Tumor Heterogeneity in Breast Cancer

Gulisa Turashvili, Edi Brogi, Gulisa Turashvili, Edi Brogi

Abstract

Breast cancer is a heterogeneous disease and differs greatly among different patients (intertumor heterogeneity) and even within each individual tumor (intratumor heterogeneity). Clinical and morphologic intertumor heterogeneity is reflected by staging systems and histopathologic classification of breast cancer. Heterogeneity in the expression of established prognostic and predictive biomarkers, hormone receptors, and human epidermal growth factor receptor 2 oncoprotein is the basis for targeted treatment. Molecular classifications are indicators of genetic tumor heterogeneity, which is probed with multigene assays and can lead to improved stratification into low- and high-risk groups for personalized therapy. Intratumor heterogeneity occurs at the morphologic, genomic, transcriptomic, and proteomic levels, creating diagnostic and therapeutic challenges. Understanding the molecular and cellular mechanisms of tumor heterogeneity that are relevant to the development of treatment resistance is a major area of research. Despite the improved knowledge of the complex genetic and phenotypic features underpinning tumor heterogeneity, there has been only limited advancement in diagnostic, prognostic, or predictive strategies for breast cancer. The current guidelines for reporting of biomarkers aim to maximize patient eligibility for targeted therapy, but do not take into account intratumor heterogeneity. The molecular classification of breast cancer is not implemented in routine clinical practice. Additional studies and in-depth analysis are required to understand the clinical significance of rapidly accumulating data. This review highlights inter- and intratumor heterogeneity of breast carcinoma with special emphasis on pathologic findings, and provides insights into the clinical significance of molecular and cellular mechanisms of heterogeneity.

Keywords: biomarkers; breast cancer; genetic markers; histopathology; tumor heterogeneity.

Figures

Figure 1
Figure 1
Histopathologic heterogeneity of breast cancer: invasive mammary carcinoma with mixed morphology (A), composed of basaloid areas with osteoid production (B) and ductal not otherwise specified (C) components. Lymph node metastasis showing a diffuse pattern of tumor growth (D). Magnification: 100× (A), 200× (B–D); Hematoxylin-eosin staining.
Figure 2
Figure 2
Biomarker heterogeneity of breast cancer: mucinous carcinoma (A) with variable expression of estrogen receptor from no immunoreactivity to nuclear staining with weak to strong intensity (B); invasive ductal carcinoma (C) with areas of 3+ (positive) and 1+ (negative) membranous staining for human epidermal growth factor receptor 2 (D). Magnification: 200× (A–D); hematoxylin-eosin staining (A,C) and immunohistochemistry (B,D).

References

    1. Ellsworth RE, Blackburn HL, Shriver CD, Soon-Shiong P, Ellsworth DL. Molecular heterogeneity in breast cancer: state of the science and implications for patient care. Semin Cell Dev Biol (2016) 64:65–72.10.1016/j.semcdb.2016.08.025
    1. Fidler IJ, Kripke ML. Metastasis results from preexisting variant cells within a malignant tumor. Science (1977) 197:893–5.10.1126/science.887927
    1. Fidler IJ. Tumor heterogeneity and the biology of cancer invasion and metastasis. Cancer Res (1978) 38:2651–60.
    1. Miller FR, Miller BE, Heppner GH. Characterization of metastatic heterogeneity among subpopulations of a single mouse mammary tumor: heterogeneity in phenotypic stability. Invasion Metastasis (1983) 3:22–31.
    1. Young RH, Louis DN. The Warrens and other pioneering clinician pathologists of the Massachusetts General Hospital during its early years: an appreciation on the 200th anniversary of the hospital founding. Mod Pathol (2011) 24:1285–94.10.1038/modpathol.2011.132
    1. Hawkins RA, Killen E, Tesdale AL, Sangster K, Thomson M, Steele RJ, et al. Oestrogen receptors, lactate dehydrogenase and cellularity in human breast cancer. Clin Chim Acta (1988) 175:89–96.10.1016/0009-8981(88)90038-1
    1. Hortobagyi GN, D’orsi CJ, Edge SB, Mittendorf EA, Rugo HS, Solin LJ, et al. AJCC Cancer Staging Manual – Breast. 8th ed Chicago: Springer; (2017).
    1. Harris LN, Ismaila N, Mcshane LM, Andre F, Collyar DE, Gonzalez-Angulo AM, et al. 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) 34:1134–50.10.1200/JCO.2015.65.2289
    1. Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, Van De Vijver MJ, editors. WHO Classification of Tumours of the Breast. Lyon: France International Agency for Research on Cancer; (2012).
    1. Page DL. Special types of invasive breast cancer, with clinical implications. Am J Surg Pathol (2003) 27:832–5.10.1097/00000478-200306000-00016
    1. Yerushalmi R, Hayes MM, Gelmon KA. Breast carcinoma – rare types: review of the literature. Ann Oncol (2009) 20:1763–70.10.1093/annonc/mdp245
    1. Weigelt B, Geyer FC, Reis-Filho JS. Histological types of breast cancer: how special are they? Mol Oncol (2010) 4:192–208.10.1016/j.molonc.2010.04.004
    1. Nagao T, Kinoshita T, Hojo T, Tsuda H, Tamura K, Fujiwara Y. The differences in the histological types of breast cancer and the response to neoadjuvant chemotherapy: the relationship between the outcome and the clinicopathological characteristics. Breast (2012) 21:289–95.10.1016/j.breast.2011.12.011
    1. Rosen PP, Groshen S, Kinne DW, Norton L. Factors influencing prognosis in node-negative breast carcinoma: analysis of 767 T1N0M0/T2N0M0 patients with long-term follow-up. J Clin Oncol (1993) 11:2090–100.10.1200/JCO.1993.11.11.2090
    1. Rakha EA, Lee AH, Evans AJ, Menon S, Assad NY, Hodi Z, et al. Tubular carcinoma of the breast: further evidence to support its excellent prognosis. J Clin Oncol (2010) 28:99–104.10.1200/JCO.2009.23.5051
    1. Colleoni M, Russo L, Dellapasqua S. Adjuvant therapies for special types of breast cancer. Breast (2011) 20(Suppl 3):S153–7.10.1016/S0960-9776(11)70315-0
    1. 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 (1991) 19:403–10.10.1111/j.1365-2559.1991.tb00229.x
    1. Rakha EA, Reis-Filho JS, Baehner F, Dabbs DJ, Decker T, Eusebi V, et al. Breast cancer prognostic classification in the molecular era: the role of histological grade. Breast Cancer Res (2010) 12:207.10.1186/bcr2607
    1. Abd El-Rehim DM, Ball G, Pinder SE, Rakha E, Paish C, Robertson JF, et al. High-throughput protein expression analysis using tissue microarray technology of a large well-characterised series identifies biologically distinct classes of breast cancer confirming recent cDNA expression analyses. Int J Cancer (2005) 116:340–50.10.1002/ijc.21004
    1. Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, et al. Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst (2006) 98:262–72.10.1093/jnci/djj052
    1. Natrajan R, Lambros MB, Rodriguez-Pinilla SM, Moreno-Bueno G, Tan DS, Marchio C, et al. Tiling path genomic profiling of grade 3 invasive ductal breast cancers. Clin Cancer Res (2009) 15:2711–22.10.1158/1078-0432.CCR-08-1878
    1. Yu K, Lee CH, Tan PH, Hong GS, Wee SB, Wong CY, et al. A molecular signature of the Nottingham prognostic index in breast cancer. Cancer Res (2004) 64:2962–8.10.1158/0008-5472.CAN-03-2430
    1. Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, 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) 28:2784–95.10.1200/JCO.2009.25.6529
    1. Wolff AC, Hammond ME, Hicks DG, Dowsett M, Mcshane LM, Allison KH, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol (2013) 31:3997–4013.10.1200/JCO.2013.50.9984
    1. Early Breast Cancer Trialists’ Collaborative Group. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet (2005) 365:1687–717.10.1016/S0140-6736(05)66544-0
    1. 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) 17:1474–81.10.1200/JCO.1999.17.5.1474
    1. 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) 21:1973–9.10.1200/JCO.2003.09.099
    1. Dean-Colomb W, Esteva FJ. Her2-positive breast cancer: herceptin and beyond. Eur J Cancer (2008) 44:2806–12.10.1016/j.ejca.2008.09.013
    1. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet (2014) 384:164–72.10.1016/S0140-6736(13)62422-8
    1. Qi JP, Yang YL, Zhu H, Wang J, Jia Y, Liu N, et al. Expression of the androgen receptor and its correlation with molecular subtypes in 980 Chinese breast cancer patients. Breast Cancer (Auckl) (2012) 6:1–8.10.4137/BCBCR.S8323
    1. Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA, et al. Phase II trial of bicalutamide in patients with androgen receptor-positive, estrogen receptor-negative metastatic breast cancer. Clin Cancer Res (2013) 19:5505–12.10.1158/1078-0432.CCR-12-3327
    1. Barton VN, D’amato NC, Gordon MA, Christenson JL, Elias A, Richer JK. Androgen receptor biology in triple negative breast cancer: a case for classification as AR+ or quadruple negative disease. Horm Cancer (2015) 6:206–13.10.1007/s12672-015-0232-3
    1. Park S, Koo JS, Kim MS, Park HS, Lee JS, Lee JS, et al. Androgen receptor expression is significantly associated with better outcomes in estrogen receptor-positive breast cancers. Ann Oncol (2011) 22:1755–62.10.1093/annonc/mdq678
    1. Gucalp A, Traina TA. Targeting the androgen receptor in triple-negative breast cancer. Curr Probl Cancer (2016) 40:141–50.10.1016/j.currproblcancer.2016.09.004
    1. Lee E, Moon A. Identification of biomarkers for breast cancer using databases. J Cancer Prev (2016) 21:235–42.10.15430/JCP.2016.21.4.235
    1. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A (2001) 98:10869–74.10.1073/pnas.191367098
    1. Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A (2003) 100:8418–23.10.1073/pnas.0932692100
    1. Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res (2010) 12:R68.10.1186/bcr2635
    1. Farmer P, Bonnefoi H, Becette V, Tubiana-Hulin M, Fumoleau P, Larsimont D, et al. Identification of molecular apocrine breast tumours by microarray analysis. Oncogene (2005) 24:4660–71.10.1038/sj.onc.1208561
    1. Wirapati P, Sotiriou C, Kunkel S, Farmer P, Pradervand S, Haibe-Kains B, et al. Meta-analysis of gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res (2008) 10:R65.10.1186/bcr2124
    1. Korde LA, Lusa L, Mcshane L, Lebowitz PF, Lukes L, Camphausen K, et al. Gene expression pathway analysis to predict response to neoadjuvant docetaxel and capecitabine for breast cancer. Breast Cancer Res Treat (2010) 119:685–99.10.1007/s10549-009-0651-3
    1. Early Breast Cancer Trialists’ Collaborative Group. Davies C, Godwin J, Gray R, Clarke M, Cutter D, 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) 378:771–84.10.1016/S0140-6736(11)60993-8
    1. Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature (2012) 490:61–70.10.1038/nature11412
    1. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature (2012) 486:346–52.10.1038/nature10983
    1. Ali HR, Rueda OM, Chin SF, Curtis C, Dunning MJ, Aparicio SA, et al. Genome-driven integrated classification of breast cancer validated in over 7,500 samples. Genome Biol (2014) 15:431.10.1186/s13059-014-0431-1
    1. Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med (2004) 351:2817–26.10.1056/NEJMoa041588
    1. Dubsky P, Brase JC, Jakesz R, Rudas M, Singer CF, Greil R, et al. The EndoPredict score provides prognostic information on late distant metastases in ER+/HER2- breast cancer patients. Br J Cancer (2013) 109:2959–64.10.1038/bjc.2013.671
    1. Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol (2009) 27:1160–7.10.1200/JCO.2008.18.1370
    1. Nielsen TO, Parker JS, Leung S, Voduc D, Ebbert M, Vickery T, et al. A comparison of PAM50 intrinsic subtyping with immunohistochemistry and clinical prognostic factors in tamoxifen-treated estrogen receptor-positive breast cancer. Clin Cancer Res (2010) 16:5222–32.10.1158/1078-0432.CCR-10-1282
    1. Nielsen T, Wallden B, Schaper C, Ferree S, Liu S, Gao D, et al. Analytical validation of the PAM50-based prosigna breast cancer prognostic gene signature assay and nCounter Analysis System using formalin-fixed paraffin-embedded breast tumor specimens. BMC Cancer (2014) 14:177.10.1186/1471-2407-14-177
    1. Sgroi DC, Sestak I, Cuzick J, Zhang Y, Schnabel CA, Schroeder B, 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) 14:1067–76.10.1016/S1470-2045(13)70387-5
    1. Beca F, Polyak K. Intratumor heterogeneity in breast cancer. Adv Exp Med Biol (2016) 882:169–89.10.1007/978-3-319-22909-6_7
    1. Paik S, Tang G, Shak S, Kim C, Baker J, Kim W, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol (2006) 24:3726–34.10.1200/JCO.2005.04.7985
    1. Partin JF, Mamounas EP. Impact of the 21-gene recurrence score assay compared with standard clinicopathologic guidelines in adjuvant therapy selection for node-negative, estrogen receptor-positive breast cancer. Ann Surg Oncol (2011) 18:3399–406.10.1245/s10434-011-1698-z
    1. Cheang MC, Voduc D, Bajdik C, Leung S, Mckinney S, Chia SK, et al. Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin Cancer Res (2008) 14:1368–76.10.1158/1078-0432.CCR-07-1658
    1. Tang P, Skinner KA, Hicks DG. Molecular classification of breast carcinomas by immunohistochemical analysis: are we ready? Diagn Mol Pathol (2009) 18:125–32.10.1097/PDM.0b013e31818d107b
    1. Geyer FC, Weigelt B, Natrajan R, Lambros MB, De Biase D, Vatcheva R, et al. Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas. J Pathol (2010) 220:562–73.10.1002/path.2675
    1. Patani N, Barbashina V, Lambros MB, Gauthier A, Mansour M, Mackay A, et al. Direct evidence for concurrent morphological and genetic heterogeneity in an invasive ductal carcinoma of triple-negative phenotype. J Clin Pathol (2011) 64:822–8.10.1136/jclinpath-2011-200135
    1. Nik-Zainal S, Van Loo P, Wedge DC, Alexandrov LB, Greenman CD, Lau KW, et al. The life history of 21 breast cancers. Cell (2012) 149:994–1007.10.1016/j.cell.2012.04.023
    1. Balko JM, Giltnane JM, Wang K, Schwarz LJ, Young CD, Cook RS, et al. Molecular profiling of the residual disease of triple-negative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets. Cancer Discov (2014) 4:232–45.10.1158/-13-0286
    1. Zardavas D, Maetens M, Irrthum A, Goulioti T, Engelen K, Fumagalli D, et al. The AURORA initiative for metastatic breast cancer. Br J Cancer (2014) 111:1881–7.10.1038/bjc.2014.341
    1. Kuukasjarvi T, Karhu R, Tanner M, Kahkonen M, Schaffer A, Nupponen N, et al. Genetic heterogeneity and clonal evolution underlying development of asynchronous metastasis in human breast cancer. Cancer Res (1997) 57:1597–604.
    1. Shah SP, Morin RD, Khattra J, Prentice L, Pugh T, Burleigh A, et al. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature (2009) 461:809–13.10.1038/nature08489
    1. Simpson PT, Reis-Filho JS, Gale T, Lakhani SR. Molecular evolution of breast cancer. J Pathol (2005) 205:248–54.10.1002/path.1691
    1. Kalinsky K, Heguy A, Bhanot UK, Patil S, Moynahan ME. PIK3CA mutations rarely demonstrate genotypic intratumoral heterogeneity and are selected for in breast cancer progression. Breast Cancer Res Treat (2011) 129:635–43.10.1007/s10549-011-1601-4
    1. Ding L, Ellis MJ, Li S, Larson DE, Chen K, Wallis JW, et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature (2010) 464:999–1005.10.1038/nature08989
    1. Hull DF, III, Clark GM, Osborne CK, Chamness GC, Knight WA, III, Mcguire WL. Multiple estrogen receptor assays in human breast cancer. Cancer Res (1983) 43:413–6.
    1. Amir E, Miller N, Geddie W, Freedman O, Kassam F, Simmons C, et al. Prospective study evaluating the impact of tissue confirmation of metastatic disease in patients with breast cancer. J Clin Oncol (2012) 30:587–92.10.1200/JCO.2010.33.5232
    1. Lindstrom LS, Karlsson E, Wilking UM, Johansson U, Hartman J, Lidbrink EK, et al. Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J Clin Oncol (2012) 30:2601–8.10.1200/JCO.2011.37.2482
    1. Allegra JC, Barlock A, Huff KK, Lippman ME. Changes in multiple or sequential estrogen receptor determinations in breast cancer. Cancer (1980) 45:792–4.10.1002/1097-0142(19800215)45:4<792::AID-CNCR2820450430>;2-X
    1. Wicha MS. Cancer stem cell heterogeneity in hereditary breast cancer. Breast Cancer Res (2008) 10:105.10.1186/bcr1990
    1. Wu JM, Fackler MJ, Halushka MK, Molavi DW, Taylor ME, Teo WW, et al. Heterogeneity of breast cancer metastases: comparison of therapeutic target expression and promoter methylation between primary tumors and their multifocal metastases. Clin Cancer Res (2008) 14:1938–46.10.1158/1078-0432.CCR-07-4082
    1. Becker TE, Ellsworth RE, Deyarmin B, Patney HL, Jordan RM, Hooke JA, et al. The genomic heritage of lymph node metastases: implications for clinical management of patients with breast cancer. Ann Surg Oncol (2008) 15:1056–63.10.1245/s10434-008-9815-3
    1. Almendro V, Kim HJ, Cheng YK, Gonen M, Itzkovitz S, Argani P, et al. Genetic and phenotypic diversity in breast tumor metastases. Cancer Res (2014) 74:1338–48.10.1158/0008-5472.CAN-13-2357-T
    1. Blau CA, Ramirez AB, Blau S, Pritchard CC, Dorschner MO, Schmechel SC, et al. A distributed network for intensive longitudinal monitoring in metastatic triple-negative breast cancer. J Natl Compr Canc Netw (2016) 14:8–17.10.6004/jnccn.2016.0003
    1. Van Poznak C, Somerfield MR, Bast RC, Cristofanilli M, Goetz MP, Gonzalez-Angulo AM, et al. Use of biomarkers to guide decisions on systemic therapy for women with metastatic breast cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol (2015) 33:2695–704.10.1200/JCO.2015.61.1459
    1. Davis BW, Zava DT, Locher GW, Goldhirsch A, Hartmann WH. Receptor heterogeneity of human breast cancer as measured by multiple intratumoral assays of estrogen and progesterone receptor. Eur J Cancer Clin Oncol (1984) 20:375–82.10.1016/0277-5379(84)90084-1
    1. Nassar A, Radhakrishnan A, Cabrero IA, Cotsonis GA, Cohen C. Intratumoral heterogeneity of immunohistochemical marker expression in breast carcinoma: a tissue microarray-based study. Appl Immunohistochem Mol Morphol (2010) 18:433–41.10.1097/PAI.0b013e3181dddb20
    1. Pertschuk LP, Axiotis CA, Feldman JG, Kim YD, Karavattayhayyil SJ, Braithwaite L. Marked intratumoral heterogeneity of the proto-oncogene Her-2/neu determined by three different detection systems. Breast J (1999) 5:369–74.10.1046/j.1524-4741.1999.97088.x
    1. Allison KH, Dintzis SM, Schmidt RA. Frequency of HER2 heterogeneity by fluorescence in situ hybridization according to CAP expert panel recommendations: time for a new look at how to report heterogeneity. Am J Clin Pathol (2011) 136:864–71.10.1309/AJCPXTZSKBRIP07W
    1. Seol H, Lee HJ, Choi Y, Lee HE, Kim YJ, Kim JH, et al. Intratumoral heterogeneity of HER2 gene amplification in breast cancer: its clinicopathological significance. Mod Pathol (2012) 25:938–48.10.1038/modpathol.2012.36
    1. Glockner S, Buurman H, Kleeberger W, Lehmann U, Kreipe H. Marked intratumoral heterogeneity of c-myc and cyclinD1 but not of c-erbB2 amplification in breast cancer. Lab Invest (2002) 82:1419–26.10.1097/01.LAB.0000032371.16521.40
    1. Andersson J, Linderholm B, Bergh J, Elmberger G. HER-2/neu (c-erbB-2) evaluation in primary breast carcinoma by fluorescent in situ hybridization and immunohistochemistry with special focus on intratumor heterogeneity and comparison of invasive and in situ components. Appl Immunohistochem Mol Morphol (2004) 12:14–20.10.1097/00129039-200403000-00003
    1. Vance GH, Barry TS, Bloom KJ, Fitzgibbons PL, Hicks DG, Jenkins RB, et al. Genetic heterogeneity in HER2 testing in breast cancer: panel summary and guidelines. Arch Pathol Lab Med (2009) 133:611–2.10.1043/1543-2165-133.4.611
    1. Chhieng DC, Frost AR, Niwas S, Weiss H, Grizzle WE, Beeken S. Intratumor heterogeneity of biomarker expression in breast carcinomas. Biotech Histochem (2004) 79:25–36.10.1080/10520290410001715237
    1. Focke CM, Decker T, Van Diest PJ. Intratumoral heterogeneity of Ki67 expression in early breast cancers exceeds variability between individual tumours. Histopathology (2016) 69:849–61.10.1111/his.13007
    1. Siitonen SM, Isola JJ, Rantala IS, Helin HJ. Intratumor variation in cell proliferation in breast carcinoma as determined by antiproliferating cell nuclear antigen monoclonal antibody and automated image analysis. Am J Clin Pathol (1993) 99:226–31.10.1093/ajcp/99.3.226
    1. de Azambuja E, Cardoso F, De Castro G, Jr, Colozza M, Mano MS, Durbecq V, 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) 96:1504–13.10.1038/sj.bjc.6603756
    1. Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA. Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol (2010) 11:174–83.10.1016/S1470-2045(09)70262-1
    1. Ingolf JB, Russalina M, Simona M, Julia R, Gilda S, Bohle RM, et al. Can ki-67 play a role in prediction of breast cancer patients’ response to neoadjuvant chemotherapy? Biomed Res Int (2014) 2014:628217.10.1155/2014/628217
    1. Cserni G, Voros A, Liepniece-Karele I, Bianchi S, Vezzosi V, Grabau D, et al. Distribution pattern of the Ki67 labelling index in breast cancer and its implications for choosing cut-off values. Breast (2014) 23:259–63.10.1016/j.breast.2014.02.003
    1. Aleskandarany MA, Green AR, Ashankyty I, Elmouna A, Diez-Rodriguez M, Nolan CC, et al. Impact of intratumoural heterogeneity on the assessment of Ki67 expression in breast cancer. Breast Cancer Res Treat (2016) 158:287–95.10.1007/s10549-016-3893-x
    1. Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol (2015) 12:381–94.10.1038/nrclinonc.2015.73
    1. Fehm T, Sagalowsky A, Clifford E, Beitsch P, Saboorian H, Euhus D, et al. Cytogenetic evidence that circulating epithelial cells in patients with carcinoma are malignant. Clin Cancer Res (2002) 8:2073–84.
    1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, 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) 10:6897–904.10.1158/1078-0432.CCR-04-0378
    1. Hartkopf AD, Wagner P, Wallwiener D, Fehm T, Rothmund R. Changing levels of circulating tumor cells in monitoring chemotherapy response in patients with metastatic breast cancer. Anticancer Res (2011) 31:979–84.
    1. Aurilio G, Sciandivasci A, Munzone E, Sandri MT, Zorzino L, Cassatella MC, et al. Prognostic value of circulating tumor cells in primary and metastatic breast cancer. Expert Rev Anticancer Ther (2012) 12:203–14.10.1586/era.11.208
    1. Hayashi N, Nakamura S, Tokuda Y, Shimoda Y, Yagata H, Yoshida A, et al. Prognostic value of HER2-positive circulating tumor cells in patients with metastatic breast cancer. Int J Clin Oncol (2012) 17:96–104.10.1007/s10147-011-0260-0
    1. Mego M, Gao H, Lee BN, Cohen EN, Tin S, Giordano A, et al. Prognostic value of EMT-circulating tumor cells in metastatic breast cancer patients undergoing high-dose chemotherapy with autologous hematopoietic stem cell transplantation. J Cancer (2012) 3:369–80.10.7150/jca.5111
    1. Pierga JY, Hajage D, Bachelot T, Delaloge S, Brain E, Campone M, et al. High independent prognostic and predictive value of circulating tumor cells compared with serum tumor markers in a large prospective trial in first-line chemotherapy for metastatic breast cancer patients. Ann Oncol (2012) 23:618–24.10.1093/annonc/mdr263
    1. Zhao L, Li P, Li F, Yang Y, Liu N, Cai L. The prognostic value of circulating tumor cells lacking cytokeratins in metastatic breast cancer patients. J Cancer Res Ther (2013) 9:29–37.10.4103/0973-1482.110353
    1. Smerage JB, Barlow WE, Hortobagyi GN, Winer EP, Leyland-Jones B, Srkalovic G, et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol (2014) 32:3483–9.10.1200/JCO.2014.56.2561
    1. Janni WJ, Rack B, Terstappen LW, Pierga JY, Taran FA, Fehm T, et al. Pooled analysis of the prognostic relevance of circulating tumor cells in primary breast cancer. Clin Cancer Res (2016) 22:2583–93.10.1158/1078-0432.CCR-15-1603
    1. Lv Q, Gong L, Zhang T, Ye J, Chai L, Ni C, et al. Prognostic value of circulating tumor cells in metastatic breast cancer: a systemic review and meta-analysis. Clin Transl Oncol (2016) 18:322–30.10.1007/s12094-015-1372-1
    1. Wicha MS, Hayes DF. Circulating tumor cells: not all detected cells are bad and not all bad cells are detected. J Clin Oncol (2011) 29:1508–11.10.1200/JCO.2010.34.0026
    1. Hayes DF, Paoletti C. Circulating tumour cells: insights into tumour heterogeneity. J Intern Med (2013) 274:137–43.10.1111/joim.12047
    1. Bidard FC, Proudhon C, Pierga JY. Circulating tumor cells in breast cancer. Mol Oncol (2016) 10:418–30.10.1016/j.molonc.2016.01.001
    1. Sieuwerts AM, Mostert B, Bolt-De Vries J, Peeters D, De Jongh FE, Stouthard JM, et al. mRNA and microRNA expression profiles in circulating tumor cells and primary tumors of metastatic breast cancer patients. Clin Cancer Res (2011) 17:3600–18.10.1158/1078-0432.CCR-11-0255
    1. Powell AA, Talasaz AH, Zhang H, Coram MA, Reddy A, Deng G, et al. Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines. PLoS One (2012) 7:e33788.10.1371/journal.pone.0033788
    1. Babayan A, Hannemann J, Spotter J, Muller V, Pantel K, Joosse SA. Heterogeneity of estrogen receptor expression in circulating tumor cells from metastatic breast cancer patients. PLoS One (2013) 8:e75038.10.1371/journal.pone.0075038
    1. Markou A, Farkona S, Schiza C, Efstathiou T, Kounelis S, Malamos N, et al. PIK3CA mutational status in circulating tumor cells can change during disease recurrence or progression in patients with breast cancer. Clin Cancer Res (2014) 20:5823–34.10.1158/1078-0432.CCR-14-0149
    1. Gasch C, Plummer PN, Jovanovic L, Mcinnes LM, Wescott D, Saunders CM, et al. Heterogeneity of miR-10b expression in circulating tumor cells. Sci Rep (2015) 5:15980.10.1038/srep15980
    1. Pestrin M, Salvianti F, Galardi F, De Luca F, Turner N, Malorni L, et al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients. Mol Oncol (2015) 9:749–57.10.1016/j.molonc.2014.12.001
    1. De Luca F, Rotunno G, Salvianti F, Galardi F, Pestrin M, Gabellini S, et al. Mutational analysis of single circulating tumor cells by next generation sequencing in metastatic breast cancer. Oncotarget (2016) 7:26107–19.10.18632/oncotarget.8431
    1. Grover PK, Cummins AG, Price TJ, Roberts-Thomson IC, Hardingham JE. Circulating tumour cells: the evolving concept and the inadequacy of their enrichment by EpCAM-based methodology for basic and clinical cancer research. Ann Oncol (2014) 25:1506–16.10.1093/annonc/mdu018
    1. Hyun KA, Koo GB, Han H, Sohn J, Choi W, Kim SI, et al. Epithelial-to-mesenchymal transition leads to loss of EpCAM and different physical properties in circulating tumor cells from metastatic breast cancer. Oncotarget (2016) 7:24677–87.10.18632/oncotarget.8250
    1. Bulfoni M, Gerratana L, Del Ben F, Marzinotto S, Sorrentino M, Turetta M, et al. In patients with metastatic breast cancer the identification of circulating tumor cells in epithelial-to-mesenchymal transition is associated with a poor prognosis. Breast Cancer Res (2016) 18:30.10.1186/s13058-016-0687-3
    1. Hayes DF, Walker TM, Singh B, Vitetta ES, Uhr JW, Gross S, et al. Monitoring expression of HER-2 on circulating epithelial cells in patients with advanced breast cancer. Int J Oncol (2002) 21:1111–7.
    1. Georgoulias V, Bozionelou V, Agelaki S, Perraki M, Apostolaki S, Kallergi G, 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) 23:1744–50.10.1093/annonc/mds020
    1. Mikulova V, Cabinakova M, Janatkova I, Mestek O, Zima T, Tesarova P. Detection of circulating tumor cells during follow-up of patients with early breast cancer: clinical utility for monitoring of therapy efficacy. Scand J Clin Lab Invest (2014) 74:132–42.10.3109/00365513.2013.864784
    1. Agelaki S, Kalykaki A, Markomanolaki H, Papadaki MA, Kallergi G, Hatzidaki D, et al. Efficacy of lapatinib in therapy-resistant HER2-positive circulating tumor cells in metastatic breast cancer. PLoS One (2015) 10:e0123683.10.1371/journal.pone.0123683
    1. Bidard FC, Fehm T, Ignatiadis M, Smerage JB, Alix-Panabieres C, Janni W, et al. Clinical application of circulating tumor cells in breast cancer: overview of the current interventional trials. Cancer Metastasis Rev (2013) 32:179–88.10.1007/s10555-012-9398-0
    1. Schramm A, Friedl TW, Schochter F, Scholz C, De Gregorio N, Huober J, et al. Therapeutic intervention based on circulating tumor cell phenotype in metastatic breast cancer: concept of the DETECT study program. Arch Gynecol Obstet (2016) 293:271–81.10.1007/s00404-015-3879-7
    1. Fiegl M, Tueni C, Schenk T, Jakesz R, Gnant M, Reiner A, et al. Interphase cytogenetics reveals a high incidence of aneuploidy and intra-tumour heterogeneity in breast cancer. Br J Cancer (1995) 72:51–5.10.1038/bjc.1995.276
    1. Torres L, Ribeiro FR, Pandis N, Andersen JA, Heim S, Teixeira MR. Intratumor genomic heterogeneity in breast cancer with clonal divergence between primary carcinomas and lymph node metastases. Breast Cancer Res Treat (2007) 102:143–55.10.1007/s10549-006-9317-6
    1. Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature (2012) 486:405–9.10.1038/nature11154
    1. Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW, et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature (2012) 486:353–60.10.1038/nature11143
    1. Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature (2012) 486:395–9.10.1038/nature10933
    1. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature (2012) 486:400–4.10.1038/nature11017
    1. Ellsworth RE, Toro AL, Blackburn HL, Decewicz A, Deyarmin B, Mamula KA, et al. Molecular heterogeneity in primary breast carcinomas and axillary lymph node metastases assessed by genomic fingerprinting analysis. Cancer Growth Metastasis (2015) 8:15–24.10.4137/CGM.S29490
    1. Navin N, Krasnitz A, Rodgers L, Cook K, Meth J, Kendall J, et al. Inferring tumor progression from genomic heterogeneity. Genome Res (2010) 20:68–80.10.1101/gr.099622.109
    1. Martelotto LG, Ng CK, Piscuoglio S, Weigelt B, Reis-Filho JS. Breast cancer intra-tumor heterogeneity. Breast Cancer Res (2014) 16:210.10.1186/bcr3658
    1. Juric D, Castel P, Griffith M, Griffith OL, Won HH, Ellis H, et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor. Nature (2015) 518:240–4.10.1038/nature13948
    1. Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer (2012) 12:323–34.10.1038/nrc3261
    1. Almendro V, Marusyk A, Polyak K. Cellular heterogeneity and molecular evolution in cancer. Annu Rev Pathol (2013) 8:277–302.10.1146/annurev-pathol-020712-163923
    1. Reynolds PA, Sigaroudinia M, Zardo G, Wilson MB, Benton GM, Miller CJ, et al. Tumor suppressor p16INK4A regulates polycomb-mediated DNA hypermethylation in human mammary epithelial cells. J Biol Chem (2006) 281:24790–802.10.1074/jbc.M604175200
    1. Pasquali L, Bedeir A, Ringquist S, Styche A, Bhargava R, Trucco G. Quantification of CpG island methylation in progressive breast lesions from normal to invasive carcinoma. Cancer Lett (2007) 257:136–44.10.1016/j.canlet.2007.07.010
    1. Sunami E, Shinozaki M, Sim MS, Nguyen SL, Vu AT, Giuliano AE, et al. Estrogen receptor and HER2/neu status affect epigenetic differences of tumor-related genes in primary breast tumors. Breast Cancer Res (2008) 10:R46.10.1186/bcr2098
    1. Corre G, Stockholm D, Arnaud O, Kaneko G, Vinuelas J, Yamagata Y, et al. Stochastic fluctuations and distributed control of gene expression impact cellular memory. PLoS One (2014) 9:e115574.10.1371/journal.pone.0115574
    1. Kendrick H, Regan JL, Magnay FA, Grigoriadis A, Mitsopoulos C, Zvelebil M, et al. Transcriptome analysis of mammary epithelial subpopulations identifies novel determinants of lineage commitment and cell fate. BMC Genomics (2008) 9:591.10.1186/1471-2164-9-591
    1. Koren S, Bentires-Alj M. Breast tumor heterogeneity: source of fitness, hurdle for therapy. Mol Cell (2015) 60:537–46.10.1016/j.molcel.2015.10.031
    1. Ince TA, Richardson AL, Bell GW, Saitoh M, Godar S, Karnoub AE, et al. Transformation of different human breast epithelial cell types leads to distinct tumor phenotypes. Cancer Cell (2007) 12:160–70.10.1016/j.ccr.2007.06.013
    1. Van Keymeulen A, Rocha AS, Ousset M, Beck B, Bouvencourt G, Rock J, et al. Distinct stem cells contribute to mammary gland development and maintenance. Nature (2011) 479:189–93.10.1038/nature10573
    1. Keller PJ, Arendt LM, Skibinski A, Logvinenko T, Klebba I, Dong S, et al. Defining the cellular precursors to human breast cancer. Proc Natl Acad Sci U S A (2012) 109:2772–7.10.1073/pnas.1017626108
    1. Melchor L, Molyneux G, Mackay A, Magnay FA, Atienza M, Kendrick H, et al. Identification of cellular and genetic drivers of breast cancer heterogeneity in genetically engineered mouse tumour models. J Pathol (2014) 233:124–37.10.1002/path.4345
    1. Tao L, Van Bragt MP, Li Z. A long-lived luminal subpopulation enriched with alveolar progenitors serves as cellular origin of heterogeneous mammary tumors. Stem Cell Reports (2015) 5:60–74.10.1016/j.stemcr.2015.05.014
    1. Koren S, Reavie L, Couto JP, De Silva D, Stadler MB, Roloff T, et al. PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature (2015) 525:114–8.10.1038/nature14669
    1. Gupta PB, Fillmore CM, Jiang G, Shapira SD, Tao K, Kuperwasser C, et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell (2011) 146:633–44.10.1016/j.cell.2011.07.026
    1. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell (2014) 14:275–91.10.1016/j.stem.2014.02.006
    1. Chaffer CL, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA, Rodrigues LO, et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci U S A (2011) 108:7950–5.10.1073/pnas.1102454108
    1. Skibinski A, Kuperwasser C. The origin of breast tumor heterogeneity. Oncogene (2015) 34:5309–16.10.1038/onc.2014.475
    1. Van Keymeulen A, Lee MY, Ousset M, Brohee S, Rorive S, Giraddi RR, et al. Reactivation of multipotency by oncogenic PIK3CA induces breast tumour heterogeneity. Nature (2015) 525:119–23.10.1038/nature14665
    1. Nowell PC. The clonal evolution of tumor cell populations. Science (1976) 194:23–8.10.1126/science.959840
    1. Bissell MJ, Hines WC. Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med (2011) 17:320–9.10.1038/nm.2328
    1. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med (2013) 19:1423–37.10.1038/nm.3394
    1. Ghajar CM, Peinado H, Mori H, Matei IR, Evason KJ, Brazier H, et al. The perivascular niche regulates breast tumour dormancy. Nat Cell Biol (2013) 15:807–17.10.1038/ncb2767
    1. Ng CK, Pemberton HN, Reis-Filho JS. Breast cancer intratumor genetic heterogeneity: causes and implications. Expert Rev Anticancer Ther (2012) 12:1021–32.10.1586/era.12.85
    1. Aparicio S, Caldas C. The implications of clonal genome evolution for cancer medicine. N Engl J Med (2013) 368:842–51.10.1056/NEJMra1204892
    1. Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic. Nature (2013) 501:355–64.10.1038/nature12627

Source: PubMed

3
Se inscrever