. A nationwide registry-based cohort study of the MammaPrint genomic risk classifier in invasive breast cancer. Breast 2018; 38: 125-31.
[http://dx.doi.org/10.1016/j.breast.2017.12.015] [PMID: 29310037]
[70]
Exner R, Bago-Horvath Z, Bartsch R, et al. The multigene signature MammaPrint impacts on multidisciplinary team decisions in ER+, HER2- early breast cancer. Br J Cancer 2014; 111(5): 837-42.
[http://dx.doi.org/10.1038/bjc.2014.339] [PMID: 25003667]
[71]
Hall PS, Smith A, Hulme C, et al. Value of information analysis of multiparameter tests for chemotherapy in early breast cancer: the optima prelim trial. Value Health 2017; 20(10): 1311-8.
[http://dx.doi.org/10.1016/j.jval.2017.04.021] [PMID: 29241890]
[72]
Chen X, Fan Y, Xu B. Distinct characteristics and metastatic behaviors of late recurrence in patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative breast cancer: a single institute experience of more than 10 years. Clin Breast Cancer 2018; 18(6): e1353-60.
[http://dx.doi.org/10.1016/j.clbc.2018.07.014] [PMID: 30201586]
[73]
Mittempergher L, Delahaye LJ, Witteveen A, et al. Mamma print and blueprint molecular diagnostics using targeted rna nextgeneration sequencing technology. J Mol Diagn 2019; S1525-1578(18): 30483-5.
[74]
Nam KJ, Park H, Ko ES, Lim Y, Cho HH, Lee JE. Radiomics signature on 3T dynamic contrast-enhanced magnetic resonance imaging for estrogen receptor-positive invasive breast cancers: Preliminary results for correlation with Oncotype DX recurrence scores. Medicine (Baltimore) 2019; 98(23) e15871
[http://dx.doi.org/10.1097/MD.0000000000015871] [PMID: 31169691]
[75]
Ribnikar D, Cardoso F. Tailoring chemotherapy in early-stage breast cancer: based on tumor biology or tumor burden? Am Soc Clin Oncol Educ Book 2016; 35: e31-8.
[http://dx.doi.org/10.14694/EDBK_159077] [PMID: 27249737]
[76]
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(27): 2817-26.
[http://dx.doi.org/10.1056/NEJMoa041588] [PMID: 15591335]
[77]
Cronin M, Sangli C, Liu ML, et al. Analytical validation of the Oncotype DX genomic diagnostic test for recurrence prognosis and therapeutic response prediction in node-negative, estrogen receptor-positive breast cancer. Clin Chem 2007; 53(6): 1084-91.
[http://dx.doi.org/10.1373/clinchem.2006.076497] [PMID: 17463177]
[78]
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(23): 3726-34.
[http://dx.doi.org/10.1200/JCO.2005.04.7985] [PMID: 16720680]
[79]
Zheng A, Zhang L, Ji Z, Fan L, Jin F. Oncotype DX for Comprehensive Treatment in Male Breast Cancer: A Case Report and Literature Review. Am J Men Health 2019; 13(3) 1557988319847856
[http://dx.doi.org/10.1177/1557988319847856] [PMID: 31068065]
[80]
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(22): 1956-64.
[http://dx.doi.org/10.1200/JCO.19.00945] [PMID: 31150316]
[81]
Rath MG, Uhlmann L, Fiedler M, et al. Oncotype DX® in breast cancer patients: clinical experience, outcome and follow-up-a case-control study. Arch Gynecol Obstet 2018; 297(2): 443-7.
[http://dx.doi.org/10.1007/s00404-017-4618-z] [PMID: 29236174]
[82]
Thibodeau S, Voutsadakis IA. The Oncotype Dx Assay in ER-Positive, HER2-Negative Breast Cancer Patients: A Real Life Experience from a Single Cancer Center. Eur J Breast Health 2019; 15(3): 163-70.
[http://dx.doi.org/10.5152/ejbh.2019.4901] [PMID: 31312792]
[83]
Wan T, Bloch BN, Plecha D, et al. A radio-genomics approach for identifying high risk estrogen receptor-positive breast cancers on dce-mri: preliminary results in predicting oncotypedx risk scores. Sci Rep 2016; 6: 21394.
[http://dx.doi.org/10.1038/srep21394] [PMID: 26887643]
[84]
Sutton EJ, Oh JH, Dashevsky BZ, et al. Breast cancer subtype intertumor heterogeneity: MRI-based features predict results of a genomic assay. J Magn Reson Imaging 2015; 42(5): 1398-406.
[http://dx.doi.org/10.1002/jmri.24890] [PMID: 25850931]
[85]
Flatley MJ, Dodwell DJ. Adjuvant treatment for breast cancer. Surgery 2016; 34(1): 43-6.
[PMID: 27401339]
[86]
Blok EJ, Bastiaannet E, van den Hout WB, et al. Systematic review of the clinical and economic value of gene expression profiles for invasive early breast cancer available in Europe. Cancer Treat Rev 2018; 62: 74-90.
[http://dx.doi.org/10.1016/j.ctrv.2017.10.012] [PMID: 29175678]
[87]
Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 2009; 27(8): 1160-7.
[http://dx.doi.org/10.1200/JCO.2008.18.1370] [PMID: 19204204]
[88]
Nielsen T, Wallden B, Schaper C, et al. Analytical validation of the pam50-based prosigna breast cancer prognostic gene signature assay and incounter analysis system using formalin-fixed paraffin-embedded breast tumor specimens. BMC Cancer 2014; 14(1): 177.
[http://dx.doi.org/10.1186/1471-2407-14-177] [PMID: 24625003]
[89]
Hannouf MB, Brezden-Masley C, Raphael J, Brackstone M. A review of gene expression profiling in early-stage er+/her2- breast cancer with a focus on the pam50 risk of recurrence assay. EMJ Oncol 2019; 7(Suppl. 1): 2-13.
[90]
Wallden B, Storhoff J, Nielsen T, et al. DCavies SR, Mardis ER, Gnant M, Sestak I, Ellis MJ, Perou CM, Bernard PS, Parker JS. Development and verification of the PAM50-based Prosigna® PAM50 breast cancer gene signature assay. BMC Med Genomics 2015; 8: 54.
[http://dx.doi.org/10.1186/s12920-015-0129-6] [PMID: 26297356]
[91]
Ohara AM, Naoi Y, Shimazu K, et al. PAM50 for prediction of response to neoadjuvant chemotherapy for ER-positive breast cancer. Breast Cancer Res Treat 2019; 173(3): 533-43.
[http://dx.doi.org/10.1007/s10549-018-5020-7] [PMID: 30361874]
[92]
Laenkholm AV, Jensen MB, Eriksen JO, et al. The ability of PAM50 risk of recurrence score to predict 10-year distant recurrence in hormone receptor-positive postmenopausal women with special histological subtypes. Acta Oncol 2018; 57(1): 44-50.
[http://dx.doi.org/10.1080/0284186X.2017.1403044] [PMID: 29202609]
[93]
Ohnstad HO, Borgen E, Falk RS, et al. Prognostic value of PAM50 and risk of recurrence score in patients with early-stage breast cancer with long-term follow-up. Breast Cancer Res 2017; 19(1): 120.
[http://dx.doi.org/10.1186/s13058-017-0911-9] [PMID: 29137653]
[94]
Rodriguez CS, Garcia-Muñoz M, Sancho M, et al. Impact of the Prosigna® PAM50 (PAM50) assay on adjuvant clinical decision making in patients with early stage breast cancer: Results of a prospective multicenter public program. J Clin Oncol 2017; 35(15_suppl): e12062-2.
[95]
Esin E, Oksuzoglu BO, Markoc F, et al. Prosigna® PAM50 assay for treatment decisions in early breast cancer: A single center, decision impact study [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; San Antonio, TX; Philadelphia (PA): AACR. 2019 Cancer Res.; 79.(4 Suppl) Abstract nr P3-08.
[96]
Jensen MB, Lænkholm AV, Nielsen TO, et al. The Prosigna gene expression assay and responsiveness to adjuvant cyclophosphamide-based chemotherapy in premenopausal high-risk patients with breast cancer. Breast Cancer Res 2018; 20(1): 79.
[http://dx.doi.org/10.1186/s13058-018-1012-0] [PMID: 30053900]
[97]
Bustamante Eduardo M, Popovici V, Imboden S, et al. Characterization of molecular scores and gene expression signatures in primary breast cancer, local recurrences and brain metastases. BMC Cancer 2019; 19(1): 549.
[http://dx.doi.org/10.1186/s12885-019-5752-8] [PMID: 31174485]
[98]
Sestak I, Martín M, Dubsky P, et al. Prediction of chemotherapy benefit by EndoPredict in patients with breast cancer who received adjuvant endocrine therapy plus chemotherapy or endocrine therapy alone. Breast Cancer Res Treat 2019; 176(2): 377-86.
[http://dx.doi.org/10.1007/s10549-019-05226-8] [PMID: 31041683]
[99]
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(12): 2959-64.
[http://dx.doi.org/10.1038/bjc.2013.671] [PMID: 24157828]
[100]
Lee J, Lee EH, Park HY, et al. Efficacy of an RNA-based multigene assay with core needle biopsy samples for risk evaluation in hormone-positive early breast cancer. BMC Cancer 2019; 19(1): 388.
[http://dx.doi.org/10.1186/s12885-019-5608-2] [PMID: 31023265]
[101]
Filipits M, Rudas M, Jakesz R, et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res 2011; 17(18): 6012-20.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-0926] [PMID: 21807638]
[102]
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(2): R38.
[http://dx.doi.org/10.1186/bcr3642] [PMID: 24725534]
[103]
Filipits M, Dubsky P, Rudas M, et al. Prediction of Distant Recurrence Using EndoPredict Among Women with ER+, HER2− Node-Positive and Node-Negative Breast Cancer Treated with Endocrine Therapy Only. Clin Can Res 2019.
[http://dx.doi.org/10.1158/1078-0432.CCR-19-0376]
[104]
Bartlett JM, Thomas J, Ross DT, et al. Mammostrat as a tool to stratify breast cancer patients at risk of recurrence during endocrine therapy. Breast Cancer Res 2010; 12(4): R47.
[http://dx.doi.org/10.1186/bcr2604] [PMID: 20615243]
[105]
Bartlett JM, Bloom KJ, Piper T, et al. Mammostrat as an immunohistochemical multigene assay for prediction of early relapse risk in the tamoxifen versus exemestane adjuvant multicenter trial pathology study. J Clin Oncol 2012; 30(36): 4477-84.
[http://dx.doi.org/10.1200/JCO.2012.42.8896] [PMID: 23045591]
[106]
Benarba B, Elmallah A, Pandiella A. Bryonia dioica aqueous extract induces apoptosis and G2/M cell cycle arrest in MDA-MB 231 breast cancer cells. Mol Med Rep 2019; 20(1): 73-80.
[http://dx.doi.org/10.3892/mmr.2019.10220] [PMID: 31115513]
[107]
Sharma A, Boise LH, Shanmugam M. Cancer Metabolism and the Evasion of Apoptotic Cell Death. Cancers (Basel) 2019; 11(8) E1144
[http://dx.doi.org/10.3390/cancers11081144] [PMID: 31405035]
[108]
Pu X, Storr SJ, Zhang Y, et al. Caspase-3 and caspase-8 expression in breast cancer: caspase-3 is associated with survival. Apoptosis 2017; 22(3): 357-68.
[http://dx.doi.org/10.1007/s10495-016-1323-5] [PMID: 27798717]
[109]
Yang X, Zhong DN, Qin H, et al. Caspase-3 over-expression is associated with poor overall survival and clinicopathological parameters in breast cancer: a meta-analysis of 3091 cases. Oncotarget 2017; 9(9): 8629-41.
[PMID: 29492222]
[110]
Liu X, Jiang S, Tian X, Jiang Y. Expression of cleaved caspase-3 predicts good chemotherapy response but poor survival for patients with advanced primary triple-negative breast cancer. Int J Clin Exp Pathol 2018; 11(9): 4363-73.
[PMID: 31949833]
[111]
Hammoud H, Saleh J, Bachour M, Salamoon M. Serum caspase-3 and caspase-7 as predictive factors of response in locally advanced and metastatic breast carcinoma. J Cancer Ther 2014; 5(6): 584.
[http://dx.doi.org/10.4236/jct.2014.56067]
[112]
Stanley J, Klepczyk L, Keene K, et al. PARP1 and phospho-p65 protein expression is increased in human HER2-positive breast cancers. Breast Cancer Res Treat 2015; 150(3): 569-79.
[http://dx.doi.org/10.1007/s10549-015-3359-6] [PMID: 25833211]
[113]
Ossovskaya V, Koo IC, Kaldjian EP, Alvares C, Sherman BM. Upregulation of poly (adp-ribose) polymerase-1 (parp1) in triple-negative breast cancer and other primary human tumor types. Genes Cancer 2010; 1(8): 812-21.
[http://dx.doi.org/10.1177/1947601910383418] [PMID: 21779467]
[114]
Siraj AK, Pratheeshkumar P, Parvathareddy SK, et al. Overexpression of PARP is an independent prognostic marker for poor survival in Middle Eastern breast cancer and its inhibition can be enhanced with embelin co-treatment. Oncotarget 2018; 9(99): 37319-32.
[http://dx.doi.org/10.18632/oncotarget.26470] [PMID: 30647872]
[115]
Rojo F, García-Parra J, Zazo S, et al. Nuclear PARP-1 protein overexpression is associated with poor overall survival in early breast cancer. Ann Oncol 2012; 23(5): 1156-64.
[http://dx.doi.org/10.1093/annonc/mdr361] [PMID: 21908496]
[116]
Park SH, Noh SJ, Kim KM, et al. Expression of DNA damage response molecules PARP1, γH2AX, BRCA1, and BRCA2 predicts poor survival of breast carcinoma patients. Transl Oncol 2015; 8(4): 239-49.
[http://dx.doi.org/10.1016/j.tranon.2015.04.004] [PMID: 26310369]
[117]
Zhai L, Li S, Li H, et al. Polymorphisms in poly (ADP-ribose) polymerase-1 (PARP1) promoter and 3′ untranslated region and their association with PARP1 expression in breast cancer patients. Int J Clin Exp Pathol 2015; 8(6): 7059-71.
[PMID: 26261599]
[118]
Green AR, Caracappa D, Benhasouna AA, et al. Biological and clinical significance of PARP1 protein expression in breast cancer. Breast Cancer Res Treat 2015; 149(2): 353-62.
[http://dx.doi.org/10.1007/s10549-014-3230-1] [PMID: 25528020]
[119]
von Minckwitz G, Müller BM, Loibl S, et al. Cytoplasmic poly(adenosine diphosphate-ribose) polymerase expression is predictive and prognostic in patients with breast cancer treated with neoadjuvant chemotherapy. J Clin Oncol 2011; 29(16): 2150-7.
[http://dx.doi.org/10.1200/JCO.2010.31.9079] [PMID: 21519019]
[120]
Song Z, Wang Y, Xiao Q, et al. Poly(ADP-ribose) polymerase-3 overexpression is associated with poor prognosis in patients with breast cancer following chemotherapy. Oncol Lett 2018; 16(5): 5621-30.
[http://dx.doi.org/10.3892/ol.2018.9398] [PMID: 30344717 ]
[121]
Eom YH, Kim HS, Lee A, Song BJ, Chae BJ. BCL2 as a Subtype-Specific Prognostic Marker for Breast Cancer. J Breast Cancer 2016; 19(3): 252-60.
[http://dx.doi.org/10.4048/jbc.2016.19.3.252] [PMID: 27721874]
[122]
Abdel-Fatah TMA, Perry C, Dickinson P, et al. Bcl2 is an independent prognostic marker of triple negative breast cancer (TNBC) and predicts response to anthracycline combination (ATC) chemotherapy (CT) in adjuvant and neoadjuvant settings. Ann Oncol 2013; 24(11): 2801-7.
[http://dx.doi.org/10.1093/annonc/mdt277] [PMID: 23908177]
[123]
Callagy GM, Pharoah PD, Pinder SE, et al. Bcl-2 is a prognostic marker in breast cancer independently of the Nottingham Prognostic Index. Clin Cancer Res 2006; 12(8): 2468-75.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-2719] [PMID: 16638854]
[124]
Dawson SJ, Makretsov N, Blows FM, et al. BCL2 in breast cancer: a favourable prognostic marker across molecular subtypes and independent of adjuvant therapy received. Br J Cancer 2010; 103(5): 668-75.
[http://dx.doi.org/10.1038/sj.bjc.6605736] [PMID: 20664598]
[125]
Tawfik K, Kimler BF, Davis MK, Fan F, Tawfik O. Prognostic significance of Bcl-2 in invasive mammary carcinomas: a comparative clinicopathologic study between “triple-negative” and non-“triple-negative” tumors. Hum Pathol 2012; 43(1): 23-30.
[http://dx.doi.org/10.1016/j.humpath.2011.04.011] [PMID: 21777944]
[126]
Callagy GM, Webber MJ, Pharoah PD, Caldas C. Meta-analysis confirms BCL2 is an independent prognostic marker in breast cancer. BMC Cancer 2008; 8: 153.
[http://dx.doi.org/10.1186/1471-2407-8-153] [PMID: 18510726]
[127]
Zheng HC, Xu XY, Xing YN, et al. Nuclear or cytoplasmic localization of Bag-1 distinctly correlates with pathologic behavior and outcome of gastric carcinomas. Hum Pathol 2010; 41(5): 724-36.
[http://dx.doi.org/10.1016/j.humpath.2009.10.017] [PMID: 20096920]
[128]
Kilbas PO, Akcay IM, Doganay GD, Arisan ED. Bag-1 silencing enhanced chemotherapeutic drug-induced apoptosis in MCF-7 breast cancer cells affecting PI3K/Akt/mTOR and MAPK signaling pathways. Mol Biol Rep 2019; 46(1): 847-60.
[http://dx.doi.org/10.1007/s11033-018-4540-x] [PMID: 30661182]
[129]
Wang YD, Ha MW, Cheng J, et al. The role of expression and polymorphism of the BAG-1 gene in response to platinum-based chemotherapeutics in NSCLC. Oncol Rep 2012; 27(4): 979-86.
[http://dx.doi.org/10.3892/or.2011.1591] [PMID: 22179630]
[130]
Mosly D, Turnbull A, Sims A, Ward C, Langdon S. Predictive markers of endocrine response in breast cancer. World J Exp Med 2018; 8(1): 1-7.
[http://dx.doi.org/10.5493/wjem.v8.i1.1] [PMID: 30191138]
[131]
Papadakis ES, Reeves T, Robson NH, Maishman T, Packham G, Cutress RI. BAG-1 as a biomarker in early breast cancer prognosis: a systematic review with meta-analyses. Br J Cancer 2017; 116(12): 1585-94.
[http://dx.doi.org/10.1038/bjc.2017.130] [PMID: 28510570]
[132]
Zhang D, Yang H, Wang M. Expressions of bcl-2-associated athanogene 1 and epithelial growth factor receptor gene in triple negative breast cancer. Can Res Clin 2018; 30(3): 165-8.
[http://dx.doi.org/10.1007/s00432-017-2526-z]
[133]
Nadler Y, Camp RL, Giltnane JM, et al. Expression patterns and prognostic value of Bag-1 and Bcl-2 in breast cancer. Breast Cancer Res 2008; 10(2): R35.
[http://dx.doi.org/10.1186/bcr1998] [PMID: 18430249]
[134]
Turner BC, Krajewski S, Krajewska M, et al. BAG-1: a novel biomarker predicting long-term survival in early-stage breast cancer. J Clin Oncol 2001; 19(4): 992-1000.
[http://dx.doi.org/10.1200/JCO.2001.19.4.992] [PMID: 11181661]
[135]
Wang W, Chen Q, Wang RB, et al. Correlation between the expression of BRCA-1 and BAG-1 proteins in triple negative breast cancer and its sensitivity to platinum-based chemotherapy. Biomed Res 2017; 28(15): 6654-61.
[136]
Liu H, Lu S, Gu L, et al. Modulation of BAG-1 expression alters the sensitivity of breast cancer cells to tamoxifen. Cell Physiol Biochem 2014; 33(2): 365-74.
[http://dx.doi.org/10.1159/000356676] [PMID: 24557447]
[137]
Rodriguez BJ, Córdoba GD, Aranda AG, et al. Detection of tp53 and pik3ca mutations in circulating tumor dna using next-generation sequencing in the screening process for early breast cancer diagnosis. J Clin Med 2019; 8(8): 1183.
[http://dx.doi.org/10.3390/jcm8081183] [PMID: 31394872]
[138]
Maltoni R, Gallerani G, Fici P, Rocca A, Fabbri F. CTCs in early breast cancer: A path worth taking. Cancer Lett 2016; 376(2): 205-10.
[http://dx.doi.org/10.1016/j.canlet.2016.03.051] [PMID: 27060205]
[139]
Krawczyk N, Fehm T, Banys-Paluchowski M, Janni W, Schramm A. Liquid biopsy in metastasized breast cancer as basis for treatment decisions. Oncol Res Treat 2016; 39(3): 112-6.
[http://dx.doi.org/10.1159/000444605] [PMID: 27031542]
[140]
Ye Q, Ling S, Zheng S, Xu X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer 2019; 18(1): 114.
[http://dx.doi.org/10.1186/s12943-019-1043-x] [PMID: 31269959]
[141]
Li H, Jing C, Wu J, et al. Circulating tumor DNA detection: A potential tool for colorectal cancer management. Oncol Lett 2019; 17(2): 1409-16.
[PMID: 30675194]
[142]
Rohanizadegan M. Analysis of circulating tumor DNA in breast cancer as a diagnostic and prognostic biomarker. Cancer Genet 2018; 228-229: 159-68.
[http://dx.doi.org/10.1016/j.cancergen.2018.02.002] [PMID: 29572011]
[143]
Zhou Y, Xu Y, Gong Y, et al. Clinical factors associated with circulating tumor DNA (ctDNA) in primary breast cancer. Mol Oncol 2019; 13(5): 1033-46.
[PMID: 30672098]
[144]
Garcia-Murillas I, Chopra N, Comino-Méndez I, et al. Assessment of Molecular Relapse Detection in Early-Stage Breast Cancer. JAMA Oncol 2019.
[http://dx.doi.org/10.1001/jamaoncol.2019.1838] [PMID: 31369045]
[145]
Coombes RC, Page K, Salari R, et al. Personalized detection of circulating tumor dna antedates breast cancer metastatic recurrence. Clin Cancer Res 2019; 25(14): 4255-63.
[http://dx.doi.org/10.1158/1078-0432.CCR-18-3663] [PMID: 30992300]
[146]
Olsson E, Winter C, George A, et al. Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol Med 2015; 7(8): 1034-47.
[http://dx.doi.org/10.15252/emmm.201404913] [PMID: 25987569]
[147]
Butler TM, Boniface CT, Johnson-Camacho K, et al. Circulating tumor DNA dynamics using patient-customized assays are associated with outcome in neoadjuvantly treated breast cancer. Cold Spring Harb Mol Case Stud 2019; 5(2) a003772
[http://dx.doi.org/10.1101/mcs.a003772] [PMID: 30833418]
[148]
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(313) 313ra182
[http://dx.doi.org/10.1126/scitranslmed.aac7551] [PMID: 26560360]
[149]
Takahashi H, Kagara N, Tanei T, et al. Correlation of methylated circulating tumor dna with response to neoadjuvant chemotherapy in breast cancer patients. Clin Breast Cancer 2017; 17(1): 61-69.e3.
[http://dx.doi.org/10.1016/j.clbc.2016.06.006] [PMID: 27395416]
[150]
Bhat SA, Majid S, Hassan T. MicroRNAs and its emerging role as breast cancer diagnostic marker-A review. Adv Biomark Sci Technol 2019; 1: 1-8.
[http://dx.doi.org/10.1016/j.abst.2019.05.001]
[151]
Asiaf A, Ahmad ST, Arjumand W, Zargar MA. MicroRNAs in Breast Cancer: Diagnostic and Therapeutic Potential. Methods Mol Biol 2018; 1699: 23-43.
[http://dx.doi.org/10.1007/978-1-4939-7435-1_2] [PMID: 29086366]
[152]
Hamam R, Hamam D, Alsaleh KA, et al. Circulating microRNAs in breast cancer: novel diagnostic and prognostic biomarkers. Cell Death Dis 2017; 8(9) e3045
[http://dx.doi.org/10.1038/cddis.2017.440] [PMID: 28880270]
[153]
Amorim M, Lobo J, Fontes-Sousa M, et al. Predictive and Prognostic Value of Selected MicroRNAs in Luminal Breast Cancer. Front Genet 2019; 10: 815.
[http://dx.doi.org/10.3389/fgene.2019.00815] [PMID: 31572437]
[154]
Lai J, Wang H, Pan Z, Su F. A novel six-microRNA-based model to improve prognosis prediction of breast cancer. Aging (Albany NY) 2019; 11(2): 649-62.
[http://dx.doi.org/10.18632/aging.101767] [PMID: 30696800]
[155]
Nama S, Muhuri M, Di Pascale F, et al. MicroRNA-138 is a prognostic biomarker for triple-negative breast cancer and promotes tumorigenesis via tusc2 repression. Sci Rep 2019; 9(1): 12718.
[http://dx.doi.org/10.1038/s41598-019-49155-4] [PMID: 31481748]
[156]
Blackley EF, Loi S. Targeting immune pathways in breast cancer: review of the prognostic utility of TILs in early stage triple negative breast cancer (TNBC). Breast 2019; 48(Suppl. 1): S44-8.
[http://dx.doi.org/10.1016/S0960-9776(19)31122-1] [PMID: 31839159]
[157]
Salgado R, Denkert C, Demaria S, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2015; 26(2): 259-71.
[http://dx.doi.org/10.1093/annonc/mdu450] [PMID: 25214542]
[158]
Schirosi L, Saponaro C, Giotta F, et al. Tumor infiltrating lymphocytes and nherf1 impact on prognosis of breast cancer patients. Transl Oncol 2019; 13(2): 186-92.
[http://dx.doi.org/10.1016/j.tranon.2019.10.020] [PMID: 31865181]
[159]
Ochi T, Bianchini G, Ando M, et al. Predictive and prognostic value of stromal tumour-infiltrating lymphocytes before and after neoadjuvant therapy in triple negative and HER2-positive breast cancer. Eur J Cancer 2019; 118: 41-8.
[http://dx.doi.org/10.1016/j.ejca.2019.05.014] [PMID: 31302586]
[160]
Miyashita M, Sasano H, Tamaki K, et al. Prognostic significance of tumor-infiltrating CD8+ and FOXP3+ lymphocytes in residual tumors and alterations in these parameters after neoadjuvant chemotherapy in triple-negative breast cancer: a retrospective multicenter study. Breast Cancer Res 2015; 17: 124.
[http://dx.doi.org/10.1186/s13058-015-0632-x] [PMID: 26341640]
[161]
Lee J, Kim DM, Lee A. Prognostic role and clinical association of tumor-infiltrating lymphocyte, programmed death ligand-1 expression with neutrophil-lymphocyte ratio in locally advanced triple-negative breast cancer. Cancer Res Treat 2019; 51(2): 649-63.
[http://dx.doi.org/10.4143/crt.2018.270] [PMID: 30064200]
[162]
Truntzer C, Isambert N, Arnould L, Ladoire S, Ghiringhelli F. Prognostic value of transcriptomic determination of tumour-infiltrating lymphocytes in localised breast cancer. Eur J Cancer 2019; 120: 97-106.
[http://dx.doi.org/10.1016/j.ejca.2019.07.020] [PMID: 31499385]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy