1
|
Rohan TE, Wang Y, Couch F, Feigelson HS, Greenlee RT, Honda S, Stark A, Chitale D, Zhang C, Xue X, Ginsberg M, Loudig O. Clinicopathologic characteristics of ductal carcinoma in situ and risk of subsequent invasive breast cancer: a multicenter, population-based cohort study. Breast Cancer Res Treat 2025; 210:615-625. [PMID: 39832051 PMCID: PMC11953078 DOI: 10.1007/s10549-024-07599-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE To study the association between clinicopathologic characteristics of ductal carcinoma in situ (DCIS) and risk of subsequent invasive breast cancer (IBC). METHODS We conducted a case-control study nested in a multicenter, population-based cohort of 8175 women aged ≥ 18 years with DCIS diagnosed between 1987 and 2016 and followed for a median duration of 83 months. Cases (n = 497) were women with a first diagnosis of DCIS who developed a subsequent IBC ≥ 6 months later; controls (2/case; n = 959) were matched to cases on age at and calendar year of DCIS diagnosis. Univariable and multivariable conditional logistic regression models were used to examine the associations between the DCIS characteristics of interest (non-screen detection of DCIS, tumor size, positive margins, grade of DCIS, necrosis, architectural pattern, microcalcification, and estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status) and risk of IBC. RESULTS In the total study population, the associations were largely null. In subgroup analyses, there were strong position associations with punctate necrosis (pre/perimenopausal women), detection by physical exam (postmenopausal women), architectural patterns other than the main types (breast-conserving surgery [BCS]), and DCIS margins (ipsilateral cases), and inverse associations with HER2 positivity (BCS) and microcalcification (mastectomy); however, the associated confidence intervals were mostly very wide. CONCLUSION The results of this study provide limited support for associations of the DCIS clinicopathologic characteristics studied here and risk of IBC.
Collapse
Affiliation(s)
- Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Fergus Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Robert T Greenlee
- Center for Clinical Epidemiology and Population Health, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Stacey Honda
- Center for Integrated Healthcare, Kaiser Permanente, Hawaii Permanente Medical Group, Honolulu, HI, USA
| | - Azadeh Stark
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, MI, USA
- Breast Oncology Program and Department of Pathology, Henry Ford Health System, Detroit, MI, USA
| | - Dhananjay Chitale
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, MI, USA
- Breast Oncology Program and Department of Pathology, Henry Ford Health System, Detroit, MI, USA
| | - Chenxin Zhang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mindy Ginsberg
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Olivier Loudig
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, Nutley, NJ, USA
| |
Collapse
|
2
|
Sobral-Leite M, Castillo SP, Vonk S, Messal HA, Melillo X, Lam N, de Bruijn B, Hagos YB, van den Bos M, Sanders J, Almekinders M, Visser LL, Groen EJ, Kristel P, Ercan C, Azarang L, van Rheenen J, Hwang ES, Yuan Y, Menezes R, Lips EH, Wesseling J. A morphometric signature to identify ductal carcinoma in situ with a low risk of progression. NPJ Precis Oncol 2025; 9:25. [PMID: 39875514 PMCID: PMC11775207 DOI: 10.1038/s41698-024-00769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 11/21/2024] [Indexed: 01/30/2025] Open
Abstract
Ductal carcinoma in situ (DCIS) may progress to ipsilateral invasive breast cancer (iIBC), but often never will. Because DCIS is treated as early breast cancer, many women with harmless DCIS face overtreatment. To identify features associated with progression, we developed an artificial intelligence-based DCIS morphometric analysis pipeline (AIDmap) on hematoxylin-eosin-stained (H&E) tissue sections. We analyzed 689 digitized H&Es of pure primary DCIS of which 226 were diagnosed with subsequent iIBC and 463 were not. The distribution of 15 duct morphological measurements was summarized in 55 morphometric variables. A ridge regression classifier with cross validation predicted 5-years-free of iIBC with an area-under the curve of 0.67 (95% CI 0.57-0.77). A combined clinical-morphometric signature, characterized by small-sized ducts, a low number of cells and a low DCIS/stroma ratio, was associated with outcome (HR = 0.56; 95% CI 0.28-0.78). AIDmap has potential to identify harmless DCIS that may not need treatment.
Collapse
Affiliation(s)
- Marcelo Sobral-Leite
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Simon P Castillo
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiva Vonk
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hendrik A Messal
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Xenia Melillo
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Noomie Lam
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Brandi de Bruijn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yeman B Hagos
- Sarcoma Molecular Pathology Team, The Institute of Cancer Research, London, UK
| | - Myrna van den Bos
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joyce Sanders
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mathilde Almekinders
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lindy L Visser
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emma J Groen
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petra Kristel
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Caner Ercan
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Leyla Azarang
- Biostatistics Centre and Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - E Shelley Hwang
- Department of Surgery, Duke University Comprehensive Cancer Center, Durham, NC, USA
| | - Yinyin Yuan
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renee Menezes
- Biostatistics Centre and Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Pathology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
3
|
Yoon KH, Kim EK, Shin HC. Prognostic implications of ductal carcinoma in situ components in BRCA1/2-positive breast cancer: a retrospective cohort study. Ann Surg Treat Res 2024; 107:327-335. [PMID: 39669383 PMCID: PMC11634396 DOI: 10.4174/astr.2024.107.6.327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose Although the breast cancer susceptibility gene (BRCA)-associated invasive breast cancer is well studied, there are limited reports on ductal carcinoma in situ (DCIS) in patients with BRCA1/2 mutations. This study aims to evaluate the differential prognostic effect of DCIS in breast cancer patients with pathologic variants of BRCA1/2 genes. Methods Breast cancer patients who tested positive for BRCA1/2 mutations between August 2003 and January 2022 at a single tertiary referral center were retrospectively analyzed. Survival outcomes were compared between patients with both invasive ductal carcinoma (IDC) and DCIS (IDC-DCIS group, n = 121) and those with IDC alone (IDC group, n = 36). Results Of the 157 patients, 65 (41.4%) exhibited mutations in BRCA1, 90 (57.3%) in BRCA2, and 2 (1.3%) in both BRCA1/2. DCIS components were more frequently found in BRCA2 pathological variants (BRCA1, 46 [38.0%] vs. BRCA2, 76 [62.4%]; P = 0.030). No statistically significant difference was found in 10-year recurrence-free survival (IDC-DCIS, 89.3% vs. IDC, 83.6%; P = 0.989). Subgroup analysis indicated that the DCIS component correlated with improved survival outcomes in the BRCA1 subgroup (BRCA1 IDC-DCIS, 85.5% vs. BRCA1 IDC, 51.0%; P = 0.024). Conversely, in the BRCA2 subgroup, IDC-DCIS patients exhibited a worse prognosis (BRCA1 IDC-DCIS, 85.5% vs. BRCA2 IDC-DCIS, 65.8%; P = 0.045). Conclusion The presence of a DCIS component carries varied prognostic significance in BRCA1 and BRCA2 mutations. A tailored approach may be necessary when determining treatment options for breast cancer patients with BRCA1/2 mutations based on the presence of DCIS.
Collapse
Affiliation(s)
- Kyung-Hwak Yoon
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Eun-Kyu Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hee-Chul Shin
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
4
|
Byng D, Schaapveld M, Lips EH, van Duijnhoven FH, Wesseling J, van Harten WH, Retèl VP. An early economic evaluation of active surveillance for low-risk ductal carcinoma in situ. Future Oncol 2024; 20:3451-3462. [PMID: 39676693 DOI: 10.1080/14796694.2024.2421152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/22/2024] [Indexed: 12/17/2024] Open
Abstract
Aim: Perform early economic evaluation comparing active surveillance (AS) to surgery for women with low-risk ductal carcinoma in situ, a precursor of invasive breast cancer.Materials & methods: A 10-year incremental costs (€) and quality-adjusted life years (QALYs) were compared between a simulated cohort of women undergoing breast conserving surgery ± radiotherapy, and a cohort with a low-risk subgroup undergoing AS using a semi-Markov model. Scenario and headroom analyses evaluated a better-performing biomarker to select low-risk women for AS.Results: AS resulted in lower costs and survival, but higher QALYs (±0.40). Scenario analyses maintained survival outcomes and maximized QALYs.Conclusion: AS for low-risk ductal carcinoma in situ is cost-effective, but a better-performing biomarker to select low-risk women can maximize quality-adjusted outcomes.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/economics
- Breast Neoplasms/mortality
- Breast Neoplasms/therapy
- Breast Neoplasms/diagnosis
- Breast Neoplasms/pathology
- Cost-Benefit Analysis
- Carcinoma, Intraductal, Noninfiltrating/economics
- Carcinoma, Intraductal, Noninfiltrating/mortality
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Carcinoma, Intraductal, Noninfiltrating/diagnosis
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Quality-Adjusted Life Years
- Watchful Waiting/economics
- Middle Aged
- Mastectomy, Segmental/economics
- Markov Chains
- Aged
Collapse
Affiliation(s)
- Danalyn Byng
- Division of Psychosocial Research & Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, PA 3062, The Netherlands
| | - Michael Schaapveld
- Division of Psychosocial Research & Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
| | - Frederieke H van Duijnhoven
- Division of Surgical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, ZA 2333, The Netherlands
| | - Wim H van Harten
- Division of Psychosocial Research & Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, PA 3062, The Netherlands
| | - Valesca P Retèl
- Division of Psychosocial Research & Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, CX 1066, The Netherlands
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, PA 3062, The Netherlands
| |
Collapse
|
5
|
Van Bockstal MR, Wesseling J, Lips EH, Smidt M, Galant C, van Deurzen CHM. Systematic assessment of HER2 status in ductal carcinoma in situ of the breast: a perspective on the potential clinical relevance. Breast Cancer Res 2024; 26:125. [PMID: 39192322 DOI: 10.1186/s13058-024-01875-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024] Open
Abstract
In many countries, hormone receptor status assessment of ductal carcinoma in situ (DCIS) is routinely performed, as hormone receptor-positive DCIS patients are eligible for adjuvant anti-hormonal treatment, aiming to reduce the ipsilateral and contralateral breast cancer risk. Although HER2 gene amplification and its associated HER2 protein overexpression constitute a major prognostic and predictive marker in invasive breast carcinoma, its use in the diagnosis and treatment of DCIS is less straightforward. HER2 immunohistochemistry is not routinely performed yet, as the role of HER2-positivity in DCIS biology is unclear. Nonetheless, recent data challenge this practice. Here, we discuss the value of routine HER2 assessment for DCIS. HER2-positivity correlates strongly with DCIS grade: around four in five HER2-positive DCIS show high grade atypia. As morphological DCIS grading is prone to interobserver variability, HER2 immunohistochemistry could render grading more robust. Several studies showed an association between HER2-positive DCIS and ipsilateral recurrence risk, albeit currently unclear whether this is for overall, in situ or invasive recurrence. HER2-positive DCIS tends to be larger, with a higher risk of involved surgical margins. HER2-positive DCIS patients benefit more from adjuvant radiotherapy: it substantially decreases the local recurrence risk after lumpectomy, without impact on overall survival. HER2-positivity in pure biopsy-diagnosed DCIS is associated with increased upstaging to invasive carcinoma after surgery. HER2 immunohistochemistry on preoperative biopsies might therefore provide useful information to surgeons, favoring wider excisions. The time seems right to consider DCIS subtype-dependent treatment, comprising appropriate local treatment for HER2-positive DCIS patients and de-escalation for hormone receptor-positive, HER2-negative DCIS patients.
Collapse
MESH Headings
- Humans
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/therapy
- Breast Neoplasms/mortality
- Breast Neoplasms/diagnosis
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Biomarkers, Tumor/metabolism
- Prognosis
- Immunohistochemistry
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/genetics
- Neoplasm Grading
- Clinical Relevance
Collapse
Affiliation(s)
- Mieke R Van Bockstal
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.
- Pôle de Morphologie (MORF), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Ester H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marjolein Smidt
- Department of Surgery, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Christine Galant
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
- Pôle de Morphologie (MORF), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Carolien H M van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute Rotterdam, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
6
|
Hutten SJ, Messal HA, Lips EH, Sheinman M, Ciwinska M, Braams E, van der Borden C, Kristel P, Stoffers S, Wessels LF, Jonkers J, van Rheenen J, Wesseling J, Scheele CL. Ductal carcinoma in situ develops within clonal fields of mutant cells in morphologically normal ducts. J Pathol 2024; 263:360-371. [PMID: 38779852 DOI: 10.1002/path.6289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/28/2024] [Accepted: 03/30/2024] [Indexed: 05/25/2024]
Abstract
Mutations are abundantly present in tissues of healthy individuals, including the breast epithelium. Yet it remains unknown whether mutant cells directly induce lesion formation or first spread, leading to a field of mutant cells that is predisposed towards lesion formation. To study the clonal and spatial relationships between morphologically normal breast epithelium adjacent to pre-cancerous lesions, we developed a three-dimensional (3D) imaging pipeline combined with spatially resolved genomics on archival, formalin-fixed breast tissue with the non-obligate breast cancer precursor ductal carcinoma in situ (DCIS). Using this 3D image-guided characterization method, we built high-resolution spatial maps of DNA copy number aberration (CNA) profiles within the DCIS lesion and the surrounding normal mammary ducts. We show that the local heterogeneity within a DCIS lesion is limited. However, by mapping the CNA profiles back onto the 3D reconstructed ductal subtree, we find that in eight out of 16 cases the healthy epithelium adjacent to the DCIS lesions has overlapping structural variations with the CNA profile of the DCIS. Together, our study indicates that pre-malignant breast transformations frequently develop within mutant clonal fields of morphologically normal-looking ducts. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stefan J Hutten
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Hendrik A Messal
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michael Sheinman
- Oncode Institute, Amsterdam, The Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marta Ciwinska
- Department of Oncology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Esmee Braams
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Carolien van der Borden
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petra Kristel
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Saskia Stoffers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lodewyk Fa Wessels
- Oncode Institute, Amsterdam, The Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Colinda Lgj Scheele
- Department of Oncology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Leonardi MC, Zerella MA, Lazzeroni M, Fusco N, Veronesi P, Galimberti VE, Corso G, Dicuonzo S, Rojas DP, Morra A, Gerardi MA, Lorubbio C, Zaffaroni M, Vincini MG, Orecchia R, Jereczek-Fossa BA, Magnoni F. Tools to Guide Radiation Oncologists in the Management of DCIS. Healthcare (Basel) 2024; 12:795. [PMID: 38610216 PMCID: PMC11011767 DOI: 10.3390/healthcare12070795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Similar to invasive breast cancer, ductal carcinoma in situ is also going through a phase of changes not only from a technical but also a conceptual standpoint. From prescribing radiotherapy to everyone to personalized approaches, including radiotherapy omission, there is still a lack of a comprehensive framework to guide radiation oncologists in decision making. Many pieces of the puzzle are finding their place as high-quality data mature and are disseminated, but very often, the interpretation of risk factors and the perception of risk remain very highly subjective. Sharing the therapeutic choice with patients requires effective communication for an understanding of risks and benefits, facilitating an informed decision that does not increase anxiety and concerns about prognosis. The purpose of this narrative review is to summarize the current state of knowledge to highlight the tools available to radiation oncologists for managing DCIS, with an outlook on future developments.
Collapse
Affiliation(s)
- Maria Cristina Leonardi
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Maria Alessia Zerella
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Matteo Lazzeroni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (N.F.); (P.V.); (G.C.)
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Paolo Veronesi
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (N.F.); (P.V.); (G.C.)
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (V.E.G.); (F.M.)
| | - Viviana Enrica Galimberti
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (V.E.G.); (F.M.)
| | - Giovanni Corso
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (N.F.); (P.V.); (G.C.)
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (V.E.G.); (F.M.)
| | - Samantha Dicuonzo
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Damaris Patricia Rojas
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Anna Morra
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Marianna Alessandra Gerardi
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Chiara Lorubbio
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (N.F.); (P.V.); (G.C.)
| | - Mattia Zaffaroni
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Maria Giulia Vincini
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
| | - Roberto Orecchia
- Scientific Directorate, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.C.L.); (S.D.); (D.P.R.); (A.M.); (M.A.G.); (C.L.); (M.Z.); (M.G.V.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (N.F.); (P.V.); (G.C.)
| | - Francesca Magnoni
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (V.E.G.); (F.M.)
| |
Collapse
|
8
|
Alaeikhanehshir S, Voets MM, van Duijnhoven FH, Lips EH, Groen EJ, van Oirsouw MCJ, Hwang SE, Lo JY, Wesseling J, Mann RM, Teuwen J. Application of deep learning on mammographies to discriminate between low and high-risk DCIS for patient participation in active surveillance trials. Cancer Imaging 2024; 24:48. [PMID: 38576031 PMCID: PMC10996224 DOI: 10.1186/s40644-024-00691-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Ductal Carcinoma In Situ (DCIS) can progress to invasive breast cancer, but most DCIS lesions never will. Therefore, four clinical trials (COMET, LORIS, LORETTA, AND LORD) test whether active surveillance for women with low-risk Ductal carcinoma In Situ is safe (E. S. Hwang et al., BMJ Open, 9: e026797, 2019, A. Francis et al., Eur J Cancer. 51: 2296-2303, 2015, Chizuko Kanbayashi et al. The international collaboration of active surveillance trials for low-risk DCIS (LORIS, LORD, COMET, LORETTA), L. E. Elshof et al., Eur J Cancer, 51, 1497-510, 2015). Low-risk is defined as grade I or II DCIS. Because DCIS grade is a major eligibility criteria in these trials, it would be very helpful to assess DCIS grade on mammography, informed by grade assessed on DCIS histopathology in pre-surgery biopsies, since surgery will not be performed on a significant number of patients participating in these trials. OBJECTIVE To assess the performance and clinical utility of a convolutional neural network (CNN) in discriminating high-risk (grade III) DCIS and/or Invasive Breast Cancer (IBC) from low-risk (grade I/II) DCIS based on mammographic features. We explored whether the CNN could be used as a decision support tool, from excluding high-risk patients for active surveillance. METHODS In this single centre retrospective study, 464 patients diagnosed with DCIS based on pre-surgery biopsy between 2000 and 2014 were included. The collection of mammography images was partitioned on a patient-level into two subsets, one for training containing 80% of cases (371 cases, 681 images) and 20% (93 cases, 173 images) for testing. A deep learning model based on the U-Net CNN was trained and validated on 681 two-dimensional mammograms. Classification performance was assessed with the Area Under the Curve (AUC) receiver operating characteristic and predictive values on the test set for predicting high risk DCIS-and high-risk DCIS and/ or IBC from low-risk DCIS. RESULTS When classifying DCIS as high-risk, the deep learning network achieved a Positive Predictive Value (PPV) of 0.40, Negative Predictive Value (NPV) of 0.91 and an AUC of 0.72 on the test dataset. For distinguishing high-risk and/or upstaged DCIS (occult invasive breast cancer) from low-risk DCIS a PPV of 0.80, a NPV of 0.84 and an AUC of 0.76 were achieved. CONCLUSION For both scenarios (DCIS grade I/II vs. III, DCIS grade I/II vs. III and/or IBC) AUCs were high, 0.72 and 0.76, respectively, concluding that our convolutional neural network can discriminate low-grade from high-grade DCIS.
Collapse
MESH Headings
- Humans
- Female
- Carcinoma, Intraductal, Noninfiltrating/diagnostic imaging
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Retrospective Studies
- Deep Learning
- Patient Participation
- Watchful Waiting
- Breast Neoplasms/diagnostic imaging
- Breast Neoplasms/pathology
- Mammography
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/surgery
Collapse
Affiliation(s)
- Sena Alaeikhanehshir
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Surgery, the Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Madelon M Voets
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Health Services and Technology Research, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | | | - Esther H Lips
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Emma J Groen
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Shelley E Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Joseph Y Lo
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Jelle Wesseling
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pathology, the Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ritse M Mann
- Department of Radiology, the Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jonas Teuwen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands.
- Department of Radiation Oncology, the Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, USA.
- Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
| |
Collapse
|
9
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
10
|
Sobral-Leite M, Castillo S, Vonk S, Melillo X, Lam N, de Bruijn B, Hagos Y, Sanders J, Almekinders M, Visser L, Groen E, Kristel P, Ercan C, Azarang L, Yuan Y, Menezes R, Lips E, Wesseling J. Artificial intelligence-based morphometric signature to identify ductal carcinoma in situ with low risk of progression to invasive breast cancer. RESEARCH SQUARE 2023:rs.3.rs-3639521. [PMID: 38168198 PMCID: PMC10760295 DOI: 10.21203/rs.3.rs-3639521/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Ductal carcinoma in situ (DCIS) may progress to ipsilateral invasive breast cancer (iIBC), but often never will. Because DCIS is treated as early breast cancer, many women with harmless DCIS face overtreatment. To identify these women that may forego treatment, we hypothesized that DCIS morphometric features relate to the risk of subsequent iIBC. We developed an artificial intelligence-based DCIS morphometric analysis pipeline (AIDmap) to detect DCIS as a pathologist and measure morphological structures in hematoxylin-eosin-stained (H&E) tissue sections. These were from a case-control study of patients diagnosed with primary DCIS, treated by breast-conserving surgery without radiotherapy. We analyzed 689 WSIs of DCIS of which 226 were diagnosed with subsequent iIBC (cases) and 463 were not (controls). The distribution of 15 duct morphological measurements in each H&E was summarized in 55 morphometric variables. A ridge regression classifier with cross validation predicted 5-years-free of iIBC with an area-under the curve of 0.65 (95% CI 0.55-0.76). A morphometric signature based on the 30 variables most associated with outcome, identified lesions containing small-sized ducts, low number of cells and low DCIS/stroma area ratio. This signature was associated with lower iIBC risk in a multivariate regression model including grade, ER, HER2 and COX-2 expression (HR = 0.56; 95% CI 0.28-0.78). AIDmap has potential to identify harmless DCIS that may not need treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Caner Ercan
- The University of Texas MD Anderson Cancer Center
| | | | | | | | | | | |
Collapse
|
11
|
Gibson SV, Roozitalab RM, Allen MD, Jones JL, Carter EP, Grose RP. Everybody needs good neighbours: the progressive DCIS microenvironment. Trends Cancer 2023; 9:326-338. [PMID: 36739265 DOI: 10.1016/j.trecan.2023.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/05/2023]
Abstract
Ductal carcinoma in situ (DCIS) is a pre-invasive form of breast cancer where neoplastic luminal cells are confined to the ductal tree. While as many as 70% of DCIS cases will remain indolent, most women are treated with surgery, often combined with endocrine and radiotherapies. Overtreatment is therefore a major issue, demanding new methods to stratify patients. Somewhat paradoxically, the neoplastic cells in DCIS are genetically comparable to those in invasive disease, suggesting the tumour microenvironment is the driving force for progression. Clinical and mechanistic studies highlight the complex DCIS microenvironment, with multiple cell types competing to regulate progression. Here, we examine recent studies detailing distinct aspects of the DCIS microenvironment and discuss how these may inform more effective care.
Collapse
Affiliation(s)
- Shayin V Gibson
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Reza M Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Michael D Allen
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
12
|
Udayasiri RI, Luo T, Gorringe KL, Fox SB. Identifying recurrences and metastasis after ductal carcinoma in situ (DCIS) of the breast. Histopathology 2023; 82:106-118. [PMID: 36482277 PMCID: PMC10953414 DOI: 10.1111/his.14804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 12/13/2022]
Abstract
Ductal carcinoma in situ (DCIS) of the breast is a non-invasive tumour that has the potential to progress to invasive ductal carcinoma (IDC). Thus, it represents a treatment dilemma: alone it does not present a risk to life, however, left untreated it may progress to a life-threatening condition. Current clinico-pathological features cannot accurately predict which patients with DCIS have invasive potential, and therefore clinicians are unable to quantify the risk of progression for an individual patient. This leads to many women being over-treated, while others may not receive sufficient treatment to prevent invasive recurrence. A better understanding of the molecular features of DCIS, both tumour-intrinsic and the microenvironment, could offer the ability to better predict which women need aggressive treatment, and which can avoid therapies carrying significant side-effects and such as radiotherapy. In this review, we summarise the current knowledge of DCIS, and consider future research directions.
Collapse
Affiliation(s)
- Ruwangi I Udayasiri
- Peter MacCallum Cancer Centre and the Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVICAustralia
| | - Tongtong Luo
- Peter MacCallum Cancer Centre and the Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVICAustralia
| | - Kylie L Gorringe
- Peter MacCallum Cancer Centre and the Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVICAustralia
| | - Stephen B Fox
- Peter MacCallum Cancer Centre and the Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVICAustralia
| |
Collapse
|
13
|
Akrida I, Mulita F. The clinical significance of HER2 expression in DCIS. Med Oncol 2022; 40:16. [PMID: 36352293 DOI: 10.1007/s12032-022-01876-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/15/2022] [Indexed: 11/11/2022]
Abstract
HER2 is an established prognostic and predictive marker for patients with invasive breast cancer. The clinical and biological significance of HER2 overexpression in patients with ductal carcinoma in situ (DCIS) remains poorly defined. DCIS is a heterogeneous disease and some patients with DCIS will not progress to invasive breast cancer. However, clinically significant recurrence rates have been reported after breast-conserving surgery for DCIS and approximately half of these cases will be life-threatening invasive recurrences. Since the incidence of DCIS is rising due to the widespread use of screening mammography, there is robust interest in selecting high-risk DCIS patients that may benefit from adjuvant therapies. Molecular prognostic and predictive models in early invasive breast cancer help clinicians identify patients that will benefit from chemotherapy. Molecular subtyping and profiling could also be useful in treating DCIS patients. According to current practice guidelines, HER2 testing is not recommended in DCIS patients. Nevertheless, evidence suggests that HER2-positive DCIS cases may be associated with adverse clinicopathological parameters and increased recurrence rates. This review summarizes the existing body of evidence linking HER2 expression and ipsilateral breast cancer recurrence in DCIS. HER2, as well as its correlation with other clinicopathological markers might be a useful prognostic and predictive marker, helping clinical decision-making in DCIS patients.
Collapse
Affiliation(s)
- Ioanna Akrida
- Department of General Surgery, University General Hospital of Patras, Rion, Greece.
- Department of Anatomy-Histology-Embryology, University of Patras Medical School, 26504, Rion, Greece.
| | - Francesk Mulita
- Department of General Surgery, University General Hospital of Patras, Rion, Greece
| |
Collapse
|
14
|
Almekinders MM, Bismeijer T, Kumar T, Yang F, Thijssen B, van der Linden R, van Rooijen C, Vonk S, Sun B, Parra Cuentas ER, Wistuba II, Krishnamurthy S, Visser LL, Seignette IM, Hofland I, Sanders J, Broeks A, Love JK, Menegaz B, Wessels L, Thompson AM, de Visser KE, Hooijberg E, Lips E, Futreal A, Wesseling J. Comprehensive multiplexed immune profiling of the ductal carcinoma in situ immune microenvironment regarding subsequent ipsilateral invasive breast cancer risk. Br J Cancer 2022; 127:1201-1213. [PMID: 35768550 PMCID: PMC9519539 DOI: 10.1038/s41416-022-01888-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 12/25/2022] Open
Abstract
Background Ductal carcinoma in situ (DCIS) is treated to prevent subsequent ipsilateral invasive breast cancer (iIBC). However, many DCIS lesions will never become invasive. To prevent overtreatment, we need to distinguish harmless from potentially hazardous DCIS. We investigated whether the immune microenvironment (IME) in DCIS correlates with transition to iIBC. Methods Patients were derived from a Dutch population-based cohort of 10,090 women with pure DCIS with a median follow-up time of 12 years. Density, composition and proximity to the closest DCIS cell of CD20+ B-cells, CD3+CD8+ T-cells, CD3+CD8− T-cells, CD3+FOXP3+ regulatory T-cells, CD68+ cells, and CD8+Ki67+ T-cells was assessed with multiplex immunofluorescence (mIF) with digital whole-slide analysis and compared between primary DCIS lesions of 77 women with subsequent iIBC (cases) and 64 without (controls). Results Higher stromal density of analysed immune cell subsets was significantly associated with higher grade, ER negativity, HER-2 positivity, Ki67 ≥ 14%, periductal fibrosis and comedonecrosis (P < 0.05). Density, composition and proximity to the closest DCIS cell of all analysed immune cell subsets did not differ between cases and controls. Conclusion IME features analysed by mIF in 141 patients from a well-annotated cohort of pure DCIS with long-term follow-up are no predictors of subsequent iIBC, but do correlate with other factors (grade, ER, HER2 status, Ki-67) known to be associated with invasive recurrences. ![]()
Collapse
Affiliation(s)
- Mathilde M Almekinders
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tycho Bismeijer
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tapsi Kumar
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA.,Department of Genetics, MD Anderson Cancer Center, Houston, TX, USA.,MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Fei Yang
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Bram Thijssen
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rianne van der Linden
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Charlotte van Rooijen
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Shiva Vonk
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.,Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Baohua Sun
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra Cuentas
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Lindy L Visser
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Iris M Seignette
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Joyce Sanders
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jason K Love
- Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Brian Menegaz
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Lodewyk Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | | | - Karin E de Visser
- Oncode Institute, Utrecht, The Netherlands.,Division of Tumour Biology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik Hooijberg
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Esther Lips
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andrew Futreal
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands. .,Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands. .,Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|
15
|
Zheng L, Gökmen-Polar Y, Badve SS. Is conservative management of ductal carcinoma in situ risky? NPJ Breast Cancer 2022; 8:55. [PMID: 35484283 PMCID: PMC9050725 DOI: 10.1038/s41523-022-00420-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/22/2022] [Indexed: 12/15/2022] Open
Abstract
Nonsurgical management of ductal carcinoma in situ is controversial and little is known about the long-term consequences of this approach. In this study, we aimed to determine the risk of (a) upstaging to invasive carcinoma at excision and (b) ipsilateral breast cancer events in patients who might have been eligible for nonsurgical management of DCIS trials. Data from women aged 20 years or older with a biopsy diagnosis of DCIS between January 1, 2010 to December 31, 2014 were collated. The women underwent biopsy and surgical resection (lumpectomy or mastectomy) and were treated with radiation or endocrine therapy as per treating physicians’ choice. The development of ipsilateral breast cancer events (IBEs) was analyzed in patients with at least 5 years of follow-up after standard of care therapy for DCIS. Subset-analysis was undertaken to identify the incidence of IBEs in patients eligible for nonsurgical management trials. The study population consisted of 378 patients with matched cases of biopsy and surgical excision. The overall upstaging rate to IBC was 14.3 and 12.9% for COMET, 8.8% for LORIS, and 10.7% for LORD trial “eligible” patients. At 5 years of follow-up, ~11.5% of overall and trial eligible patients developed IBEs of which approximately half were invasive IBEs. In conclusion, women with DCIS who would have been eligible for nonsurgical management trials have a significantly high risk of developing ipsilateral breast events within 5 years of diagnosis. Better selection criteria are needed to identify DCIS patients who are at very low risk for the development of IBC.
Collapse
Affiliation(s)
- Lan Zheng
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yesim Gökmen-Polar
- Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Road, H 184, Atlanta, GA, 30322, USA
| | - Sunil S Badve
- Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Road, H 184, Atlanta, GA, 30322, USA.
| |
Collapse
|
16
|
Thorat MA, Levey PM, Jones JL, Pinder SE, Bundred NJ, Fentiman IS, Cuzick J. Prognostic and Predictive Value of HER2 Expression in Ductal Carcinoma In Situ: Results from the UK/ANZ DCIS Randomized Trial. Clin Cancer Res 2021; 27:5317-5324. [PMID: 34380636 PMCID: PMC7612534 DOI: 10.1158/1078-0432.ccr-21-1239] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/23/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE HER2 is overexpressed more frequently in ductal carcinoma in situ (DCIS) than in invasive breast cancer but its prognostic significance and predictive role for radiotherapy has not been clearly established. We investigated the prognostic and predictive value of HER2 overexpression in DCIS. EXPERIMENTAL DESIGN HER2 expression was evaluated by IHC using the HercepTest™ in samples from UK/ANZ DCIS trial participants (n = 755) with IHC 3+ expression categorized as HER2 positive for primary analyses. Sensitivity analyses included HER2 categorization as negative (IHC 0,1+), equivocal (IHC 2+), and positive (IHC 3+) and analyses restricted to a nested case-control component where 181 cases (with recurrence) were matched to 362 controls by treatment arm and age. RESULTS Two-hundred and forty-five (34.4%) of evaluable 713 samples [181 ipsilateral breast events (IBE)] were HER2 positive. HER2 overexpression was associated with significantly increased risk of IBE [HR = 2.29; 95% confidence interval (95% CI), 1.64-3.14; P < 0.0001] and in situ IBE (DCIS-IBE; HR = 2.90; 95% CI, 1.91-4.40; P < 0.0001), but not of invasive IBE (I-IBE; HR = 1.40; 95% CI, 0.81-2.42; P = 0.23; Pheterogeneity = 0.04). Inclusion of HER2 significantly improved [Δχ2 (1d.f.) 12.25; P = 0.0005] a prognostic model of clinicopathological and treatment variables, HER2 being an independent predictor of IBE (multivariate HR = 1.91; 95% CI, 1.33-2.76; P = 0.0004). Radiotherapy benefit in preventing DCIS-IBE was significantly greater (Pheterogeneity = 0.04) in HER2-positive DCIS (HR = 0.16; 95% CI, 0.07-0.41) compared with HER2-negative DCIS (HR = 0.58; 95% CI, 0.28-1.19). CONCLUSIONS HER2 overexpression is associated with significantly increased risk of in situ recurrence and is also predictive of radiotherapy benefit, with greater reductions in in situ but not invasive recurrences in HER2-positive DCIS.
Collapse
Affiliation(s)
- Mangesh A. Thorat
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
- Breast Services, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Pauline M. Levey
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - J. Louise Jones
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sarah E. Pinder
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
- Department of Pathology, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Nigel J. Bundred
- Manchester University NHS Foundation Trust, Wythenshawe, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ian S. Fentiman
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Jack Cuzick
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
17
|
Freeman DW, Rodrigues Sousa E, Karkampouna S, Zoni E, Gray PC, Salomon DS, Kruithof-de Julio M, Spike BT. Whence CRIPTO: The Reemergence of an Oncofetal Factor in 'Wounds' That Fail to Heal. Int J Mol Sci 2021; 22:10164. [PMID: 34576327 PMCID: PMC8472190 DOI: 10.3390/ijms221810164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023] Open
Abstract
There exists a set of factors termed oncofetal proteins that play key roles in ontogeny before they decline or disappear as the organism's tissues achieve homeostasis, only to then re-emerge in cancer. Although the unique therapeutic potential presented by such factors has been recognized for more than a century, their clinical utility has yet to be fully realized1. This review highlights the small signaling protein CRIPTO encoded by the tumor derived growth factor 1 (TDGF1/Tdgf1) gene, an oft cited oncofetal protein whose presence in the cancer literature as a tumor promoter, diagnostic marker and viable therapeutic target continues to grow. We touch lightly on features well established and well-reviewed since its discovery more than 30 years ago, including CRIPTO's early developmental roles and modulation of SMAD2/3 activation by a selected set of transforming growth factor β (TGF-β) family ligands. We predominantly focus instead on more recent and less well understood additions to the CRIPTO signaling repertoire, on its potential upstream regulators and on new conceptual ground for understanding its mode of action in the multicellular and often stressful contexts of neoplastic transformation and progression. We ask whence it re-emerges in cancer and where it 'hides' between the time of its fetal activity and its oncogenic reemergence. In this regard, we examine CRIPTO's restriction to rare cells in the adult, its potential for paracrine crosstalk, and its emerging role in inflammation and tissue regeneration-roles it may reprise in tumorigenesis, acting on subsets of tumor cells to foster cancer initiation and progression. We also consider critical gaps in knowledge and resources that stand between the recent, exciting momentum in the CRIPTO field and highly actionable CRIPTO manipulation for cancer therapy and beyond.
Collapse
Affiliation(s)
- David W. Freeman
- Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| | - Elisa Rodrigues Sousa
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
| | - Sofia Karkampouna
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
| | - Eugenio Zoni
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
| | - Peter C. Gray
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA;
| | - David S. Salomon
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 20893, USA;
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
- Translational Organoid Models, Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, 3010 Bern, Switzerland
- Department of Urology, Inselspital, University Hospital of Bern, 3010 Bern, Switzerland
| | - Benjamin T. Spike
- Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| |
Collapse
|
18
|
Cobleigh MA, Anderson SJ, Siziopikou KP, Arthur DW, Rabinovitch R, Julian TB, Parda DS, Seaward SA, Carter DL, Lyons JA, Dillmon MS, Magrinat GC, Kavadi VS, Zibelli AM, Tiriveedhi L, Hill ML, Melnik MK, Beriwal S, Mamounas EP, Wolmark N. Comparison of Radiation With or Without Concurrent Trastuzumab for HER2-Positive Ductal Carcinoma In Situ Resected by Lumpectomy: A Phase III Clinical Trial. J Clin Oncol 2021; 39:2367-2374. [PMID: 33739848 PMCID: PMC8462554 DOI: 10.1200/jco.20.02824] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/09/2020] [Accepted: 01/07/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Preclinical studies report that trastuzumab (T) can boost radiotherapy (RT) effectiveness. The primary aim of the B-43 trial was to assess the efficacy of RT alone vs concurrent RT plus T in preventing recurrence of ipsilateral breast cancer (IBTR) in women with ductal carcinoma in situ (DCIS). PATIENTS AND METHODS Eligibility: Eastern Cooperative Oncology Group (ECOG) performance status 0 or 1, DCIS resected by lumpectomy, known estrogen receptor (ER) and/or progesterone receptor (PgR), and human epidermal growth factor receptor 2 (HER2) status by centralized testing. Whole-breast RT was given concurrently with T. Stratification was by menopausal status, adjuvant endocrine therapy plan, and nuclear grade. Definitive intent-to-treat primary analysis was to be conducted when either 163 IBTR events occurred or all accrued patients were on study ≥ 5 years. RESULTS There were 2,014 participants who were randomly assigned. Median follow-up time as of December 31, 2019, was 79.2 months. At primary definitive analysis, 114 IBTR events occurred: RT arm, 63 and RT plus T arm, 51 (hazard ratio [HR], 0.81; 95% CI, 0.56 to 1.17; P value = .26). There were 34 who were invasive: RT arm, 18 and RT plus T arm, 20 (HR, 1.11; 95% CI, 0.59 to 2.10; P value = .71). Seventy-six were DCIS: RT arm, 45 and RT plus T arm, 31 (HR, 0.68; 95% CI, 0.43 to 1.08; P value = .11). Annual IBTR event rates were: RT arm, 0.99%/y and RT plus T arm, 0.79%/y. The study did not reach the 163 protocol-specified events, so the definitive analysis was triggered by all patients having been on study for ≥ 5 years. CONCLUSION Addition of T to RT did not achieve the objective of 36% reduction in IBTR rate but did achieve a modest but statistically nonsignificant reduction of 19%. Nonetheless, this trial had negative results. Further exploration of RT plus T is needed in HER2-positive DCIS before its routine delivery in patients with DCIS resected by lumpectomy.
Collapse
Affiliation(s)
| | | | - Kalliopi P. Siziopikou
- NRG Oncology, Pittsburgh, PA
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Douglas W. Arthur
- NRG Oncology, Pittsburgh, PA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| | - Rachel Rabinovitch
- NRG Oncology, Pittsburgh, PA
- University of Colorado Cancer Center, Aurora, CO
| | - Thomas B. Julian
- NRG Oncology, Pittsburgh, PA
- Allegheny Health Network, Pittsburgh, PA
| | - David S. Parda
- NRG Oncology, Pittsburgh, PA
- Allegheny Health Network, Pittsburgh, PA
| | - Samantha A. Seaward
- NRG Oncology, Pittsburgh, PA
- Kaiser Permanente Cancer Research Program, Vallejo, CA
| | - Dennis L. Carter
- Rocky Mountain Cancer Centers, Aurora, CO
- US Oncology, The Woodlands, TX
| | | | | | | | | | - Allison M. Zibelli
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Lavanya Tiriveedhi
- NRG Oncology, Pittsburgh, PA
- Mercy Clinic Cancer and Hematology, Springfield, MO
| | - Matthew L. Hill
- NRG Oncology, Pittsburgh, PA
- Mission Cancer and Blood, Des Moines, IA
| | - Marianne K. Melnik
- NRG Oncology, Pittsburgh, PA
- Cancer Research Consortium of West Michigan, Grand Rapids, MI
| | - Sushil Beriwal
- NRG Oncology, Pittsburgh, PA
- UPMC Hillman Cancer Center, Magee Womens Hospital, Pittsburgh, PA
| | | | - Norman Wolmark
- NRG Oncology, Pittsburgh, PA
- University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
19
|
Houghton SC, Hankinson SE. Cancer Progress and Priorities: Breast Cancer. Cancer Epidemiol Biomarkers Prev 2021; 30:822-844. [PMID: 33947744 PMCID: PMC8104131 DOI: 10.1158/1055-9965.epi-20-1193] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 02/19/2021] [Indexed: 12/24/2022] Open
Affiliation(s)
- Serena C Houghton
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, Massachusetts.
| | - Susan E Hankinson
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, Massachusetts
| |
Collapse
|
20
|
van Seijen M, Jóźwiak K, Pinder SE, Hall A, Krishnamurthy S, Thomas JSJ, Collins LC, Bijron J, Bart J, Cohen D, Ng W, Bouybayoune I, Stobart H, Hudecek J, Schaapveld M, Thompson A, Lips EH, Wesseling J, The Grand Challenge PRECISION consortium.. Variability in grading of ductal carcinoma in situ among an international group of pathologists. J Pathol Clin Res 2021; 7:233-242. [PMID: 33620141 PMCID: PMC8073001 DOI: 10.1002/cjp2.201] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/11/2020] [Accepted: 01/08/2021] [Indexed: 01/04/2023]
Abstract
The prognostic value of cytonuclear grade in ductal carcinoma in situ (DCIS) is debated, partly due to high interobserver variability and the use of multiple guidelines. The aim of this study was to evaluate interobserver agreement in grading DCIS between Dutch, British, and American pathologists. Haematoxylin and eosin-stained slides of 425 women with primary DCIS were independently reviewed by nine breast pathologists based in the Netherlands, the UK, and the USA. Chance-corrected kappa (κma ) for association between pathologists was calculated based on a generalised linear mixed model using the ordinal package in R. Overall κma for grade of DCIS (low, intermediate, or high) was estimated to be 0.50 (95% confidence interval [CI] 0.44-0.56), indicating a moderate association between pathologists. When the model was adjusted for national guidelines, the association for grade did not change (κma = 0.53; 95% CI 0.48-0.57); subgroup analysis for pathologists using the UK pathology guidelines only had significantly higher association (κma = 0.58; 95% CI 0.56-0.61). To assess if concordance of grading relates to the expression of the oestrogen receptor (ER) and HER2, archived immunohistochemistry was analysed on a subgroup (n = 106). This showed that non-high grade according to the majority opinion was associated with ER positivity and HER2 negativity (100 and 89% of non-high grade cases, respectively). In conclusion, DCIS grade showed only moderate association using whole slide images scored by nine breast pathologists. As therapeutic decisions and inclusion in ongoing clinical trials are guided by DCIS grade, there is a pressing need to reduce interobserver variability in grading. ER and HER2 might be supportive to prevent the accidental and unwanted inclusion of high-grade DCIS in such trials.
Collapse
Affiliation(s)
- Maartje van Seijen
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Katarzyna Jóźwiak
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Institute of Biostatistics and Registry ResearchBrandenburg Medical School Theodor FontaneNeuruppinGermany
| | - Sarah E Pinder
- Comprehensive Cancer Centre at Guy's Hospital, School of Cancer & Pharmaceutical SciencesKings College LondonLondonUK
- Department of Cellular PathologyGuy's and St Thomas' NHS Foundation Trust LondonLondonUK
| | - Allison Hall
- Department of PathologyDuke University Medical CenterDurhamNCUSA
| | - Savitri Krishnamurthy
- Department of PathologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | | | - Laura C Collins
- Department of PathologyBeth Israel Deaconess Medical Center and Harvard Medical SchoolBostonMAUSA
| | - Jonathan Bijron
- Department of PathologyMartini HospitalGroningenThe Netherlands
| | - Joost Bart
- Department of PathologyIsala HospitalZwolleThe Netherlands
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Danielle Cohen
- Department of PathologyLeiden University Medical CenterLeidenThe Netherlands
| | - Wen Ng
- Department of Cellular PathologyGuy's and St Thomas' NHS Foundation Trust LondonLondonUK
| | - Ihssane Bouybayoune
- Comprehensive Cancer Centre at Guy's Hospital, School of Cancer & Pharmaceutical SciencesKings College LondonLondonUK
| | | | - Jan Hudecek
- Department of Research ITThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Michael Schaapveld
- Department of Psychosocial Research and EpidemiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Alastair Thompson
- Dan L Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Esther H Lips
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jelle Wesseling
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of PathologyIsala HospitalZwolleThe Netherlands
| | | |
Collapse
|
21
|
Rakovitch E, Bonefas E, Nofech-Mozes S, Thompson AM. Ductal carcinoma in situ (DCIS)—precision medicine for de-escalation. CURRENT BREAST CANCER REPORTS 2021. [DOI: 10.1007/s12609-021-00407-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Almekinders MMM, Schaapveld M, Thijssen B, Visser LL, Bismeijer T, Sanders J, Isnaldi E, Hofland I, Mertz M, Wessels LFA, Broeks A, Hooijberg E, Zwart W, Lips EH, Desmedt C, Wesseling J. Breast adipocyte size associates with ipsilateral invasive breast cancer risk after ductal carcinoma in situ. NPJ Breast Cancer 2021; 7:31. [PMID: 33753731 PMCID: PMC7985299 DOI: 10.1038/s41523-021-00232-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/03/2021] [Indexed: 12/25/2022] Open
Abstract
Although ductal carcinoma in situ (DCIS) is a non-obligate precursor to ipsilateral invasive breast cancer (iIBC), most DCIS lesions remain indolent. Hence, overdiagnosis and overtreatment of DCIS is a major concern. There is an urgent need for prognostic markers that can distinguish harmless from potentially hazardous DCIS. We hypothesised that features of the breast adipose tissue may be associated with risk of subsequent iIBC. We performed a case-control study nested in a population-based DCIS cohort, consisting of 2658 women diagnosed with primary DCIS between 1989 and 2005, uniformly treated with breast conserving surgery (BCS) alone. We assessed breast adipose features with digital pathology (HALO®, Indica Labs) and related these to iIBC risk in 108 women that developed subsequent iIBC (cases) and 168 women who did not (controls) by conditional logistic regression, accounting for clinicopathological and immunohistochemistry variables. Large breast adipocyte size was significantly associated with iIBC risk (odds ratio (OR) 2.75, 95% confidence interval (95% CI) = 1.25-6.05). High cyclooxygenase (COX)-2 protein expression in the DCIS cells was also associated with subsequent iIBC (OR 3.70 (95% CI = 1.59-8.64). DCIS with both high COX-2 expression and large breast adipocytes was associated with a 12-fold higher risk (OR 12.0, 95% CI = 3.10-46.3, P < 0.001) for subsequent iIBC compared with women with smaller adipocyte size and low COX-2 expression. Large breast adipocytes combined with high COX-2 expression in DCIS is associated with a high risk of subsequent iIBC. Besides COX-2, adipocyte size has the potential to improve clinical management in patients diagnosed with primary DCIS.
Collapse
Affiliation(s)
- Mathilde M M Almekinders
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Michael Schaapveld
- Division of Psychosocial Research, Epidemiology and Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bram Thijssen
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lindy L Visser
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tycho Bismeijer
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joyce Sanders
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Edoardo Isnaldi
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Internal Medicine and Medical Specialties, Università degli Studi di Genova, IT-16132, Genova, Italy
| | - Ingrid Hofland
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marjolijn Mertz
- Bio-Imaging Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Erik Hooijberg
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Oncode Institute, Utrecht, The Netherlands
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Pathology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
23
|
Hewitt K, Son J, Glencer A, Borowsky AD, Cooperberg MR, Esserman LJ. The Evolution of Our Understanding of the Biology of Cancer Is the Key to Avoiding Overdiagnosis and Overtreatment. Cancer Epidemiol Biomarkers Prev 2020; 29:2463-2474. [PMID: 33033145 DOI: 10.1158/1055-9965.epi-20-0110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/06/2020] [Accepted: 10/01/2020] [Indexed: 11/16/2022] Open
Abstract
There has been a tremendous evolution in our thinking about cancer since the 1880s. Breast cancer is a particularly good example to evaluate the progress that has been made and the new challenges that have arisen due to screening that inadvertently identifies indolent lesions. The degree to which overdiagnosis is a problem depends on the reservoir of indolent disease, the disease heterogeneity, and the fraction of the tumors that have aggressive biology. Cancers span the spectrum of biological behavior, and population-wide screening increases the detection of tumors that may not cause harm within the patient's lifetime or may never metastasize or result in death. Our approach to early detection will be vastly improved if we understand, address, and adjust to tumor heterogeneity. In this article, we use breast cancer as a case study to demonstrate how the approach to biological characterization, diagnostics, and therapeutics can inform our approach to screening, early detection, and prevention. Overdiagnosis can be mitigated by developing diagnostics to identify indolent disease, incorporating biology and risk assessment in screening strategies, changing the pathology rules for tumor classification, and refining the way we classify precancerous lesions. The more the patterns of cancers can be seen across other cancers, the more it is clear that our approach should transcend organ of origin. This will be particularly helpful in advancing the field by changing both our terminology for what is cancer and also by helping us to learn how best to mitigate the risk of the most aggressive cancers.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Kelly Hewitt
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Jennifer Son
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Alexa Glencer
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Alexander D Borowsky
- Department of Pathology, University of California, Davis, Davis, California.,Athena Breast Health Network
| | - Matthew R Cooperberg
- Department of Urology, University of California, San Francisco, San Francisco, California.,Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, California
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, California. .,Athena Breast Health Network
| |
Collapse
|
24
|
Groen EJ, Hudecek J, Mulder L, van Seijen M, Almekinders MM, Alexov S, Kovács A, Ryska A, Varga Z, Andreu Navarro FJ, Bianchi S, Vreuls W, Balslev E, Boot MV, Kulka J, Chmielik E, Barbé E, de Rooij MJ, Vos W, Farkas A, Leeuwis-Fedorovich NE, Regitnig P, Westenend PJ, Kooreman LFS, Quinn C, Floris G, Cserni G, van Diest PJ, Lips EH, Schaapveld M, Wesseling J, Grand Challenge PRECISION consortium. Prognostic value of histopathological DCIS features in a large-scale international interrater reliability study. Breast Cancer Res Treat 2020; 183:759-770. [PMID: 32734520 PMCID: PMC7497690 DOI: 10.1007/s10549-020-05816-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE For optimal management of ductal carcinoma in situ (DCIS), reproducible histopathological assessment is essential to distinguish low-risk from high-risk DCIS. Therefore, we analyzed interrater reliability of histopathological DCIS features and assessed their associations with subsequent ipsilateral invasive breast cancer (iIBC) risk. METHODS Using a case-cohort design, reliability was assessed in a population-based, nationwide cohort of 2767 women with screen-detected DCIS diagnosed between 1993 and 2004, treated by breast-conserving surgery with/without radiotherapy (BCS ± RT) using Krippendorff's alpha (KA) and Gwet's AC2 (GAC2). Thirty-eight raters scored histopathological DCIS features including grade (2-tiered and 3-tiered), growth pattern, mitotic activity, periductal fibrosis, and lymphocytic infiltrate in 342 women. Using majority opinion-based scores for each feature, their association with subsequent iIBC risk was assessed using Cox regression. RESULTS Interrater reliability of grade using various classifications was fair to moderate, and only substantial for grade 1 versus 2 + 3 when using GAC2 (0.78). Reliability for growth pattern (KA 0.44, GAC2 0.78), calcifications (KA 0.49, GAC2 0.70) and necrosis (KA 0.47, GAC2 0.70) was moderate using KA and substantial using GAC2; for (type of) periductal fibrosis and lymphocytic infiltrate fair to moderate estimates were found and for mitotic activity reliability was substantial using GAC2 (0.70). Only in patients treated with BCS-RT, high mitotic activity was associated with a higher iIBC risk in univariable analysis (Hazard Ratio (HR) 2.53, 95% Confidence Interval (95% CI) 1.05-6.11); grade 3 versus 1 + 2 (HR 2.64, 95% CI 1.35-5.14) and a cribriform/solid versus flat epithelial atypia/clinging/(micro)papillary growth pattern (HR 3.70, 95% CI 1.34-10.23) were independently associated with a higher iIBC risk. CONCLUSIONS Using majority opinion-based scores, DCIS grade, growth pattern, and mitotic activity are associated with iIBC risk in patients treated with BCS-RT, but interrater variability is substantial. Semi-quantitative grading, incorporating and separately evaluating nuclear pleomorphism, growth pattern, and mitotic activity, may improve the reliability and prognostic value of these features.
Collapse
Affiliation(s)
- Emma J. Groen
- Department of Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jan Hudecek
- Department of Research IT, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Lennart Mulder
- Department of Molecular Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Maartje van Seijen
- Department of Molecular Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Mathilde M. Almekinders
- Department of Molecular Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Stoyan Alexov
- Department of Pathology, Oncology Hospital, Sofia, Bulgaria
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ales Ryska
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Zsuzsanna Varga
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | | | - Simonetta Bianchi
- Division of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Willem Vreuls
- Department of Pathology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Eva Balslev
- Department of Pathology, Herlev University Hospital, Herlev, Denmark
| | - Max V. Boot
- Department of Pathology, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Ewa Chmielik
- Tumor Pathology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Ellis Barbé
- Department of Pathology, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
| | | | - Winand Vos
- Department of Pathology, Zuyderland Medical Center, Location Sittard-Geleen, Sittard-Geleen, The Netherlands
| | - Andrea Farkas
- Department of Pathology, Gävle Hospital, Gävle, Sweden
| | | | - Peter Regitnig
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Loes F. S. Kooreman
- Department of Pathology and GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Cecily Quinn
- Department of Pathology and Laboratory Medicine, St. Vincent’s University Hospital, Dublin, Ireland
| | - Giuseppe Floris
- Laboratory of Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Gábor Cserni
- Department of Pathology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Paul J. van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Esther H. Lips
- Department of Molecular Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Michael Schaapveld
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Department of Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department of Molecular Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Grand Challenge PRECISION consortium
- Department of Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department of Research IT, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Molecular Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Pathology, Oncology Hospital, Sofia, Bulgaria
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Atryshealth Co, S.L., Barcelona, Spain
- Division of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
- Department of Pathology, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
- Department of Pathology, Herlev University Hospital, Herlev, Denmark
- Department of Pathology, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
- Tumor Pathology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
- Symbiant Pathology Expert Centre, Location ZMC, Zaandam, The Netherlands
- Department of Pathology, Zuyderland Medical Center, Location Sittard-Geleen, Sittard-Geleen, The Netherlands
- Department of Pathology, Gävle Hospital, Gävle, Sweden
- Department of Pathology, Deventer Hospital, Deventer, The Netherlands
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- Laboratory for Pathology Dordrecht, Dordrecht, The Netherlands
- Department of Pathology and GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Pathology and Laboratory Medicine, St. Vincent’s University Hospital, Dublin, Ireland
- Laboratory of Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven - University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Department of Pathology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
- Department of Pathology, University of Szeged, Szeged, Hungary
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute – Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Chuang LY, Yang CS, Yang HS, Yang CH. Identification of High-Order Single-Nucleotide Polymorphism Barcodes in Breast Cancer Using a Hybrid Taguchi-Genetic Algorithm: Case-Control Study. JMIR Med Inform 2020; 8:e16886. [PMID: 32554381 PMCID: PMC7351259 DOI: 10.2196/16886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/09/2020] [Accepted: 04/08/2020] [Indexed: 12/24/2022] Open
Abstract
Background Breast cancer has a major disease burden in the female population, and it is a highly genome-associated human disease. However, in genetic studies of complex diseases, modern geneticists face challenges in detecting interactions among loci. Objective This study aimed to investigate whether variations of single-nucleotide polymorphisms (SNPs) are associated with histopathological tumor characteristics in breast cancer patients. Methods A hybrid Taguchi-genetic algorithm (HTGA) was proposed to identify the high-order SNP barcodes in a breast cancer case-control study. A Taguchi method was used to enhance a genetic algorithm (GA) for identifying high-order SNP barcodes. The Taguchi method was integrated into the GA after the crossover operations in order to optimize the generated offspring systematically for enhancing the GA search ability. Results The proposed HTGA effectively converged to a promising region within the problem space and provided excellent SNP barcode identification. Regression analysis was used to validate the association between breast cancer and the identified high-order SNP barcodes. The maximum OR was less than 1 (range 0.870-0.755) for two- to seven-order SNP barcodes. Conclusions We systematically evaluated the interaction effects of 26 SNPs within growth factor–related genes for breast carcinogenesis pathways. The HTGA could successfully identify relevant high-order SNP barcodes by evaluating the differences between cases and controls. The validation results showed that the HTGA can provide better fitness values as compared with other methods for the identification of high-order SNP barcodes using breast cancer case-control data sets.
Collapse
Affiliation(s)
| | - Cheng-San Yang
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Huai-Shuo Yang
- Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Cheng-Hong Yang
- Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Discordant Marker Expression Between Invasive Breast Carcinoma and Corresponding Synchronous and Preceding DCIS. Am J Surg Pathol 2020; 43:1574-1582. [PMID: 31206365 DOI: 10.1097/pas.0000000000001306] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ductal carcinoma in situ (DCIS) is considered a potential precursor of invasive breast carcinoma (IBC). Studies aiming to find markers involved in DCIS progression generally have compared characteristics of IBC lesions with those of adjacent synchronous DCIS lesions. The question remains whether synchronous DCIS and IBC comparisons are a good surrogate for primary DCIS and subsequent IBC. In this study, we compared both primary DCIS and synchronous DCIS with the associated IBC lesion, on the basis of immunohistochemical marker expression. Immunohistochemical analysis of ER, PR, HER2, p53, and cyclo-oxygenase 2 (COX-2) was performed for 143 primary DCIS and subsequent IBC lesions, including 81 IBC lesions with synchronous DCIS. Agreement between DCIS and IBC was assessed using kappa, and symmetry tests were performed to assess the pattern in marker conversion. The primary DCIS and subsequent IBC more often showed discordant marker expression than synchronous DCIS and IBC. Strikingly, 18 of 49 (36%) women with HER2-positive primary DCIS developed an HER2-negative IBC. Such a difference in HER2 expression was not observed when comparing synchronous DCIS and IBC. The frequency of discordant marker expression did not increase with longer time between primary DCIS and IBC. In conclusion, comparison of primary DCIS and subsequent IBC yields different results than a comparison of synchronous DCIS and IBC, in particular with regard to HER2 status. To gain more insight into the progression of DCIS to IBC, it is essential to focus on the relationship between primary DCIS and subsequent IBC, rather than comparing IBC with synchronous DCIS.
Collapse
|
27
|
Mannu GS, Groen EJ, Wang Z, Schaapveld M, Lips EH, Chung M, Joore I, van Leeuwen FE, Teertstra HJ, Winter-Warnars GAO, Darby SC, Wesseling J. Reliability of preoperative breast biopsies showing ductal carcinoma in situ and implications for non-operative treatment: a cohort study. Breast Cancer Res Treat 2019; 178:409-418. [PMID: 31388937 PMCID: PMC6797705 DOI: 10.1007/s10549-019-05362-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/15/2019] [Indexed: 12/26/2022]
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.
Collapse
Affiliation(s)
- Gurdeep S. Mannu
- Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF UK
| | - Emma J. Groen
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Zhe Wang
- Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF UK
| | - Michael Schaapveld
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esther H. Lips
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Monica Chung
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ires Joore
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Flora E. van Leeuwen
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hendrik J. Teertstra
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Sarah C. Darby
- Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF UK
| | - Jelle Wesseling
- Antoni van Leeuwenhoek – Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Toss MS, Miligy IM, Gorringe KL, Aleskandarany MA, Alkawaz A, Mittal K, Aneja R, Ellis IO, Green AR, Rakha EA. Collagen (XI) alpha-1 chain is an independent prognostic factor in breast ductal carcinoma in situ. Mod Pathol 2019; 32:1460-1472. [PMID: 31175327 DOI: 10.1038/s41379-019-0286-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/31/2019] [Accepted: 03/31/2019] [Indexed: 12/21/2022]
Abstract
Collagen11A1 (COL11A1) is a fibrillary type collagen constituting a minor component of the extracellular matrix and plays role in tissue tensile strength. Overexpression of COL11A1 expression is associated with aggressive behavior and poor outcome in several human malignancies. In this study, we evaluated the association between COL11A1 expression and clinicopathological parameters of the breast ductal carcinoma in situ (DCIS) and its prognostic value. COL11A1 protein expression was assessed immunohistochemically in a large well-characterized cohort of DCIS including pure (n = 776) and DCIS associated with invasive carcinoma (DCIS-mixed, n = 239). COL11A1 expression was assessed in tumor cells and surrounding stromal cells, and correlated with clinicopathological parameters, immunoprofile and disease outcome. In pure DCIS, high COL11A1 expression was observed in tumor cells and surrounding stromal cells in 25 and 13% of cases, respectively. Higher COL11A1 expression within the stromal cells was associated with hormone receptor negative, HER2 enriched and triple negative molecular subtypes and showed a positive linear correlation with proliferation index, dense tumor infiltrating lymphocytes and hypoxia-inducible factor 1 alpha. COL11A1 expression in tumor and stromal cells was significantly higher in DCIS associated with invasive carcinoma than in pure DCIS, and within the DCIS-mixed cohort, the invasive component showed higher COL11A1 expression than the DCIS component (all, p < 0.0001). Overexpression of stromal COL11A1 was an independent predictor of shorter local recurrence-free interval for all recurrences (HR = 13.2, 95% CI = 6.9-25.4, p < 0.0001) and for invasive recurrences (HR = 11.2, 95% CI = 4.9-25.8, p < 0.0001). When incorporated with other risk factors, stromal COL11A1 provided better patient risk stratification. DCIS with higher stromal COL11A1 expression showed poor outcome even with adjuvant radiotherapy management. In conclusion, overexpression of stromal COL11A1 is associated with invasive recurrence in DCIS and is a potential marker to predict the response to radiotherapy.
Collapse
Affiliation(s)
- Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK.,Histopathology department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Islam M Miligy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK.,Histopathology department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Mohammed A Aleskandarany
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK.,Histopathology department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Abdulbaqi Alkawaz
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | | | - Ritu Aneja
- Georgia State University, Atlanta, GA, USA
| | - Ian O Ellis
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK. .,Histopathology department, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| |
Collapse
|
29
|
Estrogen Receptor-positive Ductal Carcinoma In Situ Frequently Overexpresses HER2 Protein Without Gene Amplification. Am J Surg Pathol 2019; 43:1221-1228. [DOI: 10.1097/pas.0000000000001300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
30
|
Liu R, Yu X, Chen X, Zhong H, Liang C, Xu X, Xu W, Cheng Y, Wang W, Yu L, Wu Y, Yan N, Hu X. Individual factors define the overall effects of dietary genistein exposure on breast cancer patients. Nutr Res 2019; 67:1-16. [DOI: 10.1016/j.nutres.2019.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/03/2019] [Accepted: 03/25/2019] [Indexed: 12/18/2022]
|
31
|
Van Bockstal MR, Agahozo MC, Koppert LB, van Deurzen CHM. A retrospective alternative for active surveillance trials for ductal carcinoma in situ of the breast. Int J Cancer 2019; 146:1189-1197. [PMID: 31018242 PMCID: PMC7004157 DOI: 10.1002/ijc.32362] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/25/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022]
Abstract
Ductal carcinoma in situ (DCIS) of the breast is a nonobligate precursor of invasive breast cancer, accounting for 20 % of screen-detected breast cancers. Little is known about the natural progression of DCIS because most patients undergo surgery upon diagnosis. Many DCIS patients are likely being overtreated, as it is believed that only around 50 % of DCIS will progress to invasive carcinoma. Robust prognostic markers for progression to invasive carcinoma are lacking. In the past, studies have investigated women who developed a recurrence after breast-conserving surgery (BCS) and compared them with those who did not. However, where there is no recurrence, the patient has probably been adequately treated. The present narrative review advocates a new research strategy, wherein only those patients with a recurrence are studied. Approximately half of the recurrences are invasive cancers, and half are DCIS. So-called "recurrences" are probably most often the result of residual disease. The new approach allows us to ask: why did some residual DCIS evolve to invasive cancers and others not? This novel strategy compares the group of patients that developed in situ recurrence with the group of patients that developed invasive recurrence after BCS. The differences between these groups could then be used to develop a robust risk stratification tool. This tool should estimate the risk of synchronous and metachronous invasive carcinoma when DCIS is diagnosed in a biopsy. Identification of DCIS patients at low risk for developing invasive carcinoma will individualize future therapy and prevent overtreatment.
Collapse
Affiliation(s)
- Mieke R Van Bockstal
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marie C Agahozo
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Linetta B Koppert
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | |
Collapse
|
32
|
Choi SH, Choi JS, Han BK, Ko EY, Ko ES, Park KW. Long-term Surveillance of Ductal Carcinoma in Situ Detected with Screening Mammography versus US: Factors Associated with Second Breast Cancer. Radiology 2019; 292:37-48. [PMID: 31038406 DOI: 10.1148/radiol.2019181844] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background The relationship between method of breast cancer screening (mammography or US) and survival outcome in patients with screening-detected ductal carcinoma in situ (DCIS) has not been determined. Purpose To investigate whether different methods of breast cancer screening are associated with different survival outcomes in patients with screening-detected DCIS and to evaluate clinical-pathologic and imaging factors associated with second breast cancer. Materials and Methods We retrospectively identified women who underwent surgery to treat DCIS initially detected with screening mammography or US between July 2004 and December 2011 in a single institution. Overall survival (OS) and disease-free survival (DFS) were assessed. Factors associated with second breast cancer (invasive carcinoma or DCIS) were found with multivariable Cox proportional hazards regression analysis. Subgroups were analyzed according to screening method. Results A total of 814 women (median age, 47 years; age range, 25-81 years) were included; 627 underwent treatment for screening mammography-detected DCIS (mammography-detected group), and 187 underwent treatment for screening US-detected DCIS (US-detected group). During follow-up (median, 7 years; interquartile range, 5-8 years), 26 ipsilateral and 26 contralateral second breast cancers (6.4%, 52 of 814) were found, with 44 in the mammography-detected group and eight in the US-detected group. The overall 5-year OS and DFS rates were 100% and 95.3%, respectively. DFS rates did not differ according to screening method (P = .21, 5-year DFS rates were 94.9% in the mammography-detected group and 96.5% in the US-detected group). In the mammography-detected group, higher nuclear grade (intermediate grade: hazard ratio [HR], 5.7; 95% confidence interval [CI]: 1.3, 24.3; P = 0.02) (high grade: HR, 8.0; 95% CI: 1.9, 34.2; P = .01) and dense breast (HR, 3.5; 95% CI: 1.1, 11.4; P = 0.04) were associated with second breast cancer. In the US-detected group, human epidermal growth factor receptor 2 positivity was associated with second breast cancer (HR, 9.2; 95% CI: 2.2, 38.5; P = .002). Conclusion Disease-free survival of patients who underwent treatment for screening-detected ductal carcinoma in situ (DCIS) did not differ according to screening detection method. In patients with screening mammography-detected DCIS, higher nuclear grade and dense breast were associated with second breast cancer, and in patients with screening US-detected DCIS, human epidermal growth factor 2 positivity was associated with second breast cancer. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Fowler in this issue.
Collapse
Affiliation(s)
- Seung Hee Choi
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Ji Soo Choi
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Boo-Kyung Han
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Eun Young Ko
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Eun Sook Ko
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Ko Woon Park
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| |
Collapse
|
33
|
Visser LL, Groen EJ, van Leeuwen FE, Lips EH, Schmidt MK, Wesseling J. Predictors of an Invasive Breast Cancer Recurrence after DCIS: A Systematic Review and Meta-analyses. Cancer Epidemiol Biomarkers Prev 2019; 28:835-845. [PMID: 31023696 DOI: 10.1158/1055-9965.epi-18-0976] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/12/2018] [Accepted: 02/13/2019] [Indexed: 11/16/2022] Open
Abstract
We performed a systematic review with meta-analyses to summarize current knowledge on prognostic factors for invasive disease after a diagnosis of ductal carcinoma in situ (DCIS). Eligible studies assessed risk of invasive recurrence in women primarily diagnosed and treated for DCIS and included at least 10 ipsilateral-invasive breast cancer events and 1 year of follow-up. Quality in Prognosis Studies tool was used for risk of bias assessment. Meta-analyses were performed to estimate the average effect size of the prognostic factors. Of 1,781 articles reviewed, 40 articles met the inclusion criteria. Highest risk of bias was attributable to insufficient handling of confounders and poorly described study groups. Six prognostic factors were statistically significant in the meta-analyses: African-American race [pooled estimate (ES), 1.43; 95% confidence interval (CI), 1.15-1.79], premenopausal status (ES, 1.59; 95% CI, 1.20-2.11), detection by palpation (ES, 1.84; 95% CI, 1.47-2.29), involved margins (ES, 1.63; 95% CI, 1.14-2.32), high histologic grade (ES, 1.36; 95% CI, 1.04-1.77), and high p16 expression (ES, 1.51; 95% CI, 1.04-2.19). Six prognostic factors associated with invasive recurrence were identified, whereas many other factors need confirmation in well-designed studies on large patient numbers. Furthermore, we identified frequently occurring biases in studies on invasive recurrence after DCIS. Avoiding these common methodological pitfalls can improve future study designs.
Collapse
Affiliation(s)
- Lindy L Visser
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Emma J Groen
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Flora E van Leeuwen
- Division of Psychosocial Research and Epidemiology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marjanka K Schmidt
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Psychosocial Research and Epidemiology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands. .,Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
34
|
Han MS, Khan SA. Clinical Trials for Ductal Carcinoma In Situ of the Breast. J Mammary Gland Biol Neoplasia 2018; 23:293-301. [PMID: 30206737 DOI: 10.1007/s10911-018-9413-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/07/2018] [Indexed: 12/26/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) of the breast is a non-obligatory precursor to invasive breast carcinoma, with a variable natural history and biological potential for progression to invasive disease. Over the past 30 years, clinical trials have applied the therapeutic principles used for invasive carcinoma to treat DCIS (surgery, with or without breast radiotherapy, and post-operative endocrine therapy), with excellent survival outcomes, and in-breast recurrence rates that range from 0.5 to 1% annually. However, half of such recurrences are again in-situ lesions, and intensive therapy is likely not necessary for all patients. Current clinical research is focused on a better characterization of the potential of individual lesions to progress to invasive disease, and to identify women who would do well with lesser treatment. Three ongoing trials in the United States and Europe randomize women to active surveillance (with or without endocrine therapy) versus usual treatment with surgery and radiotherapy. The use of pre-operative endocrine therapy has been evaluated in a recently completed trial of letrozole use in postmenopausal women with DCIS; and in on-going trials of tamoxifen, used either orally, or as a 4-hydroxytamoxifen gel formulation for application to the breast skin. This review summaries the major past and current clinical trials of DCIS, and the likely trajectories of DCIS management in the near future.
Collapse
Affiliation(s)
- Michelle S Han
- Department of Surgery, Northwestern University, 250 E. Superior Street, Suite 4-420, Chicago, IL, 60611, USA
| | - Seema A Khan
- Prentice Women's Hospital, Northwestern University, 250 E. Superior Street, Suite 4-420, Chicago, IL, 60611, USA.
| |
Collapse
|