Skip to main content
Top
Published in: Breast Cancer Research and Treatment 2/2019

Open Access 01-11-2019 | Breast Cancer | Epidemiology

Reliability of preoperative breast biopsies showing ductal carcinoma in situ and implications for non-operative treatment: a cohort study

Authors: Gurdeep S. Mannu, Emma J. Groen, Zhe Wang, Michael Schaapveld, Esther H. Lips, Monica Chung, Ires Joore, Flora E. van Leeuwen, Hendrik J. Teertstra, Gonneke A. O. Winter-Warnars, Sarah C. Darby, Jelle Wesseling

Published in: Breast Cancer Research and Treatment | Issue 2/2019

Login to get access

Abstract

Purpose

The future of non-operative management of DCIS relies on distinguishing lesions requiring treatment from those needing only active surveillance. More accurate preoperative staging and grading of DCIS would be helpful. We identified determinants of upstaging preoperative breast biopsies showing ductal carcinoma in situ (DCIS) to invasive breast cancer (IBC), or of upgrading them to higher-grade DCIS, following examination of the surgically excised specimen.

Methods

We studied all women with DCIS at preoperative biopsy in a large specialist cancer centre during 2000–2014. Information from clinical records, mammography, and pathology specimens from both preoperative biopsy and excised specimen were abstracted. Women suspected of having IBC during biopsy were excluded.

Results

Among 606 preoperative biopsies showing DCIS, 15.0% (95% confidence interval 12.3–18.1) were upstaged to IBC and a further 14.6% (11.3–18.4) upgraded to higher-grade DCIS. The risk of upstaging increased with presence of a palpable lump (21.1% vs 13.0%, pdifference = 0.04), while the risk of upgrading increased with presence of necrosis on biopsy (33.0% vs 9.5%, pdifference < 0.001) and with use of 14G core-needle rather than 9G vacuum-assisted biopsy (22.8% vs 7.0%, pdifference < 0.001). Larger mammographic size increased the risk of both upgrading (pheterogeneity = 0.01) and upstaging (pheterogeneity = 0.004).

Conclusions

The risk of upstaging of DCIS in preoperative biopsies is lower than previously estimated and justifies conducting randomized clinical trials testing the safety of active surveillance for lower grade DCIS. Selection of women with low grade DCIS for such trials, or for active surveillance, may be improved by consideration of the additional factors identified in this study.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bleyer A, Welch HG (2012) Effect of three decades of screening mammography on breast-cancer incidence. N Engl J Med 367(21):1998–2005CrossRefPubMed Bleyer A, Welch HG (2012) Effect of three decades of screening mammography on breast-cancer incidence. N Engl J Med 367(21):1998–2005CrossRefPubMed
2.
go back to reference Kerlikowske K (2010) Epidemiology of ductal carcinoma in situ. JNCI Monogr 2010(41):139–141CrossRef Kerlikowske K (2010) Epidemiology of ductal carcinoma in situ. JNCI Monogr 2010(41):139–141CrossRef
3.
go back to reference Benson JR, Wishart GC (2013) Predictors of recurrence for ductal carcinoma in situ after breast-conserving surgery. Lancet Oncol 14(9):e348–e357CrossRefPubMed Benson JR, Wishart GC (2013) Predictors of recurrence for ductal carcinoma in situ after breast-conserving surgery. Lancet Oncol 14(9):e348–e357CrossRefPubMed
4.
go back to reference Jones JL (2006) Overdiagnosis and overtreatment of breast cancer: progression of ductal carcinoma in situ: the pathological perspective. Breast Cancer Res 8(2):204CrossRefPubMedPubMedCentral Jones JL (2006) Overdiagnosis and overtreatment of breast cancer: progression of ductal carcinoma in situ: the pathological perspective. Breast Cancer Res 8(2):204CrossRefPubMedPubMedCentral
5.
go back to reference Francis A et al (2015) Addressing overtreatment of screen detected DCIS; the LORIS trial. Eur J Cancer 51(16):2296–2303CrossRefPubMed Francis A et al (2015) Addressing overtreatment of screen detected DCIS; the LORIS trial. Eur J Cancer 51(16):2296–2303CrossRefPubMed
6.
go back to reference Benson JR, Jatoi I, Toi M (2016) Treatment of low-risk ductal carcinoma in situ: is nothing better than something? Lancet Oncol 17(10):e442–e451CrossRefPubMed Benson JR, Jatoi I, Toi M (2016) Treatment of low-risk ductal carcinoma in situ: is nothing better than something? Lancet Oncol 17(10):e442–e451CrossRefPubMed
7.
go back to reference Wiechmann L, Kuerer HM (2008) The molecular journey from ductal carcinoma in situ to invasive breast cancer. Cancer 112(10):2130–2142CrossRefPubMed Wiechmann L, Kuerer HM (2008) The molecular journey from ductal carcinoma in situ to invasive breast cancer. Cancer 112(10):2130–2142CrossRefPubMed
8.
go back to reference Youngwirth LM, Boughey JC, Hwang ES (2017) Surgery versus monitoring and endocrine therapy for low-risk DCIS: the COMET trial. Bull Am Coll Surg 102:62–63PubMed Youngwirth LM, Boughey JC, Hwang ES (2017) Surgery versus monitoring and endocrine therapy for low-risk DCIS: the COMET trial. Bull Am Coll Surg 102:62–63PubMed
9.
go back to reference Francis A, Fallowfield L, Rea D (2015) The LORIS trial: addressing overtreatment of ductal carcinoma in situ. Clin Oncol 27(1):6–8CrossRef Francis A, Fallowfield L, Rea D (2015) The LORIS trial: addressing overtreatment of ductal carcinoma in situ. Clin Oncol 27(1):6–8CrossRef
10.
go back to reference Elshof LE et al (2015) Feasibility of a prospective, randomised, open-label, international multicentre, phase III, non-inferiority trial to assess the safety of active surveillance for low risk ductal carcinoma in situ—the LORD study. Eur J Cancer 51(12):1497–1510CrossRefPubMed Elshof LE et al (2015) Feasibility of a prospective, randomised, open-label, international multicentre, phase III, non-inferiority trial to assess the safety of active surveillance for low risk ductal carcinoma in situ—the LORD study. Eur J Cancer 51(12):1497–1510CrossRefPubMed
11.
go back to reference NICE guidelines [CG80] (2009) Early and locally advanced breast cancer: Diagnosis and treatment. NICE guidance NICE guidelines [CG80] (2009) Early and locally advanced breast cancer: Diagnosis and treatment. NICE guidance
12.
go back to reference van Roozendaal LM et al (2016) Sentinel lymph node biopsy can be omitted in DCIS patients treated with breast conserving therapy. Breast Cancer Res Treat 156(3):517–525CrossRefPubMedPubMedCentral van Roozendaal LM et al (2016) Sentinel lymph node biopsy can be omitted in DCIS patients treated with breast conserving therapy. Breast Cancer Res Treat 156(3):517–525CrossRefPubMedPubMedCentral
14.
go back to reference Pilewskie M et al (2016) Do LORIS trial eligibility criteria identify a ductal carcinoma in situ patient population at low risk of upgrade to invasive carcinoma? Ann Surg Oncol 23(11):3487–3493CrossRefPubMedPubMedCentral Pilewskie M et al (2016) Do LORIS trial eligibility criteria identify a ductal carcinoma in situ patient population at low risk of upgrade to invasive carcinoma? Ann Surg Oncol 23(11):3487–3493CrossRefPubMedPubMedCentral
15.
go back to reference Brennan ME et al (2011) Ductal carcinoma in situ at core-needle biopsy: meta-analysis of underestimation and predictors of invasive breast cancer. Radiology 260(1):119–128CrossRefPubMed Brennan ME et al (2011) Ductal carcinoma in situ at core-needle biopsy: meta-analysis of underestimation and predictors of invasive breast cancer. Radiology 260(1):119–128CrossRefPubMed
16.
go back to reference Park HS et al (2013) A nomogram for predicting underestimation of invasiveness in ductal carcinoma in situ diagnosed by preoperative needle biopsy. The Breast 22(5):869–873CrossRefPubMed Park HS et al (2013) A nomogram for predicting underestimation of invasiveness in ductal carcinoma in situ diagnosed by preoperative needle biopsy. The Breast 22(5):869–873CrossRefPubMed
17.
go back to reference Grimm LJ et al (2017) Surgical upstaging rates for vacuum assisted biopsy proven DCIS: implications for active surveillance trials. Ann Surg Oncol 24(12):3534–3540CrossRefPubMedPubMedCentral Grimm LJ et al (2017) Surgical upstaging rates for vacuum assisted biopsy proven DCIS: implications for active surveillance trials. Ann Surg Oncol 24(12):3534–3540CrossRefPubMedPubMedCentral
18.
19.
go back to reference D’Orsi CJ (2013) ACR BI-RADS atlas: breast imaging reporting and data system. American College of Radiology, Virginia D’Orsi CJ (2013) ACR BI-RADS atlas: breast imaging reporting and data system. American College of Radiology, Virginia
20.
go back to reference Castro NP et al (2008) Evidence that molecular changes in cells occur before morphological alterations during the progression of breast ductal carcinoma. Breast Cancer Res 10(5):R87CrossRefPubMedPubMedCentral Castro NP et al (2008) Evidence that molecular changes in cells occur before morphological alterations during the progression of breast ductal carcinoma. Breast Cancer Res 10(5):R87CrossRefPubMedPubMedCentral
21.
go back to reference Thomson JZ et al (2001) Growth pattern of ductal carcinoma in situ (DCIS): a retrospective analysis based on mammographic findings. Br J Cancer 85(2):225–227CrossRefPubMedPubMedCentral Thomson JZ et al (2001) Growth pattern of ductal carcinoma in situ (DCIS): a retrospective analysis based on mammographic findings. Br J Cancer 85(2):225–227CrossRefPubMedPubMedCentral
22.
go back to reference Pape-Zambito D et al (2014) Identifying a highly-aggressive DCIS subgroup by studying intra-individual DCIS heterogeneity among invasive breast cancer patients. PLoS ONE 9(6):e100488CrossRefPubMedPubMedCentral Pape-Zambito D et al (2014) Identifying a highly-aggressive DCIS subgroup by studying intra-individual DCIS heterogeneity among invasive breast cancer patients. PLoS ONE 9(6):e100488CrossRefPubMedPubMedCentral
23.
go back to reference Lee CW et al (2016) Preoperative clinicopathologic factors and breast magnetic resonance imaging features can predict ductal carcinoma in situ with invasive components. Eur J Radiol 85(4):780–789CrossRefPubMed Lee CW et al (2016) Preoperative clinicopathologic factors and breast magnetic resonance imaging features can predict ductal carcinoma in situ with invasive components. Eur J Radiol 85(4):780–789CrossRefPubMed
24.
go back to reference Philpotts LE et al (1999) Comparison of rebiopsy rates after stereotactic core needle biopsy of the breast with 11-gauge vacuum suction probe versus 14-gauge needle and automatic gun. Am J Roentgenol 172(3):683–687CrossRef Philpotts LE et al (1999) Comparison of rebiopsy rates after stereotactic core needle biopsy of the breast with 11-gauge vacuum suction probe versus 14-gauge needle and automatic gun. Am J Roentgenol 172(3):683–687CrossRef
25.
go back to reference Jain RK et al (2011) Atypical ductal hyperplasia: interobserver and intraobserver variability. Mod Pathol 24(7):917CrossRefPubMed Jain RK et al (2011) Atypical ductal hyperplasia: interobserver and intraobserver variability. Mod Pathol 24(7):917CrossRefPubMed
26.
go back to reference Visser LL et al (2018) Clinicopathological risk factors for an invasive breast cancer recurrence after ductal carcinoma in situ-A nested case-control study. Clin Cancer Res 24(15):3593–3601CrossRefPubMed Visser LL et al (2018) Clinicopathological risk factors for an invasive breast cancer recurrence after ductal carcinoma in situ-A nested case-control study. Clin Cancer Res 24(15):3593–3601CrossRefPubMed
27.
go back to reference van Luijt PA et al (2016) The distribution of ductal carcinoma in situ (DCIS) grade in 4232 women and its impact on overdiagnosis in breast cancer screening. Breast Cancer Res 18(1):47CrossRefPubMedPubMedCentral van Luijt PA et al (2016) The distribution of ductal carcinoma in situ (DCIS) grade in 4232 women and its impact on overdiagnosis in breast cancer screening. Breast Cancer Res 18(1):47CrossRefPubMedPubMedCentral
28.
go back to reference Sarode VR et al (2011) A comparative analysis of biomarker expression and molecular subtypes of pure ductal carcinoma in situ and invasive breast carcinoma by image analysis: relationship of the subtypes with histologic grade, Ki67, p53 overexpression, and DNA ploidy. Int J Breast Cancer 2011:217060CrossRefPubMedPubMedCentral Sarode VR et al (2011) A comparative analysis of biomarker expression and molecular subtypes of pure ductal carcinoma in situ and invasive breast carcinoma by image analysis: relationship of the subtypes with histologic grade, Ki67, p53 overexpression, and DNA ploidy. Int J Breast Cancer 2011:217060CrossRefPubMedPubMedCentral
30.
go back to reference Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674CrossRef Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674CrossRef
Metadata
Title
Reliability of preoperative breast biopsies showing ductal carcinoma in situ and implications for non-operative treatment: a cohort study
Authors
Gurdeep S. Mannu
Emma J. Groen
Zhe Wang
Michael Schaapveld
Esther H. Lips
Monica Chung
Ires Joore
Flora E. van Leeuwen
Hendrik J. Teertstra
Gonneke A. O. Winter-Warnars
Sarah C. Darby
Jelle Wesseling
Publication date
01-11-2019
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 2/2019
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-019-05362-1

Other articles of this Issue 2/2019

Breast Cancer Research and Treatment 2/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine