1
|
O'Leary RL, Duijm LEM, Boersma LJ, van der Sangen MJC, de Munck L, Wesseling J, Schipper RJ, Voogd AC. Invasive recurrence after breast conserving treatment of ductal carcinoma in situ of the breast in the Netherlands: time trends and the association with tumour grade. Br J Cancer 2024; 131:852-859. [PMID: 38982194 PMCID: PMC11369187 DOI: 10.1038/s41416-024-02785-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND The first aim of this study was to examine trends in the risk of ipsilateral invasive breast cancer (iIBC) after breast-conserving surgery (BCS) of ductal carcinoma in situ (DCIS). A second aim was to analyse the association between DCIS grade and the risk of iIBC following BCS. PATIENTS AND METHODS In this population-based, retrospective cohort study, the Netherlands Cancer Registry collected information on 25,719 women with DCIS diagnosed in the period 1989-2021 who underwent BCS. Of these 19,034 received adjuvant radiotherapy (RT). Kaplan-Meier analyses and Cox regression models were used. RESULTS A total of 1135 patients experienced iIBC. Ten-year cumulative incidence rates of iIBC for patients diagnosed in the periods 1989-1998, 1999-2008 and 2009-2021 undergoing BCS without RT, were 12.6%, 9.0% and 5.0% (P < 0.001), respectively. For those undergoing BCS with RT these figures were 5.7%, 3.7% and 2.2%, respectively (P < 0.001). In the multivariable analyses, DCIS grade was not associated with the risk of iIBC. CONCLUSION Since 1989 the risk of iIBC has decreased substantially and has become even lower than the risk of invasive contralateral breast cancer. No significant association of DCIS grade with iIBC was found, stressing the need for more powerful prognostic factors to guide the treatment of DCIS.
Collapse
MESH Headings
- Humans
- Female
- Netherlands/epidemiology
- Middle Aged
- Mastectomy, Segmental
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/radiotherapy
- Carcinoma, Intraductal, Noninfiltrating/surgery
- Carcinoma, Intraductal, Noninfiltrating/epidemiology
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Breast Neoplasms/epidemiology
- Breast Neoplasms/radiotherapy
- Breast Neoplasms/therapy
- Retrospective Studies
- Aged
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Grading
- Adult
- Radiotherapy, Adjuvant/statistics & numerical data
- Registries
- Aged, 80 and over
Collapse
Affiliation(s)
- Rebecca L O'Leary
- Department of Epidemiology, Maastricht University, Maastricht, Netherlands
| | - Lucien E M Duijm
- Department of Radiology, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
| | - Liesbeth J Boersma
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Linda de Munck
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pathology, the Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Adri C Voogd
- Department of Epidemiology, Maastricht University, Maastricht, Netherlands.
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, Netherlands.
| |
Collapse
|
2
|
Buchheit JT, Schacht D, Kulkarni SA. Update on Management of Ductal Carcinoma in Situ. Clin Breast Cancer 2024; 24:292-300. [PMID: 38216382 DOI: 10.1016/j.clbc.2023.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/14/2024]
Abstract
Ductal carcinoma in situ (DCIS) represents 18% to 25% of all diagnosed breast cancers, and is a noninvasive, nonobligate precursor lesion to invasive cancer. The diagnosis of DCIS represents a wide range of disease, including lesions with both low and high risk of progression to invasive cancer and recurrence. Over the past decade, research on the topic of DCIS has focused on the possibility of tailoring treatment for patients according to their risk for progression and recurrence, which is based on clinicopathologic, biomolecular and genetic factors. These efforts are ongoing, with recently completed and continuing clinical trials spanning the continuum of cancer care. We conducted a review to identify recent advances on the topic of diagnosis, risk stratification and management of DCIS. While novel imaging techniques have increased the rate of DCIS diagnosis, questions persist regarding the optimal management of lesions that would not be identified with conventional methods. Additionally, among trials investigating the potential for omission of surgery and use of active surveillance, 2 trials have completed accrual and 2 clinical trials are continuing to enroll patients. Identification of novel genetic patterns is expanding our potential for risk stratification and aiding our ability to de-escalate radiation and systemic therapies for DCIS. These advances provide hope for tailoring of DCIS treatment in the near future.
Collapse
Affiliation(s)
- Joanna T Buchheit
- Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - David Schacht
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Swati A Kulkarni
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| |
Collapse
|
3
|
Huang CY, Chang RF, Lin CY, Hsieh MS, Liao PC, Wang YJ, Kao YC, Porta L, Lin PY, Lee CC, Lee YH. Deep-learning model to improve histological grading and predict upstaging of atypical ductal hyperplasia / ductal carcinoma in situ on breast biopsy. Histopathology 2024; 84:983-1002. [PMID: 38288642 DOI: 10.1111/his.15144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 04/04/2024]
Abstract
AIMS Risk stratification of atypical ductal hyperplasia (ADH) and ductal carcinoma in situ (DCIS), diagnosed using breast biopsy, has great clinical significance. Clinical trials are currently exploring the possibility of active surveillance for low-risk lesions, whereas axillary lymph node staging may be considered during surgical planning for high-risk lesions. We aimed to develop a machine-learning algorithm based on whole-slide images of breast biopsy specimens and clinical information to predict the risk of upstaging to invasive breast cancer after wide excision. METHODS AND RESULTS Patients diagnosed with ADH/DCIS on breast biopsy were included in this study, comprising 592 (740 slides) and 141 (198 slides) patients in the development and independent testing cohorts, respectively. Histological grading of the lesions was independently evaluated by two pathologists. Clinical information, including biopsy method, lesion size, and Breast Imaging Reporting and Data System (BI-RADS) classification of ultrasound and mammograms, were collected. Deep DCIS consisted of three deep neural networks to evaluate nuclear grade, necrosis, and stromal reactivity. Deep DCIS output comprised five parameters: total patches, lesion extent, Deep Grade, Deep Necrosis, and Deep Stroma. Deep DCIS highly correlated with the pathologists' evaluations of both slide- and patient-level labels. All five parameters of Deep DCIS were significantly associated with upstaging to invasive carcinoma in subsequent wide excisional specimens. Using multivariate logistic regression, Deep DCIS predicted upstaging to invasive carcinoma with an area under the curve (AUC) of 0.81, outperforming pathologists' evaluation (AUC, 0.71 and 0.69). After including clinical and hormone receptor status information, performance further improved (AUC, 0.87). This combined model retained its predictive power in two subgroup analyses: the first subgroup included unequivocal DCIS (excluding cases of ADH and DCIS suspicious for microinvasion) (AUC, 0.83), while the second excluded cases of high-grade DCIS (AUC, 0.81). The model was validated in an independent testing cohort (AUC, 0.81). CONCLUSION This study demonstrated that deep-learning models can refine histological evaluation of ADH and DCIS on breast biopsies, which may help guide future treatment planning.
Collapse
Affiliation(s)
- Chung-Yen Huang
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ruey-Feng Chang
- Center for Intelligent Healthcare, National Taiwan University Hospital, Taipei, Taiwan
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Chih-Yung Lin
- Center for Intelligent Healthcare, National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Po-Chun Liao
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Jui Wang
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chien Kao
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Lorenzo Porta
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Department of Emergency Medicine, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Pin-Yu Lin
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Chang Lee
- Center for Intelligent Healthcare, National Taiwan University Hospital, Taipei, Taiwan
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
4
|
van Diest PJ, Flach RN, van Dooijeweert C, Makineli S, Breimer GE, Stathonikos N, Pham P, Nguyen TQ, Veta M. Pros and cons of artificial intelligence implementation in diagnostic pathology. Histopathology 2024; 84:924-934. [PMID: 38433288 DOI: 10.1111/his.15153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/19/2024] [Indexed: 03/05/2024]
Abstract
The rapid introduction of digital pathology has greatly facilitated development of artificial intelligence (AI) models in pathology that have shown great promise in assisting morphological diagnostics and quantitation of therapeutic targets. We are now at a tipping point where companies have started to bring algorithms to the market, and questions arise whether the pathology community is ready to implement AI in routine workflow. However, concerns also arise about the use of AI in pathology. This article reviews the pros and cons of introducing AI in diagnostic pathology.
Collapse
Affiliation(s)
- Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rachel N Flach
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Oncological Urology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Seher Makineli
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerben E Breimer
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paul Pham
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Mitko Veta
- Department of Oncological Urology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
5
|
Poelhekken K, Lin Y, Greuter MJW, van der Vegt B, Dorrius M, de Bock GH. The natural history of ductal carcinoma in situ (DCIS) in simulation models: A systematic review. Breast 2023; 71:74-81. [PMID: 37541171 PMCID: PMC10412870 DOI: 10.1016/j.breast.2023.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023] Open
Abstract
OBJECTIVE Assumptions on the natural history of ductal carcinoma in situ (DCIS) are necessary to accurately model it and estimate overdiagnosis. To improve current estimates of overdiagnosis (0-91%), the purpose of this review was to identify and analyse assumptions made in modelling studies on the natural history of DCIS in women. METHODS A systematic review of English full-text articles using PubMed, Embase, and Web of Science was conducted up to February 6, 2023. Eligibility and all assessments were done independently by two reviewers. Risk of bias and quality assessments were performed. Discrepancies were resolved by consensus. Reader agreement was quantified with Cohen's kappa. Data extraction was performed with three forms on study characteristics, model assessment, and tumour progression. RESULTS Thirty models were distinguished. The most important assumptions regarding the natural history of DCIS were addition of non-progressive DCIS of 20-100%, classification of DCIS into three grades, where high grade DCIS had an increased chance of progression to invasive breast cancer (IBC), and regression possibilities of 1-4%, depending on age and grade. Other identified risk factors of progression of DCIS to IBC were younger age, birth cohort, larger tumour size, and individual risk. CONCLUSION To accurately model the natural history of DCIS, aspects to consider are DCIS grades, non-progressive DCIS (9-80%), regression from DCIS to no cancer (below 10%), and use of well-established risk factors for progression probabilities (age). Improved knowledge on key factors to consider when studying DCIS can improve estimates of overdiagnosis and optimization of screening.
Collapse
Affiliation(s)
- Keris Poelhekken
- University of Groningen, University Medical Center Groningen, Groningen, Department of Epidemiology, P.O. Box 30 001, FA40, 9700, RB, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen, Department of Radiology, PO Box 30.001, EB44, 9700, RB, Groningen, the Netherlands.
| | - Yixuan Lin
- University of Groningen, University Medical Center Groningen, Groningen, Department of Epidemiology, P.O. Box 30 001, FA40, 9700, RB, Groningen, the Netherlands
| | - Marcel J W Greuter
- University of Groningen, University Medical Center Groningen, Groningen, Department of Radiology, PO Box 30.001, EB44, 9700, RB, Groningen, the Netherlands
| | - Bert van der Vegt
- University of Groningen, University Medical Center Groningen, Groningen, Department of Pathology and Medical Biology, PO Box 30.001, 9700, RB, Groningen, the Netherlands
| | - Monique Dorrius
- University of Groningen, University Medical Center Groningen, Groningen, Department of Radiology, PO Box 30.001, EB44, 9700, RB, Groningen, the Netherlands
| | - Geertruida H de Bock
- University of Groningen, University Medical Center Groningen, Groningen, Department of Epidemiology, P.O. Box 30 001, FA40, 9700, RB, Groningen, the Netherlands
| |
Collapse
|
6
|
Ploumen RAW, Claassens EL, Kooreman LFS, Keymeulen KBMI, van Kats MACE, Gommers S, Siesling S, van Nijnatten TJA, Smidt ML. Pathologic complete response of ductal carcinoma in situ to neoadjuvant systemic therapy in HER2-positive invasive breast cancer patients: a nationwide analysis. Breast Cancer Res Treat 2023:10.1007/s10549-023-07012-z. [PMID: 37395816 PMCID: PMC10361905 DOI: 10.1007/s10549-023-07012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/12/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) is present in more than half of HER2-positive invasive breast cancer (IBC). Recent studies show that DCIS accompanying HER2-positive IBC can be completely eradicated by neoadjuvant systemic therapy (NST). Our aim was to determine the percentage of pathologic complete response of the DCIS component in a nationwide cohort and to assess associated clinicopathologic variables. Furthermore, the impact on surgical treatment after NST was investigated. METHODS Women diagnosed with HER2-positive IBC, treated with NST and surgery, between 2010 and 2020, were selected from the Netherlands Cancer Registry. Pre-NST biopsy and postoperative pathology reports were obtained from the Dutch Nationwide Pathology Databank and assessed for the presence of DCIS. Clinicopathologic factors associated with DCIS response were assessed using logistic regression analyses. RESULTS A DCIS component was present in the pre-NST biopsy in 1403 (25.1%) of 5598 included patients. Pathologic complete response of the DCIS component was achieved in 730 patients (52.0%). Complete response of DCIS occurred more frequently in case of complete response of IBC (63.4% versus 33.8%, p < 0.001). ER-negative IBC (OR 1.79; 95%CI 1.33-2.42) and more recent years of diagnosis (2014-2016 OR 1.60; 95%CI 1.17-2.19, 2017-2019 OR 1.76; 95%CI 1.34-2.34) were associated with DCIS response. Mastectomy rates were higher in IBC+DCIS compared to IBC (53.6% versus 41.0%, p < 0.001). CONCLUSION Pathologic complete response of DCIS occurred in 52.0% of HER2-positive IBC patients and was associated with ER-negative IBC and more recent years of diagnosis. Future studies should investigate imaging evaluation of DCIS response to improve surgical decision making.
Collapse
Affiliation(s)
- Roxanne A W Ploumen
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands.
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Eva L Claassens
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Loes F S Kooreman
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Pathology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Maartje A C E van Kats
- Department of Medical Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Suzanne Gommers
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sabine Siesling
- Department of Health Technology and Services Research, Technical Medical Centre, University of Twente, Enschede, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Thiemo J A van Nijnatten
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marjolein L Smidt
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
7
|
Flach RN, Stathonikos N, Nguyen TQ, Ter Hoeve ND, van Diest PJ, van Dooijeweert C. CONFIDENT-trial protocol: a pragmatic template for clinical implementation of artificial intelligence assistance in pathology. BMJ Open 2023; 13:e067437. [PMID: 37286323 DOI: 10.1136/bmjopen-2022-067437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
INTRODUCTION Artificial intelligence (AI) has been on the rise in the field of pathology. Despite promising results in retrospective studies, and several CE-IVD certified algorithms on the market, prospective clinical implementation studies of AI have yet to be performed, to the best of our knowledge. In this trial, we will explore the benefits of an AI-assisted pathology workflow, while maintaining diagnostic safety standards. METHODS AND ANALYSIS This is a Standard Protocol Items: Recommendations for Interventional Trials-Artificial Intelligence compliant single-centre, controlled clinical trial, in a fully digital academic pathology laboratory. We will prospectively include prostate cancer patients who undergo prostate needle biopsies (CONFIDENT-P) and breast cancer patients who undergo a sentinel node procedure (CONFIDENT-B) in the University Medical Centre Utrecht. For both the CONFIDENT-B and CONFIDENT-P trials, the specific pathology specimens will be pseudo-randomised to be assessed by a pathologist with or without AI assistance in a pragmatic (bi-)weekly sequential design. In the intervention group, pathologists will assess whole slide images (WSI) of the standard hematoxylin and eosin (H&E)-stained sections assisted by the output of the algorithm. In the control group, pathologists will assess H&E WSI according to the current clinical workflow. If no tumour cells are identified or when the pathologist is in doubt, immunohistochemistry (IHC) staining will be performed. At least 80 patients in the CONFIDENT-P and 180 patients in the CONFIDENT-B trial will need to be enrolled to detect superiority, allocated as 1:1. Primary endpoint for both trials is the number of saved resources of IHC staining procedures for detecting tumour cells, since this will clarify tangible cost savings that will support the business case for AI. ETHICS AND DISSEMINATION The ethics committee (MREC NedMec) waived the need of official ethical approval, since participants are not subjected to procedures nor are they required to follow rules. Results of both trials (CONFIDENT-B and CONFIDENT-P) will be published in scientific peer-reviewed journals.
Collapse
Affiliation(s)
- Rachel N Flach
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
8
|
Kreipe H, Harbeck N, Christgen M. Clinical validity and clinical utility of Ki67 in early breast cancer. Ther Adv Med Oncol 2022; 14:17588359221122725. [PMID: 36105888 PMCID: PMC9465566 DOI: 10.1177/17588359221122725] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Ki67 represents an immunohistochemical nuclear localized marker that is widely
used in surgical pathology. Nuclear immunoreactivity for Ki67 indicates that
cells are cycling and are in G1- to S-phase. The percentage of Ki67-positive
tumor cells (Ki67 index) therefore provides an estimate of the growth fraction
in tumor specimens. In breast cancer (BC), tumor cell proliferation rate is one
of the most relevant prognostic markers and Ki67 is consequently helpful in
prognostication similar to histological grading and mRNA profiling-based BC risk
stratification. In BCs treated with short-term preoperative endocrine therapy,
Ki67 dynamics enable distinguishing between endocrine sensitive and resistant
tumors. Despite its nearly universal use in pathology laboratories worldwide, no
internationally accepted consensus has yet been achieved for some methodological
details related to Ki67 immunohistochemistry (IHC). Controversial issues refer
to choice of IHC antibody clones, scoring methods, inter-laboratory
reproducibility, and the potential value of computer-assisted imaging analysis
and/or artificial intelligence for Ki67 assessment. Prospective clinical trials
focusing on BC treatment have proven that Ki67, as determined by standardized
central pathology assessment, is of clinical validity. Clinical utility has been
demonstrated in huge observational studies.
Collapse
Affiliation(s)
- Hans Kreipe
- Institute of Pathology, Hannover Medical School, Carl-Neubergstraße 1, Hannover 30625, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München (LMU) Frauenklinik Maistrasse-Innenstadt und Klinikum Großhadern, Germany
| | | |
Collapse
|
9
|
Flach RN, Fransen NL, Sonnen AFP, Nguyen TQ, Breimer GE, Veta M, Stathonikos N, van Dooijeweert C, van Diest PJ. Implementation of Artificial Intelligence in Diagnostic Practice as a Next Step after Going Digital: The UMC Utrecht Perspective. Diagnostics (Basel) 2022; 12:diagnostics12051042. [PMID: 35626198 PMCID: PMC9140005 DOI: 10.3390/diagnostics12051042] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 01/31/2023] Open
Abstract
Building on a growing number of pathology labs having a full digital infrastructure for pathology diagnostics, there is a growing interest in implementing artificial intelligence (AI) algorithms for diagnostic purposes. This article provides an overview of the current status of the digital pathology infrastructure at the University Medical Center Utrecht and our roadmap for implementing AI algorithms in the next few years.
Collapse
Affiliation(s)
- Rachel N. Flach
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Nina L. Fransen
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Andreas F. P. Sonnen
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Tri Q. Nguyen
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Gerben E. Breimer
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Mitko Veta
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Carmen van Dooijeweert
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
| | - Paul J. van Diest
- Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (R.N.F.); (N.L.F.); (A.F.P.S.); (T.Q.N.); (G.E.B.); (M.V.); (N.S.); (C.v.D.)
- Correspondence:
| |
Collapse
|
10
|
Kelly C, Fitzpatrick P, Quinn C, Flanagan F, Connors A, Larke A, Mooney T, Kennedy M, Sheehan M, Bennett MW, Brodie C, O'Doherty A. Screen-detected ductal carcinoma in situ, 2008-2020: An observational study. J Med Screen 2022; 29:172-177. [PMID: 35341364 DOI: 10.1177/09691413221090739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The purpose of this study was to evaluate the grade distribution of screen-detected ductal carcinoma in situ (DCIS) diagnosed in Ireland, in the context of the clinical trials currently underway to determine if active surveillance is a feasible management option for low-risk DCIS. SETTING BreastCheck is the national breast screening programme in Ireland, offering screening to women aged 50 to 69 every two years. METHODS This study was a secondary analysis of data collected by BreastCheck on all screen-detected DCIS diagnosed in the 12 years of nationwide screening. Incidence and detection rates were calculated. Descriptive analysis of the cases was performed and, for comparative analysis, grade of DCIS was analysed as a binary variable (high vs. low/intermediate) in keeping with the inclusion criteria for active surveillance trials. Analysis was performed in IBM Statistical Package for Social Sciences, version 26. RESULTS Between 2008 and 2020, 2240 women were diagnosed with DCIS through BreastCheck; 876 (39.1%) were low/intermediate-grade. The overall incidence rate has remained relatively stable during this period. Women with low/intermediate-grade DCIS were younger than women with high-grade DCIS (56 (interquartile range: 56-61) years v 57 (interquartile range: 53-61) years; p < 0.001). They were also more likely to have been diagnosed at an initial screening episode compared with those who had high-grade lesions (42.5% v 29.0%; p < 0.001). CONCLUSION If current clinical trials recommend active surveillance as a feasible option for DCIS, up to 40% of women with screen-detected DCIS may be eligible. These women are younger and often diagnosed on initial screening episode, so may require longer active follow-up.
Collapse
Affiliation(s)
- Caitriona Kelly
- School of Public Health, Physiotherapy and Sports Science, 8797University College Dublin, Dublin, Ireland.,Department of Public Health HSE North East, Navan, Ireland
| | - Patricia Fitzpatrick
- School of Public Health, Physiotherapy and Sports Science, 8797University College Dublin, Dublin, Ireland.,155307National Screening Service, Dublin, Ireland
| | - Cecily Quinn
- BreastCheck, 155307National Screening Service, Dublin, Ireland
| | | | - Alissa Connors
- BreastCheck, 155307National Screening Service, Dublin, Ireland
| | - Aideen Larke
- BreastCheck, 155307National Screening Service, Dublin, Ireland
| | | | - Maria Kennedy
- BreastCheck, 155307National Screening Service, Dublin, Ireland
| | | | - Michael W Bennett
- 155307National Screening Service, Dublin, Ireland.,Department of Pathology, 57983Cork University Hospital, Cork, Ireland
| | - Caroline Brodie
- BreastCheck, 155307National Screening Service, Dublin, Ireland.,Department of Anatomic Pathology, 58040Galway University Hospital and National University of Ireland, Galway, Ireland
| | - Ann O'Doherty
- BreastCheck, 155307National Screening Service, Dublin, Ireland
| |
Collapse
|
11
|
A deep learning model for breast ductal carcinoma in situ classification in whole slide images. Virchows Arch 2022; 480:1009-1022. [PMID: 35076741 DOI: 10.1007/s00428-021-03241-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/12/2021] [Accepted: 11/20/2021] [Indexed: 02/06/2023]
Abstract
The pathological differential diagnosis between breast ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) is of pivotal importance for determining optimum cancer treatment(s) and clinical outcomes. Since conventional diagnosis by pathologists using microscopes is limited in terms of human resources, it is necessary to develop new techniques that can rapidly and accurately diagnose large numbers of histopathological specimens. Computational pathology tools which can assist pathologists in detecting and classifying DCIS and IDC from whole slide images (WSIs) would be of great benefit for routine pathological diagnosis. In this paper, we trained deep learning models capable of classifying biopsy and surgical histopathological WSIs into DCIS, IDC, and benign. We evaluated the models on two independent test sets (n= 1382, n= 548), achieving ROC areas under the curves (AUCs) up to 0.960 and 0.977 for DCIS and IDC, respectively.
Collapse
|
12
|
Preneoplastic Low-Risk Mammary Ductal Lesions (Atypical Ductal Hyperplasia and Ductal Carcinoma In Situ Spectrum): Current Status and Future Directions. Cancers (Basel) 2022; 14:cancers14030507. [PMID: 35158775 PMCID: PMC8833401 DOI: 10.3390/cancers14030507] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Intraepithelial mammary ductal neoplasia is a spectrum of disease that varies from atypical ductal hyperplasia (ADH), low-grade (LG), intermediate-grade (IG), to high-grade (HG) ductal carcinoma in situ (DCIS). While ADH has the lowest prognostic significance, HG-DCIS carries the highest risk. Due to widely used screening mammography, the number of intraepithelial mammary ductal neoplastic lesions has increased. The consequence of this practice is the increase in the number of patients who are overdiagnosed and, therefore, overtreated. The active surveillance (AS) trials are initiated to separate lesions that require active treatment from those that can be safely monitored and only be treated when they develop a change in the clinical/radiologic characteristics. At the same time, the natural history of these lesions can be evaluated. This review aims to evaluate ADH/DCIS as a spectrum of intraductal neoplastic disease (risk and histomorphology); examine the controversies of distinguishing ADH vs. DCIS and the grading of DCIS; review the upgrading for both ADH and DCIS with emphasis on the variation of methods of detection and the definitions of upgrading; and evaluate the impact of all these variables on the AS trials.
Collapse
|
13
|
van Seijen M, Lips EH, Fu L, Giardiello D, van Duijnhoven F, de Munck L, Elshof LE, Thompson A, Sawyer E, Ryser MD, Hwang ES, Schmidt MK, Elkhuizen PHM, Grand Challenge PRECISION Consortium, Wesseling J, Schaapveld M. Long-term risk of subsequent ipsilateral lesions after surgery with or without radiotherapy for ductal carcinoma in situ of the breast. Br J Cancer 2021; 125:1443-1449. [PMID: 34408284 PMCID: PMC8575990 DOI: 10.1038/s41416-021-01496-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/20/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiotherapy (RT) following breast-conserving surgery (BCS) for ductal carcinoma in situ (DCIS) reduces ipsilateral breast event rates in clinical trials. This study assessed the impact of DCIS treatment on a 20-year risk of ipsilateral DCIS (iDCIS) and ipsilateral invasive breast cancer (iIBC) in a population-based cohort. METHODS The cohort comprised all women diagnosed with DCIS in the Netherlands during 1989-2004 with follow-up until 2017. Cumulative incidence of iDCIS and iIBC following BCS and BCS + RT were assessed. Associations of DCIS treatment with iDCIS and iIBC risk were estimated in multivariable Cox models. RESULTS The 20-year cumulative incidence of any ipsilateral breast event was 30.6% (95% confidence interval (CI): 28.9-32.6) after BCS compared to 18.2% (95% CI 16.3-20.3) following BCS + RT. Women treated with BCS compared to BCS + RT had higher risk of developing iDCIS and iIBC within 5 years after DCIS diagnosis (for iDCIS: hazard ratio (HR)age < 50 3.2 (95% CI 1.6-6.6); HRage ≥ 50 3.6 (95% CI 2.6-4.8) and for iIBC: HRage<50 2.1 (95% CI 1.4-3.2); HRage ≥ 50 4.3 (95% CI 3.0-6.0)). After 10 years, the risk of iDCIS and iIBC no longer differed for BCS versus BCS + RT (for iDCIS: HRage < 50 0.7 (95% CI 0.3-1.5); HRage ≥ 50 0.7 (95% CI 0.4-1.3) and for iIBC: HRage < 50 0.6 (95% CI 0.4-0.9); HRage ≥ 50 1.2 (95% CI 0.9-1.6)). CONCLUSION RT is associated with lower iDCIS and iIBC risk up to 10 years after BCS, but this effect wanes thereafter.
Collapse
Affiliation(s)
- Maartje van Seijen
- grid.430814.a0000 0001 0674 1393Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Esther H. Lips
- grid.430814.a0000 0001 0674 1393Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Liping Fu
- grid.430814.a0000 0001 0674 1393Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Daniele Giardiello
- grid.430814.a0000 0001 0674 1393Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Frederieke van Duijnhoven
- grid.430814.a0000 0001 0674 1393Department of Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Linda de Munck
- grid.470266.10000 0004 0501 9982Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Lotte E. Elshof
- grid.414725.10000 0004 0368 8146Department of radiology, Meander Medical Centre, Amersfoort, The Netherlands
| | - Alastair Thompson
- grid.39382.330000 0001 2160 926XDan L Duncan Comprehensive Cancer Centre, Baylor College of Medicine, Houston, TX USA
| | - Elinor Sawyer
- grid.239826.40000 0004 0391 895XDivision of Cancer Studies, King’s College London, Comprehensive Cancer Centre, Guy’s Hospital, London, UK
| | - Marc D. Ryser
- grid.26009.3d0000 0004 1936 7961Department of Population Health Sciences, Duke University, Durham, NC USA ,grid.26009.3d0000 0004 1936 7961Department of Mathematics, Duke University, Durham, NC USA
| | - E. Shelley Hwang
- grid.26009.3d0000 0004 1936 7961Department of Surgery, Duke University, Durham, NC USA
| | - Marjanka K. Schmidt
- grid.430814.a0000 0001 0674 1393Division of Molecular Pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands ,grid.10419.3d0000000089452978Department of clinical genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Paula H. M. Elkhuizen
- grid.430814.a0000 0001 0674 1393Department of radiotherapy, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - Jelle Wesseling
- grid.430814.a0000 0001 0674 1393Department of pathology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands ,grid.10419.3d0000000089452978Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael Schaapveld
- grid.430814.a0000 0001 0674 1393Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Significant Inter- and Intralaboratory Variation in Gleason Grading of Prostate Cancer: A Nationwide Study of 35,258 Patients in The Netherlands. Cancers (Basel) 2021; 13:cancers13215378. [PMID: 34771542 PMCID: PMC8582481 DOI: 10.3390/cancers13215378] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Gleason grading of prostate cancer is essential for treatment strategies and patient prognosis. Previous studies showed grading variation between pathologists when grading prostate cancer. Our study analyzed the presence and extent of grading variation between and within pathology laboratories in The Netherlands. In our nationwide retrospective study, we analyzed prostate needle biopsy reports of 35,258 patients in The Netherlands graded by 40 pathology laboratories. We found a considerable variation between and within pathology laboratories, as over half of the laboratories graded significantly different from the national mean. This likely affects treatment strategy and prognosis assessment of prostate cancer patients. Abstract Purpose: Our aim was to analyze grading variation between pathology laboratories and between pathologists within individual laboratories using nationwide real-life data. Methods: We retrieved synoptic (n = 13,397) and narrative (n = 29,377) needle biopsy reports from the Dutch Pathology Registry and prostate-specific antigen values from The Netherlands Cancer Registration for prostate cancer patients diagnosed between January 2017 and December 2019. We determined laboratory-specific proportions per histologic grade and unadjusted odds ratios (ORs) for International Society of Urological Pathologists Grades 1 vs. 2–5 for 40 laboratories due to treatment implications for higher grades. Pathologist-specific proportions were determined for 21 laboratories that consented to this part of analysis. The synoptic reports of 21 laboratories were used for analysis of case-mix correction for PSA, age, year of diagnosis, number of biopsies and positive cores. Results: A total of 38,321 reports of 35,258 patients were included. Grade 1 ranged between 19.7% and 44.3% per laboratory (national mean = 34.1%). Out of 40 laboratories, 22 (55%) reported a significantly deviant OR, ranging from 0.48 (95% confidence interval (CI) 0.39–0.59) to 1.54 (CI 1.22–1.93). Case-mix correction was performed for 10,294 reports, altering the status of 3/21 (14%) laboratories, but increasing the observed variation (20.8% vs. 17.7%). Within 15/21 (71%) of laboratories, significant inter-pathologist variation existed. Conclusion: Substantial variation in prostate cancer grading was observed between and within Dutch pathology laboratories. Case-mix correction did not explain the variation. Better standardization of prostate cancer grading is warranted to optimize and harmonize treatment.
Collapse
|
15
|
Koomen BM, Voorham QJM, Epskamp-Kuijpers CCHJ, van Dooijeweert C, van Lindert ASR, Deckers IAG, Willems SM. Considerable interlaboratory variation in PD-L1 positivity in a nationwide cohort of non-small cell lung cancer patients. Lung Cancer 2021; 159:117-126. [PMID: 34332333 DOI: 10.1016/j.lungcan.2021.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/14/2021] [Accepted: 07/16/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Immunohistochemical expression of programmed death-ligand 1 (PD-L1) is used as a predictive biomarker for prescription of immunotherapy to non-small cell lung cancer (NSCLC) patients. Accurate assessment of PD-L1 expression is therefore crucial. In this study, the extent of interlaboratory variation in PD-L1 positivity in the Netherlands was assessed, using real-world clinical pathology data. MATERIALS AND METHODS Data on all NSCLC patients in the Netherlands with a mention of PD-L1 testing in their pathology report from July 2017 to December 2018 were extracted from PALGA, the nationwide network and registry of histo- and cytopathology in the Netherlands. PD-L1 positivity rates were determined for each laboratory that performed PD-L1 testing, with separate analyses for histological and cytological material. Two cutoffs (1% and 50%) were used to determine PD-L1 positivity. Differences between laboratories were assessed using funnel plots with 95% confidence limits around the overall mean. RESULTS 6,354 patients from 30 laboratories were included in the analysis of histology data. At the 1% cutoff, maximum interlaboratory variation was 39.1% (32.7%-71.8%) and ten laboratories (33.3%) differed significantly from the mean. Using the 50% cutoff, four laboratories (13.3%) differed significantly from the mean and maximum variation was 23.1% (17.2%-40.3%). In the analysis of cytology data, 1,868 patients from 23 laboratories were included. Eight laboratories (34.8%) differed significantly from the mean in the analyses of both cutoffs. Maximum variation was 41.2% (32.2%-73.4%) and 29.2% (14.7%-43.9%) using the 1% and 50% cutoffs, respectively. CONCLUSION Considerable interlaboratory variation in PD-L1 positivity was observed. Variation was largest using the 1% cutoff. At the 50% cutoff, analysis of cytology data demonstrated a higher degree of variation than the analysis of histology data.
Collapse
Affiliation(s)
- Bregje M Koomen
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| | | | - Chantal C H J Epskamp-Kuijpers
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands; PALGA Foundation, De Bouw 123, 3991 SZ, Houten, the Netherlands
| | - Carmen van Dooijeweert
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Anne S R van Lindert
- Department of Pulmonology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | | | - Stefan M Willems
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
16
|
Wetstein SC, Stathonikos N, Pluim JPW, Heng YJ, Ter Hoeve ND, Vreuls CPH, van Diest PJ, Veta M. Deep learning-based grading of ductal carcinoma in situ in breast histopathology images. J Transl Med 2021; 101:525-533. [PMID: 33608619 PMCID: PMC7985025 DOI: 10.1038/s41374-021-00540-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 11/08/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-invasive breast cancer that can progress into invasive ductal carcinoma (IDC). Studies suggest DCIS is often overtreated since a considerable part of DCIS lesions may never progress into IDC. Lower grade lesions have a lower progression speed and risk, possibly allowing treatment de-escalation. However, studies show significant inter-observer variation in DCIS grading. Automated image analysis may provide an objective solution to address high subjectivity of DCIS grading by pathologists. In this study, we developed and evaluated a deep learning-based DCIS grading system. The system was developed using the consensus DCIS grade of three expert observers on a dataset of 1186 DCIS lesions from 59 patients. The inter-observer agreement, measured by quadratic weighted Cohen's kappa, was used to evaluate the system and compare its performance to that of expert observers. We present an analysis of the lesion-level and patient-level inter-observer agreement on an independent test set of 1001 lesions from 50 patients. The deep learning system (dl) achieved on average slightly higher inter-observer agreement to the three observers (o1, o2 and o3) (κo1,dl = 0.81, κo2,dl = 0.53 and κo3,dl = 0.40) than the observers amongst each other (κo1,o2 = 0.58, κo1,o3 = 0.50 and κo2,o3 = 0.42) at the lesion-level. At the patient-level, the deep learning system achieved similar agreement to the observers (κo1,dl = 0.77, κo2,dl = 0.75 and κo3,dl = 0.70) as the observers amongst each other (κo1,o2 = 0.77, κo1,o3 = 0.75 and κo2,o3 = 0.72). The deep learning system better reflected the grading spectrum of DCIS than two of the observers. In conclusion, we developed a deep learning-based DCIS grading system that achieved a performance similar to expert observers. To the best of our knowledge, this is the first automated system for the grading of DCIS that could assist pathologists by providing robust and reproducible second opinions on DCIS grade.
Collapse
Affiliation(s)
- Suzanne C Wetstein
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Josien P W Pluim
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Yujing J Heng
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Celien P H Vreuls
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Mitko Veta
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
17
|
Patterns of treatment and outcome of ductal carcinoma in situ in the Netherlands. Breast Cancer Res Treat 2021; 187:245-254. [PMID: 33385265 PMCID: PMC8062340 DOI: 10.1007/s10549-020-06055-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/07/2020] [Indexed: 01/03/2023]
Abstract
Purpose To spare DCIS patients from overtreatment, treatment de-escalated over the years. This study evaluates the influence of these developments on the patterns of care in the treatment of DCIS with particular interest in the use of breast conserving surgery (BCS), radiotherapy following BCS and the use and type of axillary staging. Methods In this large population-based cohort study all women, aged 50–74 years diagnosed with DCIS from January 1989 until January 2019, were analyzed per two-year cohort. Results A total of 30,417 women were diagnosed with DCIS. The proportion of patients undergoing BCS increased from 47.7% in 1995–1996 to 72.7% in 2017–2018 (p < 0.001). Adjuvant radiotherapy following BCS increased from 28.9% (1995–1996) to 89.6% (2011–2012) and subsequently decreased to 74.9% (2017–2018; p < 0.001). Since its introduction, the use of sentinel lymph node biopsy (SLNB) increased to 63.1% in 2013–2014 and subsequently decreased to 52.8% in 2017–2018 (p < 0.001). Axillary surgery is already omitted in 55.8% of the patients undergoing BCS nowadays. The five-year invasive relapse-free survival (iRFS) for BCS with adjuvant radiotherapy in the period 1989–2010, was 98.7% [CI 98.4% – 99.0%], compared to 95.0% [CI 94.1% –95.8%] for BCS only (p < 0.001). In 2011–2018, this was 99.3% [CI 99.1% – 99.5%] and 98.8% [CI 98.2% – 99.4%] respectively (p = 0.01). Conclusions This study shows a shift toward less extensive treatment. DCIS is increasingly treated with BCS and less often followed by additional radiotherapy. The absence of radiotherapy still results in excellent iRFS. Axillary surgery is increasingly omitted in DCIS patients.
Collapse
|
18
|
Shaaban AM, Hilton B, Clements K, Provenzano E, Cheung S, Wallis MG, Sawyer E, Thomas JS, Hanby AM, Pinder SE, Thompson AM. Pathological features of 11,337 patients with primary ductal carcinoma in situ (DCIS) and subsequent events: results from the UK Sloane Project. Br J Cancer 2020; 124:1009-1017. [PMID: 33199800 PMCID: PMC7921398 DOI: 10.1038/s41416-020-01152-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/28/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background The Sloane audit compares screen-detected ductal carcinoma in situ (DCIS) pathology with subsequent management and outcomes. Methods This was a national, prospective cohort study of DCIS diagnosed during 2003–2012. Results Among 11,337 patients, 7204 (64%) had high-grade DCIS. Over time, the proportion of high-grade disease increased (from 60 to 65%), low-grade DCIS decreased (from 10 to 6%) and mean size increased (from 21.4 to 24.1 mm). Mastectomy was more common for high-grade (36%) than for low-grade DCIS (15%). Few (6%) patients treated with breast-conserving surgery (BCS) had a surgical margin <1 mm. Of the 9191 women diagnosed in England (median follow-up 9.4 years), 7% developed DCIS or invasive malignancy in the ipsilateral and 5% in the contralateral breast. The commonest ipsilateral event was invasive carcinoma (n = 413), median time 62 months, followed by DCIS (n = 225), at median 37 months. Radiotherapy (RT) was most protective against recurrence for high-grade DCIS (3.2% for high-grade DCIS with RT compared to 6.9% without, compared with 2.3 and 3.0%, respectively, for low/intermediate-grade DCIS). Ipsilateral DCIS events lessened after 5 years, while the risk of ipsilateral invasive cancer remained consistent to beyond 10 years. Conclusion DCIS pathology informs patient management and highlights the need for prolonged follow-up of screen-detected DCIS.
Collapse
Affiliation(s)
- Abeer M Shaaban
- Queen Elizabeth Hospital Birmingham and University of Birmingham, Birmingham, UK.
| | - Bridget Hilton
- Screening Quality Assurance Service, Public Health England, Birmingham, UK
| | - Karen Clements
- Screening Quality Assurance Service, Public Health England, Birmingham, UK
| | - Elena Provenzano
- Addenbrookes Hospital, Cambridge, UK.,Cambridge Breast Unit, and NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Shan Cheung
- Screening Quality Assurance Service, Public Health England, Birmingham, UK
| | - Matthew G Wallis
- Addenbrookes Hospital, Cambridge, UK.,Cambridge Breast Unit, and NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Elinor Sawyer
- School of Cancer & Pharmaceutical Sciences, King's College London and Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | | | - Andrew M Hanby
- Leeds Institute of Medical Research at St. James's, St James's University Hospital, Leeds, UK
| | - Sarah E Pinder
- School of Cancer & Pharmaceutical Sciences, King's College London and Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | | | | |
Collapse
|
19
|
Van Bockstal MR, Berlière M, Duhoux FP, Galant C. Interobserver Variability in Ductal Carcinoma In Situ of the Breast. Am J Clin Pathol 2020; 154:596-609. [PMID: 32566938 DOI: 10.1093/ajcp/aqaa077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Since most patients with ductal carcinoma in situ (DCIS) of the breast are treated upon diagnosis, evidence on its natural progression to invasive carcinoma is limited. It is estimated that around half of the screen-detected DCIS lesions would have remained indolent if they had never been detected. Many patients with DCIS are therefore probably overtreated. Four ongoing randomized noninferiority trials explore active surveillance as a treatment option. Eligibility for these trials is mainly based on histopathologic features. Hence, the call for reproducible histopathologic assessment has never sounded louder. METHODS Here, the available classification systems for DCIS are discussed in depth. RESULTS This comprehensive review illustrates that histopathologic evaluation of DCIS is characterized by significant interobserver variability. Future digitalization of pathology, combined with development of deep learning algorithms or so-called artificial intelligence, may be an innovative solution to tackle this problem. However, implementation of digital pathology is not within reach for each laboratory worldwide. An alternative classification system could reduce the disagreement among histopathologists who use "conventional" light microscopy: the introduction of dichotomous histopathologic assessment is likely to increase interobserver concordance. CONCLUSIONS Reproducible histopathologic assessment is a prerequisite for robust risk stratification and adequate clinical decision-making. Two-tier histopathologic assessment might enhance the quality of care.
Collapse
Affiliation(s)
- Mieke R Van Bockstal
- Department of Pathology, Brussels, Belgium
- Breast Clinic, Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Martine Berlière
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Francois P Duhoux
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
- Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christine Galant
- Department of Pathology, Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
20
|
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
|
21
|
The effect of an e-learning module on grading variation of (pre)malignant breast lesions. Mod Pathol 2020; 33:1961-1967. [PMID: 32404951 DOI: 10.1038/s41379-020-0556-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 12/14/2022]
Abstract
Histologic grade is a biomarker that is widely used to guide treatment of invasive breast cancer (IBC) and ductal carcinoma in situ of the breast (DCIS). Yet, currently, substantial grading variation between laboratories and pathologists exists in daily pathology practice. This study was conducted to evaluate whether an e-learning may be a feasible tool to decrease grading variation of (pre)malignant breast lesions. An e-learning module, representing the key-concepts of grading (pre)malignant breast lesions through gold standard digital images, was designed. Pathologists and residents could take part in either or both the separate modules on DCIS and IBC. Variation in grading of a digital set of lesions before and after the e-learning was compared in a fully-crossed study-design. Multiple outcome measures were assessed: inter-rater reliability (IRR) by Light's kappa, the number of images graded unanimously, the number of images with both extreme scores (i.e., grade I and grade III), and the average number of discrepancies from expert-consensus. Participants were included as they completed both the pre- and post-e-learning set (DCIS-module: n = 36, IBC-module: n = 21). For DCIS, all outcome measures improved after e-learning, with the IRR improving from fair (kappa: 0.532) to good (kappa: 0.657). For IBC, all outcome measures for the subcategories tubular differentiation and mitosis improved, with >90% of participants agreeing on almost 90% of the images after the e-learning. In contrast, the IRR for the subcategory of nuclear pleomorphism remained fair (kappa: 0.523 vs. kappa: 0.571). This study shows that an e-learning module, in which pathologists and residents are trained in histologic grading of DCIS and IBC, is a feasible and promising tool to decrease grading variation of (pre)malignant breast lesions. This is highly relevant given the important role of histologic grading in clinical decision making of (pre)malignant breast lesions.
Collapse
|
22
|
van Dooijeweert C, van Diest PJ, Baas IO, van der Wall E, Deckers IAG. Grading variation in 2,934 patients with ductal carcinoma in situ of the breast: the effect of laboratory- and pathologist-specific feedback reports. Diagn Pathol 2020; 15:52. [PMID: 32393303 PMCID: PMC7216330 DOI: 10.1186/s13000-020-00970-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Histologic grade of ductal carcinoma in situ of the breast (DCIS) may become the single biomarker that decides whether patients will be treated. Yet, evidence shows that grading variation in daily practice is substantial. To facilitate quality improvement, feedback reports, in which laboratory-specific case-mix adjusted proportions per grade were benchmarked against other laboratories, were sent to the individual laboratories by March 1, 2018. One year later, the effect of these feedback reports on inter-laboratory variation was studied. METHODS Synoptic pathology reports of all pure DCIS resection specimens between March 1, 2017 and March 1, 2019 were retrieved from PALGA (the nationwide Dutch pathology registry). Laboratory-specific proportions per grade were compared to the overall proportion in the year before and after feedback. The absolute deviation for all three grades at once, represented by the overall deviation score (ODS), was calculated as the sum of deviations from the grade-specific overall proportions. Case-mix adjusted, laboratory-specific odds ratios (ORs) for high- (grade III) versus low-grade (grade I-II) DCIS were obtained by multivariable logistic regression. RESULTS Overall, 2954 DCIS reports from 31 laboratories were included. After feedback, the range between laboratories decreased by 22 and 6.5% for grades II and III, while an increase of 6.2% was observed for grade I. Both the mean ODS (27.2 to 24.1%) and maximum ODS (87.7 to 59.6%) decreased considerably. However, the range of case-mix adjusted ORs remained fairly stable and substantial (0.39 (95% CI: 0.18-0.86) to 3.69 (95% CI: 1.30-10.51)). CONCLUSION A promising decrease in grading variation was observed after laboratory-specific feedback for DCIS grades II-III, while this was not observed for DCIS grade I. Overall, grading variation remained substantial which needs to be addressed considering its clinical implications. Nationwide consensus on a classification, and training of (expert breast) pathologists, for example by e-learning, may help to further improve grading standardization.
Collapse
Affiliation(s)
- Carmen van Dooijeweert
- Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508, GA, Utrecht, the Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, PO Box 85500, 3508, GA, Utrecht, the Netherlands.
| | - Inge O Baas
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Elsken van der Wall
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ivette A G Deckers
- Foundation PALGA (the nationwide network and registry of histo- and cytopathology in the Netherlands), Houten, the Netherlands
| |
Collapse
|
23
|
Cutuli B, Lemanski C, De Lafontan B, Chauvet MP, De Lara CT, Mege A, Fric D, Richard-Molard M, Mazouni C, Cuvier C, Carre A, Kirova Y. Ductal Carcinoma in Situ: A French National Survey. Analysis of 2125 Patients. Clin Breast Cancer 2020; 20:e164-e172. [DOI: 10.1016/j.clbc.2019.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 12/27/2022]
|
24
|
Interobserver variability in upfront dichotomous histopathological assessment of ductal carcinoma in situ of the breast: the DCISion study. Mod Pathol 2020; 33:354-366. [PMID: 31534203 PMCID: PMC7983551 DOI: 10.1038/s41379-019-0367-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/01/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022]
Abstract
Histopathological assessment of ductal carcinoma in situ, a nonobligate precursor of invasive breast cancer, is characterized by considerable interobserver variability. Previously, post hoc dichotomization of multicategorical variables was used to determine the "ideal" cutoffs for dichotomous assessment. The present international multicenter study evaluated interobserver variability among 39 pathologists who performed upfront dichotomous evaluation of 149 consecutive ductal carcinomas in situ. All pathologists independently assessed nuclear atypia, necrosis, solid ductal carcinoma in situ architecture, calcifications, stromal architecture, and lobular cancerization in one digital slide per lesion. Stromal inflammation was assessed semiquantitatively. Tumor-infiltrating lymphocytes were quantified as percentages and dichotomously assessed with a cutoff at 50%. Krippendorff's alpha (KA), Cohen's kappa and intraclass correlation coefficient were calculated for the appropriate variables. Lobular cancerization (KA = 0.396), nuclear atypia (KA = 0.422), and stromal architecture (KA = 0.450) showed the highest interobserver variability. Stromal inflammation (KA = 0.564), dichotomously assessed tumor-infiltrating lymphocytes (KA = 0.520), and comedonecrosis (KA = 0.539) showed slightly lower interobserver disagreement. Solid ductal carcinoma in situ architecture (KA = 0.602) and calcifications (KA = 0.676) presented with the lowest interobserver variability. Semiquantitative assessment of stromal inflammation resulted in a slightly higher interobserver concordance than upfront dichotomous tumor-infiltrating lymphocytes assessment (KA = 0.564 versus KA = 0.520). High stromal inflammation corresponded best with dichotomously assessed tumor-infiltrating lymphocytes when the cutoff was set at 10% (kappa = 0.881). Nevertheless, a post hoc tumor-infiltrating lymphocytes cutoff set at 20% resulted in the highest interobserver agreement (KA = 0.669). Despite upfront dichotomous evaluation, the interobserver variability remains considerable and is at most acceptable, although it varies among the different histopathological features. Future studies should investigate its impact on ductal carcinoma in situ prognostication. Forthcoming machine learning algorithms may be useful to tackle this substantial diagnostic challenge.
Collapse
|
25
|
van Dooijeweert C, van Diest PJ, Willems SM, Kuijpers CCHJ, van der Wall E, Overbeek LIH, Deckers IAG. Significant inter- and intra-laboratory variation in grading of invasive breast cancer: A nationwide study of 33,043 patients in the Netherlands. Int J Cancer 2020; 146:769-780. [PMID: 30977119 PMCID: PMC6916412 DOI: 10.1002/ijc.32330] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022]
Abstract
Accurate, consistent and reproducible grading by pathologists is of key-importance for identification of individual patients with invasive breast cancer (IBC) that will or will not benefit from adjuvant systemic treatment. We studied the laboratory-specific grading variation using nationwide real-life data to create insight and awareness in grading variation. Synoptic pathology reports of all IBC resection-specimens, obtained between 2013 and 2016, were retrieved from the nationwide Dutch Pathology Registry (PALGA). Absolute differences in laboratory-proportions of Grades I-III were compared to the national reference. Multivariable logistic regression provided laboratory-specific odds ratios (ORs) for high- vs. low-grade IBC. 33,792 IBC pathology reports of 33,043 patients from 39 laboratories were included, of which 28.1% were reported as Grade I (range between laboratories 16.3-43.3%), 47.6% as Grade II (38.4-57.8%), and 24.3% as Grade III (15.5-34.3%). Based on national guidelines, the indication for adjuvant chemotherapy was dependent on histologic grade in 29.9% of patients. After case-mix correction, 20 laboratories (51.3%) showed a significantly deviant OR. Significant grading differences were also observed among pathologists within laboratories. In this cohort of 33,043 breast cancer patients, we observed substantial inter- and intra-laboratory variation in histologic grading. It can be anticipated that this has influenced outcome including exposure to unnecessary toxicity, since choice of adjuvant chemotherapy was dependent on grade in nearly a third of patients. Better standardization and training seems warranted.
Collapse
Affiliation(s)
| | - Paul J. van Diest
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Stefan M. Willems
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Elsken van der Wall
- Department of Medical OncologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Lucy I. H. Overbeek
- Foundation PALGA (the nationwide network and registry of histo‐ and cytopathology in The Netherlands)HoutenThe Netherlands
| | - Ivette A. G. Deckers
- Foundation PALGA (the nationwide network and registry of histo‐ and cytopathology in The Netherlands)HoutenThe Netherlands
| |
Collapse
|
26
|
Grading Ductal Carcinoma In Situ (DCIS) of the Breast - What's Wrong with It? Pathol Oncol Res 2019; 26:665-671. [PMID: 31776839 PMCID: PMC7242244 DOI: 10.1007/s12253-019-00760-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022]
Abstract
Ductal carcinoma in situ of the breast is a non-obligate precursor of invasive breast cancer, and at its lower risk end might not need treatment, a hypothesis tested in several currently running randomized clinical trials. This review describes the heterogeneity of grading ductal carcinoma in situ (DCIS). First it considers differences between low and high grade DCIS, and then it looks at several grading schemes and highlights how different these are, not only in the features considered for defining a given grade but also in their wording of a given variable seen in the grade in question. Rather than being fully comprehensive, the review aims to illustrate the inconsistencies. Reproducibility studies on grading mostly suggestive of moderate agreement on DCIS differentiation are also illustrated. The need for a well structured, more uniform and widely accepted language for grading DCIS is urged to avoid misunderstanding based misclassifications and improper treatment selection.
Collapse
|
27
|
Ponti A, Ronco G, Lynge E, Tomatis M, Anttila A, Ascunce N, Broeders M, Bulliard JL, Castellano I, Fitzpatrick P, Frigerio A, Hofvind S, Májek O, Segnan N, Taplin S. Low-grade screen-detected ductal carcinoma in situ progresses more slowly than high-grade lesions: evidence from an international multi-centre study. Breast Cancer Res Treat 2019; 177:761-765. [PMID: 31250357 DOI: 10.1007/s10549-019-05333-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/18/2019] [Indexed: 12/27/2022]
Abstract
PURPOSE Nuclear grade is an important indicator of the biological behaviour of ductal carcinoma in situ (DCIS). De-escalation of treatment has been suggested for low-grade DCIS. Our aim is to estimate the relative rate of progression of DCIS by nuclear grade by analysing the distribution of nuclear grade by detection at initial or subsequent screening. METHODS We asked International Cancer Screening Network sites to complete, based on their screening and clinical databases, an aggregated data file on DCIS detection, diagnosis and treatment. RESULTS Eleven screening programs reported 5068 screen-detected pure DCIS in nearly 7 million screening tests in women 50-69 years of age. For all programs combined, low-grade DCIS were 20.1% (range 11.4-31.8%) of graded DCIS, intermediate grade 31.0% and high grade 48.9%. Detection rates decreased more steeply from initial to subsequent screening in low compared to high-grade DCIS: the ratios of subsequent to initial detection rates were 0.39 for low grade, 0.51 for intermediate grade, and 0.75 for high grade (p < 0.001). CONCLUSIONS These results suggest that the duration of the preclinical detectable phase is longer for low than for high-grade DCIS. The findings from this large multi-centre, international study emphasize that the management of low-grade DCIS should be carefully scrutinized in order to minimize overtreatment of screen-detected slow-growing or indolent lesions. The high variation by site in the proportion of low grade suggests that further pathology standardization and training would be beneficial.
Collapse
Affiliation(s)
- Antonio Ponti
- CPO Piemonte, AOU Città della Salute e della Scienza, Via Cavour 31, 10123, Torino, Italy.
| | - Guglielmo Ronco
- CPO Piemonte, AOU Città della Salute e della Scienza, Via Cavour 31, 10123, Torino, Italy
| | - Elsebeth Lynge
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mariano Tomatis
- CPO Piemonte, AOU Città della Salute e della Scienza, Via Cavour 31, 10123, Torino, Italy
| | - Ahti Anttila
- Mass Screening Registry, Finnish Cancer Registry, Helsinki, Finland
| | - Nieves Ascunce
- Breast Cancer Screening Program, Public Health and Labour Institute of Navarra, Pamplona, Spain
| | - Mireille Broeders
- Dutch Expert Centre for Screening and Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Jean-Luc Bulliard
- Centre for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Isabella Castellano
- Unit of Pathology, Department of Medical Sciences, University of Torino, Torino, Italy
| | | | - Alfonso Frigerio
- Breast Cancer Screening Reference Centre, AOU Città della Salute e della Scienza, Torino, Italy
| | | | - Ondřej Májek
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Nereo Segnan
- CPO Piemonte, AOU Città della Salute e della Scienza, Via Cavour 31, 10123, Torino, Italy
| | - Stephen Taplin
- Centre for Global Health, National Cancer Institute, Rockville, MD, USA
| | | |
Collapse
|
28
|
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
|