Geburtshilfe Frauenheilkd 2014; 74(11): 1016-1022
DOI: 10.1055/s-0034-1383033
Original Article
GebFra Science
Georg Thieme Verlag KG Stuttgart · New York

Molecular Markers as Prognostic Factors in DCIS and Small Invasive Breast Cancers

Molekulare Marker als Prognosefaktoren für DCIS und invasive T1a Mammakarzinome
N. Sänger
1   Dept. of Gynecology and Obstetrics, Goethe University Frankfurt, Frankfurt
,
K. Engels
2   Zentrum für Pathologie, Zytologie und Molekularpathologie, Neuss
,
A. Graf
1   Dept. of Gynecology and Obstetrics, Goethe University Frankfurt, Frankfurt
,
E. Ruckhäberle
3   Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf
,
K. E. Effenberger
4   University Medical Center Hamburg-Eppendorf, Hamburg
,
T. Fehm
3   Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf
,
U. Holtrich
1   Dept. of Gynecology and Obstetrics, Goethe University Frankfurt, Frankfurt
,
S. Becker
,
T. Karn
1   Dept. of Gynecology and Obstetrics, Goethe University Frankfurt, Frankfurt
› Author Affiliations
Further Information

Publication History

received 28 July 2014
revised 11 August 2014

accepted 18 August 2014

Publication Date:
26 November 2014 (online)

Abstract

Ductal carcinoma in situ (DCIS) accounts for up to half of screen-detected breast cancers and thus constitutes a major public health problem. Despite effective current treatment many patients with DCIS are either over- or undertreated because of the paucity of precise models to predict recurrence or progression. The combination of clinical and molecular factors as already applied for invasive disease may help to build such models also for DCIS. We compared 53 DCIS (36.6 %) and 92 (63.4 %) invasive breast cancer cases and found no significant differences in age, receptor status of ER, PR, and HER2, and the use of radiotherapy. Interestingly, the proportion of disseminated tumor cells (DTC) did also not significantly differ between DCIS and invasive cases (p = 0.57). A negative PR status was associated with the detection of DTCs (p = 0.026). We then compared relationships of clinical parameters and biomarkers with patientsʼ prognosis in 43 DCIS and 40 small invasive tumors ≤ 5 mm (T1a). ER negativity was associated with shorter relapse free survival in the complete cohort (p = 0.004) and showed a trend in both subgroups (p = 0.053 for DCIS and p = 0.046 for T1a, respectively). In conclusion, we found markedly similar properties of both DCIS and small invasive breast cancers with respect to the distribution of several parameters as well as to the prognostic value of biomarkers. DCIS with a luminal phenotype seem to be characterized by a favourable prognosis.

Zusammenfassung

Das duktale Carcinoma in situ (DCIS) ist aufgrund seiner durch das Mammografie-Screening stark angestiegenen Häufigkeit in den letzten Dekaden deutlicher ins Blickfeld von Forschung und Praxis gerückt. Trotz effektiver Behandlung stellt sich für viele Patientinnen die Frage einer Über- oder Untertherapie, da sich der Verlauf der Erkrankung individuell nicht vorhersagen lässt. Eine Kombination von klinischen und molekularen Parametern, wie sie bereits vielfach für das invasive Mammakarzinom angewandt wird, könnte hier möglicherweise helfen, entsprechende Prädiktoren zu entwickeln. Bei einem Vergleich von 53 DCIS (36,6 %) und 92 (63,4 %) invasiven Mammakarzinomen bezüglich klinischer und molekularer Parameter fanden wir keine signifikanten Unterschiede bez. Alter, Hormonrezepor- und HER2-Status sowie dem Einsatz adjuvanter Bestrahlung. Interessanterweise unterschied sich auch die Häufigkeit der Detektion disseminierter Tumorzellen (DTZ) nicht signifikant zwischen DCIS und invasiven Fällen (p = 0,57). Ein negativer Progesteronrezeptor-Status war mit dem DTZ-Nachweis assoziiert (p = 0,026). Untersucht wurde ebenfalls der Zusammenhang von klinischen Parametern und Biomarkern mit der Prognose bei 43 DCIS und 40 invasiven T1a-Karzinomen. Negativität für den Östrogenrezeptor zeigte hierbei einen signifikanten Zusammenhang zu einem kürzeren krankheitsfreien Intervall in der Gesamtkohorte (p = 0,004) und einen Trend in beiden Subgruppen (p = 0,053 bei DCIS bzw. p = 0,046 bei T1a). Zusammenfassend fanden wir sehr ähnliche Charakteristika bez. der Verteilung verschiedener Parameter und des prognostischen Werts von Biomarkern sowohl bei DCIS als auch invasiven Karzinomen. DCIS mit einem luminalen Phänotyp scheint durch eine günstigere Prognose gekennzeichnet zu sein.

 
  • 1 Benson JR, Wishart GC. Predictors of recurrence for ductal carcinoma in situ after breast-conserving surgery. Lancet Oncol 2013; 14: e348-e357
  • 2 Heywang-Koebrunner S, Bock K, Heindel W et al. Mammography screening – as of 2013. Geburtsh Frauenheilk 2013; 73: 1007-1016
  • 3 Bartlett JMS, Nofech-Moses S, Rakovitch E. Ductal carcinoma in situ of the breast: can biomarkers improve current management?. Clin Chem 2014; 60: 60-67
  • 4 Cowell CF, Weigelt B, Sakr RA et al. Progression from ductal carcinoma in situ to invasive breast cancer: revisited. Mol Oncol 2013; 7: 859-869
  • 5 Fasching PA, Ekici AB, Wachter DL et al. Breast cancer risk – from genetics to molecular understanding of pathogenesis. Geburtsh Frauenheilk 2013; 73: 1228-1235
  • 6 Solin LJ, Gray R, Baehner FL et al. A multigene expression assay to predict local recurrence risk for ductal carcinoma in situ of the breast. J Natl Cancer Inst 2013; 105: 701-710
  • 7 Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet 2011; 378: 1812-1823
  • 8 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
  • 9 Prat A, Ellis MJ, Perou CM. Practical implications of gene-expression-based assays for breast oncologists. Nat Rev Clin Oncol 2011; 9: 48-57
  • 10 Koboldt DC, Fulton RS, McLellan MD et al. Comprehensive molecular portraits of human breast tumours. Nature 2012; 490: 61-70
  • 11 Karn T. High-throughput gene expression and mutation profiling: current methods and future perspectives. Breast Care (Basel) 2013; 8: 401-406
  • 12 Clark SE, Warwick J, Carpenter R et al. Molecular subtyping of DCIS: heterogeneity of breast cancer reflected in pre-invasive disease. Br J Cancer 2011; 104: 120-127
  • 13 Williams KE, Barnes NL, Cheema K et al. Molecular phenotypes of DCIS predict invasive and DCIS recurrence. 35th Annual San Antonio Breast Cancer Symposium. PD04-06, San Antonio, TX. 2012
  • 14 Lester SC, Bose S, Chen Y et al. Protocol for the examination of specimens from patients with invasive carcinoma of the breast. Arch Pathol Lab Med 2009; 133: 1515-1538
  • 15 Lester SC, Bose S, Chen Y et al. Protocol for the examination of specimens from patients with ductal carcinoma in situ of the breast. Arch Pathol Lab Med 2009; 133: 15-25
  • 16 Hugh J, Hanson J, Cheang MCU et al. Breast cancer subtypes and response to docetaxel in node-positive breast cancer: use of an immunohistochemical definition in the BCIRG 001 trial. J Clin Oncol 2009; 27: 1168-1176
  • 17 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
  • 18 Sänger N, Effenberger KE, Riethdorf S et al. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. Int J Cancer 2011; 129: 2522-2526
  • 19 Hüsemann Y, Geigl JB, Schubert F et al. Systemic spread is an early step in breast cancer. Cancer Cell 2008; 13: 58-68
  • 20 Banys M, Gruber I, Krawczyk N et al. Hematogenous and lymphatic tumor cell dissemination may be detected in patients diagnosed with ductal carcinoma in situ of the breast. Breast Cancer Res Treat 2012; 131: 801-808
  • 21 Banys M, Hahn M, Gruber I et al. Detection and clinical relevance of hematogenous tumor cell dissemination in patients with ductal carcinoma in situ. Breast Cancer Res Treat 2014; 144: 531-538
  • 22 Arvold ND, Punglia RS, Hughes ME et al. Pathologic characteristics of second breast cancers after breast conservation for ductal carcinoma in situ. Cancer 2012; 118: 6022-6030
  • 23 Ma X, Salunga R, Tuggle JT et al. Gene expression profiles of human breast cancer progression. Proc Natl Acad Sci U S A 2003; 100: 5974-5979
  • 24 Schuetz CS, Bonin M, Clare SE et al. Progression-specific genes identified by expression profiling of matched ductal carcinomas in situ and invasive breast tumors, combining laser capture microdissection and oligonucleotide microarray analysis. Cancer Res 2006; 66: 5278-5286
  • 25 Vincent-Salomon A, Lucchesi C, Gruel N et al. Integrated genomic and transcriptomic analysis of ductal carcinoma in situ of the breast. Clin Cancer Res 2008; 14: 1956-1965
  • 26 Allred DC, Wu Y, Mao S et al. Ductal carcinoma in situ and the emergence of diversity during breast cancer evolution. Clin Cancer Res 2008; 14: 370-378
  • 27 Muggerud AA, Hallett M, Johnsen H et al. Molecular diversity in ductal carcinoma in situ (DCIS) and early invasive breast cancer. Mol Oncol 2010; 4: 357-368
  • 28 Stoecklein NH, Klein CA. Genetic disparity between primary tumours, disseminated tumour cells, and manifest metastasis. Int J Cancer 2010; 126: 589-598
  • 29 Lari SA, Kuerer HM. Biological markers in DCIS and risk of breast recurrence: a systematic review. J Cancer 2011; 2: 232-261
  • 30 Kerlikowske K, Molinaro AM, Gauthier ML et al. Biomarker expression and risk of subsequent tumors after initial ductal carcinoma in situ diagnosis. J Natl Cancer Inst 2010; 102: 627-637
  • 31 Zhou W, Jirström K, Johansson C et al. Long-term survival of women with basal-like ductal carcinoma in situ of the breast: a population-based cohort study. BMC Cancer 2010; 10: 653
  • 32 Han K, Nofech-Mozes S, Narod S et al. Expression of HER2neu in ductal carcinoma in situ is associated with local recurrence. Clin Oncol (R Coll Radiol) 2012; 24: 183-189