1
|
Borowsky A, Glencer A, Ramalingam K, Schindler N, Mori H, Ghule P, Lee K, Nachmanson D, Officer A, Harismendy O, Stein J, Stein G, Evans M, Weaver D, Yau C, Hirst G, Campbell M, Esserman L. Tumor microenvironmental determinants of high-risk DCIS progression. RESEARCH SQUARE 2024:rs.3.rs-4126092. [PMID: 38766192 PMCID: PMC11100907 DOI: 10.21203/rs.3.rs-4126092/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Ductal carcinoma in situ (DCIS) constitutes an array of morphologically recognized intraductal neoplasms in the mammary ductal tree defined by an increased risk for subsequent invasive carcinomas at or near the site of biopsy detection. However, only 15-45% of untreated DCIS cases progress to invasive cancer, so understanding mechanisms that prevent progression is key to avoid overtreatment and provides a basis for alternative therapies and prevention. This study was designed to characterize the tumor microenvironment and molecular profile of high-risk DCIS that grew to a large size but remained as DCIS. All patients had DCIS lesions >5cm in size with at least one additional high-risk feature: young age (<45 years), high nuclear grade, hormone receptor negativity, HER2 positivity, the presence of comedonecrosis, or a palpable mass. The tumor immune microenvironment was characterized using multiplex immunofluorescence to identify immune cells and their spatial relationships within the ducts and stroma. Gene copy number analysis and whole exome DNA sequencing identified the mutational burden and driver mutations, and quantitative whole-transcriptome/gene expression analyses were performed. There was no association between the percent of the DCIS genome characterized by copy number variants (CNAs) and recurrence events (DCIS or invasive). Mutations, especially missense mutations, in the breast cancer driver genes PIK3CA and TP53 were common in this high-risk DCIS cohort (47% of evaluated lesions). Tumor infiltrating lymphocyte (TIL) density was higher in DCIS lesions with TP53 mutations (p=0.0079) compared to wildtype lesions, but not in lesions with PIK3CA mutations (p=0.44). Immune infiltrates were negatively associated with hormone receptor status and positively associated with HER2 expression. High levels of CD3+CD8- T cells were associated with good outcomes with respect to any subsequent recurrence (DCIS or invasive cancer), whereas high levels of CD3+Foxp3+ Treg cells were associated with poor outcomes. Spatial proximity analyses of immune cells and tumor cells demonstrated that close proximity of T cells with tumor cells was associated with good outcomes with respect to any recurrence as well as invasive recurrences. Interestingly, we found that myoepithelial continuity (distance between myoepithelial cells surrounding the involved ducts) was significantly lower in DCIS lesions compared to normal tissue (p=0.0002) or to atypical ductal hyperplasia (p=0.011). Gene set enrichment analysis identified several immune pathways associated with low myoepithelial continuity and a low myoepithelial continuity score was associated with better outcomes, suggesting that gaps in the myoepithelial layer may allow access/interactions between immune infiltrates and tumor cells. Our study demonstrates the immune microenvironment of DCIS, in particular the spatial proximity of tumor cells and T cells, and myoepithelial continuity are important determinants for progression of disease.
Collapse
|
2
|
Chieh AY, Willis JG, Carroll CM, Mobley AA, Li Y, Li M, Woodard S. Why Start Now? Retrospective Study Evaluating Baseline Screening Mammography in Patients Age 60 and Older. Curr Probl Diagn Radiol 2024; 53:62-67. [PMID: 37704485 DOI: 10.1067/j.cpradiol.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/17/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
PURPOSE Extensive data exist regarding the importance of baseline mammography and screening recommendations in the age range of 40-50 years old, however, less is known about women who start screening at age 60. The purpose of this retrospective study is to assess the characteristics and outcomes of women aged 60 years and older presenting for baseline mammographic screening. METHODS This is an IRB-approved single institution retrospective review of data from patients aged 60+ receiving baseline screening mammograms between 2010 and 2022 was obtained. Information regarding patient demographics, breast density, and BI-RADS assessment was acquired from Cerner EHR. Of patients with a BI-RADS 0 assessment, imaging, and chart review was performed. Family history, gynecologic history, prior breast biopsy or surgery, and hormone use was reviewed. For those with a category 4 or 5 assessment after diagnostic work-up, biopsy outcomes were reported. Cancer detection rate (CDR), recall rate (RR), positive predictive value 1 (PPV1), PPV2, and PPV3 were calculated. RESULTS Data was analyzed from 1409 women over age 60 who underwent breast cancer screening. The recall rate was 29.3% (413/1409). The CDR, PPV1, PPV2, and PPV3 were calculated as 15/1000, 5.2% (21/405), 29.2% (21/72), and 31.8% (21/66), respectively. After work-up, 224 diagnostic patients had a 1-year follow-up and none were diagnosed with breast cancer. One (1.4%, 1/71) of the BI-RADS 3 lesions was malignant at 2-year follow-up. Of the patients recalled from screening, 29.6% had a family history of breast cancer, and the majority of both recalled and nonrecalled patients had Category B breast density. There was no statistically significant difference in breast density or race of patients recalled vs not recalled. 93.2% of recalled cases were given BI-RADS descriptors, with mass and focal asymmetry being the most common lesions, and 22.1% of recalled cases included more than one lesion. CONCLUSION Initiating screening mammography for patients over 60 years old may result in higher recall rates, but also leads to a high CDR of potentially clinically relevant invasive cancers. After a diagnostic work-up, BI-RADS 3 assessments are within standard guidelines. This study provides guidance for radiologists reading baseline mammograms and clinicians making screening recommendations in patients over age 60.
Collapse
Affiliation(s)
- Angela Y Chieh
- The University of Alabama at Birmingham Marnix E, Heersink School of Medicine, Birmingham, AL
| | - Joseph G Willis
- The University of Alabama at Birmingham Marnix E, Heersink School of Medicine, Birmingham, AL
| | - Caleb M Carroll
- The University of Alabama at Birmingham Marnix E, Heersink School of Medicine, Birmingham, AL
| | - Alisa A Mobley
- The University of Alabama at Birmingham Marnix E, Heersink School of Medicine, Birmingham, AL
| | - Yufeng Li
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Mei Li
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Stefanie Woodard
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
3
|
Schandiz H, Park D, Kaiser YL, Lyngra M, Talleraas IS, Geisler J, Sauer T. Subtypes of high-grade breast ductal carcinoma in situ (DCIS): incidence and potential clinical impact. Breast Cancer Res Treat 2023:10.1007/s10549-023-07016-9. [PMID: 37453021 PMCID: PMC10361903 DOI: 10.1007/s10549-023-07016-9] [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: 04/03/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate and classify the molecular subtypes of high-grade ductal carcinoma in situ (DCIS) and identify possible high-risk subtypes. The heterogenicity of DCIS with variable clinical and histopathological presentations has been recognized. Nevertheless, only histopathological grading and diameter are currently implemented in clinical decision-making following the diagnosis of DCIS. The molecular subtypes of DCIS and their IHC surrogate markers have not been defined in conventional treatment guidelines and recommendations. We applied the definitions of molecular subtypes according to the IHC surrogate markers defined for IBC and subclassified high-grade DCIS, accordingly. METHODS Histopathological specimens were collected, revised, and regraded from 494 patients diagnosed with DCIS between 1996 and 2018. Other in situ and papillary lesions observed in breast biopsies were excluded from this study. 357 high-grade DCIS cases were submitted to IHC analysis. The markers investigated were ER, PR, HER2, and Ki67. RESULTS 45 cases were classified as grade 1, 19 as grade 2, and 430 as grade 3. Sixty patients with high-grade DCIS had an additional invasive component in the surgical specimen. Thirty-three patients were diagnosed with recurrent DCIS or invasive cancer (minimum one year after their primary DCIS diagnosis). The proportions of luminal A and luminal B HER2-negative subtypes varied depending on whether 2011 or 2013 St. Gallen Consensus Conference guidelines were adopted. Luminal A was the most prevalent subtype, according to both classifications. The luminal B HER2-positive subtype was found in 22.1% of cases, HER2-enriched subtype in 21.8%, and TPN subtype in 5.6%. There were strong indications that HER2-enriched subtype was significantly more frequent among DCIS with invasive component (p = 0.0169). CONCLUSIONS High-grade DCIS exhibits all the molecular subtypes previously identified in IBC, but with a somewhat different distribution in our cohort. HER2-enriched subtype is substantially related to the presence of an invasive component in DCIS; consequently, it is regarded as a high-risk entity.
Collapse
Affiliation(s)
- Hossein Schandiz
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway.
| | - Daehoon Park
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Yan Liu Kaiser
- Department of Clinical Molecular Biology (EpiGen), Akershus University Hospital (AHUS), Lørenskog, Norway
| | - Marianne Lyngra
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | | | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Campus AHUS, Oslo, Norway
| | - Torill Sauer
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Campus AHUS, Oslo, Norway
| |
Collapse
|
4
|
Oncoplastic Breast Surgery versus Conservative Mastectomy in the Management of Large Ductal Carcinoma In Situ (DCIS): Surgical, Oncological, and Patient-Reported Outcomes. Cancers (Basel) 2022; 14:cancers14225624. [PMID: 36428718 PMCID: PMC9688088 DOI: 10.3390/cancers14225624] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Oncoplastic level II breast-conserving surgery (OPS2) allows for wider excisions than standard breast-conserving surgery, but the literature on this technique in the treatment of DCIS is scarce. This study compares OPS2 to conservative mastectomy (CM) in patients undergoing surgery for large DCIS. The clinical, radiological, surgical, and post-operative data of 147 patients who underwent either CM or OPS2 for large DCIS between 2007 and 2021 were retrospectively reviewed. The surgical, oncological, and patient-reported outcomes (PRO) were analyzed and compared between the two groups. The surgical outcomes were similar, in terms of margin involvement (p = 0.211), complication rate (p = 0.827), and re-excision rate (p = 1). The rate of additional surgery for cosmetic optimization was significantly lower in the OPS2 group: only 1 (1.8%) patient required surgical adjustments versus 24 (26.4%) patients in the CM group (p < 0.001). The mean hospital stay was lower in the OPS2 group (p < 0.001). The oncological outcomes did not differ between the two groups (p = 0.662). The PRO analysis showed better outcomes in the OPS2 group, which achieved statistical significance in the sexual well-being module (p = 0.015). Skin sensitivity loss was also significantly lower in the OPS2 group (p < 0.001). When feasible, OPS2 should be considered in the treatment of large DCIS, as it is safe and shows high levels of patient satisfaction.
Collapse
|
5
|
Hophan SL, Odnokoz O, Liu H, Luo Y, Khan S, Gradishar W, Zhou Z, Badve S, Torres MA, Wan Y. Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management. Endocrinology 2022; 163:bqac027. [PMID: 35245349 PMCID: PMC8962444 DOI: 10.1210/endocr/bqac027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 11/19/2022]
Abstract
Ductal carcinoma in situ (DCIS) makes up a majority of noninvasive breast cancer cases. DCIS is a neoplastic proliferation of epithelial cells within the ductal structure of the breast. Currently, there is little known about the progression of DCIS to invasive ductal carcinoma (IDC), or the molecular etiology behind each DCIS lesion or grade. The DCIS lesions can be heterogeneous in morphology, genetics, cellular biology, and clinical behavior, posing challenges to our understanding of the molecular mechanisms by which approximately half of all DCIS lesions progress to an invasive status. New strategies that pinpoint molecular mechanisms are necessary to overcome this gap in understanding, which is a barrier to more targeted therapy. In this review, we will discuss the etiological factors associated with DCIS, as well as the complexity of each nuclear grade lesion. Moreover, we will discuss the possible molecular features that lead to progression of DCIS to IDC. We will highlight current therapeutic management and areas for improvement.
Collapse
Affiliation(s)
- Shelby Lynn Hophan
- Department of Obstetrics and Gynecology, Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olena Odnokoz
- Department of Pharmacology and Chemical Biology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Huiping Liu
- Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yuan Luo
- Department of Preventive Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Seema Khan
- Department of Surgery, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - William Gradishar
- Department of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Zhuan Zhou
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sunil Badve
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mylin A Torres
- Department of Hematology and Oncology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yong Wan
- Department of Pharmacology and Chemical Biology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Hematology and Oncology, Winship Cancer Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
6
|
Roles and regulation of Haspin kinase and its impact on carcinogenesis. Cell Signal 2022; 93:110303. [DOI: 10.1016/j.cellsig.2022.110303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 01/15/2023]
|
7
|
Paul K, Razmi S, Pockaj BA, Ladani L, Stromer J. Finite Element Modeling of Quantitative Ultrasound Analysis of the Surgical Margin of Breast Tumor. Tomography 2022; 8:570-584. [PMID: 35314624 PMCID: PMC8938815 DOI: 10.3390/tomography8020047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
Ultrasound is commonly used as an imaging tool in the medical sector. Compared to standard ultrasound imaging, quantitative ultrasound analysis can provide more details about a material microstructure. In this study, quantitative ultrasound analysis was conducted through computational modeling to detect various breast duct pathologies in the surgical margin tissue. Both pulse-echo and pitch-catch methods were evaluated for a high-frequency (22–41 MHz) ultrasound analysis. The computational surgical margin modeling was based on various conditions of breast ducts, such as normal duct, ductal hyperplasia, DCIS, and calcification. In each model, ultrasound pressure magnitude variation in the frequency spectrum was analyzed through peak density and mean-peak-to-valley distance (MPVD) values. Furthermore, the spectral patterns of all the margin models were compared to extract more pathology-based information. For the pitch-catch mode, only peak density provided a trend in relation to different duct pathologies. For the pulse-echo mode, only the MPVD was able to do that. From the spectral comparison, it was found that overall pressure magnitude, spectral variation, peak pressure magnitude, and corresponding frequency level provided helpful information to differentiate various pathologies in the surgical margin.
Collapse
Affiliation(s)
- Koushik Paul
- School for Engineering of Matter, Transport and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85281, USA;
- Correspondence:
| | - Samuel Razmi
- EnMed Department, Texas A&M College of Medicine, Houston, TX 77807, USA;
| | | | - Leila Ladani
- School for Engineering of Matter, Transport and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85281, USA;
| | - Jeremy Stromer
- Survivability Engineering Branch, US Army Engineer Research and Development Center, Vicksburg, MS 39180, USA;
| |
Collapse
|
8
|
Dillard C, Reis JGT, Rusten TE. RasV12; scrib-/- Tumors: A Cooperative Oncogenesis Model Fueled by Tumor/Host Interactions. Int J Mol Sci 2021; 22:ijms22168873. [PMID: 34445578 PMCID: PMC8396170 DOI: 10.3390/ijms22168873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
The phenomenon of how oncogenes and tumor-suppressor mutations can synergize to promote tumor fitness and cancer progression can be studied in relatively simple animal model systems such as Drosophila melanogaster. Almost two decades after the landmark discovery of cooperative oncogenesis between oncogenic RasV12 and the loss of the tumor suppressor scribble in flies, this and other tumor models have provided new concepts and findings in cancer biology that has remarkable parallels and relevance to human cancer. Here we review findings using the RasV12; scrib-/- tumor model and how it has contributed to our understanding of how these initial simple genetic insults cooperate within the tumor cell to set in motion the malignant transformation program leading to tumor growth through cell growth, cell survival and proliferation, dismantling of cell-cell interactions, degradation of basement membrane and spreading to other organs. Recent findings have demonstrated that cooperativity goes beyond cell intrinsic mechanisms as the tumor interacts with the immediate cells of the microenvironment, the immune system and systemic organs to eventually facilitate malignant progression.
Collapse
Affiliation(s)
- Caroline Dillard
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Correspondence: (C.D.); (T.E.R.)
| | - José Gerardo Teles Reis
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Tor Erik Rusten
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Correspondence: (C.D.); (T.E.R.)
| |
Collapse
|
9
|
Predictors of Sentinel Lymph Node Metastasis in Postoperatively Upgraded Invasive Breast Carcinoma Patients. Cancers (Basel) 2021; 13:cancers13164099. [PMID: 34439252 PMCID: PMC8392104 DOI: 10.3390/cancers13164099] [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: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
Sentinel lymph node (SLN) biopsy (SLNB) usually need not be simultaneously performed with breast-conserving surgery (BCS) for patients diagnosed with ductal carcinoma in situ (DCIS) by preoperative core needle biopsy (CNB), but must be performed once there is invasive carcinoma (IC) found postoperatively. This study aimed to investigate the factors contributing to SLN metastasis in underestimated IC patients with an initial diagnosis of DCIS by CNB. We retrospectively reviewed 1240 consecutive cases of DCIS by image-guided CNB from January 2010 to December 2017 and identified 316 underestimated IC cases with SLNB. Data on clinical characteristics, radiologic features, and final pathological findings were examined. Twenty-three patients (7.3%) had SLN metastasis. Multivariate analysis indicated that an IC tumor size > 0.5 cm (odds ratio: 3.11, p = 0.033) and the presence of lymphovascular invasion (odds ratio: 32.85, p < 0.0001) were independent risk predictors of SLN metastasis. In the absence of any predictors, the incidence of positive SLNs was very low (2.6%) in the total population and extremely low (1.3%) in the BCS subgroup. Therefore, omitting SLNB may be an acceptable option for patients who initially underwent BCS without risk predictors on final pathological assessment. Further prospective studies are necessary before clinical application.
Collapse
|
10
|
Avdan Aslan A, Gültekin S, Esendağli Yilmaz G, Kurukahvecioğlu O. Is There Any Association Between Mammographic Features of Microcalcifications and Breast Cancer Subtypes in Ductal Carcinoma In Situ? Acad Radiol 2021; 28:963-968. [PMID: 32620528 DOI: 10.1016/j.acra.2020.05.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 01/29/2023]
Abstract
RATIONALE AND OBJECTIVES To analyze the association between mammographic features of microcalcifications and histopathological prognostic factors based on estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2/neu) in ductal carcinoma in situ (DCIS). MATERIALS AND METHODS We retrospectively determined 66 patients with microcalcification-associated pure DCIS. Distribution and morphological features of the microcalcifications were described using Breast Imaging Reporting and Data System lexicon. All patients were divided into three subgroups: ER-positive, HER-2 positive, and triple-negative according to the immunohistochemical findings. RESULTS The morphological features of microcalcifications and receptor subtypes were significantly correlated (p = 0.026). Fine pleomorphic and fine linear branching microcalcifications were observed in 85.2% of HER-2 positive cases, whereas this ratio was 71.4 % in ER-positive and 25% in the triple-negative group. Fine linear branching microcalcifications with linear or segmental distribution were more frequently found with comedo necrosis (p < 0.05). Larger tumour sizes were also associated with microcalcification distribution (p < 0.001). Segmental microcalcifications more likely associated with larger tumour sizes. CONCLUSION Mammographic features in DCIS correlated with immunohistochemical and histopathological prognostic factors.
Collapse
|
11
|
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
|
12
|
Lewis GD, Haque W, Farach A, Hatch SS, Butler EB, Niravath PA, Schwartz MR, Bonefas E, Teh BS. The impact of HER2-directed targeted therapy on HER2-positive DCIS of the breast. ACTA ACUST UNITED AC 2021; 26:179-187. [PMID: 34211767 DOI: 10.5603/rpor.a2021.0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/30/2021] [Indexed: 12/19/2022]
Abstract
Background In invasive breast cancer, HER2 is a well-established negative prognostic factor. However, its significance on the prognosis of ductal carcinoma in situ (DCIS) of the breast is unclear. As a result, the impact of HER2-directed therapy on HER2-positive DCIS is unknown and is currently the subject of ongoing clinical trials. In this study, we aim to determine the possible impact of HER 2-directed targeted therapy on survival outcomes for HER2-positive DCIS patients. Materials and methods The National Cancer Data Base (NCDB) was used to retrieve patients with biopsy-proven DCIS diagnosed from 2004-2015. Patients were divided into two groups based on the adjuvant therapy they received: systemic HER2-directed targeted therapy or no systemic therapy. Statistics included multivariable logistic regression to determine factors predictive of receiving systemic therapy, Kaplan-Meier analysis to evaluate overall survival (OS), and Cox proportional hazards modeling to determine variables associated with OS. Results Altogether, 1927 patients met inclusion criteria; 430 (22.3%) received HER2-directed targeted therapy; 1497 (77.7%) did not. Patients who received HER2-directed targeted therapy had a higher 5-year OS compared to patients that did not (97.7% vs. 95.8%, p = 0.043). This survival benefit remained on multivariable analysis. Factors associated with worse OS on multivariable analysis included Charlson-Deyo Comorbidity Score ≥ 2 and no receipt of hormonal therapy. Conclusion In this large study evaluating HER2-positive DCIS patients, the receipt of HER2-directed targeted therapy was associated with an improvement in OS. The results of currently ongoing clinical trials are needed to confirm this finding.
Collapse
Affiliation(s)
- Gary D Lewis
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Waqar Haque
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Andrew Farach
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Sandra S Hatch
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas, United States
| | - E Brian Butler
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Polly A Niravath
- Department of Clinical Medicine in Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Mary R Schwartz
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, United States
| | | | - Bin S Teh
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| |
Collapse
|
13
|
Taurin S, Alkhalifa H. Breast cancers, mammary stem cells, and cancer stem cells, characteristics, and hypotheses. Neoplasia 2020; 22:663-678. [PMID: 33142233 PMCID: PMC7586061 DOI: 10.1016/j.neo.2020.09.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022]
Abstract
The cellular heterogeneity of breast cancers still represents a major therapeutic challenge. The latest genomic studies have classified breast cancers in distinct clusters to inform the therapeutic approaches and predict clinical outcomes. The mammary epithelium is composed of luminal and basal cells, and this seemingly hierarchical organization is dependent on various stem cells and progenitors populating the mammary gland. Some cancer cells are conceptually similar to the stem cells as they can self-renew and generate bulk populations of nontumorigenic cells. Two models have been proposed to explain the cell of origin of breast cancer and involve either the reprogramming of differentiated mammary cells or the dysregulation of mammary stem cells or progenitors. Both hypotheses are not exclusive and imply the accumulation of independent mutational events. Cancer stem cells have been isolated from breast tumors and implicated in the development, metastasis, and recurrence of breast cancers. Recent advances in single-cell sequencing help deciphering the clonal evolution within each breast tumor. Still, few clinical trials have been focused on these specific cancer cell populations.
Collapse
Affiliation(s)
- Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain.
| | - Haifa Alkhalifa
- New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
14
|
Imaging of Breast Cancers With Predilection for Nonmass Pattern of Growth: Invasive Lobular Carcinoma and DCIS-Does Imaging Capture It All? AJR Am J Roentgenol 2020; 215:1504-1511. [PMID: 33021831 DOI: 10.2214/ajr.19.22027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE. Invasive lobular carcinoma (ILC) and ductal carcinoma in situ (DCIS) are distinct histopathologic entities with several commonalities: both have subtle clinical and imaging presentation, have been linked with controversy regarding optimal imaging techniques and management, and exemplify the codependence of adequate imaging evaluation and optimal treatment strategies in breast care. CONCLUSION. We review molecular mechanisms and histopathologic patterns that define the biologic behavior of both ILC and DCIS and discuss how these mechanisms translate into distinct clinical and imaging presentations that affect the staging workup and patient management algorithm.
Collapse
|
15
|
Hagerling C, Owyong M, Sitarama V, Wang CY, Lin C, van den Bijgaart RJE, Koopman CD, Brenot A, Nanjaraj A, Wärnberg F, Jirström K, Klein OD, Werb Z, Plaks V. LGR5 in breast cancer and ductal carcinoma in situ: a diagnostic and prognostic biomarker and a therapeutic target. BMC Cancer 2020; 20:542. [PMID: 32522170 PMCID: PMC7285764 DOI: 10.1186/s12885-020-06986-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Novel biomarkers are required to discern between breast tumors that should be targeted for treatment from those that would never become clinically apparent and/or life threatening for patients. Moreover, therapeutics that specifically target breast cancer (BC) cells with tumor-initiating capacity to prevent recurrence are an unmet need. We investigated the clinical importance of LGR5 in BC and ductal carcinoma in situ (DCIS) to explore LGR5 as a biomarker and a therapeutic target. METHODS We stained BC (n = 401) and DCIS (n = 119) tissue microarrays with an antibody against LGR5. We examined an LGR5 knockdown ER- cell line that was orthotopically transplanted and used for in vitro colony assays. We also determined the tumor-initiating role of Lgr5 in lineage-tracing experiments. Lastly, we transplanted ER- patient-derived xenografts into mice that were subsequently treated with a LGR5 antibody drug conjugate (anti-LGR5-ADC). RESULTS LGR5 expression correlated with small tumor size, lower grade, lymph node negativity, and ER-positivity. ER+ patients with LGR5high tumors rarely had recurrence, while high-grade ER- patients with LGR5high expression recurred and died due to BC more often. Intriguingly, all the DCIS patients who later died of BC had LGR5-positive tumors. Colony assays and xenograft experiments substantiated a role for LGR5 in ER- tumor initiation and subsequent growth, which was further validated by lineage-tracing experiments in ER- /triple-negative BC mouse models. Importantly, by utilizing LGR5high patient-derived xenografts, we showed that anti-LGR5-ADC should be considered as a therapeutic for high-grade ER- BC. CONCLUSION LGR5 has distinct roles in ER- vs. ER+ BC with potential clinical applicability as a biomarker to identify patients in need of therapy and could serve as a therapeutic target for high-grade ER- BC.
Collapse
Affiliation(s)
- Catharina Hagerling
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA. .,Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, SE-221 85, Lund, Sweden. .,Present Address: Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, SE-221 85, Lund, Sweden.
| | - Mark Owyong
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA
| | - Vaishnavi Sitarama
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA
| | - Chih-Yang Wang
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA.,Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Charlene Lin
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA
| | - Renske J E van den Bijgaart
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA.,Present Address: Radiotherapy and Oncoimmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 32, 6525 GA, Nijmegen, Netherlands
| | - Charlotte D Koopman
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA.,Present Address: Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584CM, Utrecht, Netherlands.,Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Centre Utrecht, 3584CT, Utrecht, Netherlands
| | - Audrey Brenot
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA.,Present Address: ICCE Institute, School of Medicine, Department of Medicine, Washington University, St Louis, MO, 63110, USA
| | - Ankitha Nanjaraj
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA
| | - Fredrik Wärnberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, S413 45, Gothenburg, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, SE-221 85, Lund, Sweden
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143-0452, USA.,Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Zena Werb
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA
| | - Vicki Plaks
- Department of Anatomy and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143-0452, USA. .,Department of Orofacial Sciences, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143-0452, USA.
| |
Collapse
|
16
|
Does sentinel lymph node biopsy for screening high-grade ductal carcinoma in situ of the breast cause more harm than good? Breast Cancer Res Treat 2020; 182:47-54. [PMID: 32430678 DOI: 10.1007/s10549-020-05690-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) of the breast does not metastasize to axillary lymph nodes. Yet high-grade DCIS (HgDCIS) is often subjected to Sentinel Lymph Node Biopsy (SLNB) concomitant with definitive surgery. This is to avoid further axillary surgery in the event of upstaging to invasive carcinoma, which often entails Axillary Lymph Node Dissection (ALND). We wished to examine the validity of this approach. METHODS This study includes a retrospective analysis of consecutive pre-operatively diagnosed HgDCIS patients from a single screening unit between December/2014 and August/2016. The main outcomes were the overall incidence of upstaging and the independent predictors of upstaging on multivariable analysis. The rates of various complications of SLNB vs ALND in four RCTs were used to calculate the upstaging rate below which SLNB could be safely omitted. RESULTS There were 224 eligible patients of whom 26 (11.6%) were upstaged. Axillary metastasis (pN1) occurred in two patients (0.9%). On Univariable analysis, upstaged patients were significantly younger (median (IQR) = 56.0 (51.0-63) vs 60.0 (54.0-65.0); p = 0.019). Radiological size, pathological size, type of biopsy, type of operation, and comedo-necrosis were not significant (p > 0.05). On multivariable analysis, age as a continuous variable (OR 0.93; p = 0.031) and core biopsy (OR 2.62; p = 0.036) were the only independent predictors of upstaging. Chi-square test showed that patients < 55 years whose pre-operative diagnosis was made on core biopsy were at significantly higher risk of upstaging than the others (31.8% vs 9.4%; p = 0.002). CONCLUSION Upstaging of HgDCIS is infrequent. According to the known rates of complications of SLNB relative to ALND, routine SLNB concomitant with surgery seems to be more harmful than its routine omission. A selective approach based on age and type of biopsy could be considered.
Collapse
|
17
|
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
|
18
|
Nyhof BB, Wright FC, Look Hong NJ, Groot G, Helyer L, Meiers P, Quan ML, Baxter NN, Urquhart R, Warburton R, Gagliardi AR. Recommendations to improve patient-centred care for ductal carcinoma in situ: Qualitative focus groups with women. Health Expect 2019; 23:106-114. [PMID: 31532871 PMCID: PMC6978860 DOI: 10.1111/hex.12973] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/28/2019] [Accepted: 09/03/2019] [Indexed: 11/29/2022] Open
Abstract
Background Patient‐centred care (PCC) improves health‐care experiences and outcomes. Women with ductal carcinoma in situ (DCIS) and clinicians have reported communication difficulties. Little prior research has studied how to improve communication and PCC for DCIS. Objective This study explored how to achieve PCC for DCIS. Design Canadian women treated for DCIS from five provinces participated in semi‐structured focus groups based on a 6‐domain cancer‐specific PCC framework to discuss communication about DCIS. Data were analysed using constant comparative technique. Setting and Participants Thirty‐five women aged 30 to 86 participated in five focus groups at five hospitals. Results Women said their clinicians used multiple approaches for fostering a healing relationship; however, most described an absence of desired information or behaviour to exchange information, respond to emotions, manage uncertainty, make decisions and enable self‐management. Most women were confused by terminology, offered little information about the risks of progression/recurrence, uninformed about treatment benefits and risks, frustrated with lack of engagement in decision making, given little information about follow‐up plans or self‐care advice, and received no acknowledgement or offer of emotional support. Discussion and Conclusions By comparing the accounts of women with DCIS to a PCC framework, we identified limitations and inconsistencies in women's lived experience of communication about DCIS, and approaches by which clinicians can more consistently achieve PCC for DCIS. Future research should develop and evaluate informational tools to support PCC for DCIS.
Collapse
Affiliation(s)
- Bryanna B Nyhof
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | - Gary Groot
- University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | | | | | | | | | - Anna R Gagliardi
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
19
|
van Seijen M, Lips EH, Thompson AM, Nik-Zainal S, Futreal A, Hwang ES, Verschuur E, Lane J, Jonkers J, Rea DW, Wesseling J. Ductal carcinoma in situ: to treat or not to treat, that is the question. Br J Cancer 2019; 121:285-292. [PMID: 31285590 PMCID: PMC6697179 DOI: 10.1038/s41416-019-0478-6] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/27/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) now represents 20-25% of all 'breast cancers' consequent upon detection by population-based breast cancer screening programmes. Currently, all DCIS lesions are treated, and treatment comprises either mastectomy or breast-conserving surgery supplemented with radiotherapy. However, most DCIS lesions remain indolent. Difficulty in discerning harmless lesions from potentially invasive ones can lead to overtreatment of this condition in many patients. To counter overtreatment and to transform clinical practice, a global, comprehensive and multidisciplinary collaboration is required. Here we review the incidence of DCIS, the perception of risk for developing invasive breast cancer, the current treatment options and the known molecular aspects of progression. Further research is needed to gain new insights for improved diagnosis and management of DCIS, and this is integrated in the PRECISION (PREvent ductal Carcinoma In Situ Invasive Overtreatment Now) initiative. This international effort will seek to determine which DCISs require treatment and prevent the consequences of overtreatment on the lives of many women affected by DCIS.
Collapse
Affiliation(s)
- Maartje van Seijen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alastair M Thompson
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Serena Nik-Zainal
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Andrew Futreal
- Department of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Comprehensive Cancer Center, Durham, NC, USA
| | | | - Joanna Lane
- Health Cluster Net, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Daniel W Rea
- Department of Medical Oncology, University of Birmingham, Birmingham, UK
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
20
|
Ubago JM, Blanco LZ, Shen T, Siziopikou KP. The PD-1/PD-L1 Axis in HER2+ Ductal Carcinoma In Situ (DCIS) of the Breast. Am J Clin Pathol 2019; 152:169-176. [PMID: 30984969 DOI: 10.1093/ajcp/aqz020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES The aims were to evaluate the programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) axis in ductal carcinoma in situ (DCIS) of the breast. METHODS We reviewed 85 pure DCIS cases treated with surgical excision at our institution, including 51 luminal A (estrogen receptor [ER] positive/human epidermal growth factor 2 [HER2] negative), 15 luminal B (ER+/HER2+), 13 HER2 (ER-/HER2+), and six basal-like (ER-/HER2-/CK5/6+). The extent and intensity of PD-1 and PD-L1 immunohistochemical staining in the tumor-infiltrating lymphocytes (TILs) and in the tumor cells were recorded. RESULTS Our study found that moderate/severe inflammation around DCIS correlated with HER2 expression (20/28 HER2+ cases [71%] vs 21/57 HER2- cases [37%], P = .005). Of interest, over half of the TILs around the HER2 subtype expressed PD-L1 (7/13, 54%). In addition, about one-third of TILs around the HER2 subtype expressed PD-1 (4/13, 31%). CONCLUSIONS These findings suggest that immune-based therapeutic strategies may be used as a potential therapy in DCIS cases with PD-L1 overexpression, especially those of the HER2 molecular subtype.
Collapse
Affiliation(s)
- Julianne M Ubago
- Breast Pathology Section, Department of Pathology, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Luis Z Blanco
- Breast Pathology Section, Department of Pathology, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Tiansheng Shen
- Breast Pathology Section, Department of Pathology, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Kalliopi P Siziopikou
- Breast Pathology Section, Department of Pathology, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| |
Collapse
|
21
|
Prognostic role of immune infiltrates in breast ductal carcinoma in situ. Breast Cancer Res Treat 2019; 177:17-27. [PMID: 31134489 DOI: 10.1007/s10549-019-05272-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/06/2019] [Indexed: 01/13/2023]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) of the breast is often regarded as a non-obligate precursor to invasive breast carcinoma but current diagnostic tools are unable to accurately predict the invasive potential of DCIS. Infiltration of immune cells into the tumour and its microenvironment is often an early event at the site of tumourigenesis. These immune infiltrates may be potential predictive and/or prognostic biomarkers for DCIS. This review aims to discuss recent findings pertaining to the potential prognostic significance of immune infiltrates as well as their evaluation in DCIS. METHODS A literature search on PubMed was conducted up to 28th January 2019. Search terms used were "DCIS", "ductal carcinoma in situ", "immune", "immunology", "TIL", "TIL assessment", and "tumour-infiltrating lymphocyte". Search filters for "Most Recent" and "English" were applied. Information from published papers related to the research topic were synthesised and summarised for this review. RESULTS Studies have revealed that immune infiltrates play a role in the biology and microenvironment of DCIS, as well as treatment response. There is currently no consensus on the evaluation of TILs in DCIS for clinical application. CONCLUSIONS This review highlights the recent findings on the potential influence and prognostic value of immunological processes on DCIS progression, as well as the evaluation of TILs in DCIS. Further characterisation of the immune milieu of DCIS is recommended to better understand the immune response in DCIS progression and recurrence.
Collapse
|
22
|
Van Bockstal M, Lambein K, Smeets A, Slembrouck L, Neven P, Nevelsteen I, Weltens C, Van Limbergen E, Christiaens MR, Van Ongeval C, Wildiers H, Libbrecht L, Floris G. Stromal characteristics are adequate prognosticators for recurrence risk in ductal carcinoma in situ of the breast. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2018; 45:550-559. [PMID: 30454971 DOI: 10.1016/j.ejso.2018.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) of the breast constitutes a heterogeneous group of non-obligate precursors for invasive breast cancer. To date, adequate risk stratification is lacking, which is presumed to result in overtreatment. We previously identified myxoid stromal architecture as a potential prognosticator for loco-regional recurrence. In the present study, we investigated the prognostic potential of stromal characteristics. METHODS Hematoxylin and eosin stained slides from 211 DCIS patients were reviewed. The following histological features were dichotomously assessed: nuclear grade, DCIS architecture, presence of necrosis, intraductal calcifications, stromal inflammation and myxoid stromal architecture. Loco-regional recurrences constituted the primary endpoint. RESULTS Cox regression analysis showed that high nuclear grade, myxoid stromal architecture and moderate to extensive stromal inflammation were significantly associated with decreased recurrence-free survival, independent of radiotherapy. Based on these features, a combined risk score (CRS) was calculated, ranging from zero to three. A high CRS of three was associated with significantly shorter recurrence-free survival. Nineteen patients had a CRS of three, of which three relapsed (15.7%), whereas only one out of 113 patients with a CRS of zero relapsed (0.9%). CONCLUSIONS We were able to validate our previously reported findings regarding the prognostic potential of myxoid periductal stroma in an independent DCIS patient cohort. A CRS based on nuclear grade, myxoid stromal architecture and stromal inflammation might facilitate discrimination of low risk from high risk patients. Consequently, the CRS may tailor adjuvant therapy. Future research should investigate whether radiotherapy can be safely omitted in patients with a low CRS.
Collapse
Affiliation(s)
- Mieke Van Bockstal
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, the Netherlands; Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium.
| | - Kathleen Lambein
- Department of Pathology, AZ St Lucas Hospital Ghent, Groenebriel 1, 9000 Ghent, Belgium; Department of Surgical Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Laurence Slembrouck
- Department of Oncology, KUL University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Patrick Neven
- Department of Oncology, KUL University of Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Gynecology and Obstetrics, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Ines Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Caroline Weltens
- Department of Radiotherapy Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Erik Van Limbergen
- Department of Radiotherapy Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Marie-Rose Christiaens
- Department of Surgical Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Chantal Van Ongeval
- Department of Radiology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Hans Wildiers
- Department of Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Louis Libbrecht
- Department of Pathology, University Clinics St Luc, Hippokrateslaan 10, 1200 Sint-Lambrechts-Woluwe, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KUL University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
23
|
Wu R, Liu F, Peng P, Qiu H, Xiong H, Yu S, Huang X, Zhang H, Zhuang L. Tumor stress-induced phosphoprotein 1 as a prognostic biomarker for breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:302. [PMID: 30211190 DOI: 10.21037/atm.2018.06.46] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Recent studies suggested an important relationship between tumor stress-induced phosphoprotein 1 (STIP1) and cancer. However, the expression of STIP1 in breast cancer tissues and its relationship with clinical characteristics and survival have not been investigated in humans. The aim of our work was to evaluate the association of STIP1 and the prognosis of breast cancer patients. Methods The included patients were followed-up by telephone and through a review of their outpatient records. The expression of STIP1 was assessed by immunohistochemistry (IHC). The 5-year recurrence-free survival (RFS) rate and the 5-year overall survival (OS) rate were the prognostic indicators evaluated by the Kaplan-Meier method. Univariate and multivariate analyses employing a Cox regression model were used to calculate hazard ratios (HRs). Results The rate of high expression of STIP1 was 55.3% (126/228) in breast cancer tissues and 14.9% (34/228) in adjacent normal tissues (χ2=81.495, P<0.001). High expression of STIP1 was associated with tumor size, stage and human epidermal growth factor receptor 2 (HER-2) status. The 5-year RFS rate was 75.4% in the STIP1 high expression group and 87.3% in the STIP1 low expression group (χ2=5.721, P=0.017). The 5-year OS rate was 84.1% in the STIP1 high expression group and 94.1% in the STIP1 low expression group (χ2=5.814, P=0.016). STIP1 was found to be an independent relapse predictor for the adjusted HR is 1.983 (95% CI, 1.031-3.815). Conclusions High expression of STIP1 is associated with the poor prognosis of breast cancer patients and HER-2 positive expression. STIP1 may therefore serve as a prognostic biomarker for breast cancer patients.
Collapse
Affiliation(s)
- Ruxing Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fei Liu
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Peng
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong Qiu
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huihua Xiong
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiying Yu
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoyuan Huang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hanwang Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Zhuang
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
24
|
Chade MC, Piato S, Galvão MAL, Aldrighi JM, Negrini R, Mateus EF, Medeiros EM. Evaluation of survivin immunoexpression in the differentiation of high- and low-grade breast ductal carcinoma in situ. ACTA ACUST UNITED AC 2018; 16:eAO4065. [PMID: 29694611 PMCID: PMC5968810 DOI: 10.1590/s1679-45082018ao4065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/22/2017] [Indexed: 12/16/2022]
Abstract
Objective To evaluate the expression of survivin protein in low- and high-grade ductal carcinoma in situ. Methods Breast tissue fragments obtained by incisional biopsy and surgical procedures of 37 women with ductal carcinoma in situ of the breast were subdivided into two groups: Group A, composed of women with low-grade ductal carcinoma in situ, and Group B, women with high-grade ductal carcinoma in situ. Survivin protein expression test was performed by immunohistochemistry, using a monoclonal antibody clone I2C4. The criterion to evaluate survivin immunoexpression was based on the percentage of neoplastic cells that presented brown-gold staining. This criterion was positive when the percentage of stained cells was ≥10%. Results The survivin protein was expressed in 22 out of 24 cases of high-grade ductal carcinoma in situ (78%), whereas, in Group A, of low-grade ductal carcinoma in situ (n=13), it was positive in only 6 cases (21.40%; p=0.004). Conclusion The frequency of expression of survivin was significantly higher in the group of patients with high-grade ductal carcinoma in situ compared to those in the low-grade ductal carcinoma in situ group.
Collapse
Affiliation(s)
- Milca Cezar Chade
- Faculdade de Ciências Médicas, Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Sebastião Piato
- Faculdade de Ciências Médicas, Santa Casa de São Paulo, São Paulo, SP, Brazil
| | | | | | - Rômulo Negrini
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | | |
Collapse
|
25
|
Yoo J, Kim BS, Yoon HJ. Predictive significance of breast-specific gamma imaging for upstaging core-needle biopsy-detected ductal carcinoma in situ to invasive cancer. Ann Nucl Med 2018; 32:328-336. [DOI: 10.1007/s12149-018-1251-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/15/2018] [Indexed: 12/21/2022]
|
26
|
Kim S, Lee S, Kim S, Lee S, Yum H. The usefulness of fluorodeoxyglucose-PET/CT for preoperative evaluation of ductal carcinoma in situ. Ann Surg Treat Res 2018; 94:63-68. [PMID: 29441334 PMCID: PMC5801329 DOI: 10.4174/astr.2018.94.2.63] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 05/30/2017] [Accepted: 06/15/2017] [Indexed: 11/30/2022] Open
Abstract
Purpose PET/CT is useful in preoperative evaluation of invasive breast cancer (IBC) to predict axillary metastasis and staging workup. The usefulness is unclear in cases of ductal carcinoma in situ (DCIS) diagnosed at biopsy before surgery, which sometimes is upgraded to IBC after definitive surgery. The aim of this study is to find out the usefulness of PET/CT on DCIS as a preoperative evaluation tool. Methods We investigated 102 patients preoperatively diagnosed with DCIS who subsequently underwent definitive surgery between 2010 and 2015. The uptake of 18F-fluorodeoxyglucose was graded by visual and semiquantitative methods. We analyzed the maximum standardized uptake value (SUVmax) of each patient with clinicopathologic variables. We determined optimal cutoff values for SUVmax by receiver operating characteristic curve analysis. Results Fifteen cases out of 102 cases (14.7%) were upgraded to IBC after surgery. The SUVmax was higher in patients upgraded to IBC (mean: 2.56 vs. 1.36) (P = 0.007). The SUVmax was significantly higher in patients who had symptoms, palpable masses, lesions over 2 cm in size and BI-RAD category 5. Both visual and semiquantitative analysis were significant predictors of IBC underestimation. SUVmax of 2.65 was the theoretical cutoff value in ROC curve analysis in predicting the underestimation of IBC. The underestimation rate was significantly higher in patients with SUVmax >2.65 (P < 0.001), over the moderate enhanced uptake on visual analysis (P < 0.001). Conclusion PET/CT can be used as a complementary evaluation tool to predict the underestimation of DCIS combined with the lesion size, palpable mass, symptomatic lesion, and BI-RAD category.
Collapse
Affiliation(s)
- Sungchul Kim
- Department of Surgery, Mother's Hospital, Busan, Korea
| | - Seokjae Lee
- Department of Surgery, Mother's Hospital, Busan, Korea
| | - Sangwon Kim
- Department of Surgery, Mother's Hospital, Busan, Korea
| | - Seokmo Lee
- Department of Nuclear Medicine, Inje University College of Medicine, Busan, Korea
| | - Hayong Yum
- Busan PET & Dr Yum's Thyroid Clinic, Busan, Korea
| |
Collapse
|
27
|
Huang N, Si J, Yang B, Quan C, Chen J, Wu J. Trends and clinicopathological predictors of axillary evaluation in ductal carcinoma in situ patients treated with breast-conserving therapy. Cancer Med 2018; 7:56-63. [PMID: 29271113 PMCID: PMC5774004 DOI: 10.1002/cam4.1252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/07/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to investigate the trends of axillary lymph node evaluation in ductal carcinoma in situ (DCIS) patients treated with breast-conserving therapy (BCT) and to identify the clinicopathological predictors of axillary evaluation. DCIS patients treated with BCT in 2006-2015 at our institute were retrospectively included in the analysis. Patients were categorized into three groups: sentinel lymph node biopsy (SLNB), axillary lymph node dissection (ALND), and non-evaluation. Univariate and multivariate logistic regression analyses were performed to identify factors that predicted axillary evaluation. A total of 315 patients were identified, among whom 135 underwent SLNB, and 15 underwent ALND. The proportion of patients who underwent axillary evaluation increased from 33.0% in 2006-2010 to 53.8% in 2011-2015 (P < 0.001), however, no patients had lymph node metastasis based on final pathology. In multivariate analysis, high-grade tumor favored axillary evaluation (OR = 4.376, 95% CI:1.410-13.586, P = 0.011); while excision biopsy favored no axillary evaluation compared with other biopsy methods (OR = 0.418, 95% CI: 0.192-0.909, P = 0.028). Subgroup analysis of patients treated in 2011-2015 revealed that high-grade tumor (OR = 5.898, 95% CI: 1.626-21.390, P = 0.007) and palpable breast lump (OR = 2.497, 95% CI: 1.037-6.011, P = 0.041) were independent predictors of axillary lymph node evaluation. Despite the significant decrease in ALND and a concerning overuse of SLNB, we identified no axillary lymph node metastasis, which justified omitting axillary evaluation in these patients. High-grade tumor, palpable lump, and biopsy method were independent predictors of axillary evaluations. Excision biopsy of suspicious DCIS lesions may potentially preclude the invasive component of the disease and help to avoid axillary surgery.
Collapse
Affiliation(s)
- Nai‐si Huang
- Department of Breast SurgeryFudan University Shanghai Cancer CenterNo. 270, Dongan RdShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghaiChina
| | - Jing Si
- Department of Breast SurgeryFudan University Shanghai Cancer CenterNo. 270, Dongan RdShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghaiChina
| | - Ben‐long Yang
- Department of Breast SurgeryFudan University Shanghai Cancer CenterNo. 270, Dongan RdShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghaiChina
| | - Chen‐lian Quan
- Department of Breast SurgeryFudan University Shanghai Cancer CenterNo. 270, Dongan RdShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghaiChina
| | - Jia‐jian Chen
- Department of Breast SurgeryFudan University Shanghai Cancer CenterNo. 270, Dongan RdShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghaiChina
| | - Jiong Wu
- Department of Breast SurgeryFudan University Shanghai Cancer CenterNo. 270, Dongan RdShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghaiChina
- Collaborative Innovation Center for Cancer MedicineShanghaiChina
| |
Collapse
|
28
|
Sorrentino L, Sartani A, Bossi D, Amadori R, Nebuloni M, Truffi M, Bonzini M, Riggio E, Foschi D, Corsi F. Sentinel node biopsy in ductal carcinoma in situ of the breast: Never justified? Breast J 2017; 24:325-333. [PMID: 29024241 DOI: 10.1111/tbj.12928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/24/2016] [Accepted: 06/16/2016] [Indexed: 11/29/2022]
Abstract
Sentinel lymph node biopsy for ductal carcinoma in situ (DCIS) of the breast is not standard of care. However, nodal involvement for DCIS patients is reported. Aim of our study was to identify preoperative features predictive of nodal involvement in DCIS patients. We have retrospectively reviewed 175 patients with a preoperative diagnosis of DCIS following a vacuum-assisted breast biopsy, and undergoing surgery with sentinel node biopsy. Variables distribution was compared between patients upstaged to invasive cancer at final pathology and patients with a confirmed DCIS, and between positive vs negative sentinel node patients. Univariate and multivariate analyses were performed for risk of a positive node. Lymph node biopsy was positive in 13 (7.4%) patients, with 8 (61.5%) macrometastases and 5 (38.5%) micrometastases. In these patients, Breast Imaging Reporting and Data System (BI-RADS) index >4 (OR 4.69, 95% CI 1.282-17.224, P = .02), lesion extension ≥20 mm (OR 4.25, 95% CI 1.255-14.447, P = .02), multifocal disease (OR 4.12, 95% CI 0.987-17.174, P = .05), comedo type (OR 3.54, 95% CI 1.044-11.969, P = .04), and upstaging (OR 4.56, 95% CI 1.080-19.249, P = .04) were all predictive of nodal involvement, although upstaging could not be predicted preoperatively. By multivariate analysis, the only independent factor predictive for positive sentinel node was multifocal disease (OR 5.14, 95% CI 1.015-26.066, P < .05). A preoperative diagnosis of DCIS, also including advanced biopsy systems such as vacuum-assisted breast biopsy, may be not always sufficient to exclude patients from sentinel node biopsy. DCIS patients with associated BI-RADS >4, lesion extension ≥20 mm, comedo type, and above all multifocal disease should be considered for axillary evaluation.
Collapse
Affiliation(s)
- Luca Sorrentino
- Surgery Division, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milano, Italy
| | - Alessandra Sartani
- Surgery Division, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milano, Italy
| | - Daniela Bossi
- Surgery Department, Breast Unit, ICS Maugeri S.p.A. SB, Pavia, Italy
| | - Rosella Amadori
- Surgery Department, Breast Unit, ICS Maugeri S.p.A. SB, Pavia, Italy
| | - Manuela Nebuloni
- Service of Pathology, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milano, Italy.,Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, Milano, Italy
| | - Marta Truffi
- Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, Milano, Italy
| | - Matteo Bonzini
- Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Eliana Riggio
- Surgery Division, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milano, Italy
| | - Diego Foschi
- Surgery Division, ASST Fatebenefratelli-Sacco, Luigi Sacco University Hospital, Milano, Italy.,Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, Milano, Italy
| | - Fabio Corsi
- Surgery Department, Breast Unit, ICS Maugeri S.p.A. SB, Pavia, Italy.,Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, Milano, Italy
| |
Collapse
|
29
|
Lambein K, Van Bockstal M, Vandemaele L, Van den Broecke R, Cocquyt V, Geenen S, Denys H, Libbrecht L. Comparison of HER2 amplification status among breast cancer subgroups offers new insights in pathways of breast cancer progression. Virchows Arch 2017; 471:575-587. [PMID: 28567637 DOI: 10.1007/s00428-017-2161-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/09/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022]
Abstract
Although the prognostic and predictive significance of human epidermal growth factor receptor 2 (HER2) in invasive breast cancer is well established, its role in ductal carcinoma in situ (DCIS) remains unclear. Reports on combined evaluation of both HER2 protein expression and HER2 amplification status in pure DCIS and DCIS adjacent to invasive ductal carcinoma (i.e., admixed DCIS) are scarce. In this study, immunohistochemistry and fluorescence in situ hybridization (FISH) were used to assess HER2 status in 72 cases of pure DCIS, 73 cases of DCIS admixed with invasive ductal carcinoma (IDC), and 60 cases of pure IDC. HER2 copy number-based amplification was present in 49% of pure DCIS, 16% of admixed DCIS, 18% of admixed IDC, and 8% of pure IDC. Amplified pure DCIS with clusters of HER2 signals showed a significantly lower HER2 copy number than amplified admixed DCIS with clusters. Whereas pure DCIS and admixed DCIS presented significant differences, the in situ and invasive component of admixed tumors showed striking similarities regarding mean HER2 and chromosome 17 centromere (CEP17) copy number, grade, and estrogen and progesterone receptor expression. The discrepant prevalence of HER2 amplification among breast cancer subgroups indirectly suggests that HER2 may not play a crucial role in the transition of in situ to invasive breast cancer. The similarities in HER2 amplification status between the in situ and invasive component of admixed tumors hint at a common biological pathway for both components. Our data support the theory that pure DCIS, pure IDC, and admixed lesions have a common progenitor, but can progress as separate lineages.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Disease Progression
- Female
- Gene Amplification
- Humans
- Middle Aged
- Receptor, ErbB-2/genetics
Collapse
Affiliation(s)
- Kathleen Lambein
- Department of Pathology, AZ St Lucas Hospital, Groenebriel 1, 9000, Ghent, Belgium
- Department of Oncology, KU Leuven, Surgical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Mieke Van Bockstal
- Department of Medical and Forensic Pathology, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lies Vandemaele
- Department of Medical and Forensic Pathology, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Rudy Van den Broecke
- Department of Gynaecology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Veronique Cocquyt
- Department of Medical Oncology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Sofie Geenen
- Department of Medical and Forensic Pathology, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Hannelore Denys
- Department of Medical Oncology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Louis Libbrecht
- Department of Medical and Forensic Pathology, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium.
- Department of Pathology, University Clinics St Luc, Hippokrateslaan 10, 1200, Sint-Lambrechts-Woluwe, Belgium.
| |
Collapse
|
30
|
Watanabe T, Yamaguchi T, Tsunoda H, Kaoku S, Tohno E, Yasuda H, Ban K, Hirokaga K, Tanaka K, Umemoto T, Okuno T, Fujimoto Y, Nakatani S, Ito J, Ueno E. Ultrasound Image Classification of Ductal Carcinoma In Situ (DCIS) of the Breast: Analysis of 705 DCIS Lesions. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:918-925. [PMID: 28242086 DOI: 10.1016/j.ultrasmedbio.2017.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 06/06/2023]
Abstract
The Japan Association of Breast and Thyroid Sonology (JABTS) proposed, in 2003, a conceptual classification system for non-mass abnormalities to be applied in addition to the conventional concept of masses, to facilitate detecting ductal carcinoma in situ (DCIS) lesions. The aim of this study was to confirm the utility of this system and to clarify the distribution of these findings in DCIS lesions. Data on 705 surgically treated DCIS lesions from 16 institutions in Japan were retrospectively reviewed. All 705 DCIS lesions could be classified according to the JABTS classification system. The most frequent findings were hypo-echoic areas in the mammary gland (48.6%), followed by solid masses (28.0%) and duct abnormalities (10.2%) or mixed masses (8.1%). Distortion (1.3%), clustered microcysts (1.4%) and echogenic foci without a hypo-echoic area (2.5%) were uncommon. These results suggest that the concept of non-mass abnormalities is useful in detecting DCIS lesions.
Collapse
Affiliation(s)
- Takanori Watanabe
- Department of Breast Surgery, National Hospital Organization Sendai Medical Center, Sendai, Miyagi, Japan.
| | - Takuhiro Yamaguchi
- Clinical Research Data Center, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Hiroko Tsunoda
- Department of Radiology Diagnostic Breast Imaging, St Luke's International Hospital, Chuo-ku, Tokyo, Japan
| | - Setsuko Kaoku
- Department of Surgical Pathology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eriko Tohno
- Tsukuba International Breast Clinic, Tsukuba, Ibaraki, Japan
| | - Hidemitsu Yasuda
- Department of Breast Surgery, National Center for Global Health and Medicine, Shinjuku-Ku, Tokyo, Japan
| | - Kanako Ban
- Department of Cancer Detection and Diagnosis, Tokyo Health Service Association, Shinjuku-ku, Tokyo, Japan
| | - Koichi Hirokaga
- Department of Breast Surgery, Hyogo Cancer Center, Akashi, Hyogo, Japan
| | - Kumiko Tanaka
- Breast Surgery, Shonan Kamakura General Hospital, Kamakura-shi, Kanagawa, Japan
| | - Takeshi Umemoto
- Tsukuba International Breast Clinic, Tsukuba, Ibaraki, Japan
| | - Toshitaka Okuno
- Breast Surgery, Nishi-kobe Medical Center, Nishi-ku, Kobe City, Japan
| | | | - Shuichi Nakatani
- Department of Breast Surgery, Minami Osaka General Hospital, Higashi-Kagaya Suminoe-ku, Osaka City, Japan
| | - Jun Ito
- Department of Surgery I, Dokkyo Medical University, Mibu-Machi, Tochigi, Japan
| | - Ei Ueno
- Tsukuba International Breast Clinic, Tsukuba, Ibaraki, Japan
| |
Collapse
|
31
|
Lee JY, Chaudhuri O. Regulation of Breast Cancer Progression by Extracellular Matrix Mechanics: Insights from 3D Culture Models. ACS Biomater Sci Eng 2017; 4:302-313. [DOI: 10.1021/acsbiomaterials.7b00071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Joanna Y. Lee
- Department of Mechanical
Engineering, Stanford University, 452 Escondido Mall, Building 520,
Room 226, Stanford, California 94305-4038, United States
| | - Ovijit Chaudhuri
- Department of Mechanical
Engineering, Stanford University, 452 Escondido Mall, Building 520,
Room 226, Stanford, California 94305-4038, United States
| |
Collapse
|
32
|
Groen EJ, Elshof LE, Visser LL, Rutgers EJT, Winter-Warnars HA, Lips EH, Wesseling J. Finding the balance between over- and under-treatment of ductal carcinoma in situ (DCIS). Breast 2017; 31:274-283. [DOI: 10.1016/j.breast.2016.09.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/19/2016] [Accepted: 09/01/2016] [Indexed: 12/21/2022] Open
|
33
|
Ong MS, Mandl KD. National expenditure for false-positive mammograms and breast cancer overdiagnoses estimated at $4 billion a year. Health Aff (Millwood) 2016; 34:576-83. [PMID: 25847639 DOI: 10.1377/hlthaff.2014.1087] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Populationwide mammography screening has been associated with a substantial rise in false-positive mammography findings and breast cancer overdiagnosis. However, there is a lack of current data on the associated costs in the United States. We present costs due to false-positive mammograms and breast cancer overdiagnoses among women ages 40-59, based on expenditure data from a major US health care insurance plan for 702,154 women in the years 2011-13. The average expenditures for each false-positive mammogram, invasive breast cancer, and ductal carcinoma in situ in the twelve months following diagnosis were $852, $51,837 and $12,369, respectively. This translates to a national cost of $4 billion each year. The costs associated with false-positive mammograms and breast cancer overdiagnoses appear to be much higher than previously documented. Screening has the potential to save lives. However, the economic impact of false-positive mammography results and breast cancer overdiagnoses must be considered in the debate about the appropriate populations for screening.
Collapse
Affiliation(s)
- Mei-Sing Ong
- Mei-Sing Ong is a research fellow at Boston Children's Hospital, in Massachusetts, and a research fellow at the Australian Institute of Health Innovation, Macquarie University, in Sydney, Australia
| | - Kenneth D Mandl
- Kenneth D. Mandl is a professor at Harvard Medical School and director of the Children's Hospital Informatics Program at Boston Children's Hospital
| |
Collapse
|
34
|
Campbell MJ, Baehner F, O'Meara T, Ojukwu E, Han B, Mukhtar R, Tandon V, Endicott M, Zhu Z, Wong J, Krings G, Au A, Gray JW, Esserman L. Characterizing the immune microenvironment in high-risk ductal carcinoma in situ of the breast. Breast Cancer Res Treat 2016; 161:17-28. [PMID: 27785654 DOI: 10.1007/s10549-016-4036-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 02/04/2023]
Abstract
PURPOSE The recent increase in the incidence of ductal carcinoma in situ (DCIS) has sparked debate over the classification and treatment of this disease. Although DCIS is considered a precursor lesion to invasive breast cancer, some DCIS may have more or less risk than is realized. In this study, we characterized the immune microenvironment in DCIS to determine if immune infiltrates are predictive of recurrence. METHODS Fifty-two cases of high-grade DCIS (HG-DCIS), enriched for large lesions and a history of recurrence, were age matched with 65 cases of non-high-grade DCIS (nHG-DCIS). Immune infiltrates were characterized by single- or dual-color staining of FFPE sections for the following antigens: CD4, CD8, CD20, FoxP3, CD68, CD115, Mac387, MRC1, HLA-DR, and PCNA. Nuance multispectral imaging software was used for image acquisition. Protocols for automated image analysis were developed using CellProfiler. Immune cell populations associated with risk of recurrence were identified using classification and regression tree analysis. RESULTS HG-DCIS had significantly higher percentages of FoxP3+ cells, CD68+ and CD68+PCNA+ macrophages, HLA-DR+ cells, CD4+ T cells, CD20+ B cells, and total tumor infiltrating lymphocytes compared to nHG-DCIS. A classification tree, generated from 16 immune cell populations and 8 clinical parameters, identified three immune cell populations associated with risk of recurrence: CD8+HLADR+ T cells, CD8+HLADR- T cells, and CD115+ cells. CONCLUSION These findings suggest that the tumor immune microenvironment is an important factor in identifying DCIS cases with the highest risk for recurrence and that manipulating the immune microenvironment may be an efficacious strategy to alter or prevent disease progression.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/therapy
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/mortality
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Combined Modality Therapy
- Female
- Humans
- Lymphocyte Count
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Lymphocyte Subsets/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Middle Aged
- Neoplasm Grading
- Neoplasm Recurrence, Local
- Neoplasm Staging
- Patient Outcome Assessment
- Prognosis
- Tumor Burden
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Michael J Campbell
- Department of Surgery, University of California, 2340 Sutter St, N321, San Francisco, CA, 94115, USA.
| | - Frederick Baehner
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Tess O'Meara
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| | - Ekene Ojukwu
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| | - Booyeon Han
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| | - Rita Mukhtar
- Department of Surgery, University of California, 2340 Sutter St, N321, San Francisco, CA, 94115, USA
| | - Vickram Tandon
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| | - Max Endicott
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| | - Zelos Zhu
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| | - Jasmine Wong
- Department of Surgery, University of California, 2340 Sutter St, N321, San Francisco, CA, 94115, USA
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Alfred Au
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Joe W Gray
- Oregon Health and Science University, Portland, OR, USA
| | - Laura Esserman
- Department of Surgery, University of California, 2340 Sutter St, N321, San Francisco, CA, 94115, USA
- Mt Zion Carol Franc Buck Breast Care Center, University of California, San Francisco, CA, USA
| |
Collapse
|
35
|
Molecular classification of non-invasive breast lesions for personalised therapy and chemoprevention. Oncotarget 2016; 6:43244-54. [PMID: 26657114 PMCID: PMC4791229 DOI: 10.18632/oncotarget.6525] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 11/27/2015] [Indexed: 12/29/2022] Open
Abstract
Breast cancer screening has led to a dramatic increase in the detection of pre-invasive breast lesions. While mastectomy is almost guaranteed to treat the disease, more conservative approaches could be as effective if patients can be stratified based on risk of co-existing or recurrent invasive disease.Here we use a range of biomarkers to interrogate and classify purely non-invasive lesions (PNL) and those with co-existing invasive breast cancer (CEIN). Apart from Ductal Carcinoma In Situ (DCIS), relative homogeneity is observed. DCIS contained a greater spread of molecular subtypes. Interestingly, high expression of p-mTOR was observed in all PNL with lower expression in DCIS and invasive carcinoma while the opposite expression pattern was observed for TOP2A.Comparing PNL with CEIN, we have identified p53 and Ki67 as predictors of CEIN with a combined PPV and NPV of 90.48% and 43.3% respectively. Furthermore, HER2 expression showed the best concordance between DCIS and its invasive counterpart.We propose that these biomarkers can be used to improve the management of patients with pre-invasive breast lesions following further validation and clinical trials. p53 and Ki67 could be used to stratify patients into low and high-risk groups for co-existing disease. Knowledge of expression of more actionable targets such as HER2 or TOP2A can be used to design chemoprevention or neo-adjuvant strategies. Increased knowledge of the molecular profile of pre-invasive lesions can only serve to enhance our understanding of the disease and, in the era of personalised medicine, bring us closer to improving breast cancer care.
Collapse
|
36
|
Axillary evaluation and lymphedema in women with ductal carcinoma in situ. Breast Cancer Res Treat 2016; 158:373-84. [PMID: 27365080 DOI: 10.1007/s10549-016-3890-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/25/2016] [Indexed: 10/21/2022]
Abstract
Axillary evaluation in women with ductal carcinoma in situ (DCIS) is increasing; however, this may introduce additional morbidity with unclear benefit. Our objective was to examine the morbidity and mortality associated with axillary evaluation in DCIS. We conducted a retrospective cohort study of 10,504 women aged 65-90 years with DCIS who underwent breast conserving surgery between 2002 and 2012 using SEER-Medicare database. Patients were categorized by receipt of axillary evaluation with either sentinel lymph node biopsy (SLNB) or axillary node dissection (ALND). We determined the incidence of lymphedema treatment as defined by diagnostic and procedural codes, as well as 10-year breast cancer-specific and all-cause mortality. 18.3 % of those treated with BCS and 69.4 % of those treated with mastectomy had an axillary evaluation. One year after treatment, 8.2 % of women who had an axillary evaluation developed lymphedema, compared to 5.9 % of those who did not. In a multivariable Cox proportional hazard model, the incidence of lymphedema was higher among those who underwent axillary evaluation (HR 1.22, 95 % CI 1.04-1.45). Overall 10-year breast cancer-specific survival was similar between both groups (HR 0.83, 95 % CI 0.40-1.74). Only 44 (0.40 %) women died of breast cancer; receipt of axillary evaluation did not alter overall survival. Axillary evaluation is commonly performed in women with DCIS, especially those undergoing mastectomy. However, women who receive an axillary evaluation have higher rates of lymphedema, without breast cancer-specific or overall survival benefit. Efforts should be made to determine the population of women with DCIS who benefit from this procedure.
Collapse
|
37
|
Van Bockstal M, Libbrecht L, Floris G, Lambein K. The Baader-Meinhof phenomenon in ductal carcinoma in situ of the breast. Histopathology 2016; 69:522-3. [PMID: 27028714 DOI: 10.1111/his.12977] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mieke Van Bockstal
- Department of Pathology, Ghent University, Ghent, Belgium.,Department of Medical and Forensic Pathology, Ghent University, Ghent, Belgium
| | - Louis Libbrecht
- Department of Pathology, University Clinics Saint Luc, Brussels, Belgium
| | - Giuseppe Floris
- Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, University of Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Kathleen Lambein
- Department of Pathology, AZ St Lucas Hospital, Ghent, Belgium.,Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Mardekian SK, Bombonati A, Palazzo JP. Ductal carcinoma in situ of the breast: the importance of morphologic and molecular interactions. Hum Pathol 2015; 49:114-23. [PMID: 26826418 DOI: 10.1016/j.humpath.2015.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/02/2015] [Accepted: 11/05/2015] [Indexed: 12/21/2022]
Abstract
Ductal carcinoma in situ (DCIS) of the breast is a lesion characterized by significant heterogeneity, in terms of morphology, immunohistochemical staining, molecular signatures, and clinical expression. For some patients, surgical excision provides adequate treatment, but a subset of patients will experience recurrence of DCIS or progression to invasive ductal carcinoma (IDC). Recent years have seen extensive research aimed at identifying the molecular events that characterize the transition from normal epithelium to DCIS and IDC. Tumor epithelial cells, myoepithelial cells, and stromal cells undergo alterations in gene expression, which are most important in the early stages of breast carcinogenesis. Epigenetic modifications, such as DNA methylation, together with microRNA alterations, play a major role in these genetic events. In addition, tumor proliferation and invasion is facilitated by the lesional microenvironment, which includes stromal fibroblasts and macrophages that secrete growth factors and angiogenesis-promoting substances. Characterization of DCIS on a molecular level may better account for the heterogeneity of these lesions and how this manifests as differences in patient outcome and response to therapy. Molecular assays originally developed for assessing likelihood of recurrence in IDC are recently being applied to DCIS, with promising results. In the future, the classification of DCIS will likely incorporate molecular findings along with histologic and immunohistochemical features, allowing for personalized prognostic information and therapeutic options for patients with DCIS. This review summarizes current data regarding the molecular characterization of DCIS and discusses the potential clinical relevance.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biopsy
- Breast Neoplasms/chemistry
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma/chemistry
- Carcinoma/genetics
- Carcinoma/pathology
- Carcinoma, Intraductal, Noninfiltrating/chemistry
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Disease Progression
- Epigenesis, Genetic
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Humans
- Immunohistochemistry
- Mastectomy
- Molecular Diagnostic Techniques
- Neoplasm Recurrence, Local
- Phenotype
- Predictive Value of Tests
- Reproducibility of Results
- Treatment Outcome
Collapse
Affiliation(s)
- Stacey K Mardekian
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107.
| | - Alessandro Bombonati
- Department of Pathology, Albert Einstein Medical Center, Philadelphia, PA 19141.
| | - Juan P Palazzo
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107.
| |
Collapse
|
39
|
Subhedar P, Olcese C, Patil S, Morrow M, Van Zee KJ. Decreasing Recurrence Rates for Ductal Carcinoma In Situ: Analysis of 2996 Women Treated with Breast-Conserving Surgery Over 30 Years. Ann Surg Oncol 2015. [PMID: 26215193 DOI: 10.1245/s10434-015-4740-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Randomized trials of radiation after breast-conserving surgery (BCS) for ductal carcinoma in situ (DCIS) found substantial rates of recurrence, with half of the recurrences being invasive. Decreasing local recurrence rates for invasive breast carcinoma have been observed and are largely attributed to improvements in systemic therapy. In this study, we examine recurrence rates after BCS for DCIS over 3 decades at one institution. METHODS We retrospectively reviewed a prospectively maintained database of DCIS patients undergoing BCS from 1978 to 2010. Cox proportional hazard models were used to investigate the association between the treatment period and recurrence, controlling for other variables. RESULTS Overall, 363 (12%) recurrences among 2996 cases were observed. Median follow-up for patients without recurrence was 75 months (range 0-30 years); 732 patients were followed for ≥10 years. The 5-year recurrence rate for the period 1978-1998 was 13.6 versus 6.6% for the period 1999-2010 [hazard ratio (HR) 0.62, p < 0.0001]. Controlling for age, family history, presentation, nuclear grade, necrosis, number of excisions, margin status, radiation, and endocrine therapy, treatment period remained significantly associated with recurrence, with later years associated with a lower HR (0.74, p = 0.02) compared to earlier. After stratification by radiation use, association of recurrence with treatment period persisted in those treated without radiation (HR 0.62, p = 0.003). CONCLUSIONS Recurrence rates for DCIS have fallen over time, with increases in screen detection, negative margins, and use of adjuvant therapies only partially explaining this decrease. The unexplained decline persists in women not receiving radiation, suggesting it is not due to changes in radiation efficacy but may be due to improvements in radiologic detection and pathologic assessment.
Collapse
Affiliation(s)
- Preeti Subhedar
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | |
Collapse
|
40
|
Van Bockstal M, Libbrecht L, De Wever O. Fibroblast-induced matrix remodeling paves the path for invasion. Cell Cycle 2015; 14:793-4. [PMID: 25790091 PMCID: PMC4615059 DOI: 10.1080/15384101.2015.1010968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Mieke Van Bockstal
- a Department of Pathology ; Ghent University and Ghent University Hospital ; Ghent , Belgium
| | | | | |
Collapse
|
41
|
Perez AA, Balabram D, Rocha RM, da Silva Souza Á, Gobbi H. Co-Expression of p16, Ki67 and COX-2 Is Associated with Basal Phenotype in High-Grade Ductal Carcinoma In Situ of the Breast. J Histochem Cytochem 2015; 63:408-16. [PMID: 25711229 DOI: 10.1369/0022155415576540] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 02/10/2015] [Indexed: 12/21/2022] Open
Abstract
We assessed the co-expression of cell cycle-related biomarkers in a series of 121 consecutive cases of high-grade ductal carcinoma in situ (DCIS), pure or associated with invasive carcinoma, and their associations with the different immunoprofiles of DCIS. Cases were identified from the histopathology files of the Breast Pathology Laboratory, Federal University of Minas Gerais, Brazil, from 2003 to 2008. The expression of estrogen receptor, progesterone receptor, HER2 overexpression, cytokeratin 5, epidermal growth factor receptor 1, cyclooxygenase-2, p16 and Ki67 were assessed. Tumors were placed into five subgroups according to their immunohistochemical profile: luminal A, luminal B, HER2, basal-like and "not classified". We found that the basal phenotype was associated with a higher frequency of p16-positive cases (83%) and the luminal A phenotype showed a higher frequency of p16-negative cases (93%; p=0.000). The association of biomarkers p16(+)/Ki67(+)/COX2(+) was expressed in 02/06 cases (33.3%) of the basal phenotype but in only 01/70 cases (1.4%) of the luminal A phenotype (p=0.01). The co-expression of p16(+)/Ki67(+)/COX2(-) was associated with a basal phenotype (p=0.004). P16 expression, p16(+)/Ki67(+)/COX2(+) and p16(+)/Ki67(+)/COX2(-) co-expression showed significant associations with the basal phenotype and these profiles could be used to guide more aggressive treatment strategies in patients with high-grade DCIS.
Collapse
Affiliation(s)
- Amanda Arantes Perez
- Breast Pathology Laboratory, School of Medicine, Federal University of Minas Gerais, Brazil (AAP, DB, ADSS, HG)
| | - Débora Balabram
- Breast Pathology Laboratory, School of Medicine, Federal University of Minas Gerais, Brazil (AAP, DB, ADSS, HG)
| | | | - Átila da Silva Souza
- Breast Pathology Laboratory, School of Medicine, Federal University of Minas Gerais, Brazil (AAP, DB, ADSS, HG)
| | - Helenice Gobbi
- Breast Pathology Laboratory, School of Medicine, Federal University of Minas Gerais, Brazil (AAP, DB, ADSS, HG)
| |
Collapse
|
42
|
Reyes-Uribe E, Departamento de Biologia Celular, Cinvestav-IPN, Av IPN # 2508, San Pedro Zacatenco, DF 07360, Mexico, Serna-Marquez N, Perez Salazar E. DDRs: receptors that mediate adhesion, migration and invasion in breast cancer cells. AIMS BIOPHYSICS 2015. [DOI: 10.3934/biophy.2015.3.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
43
|
Ductal carcinoma in situ of the breast: correlation between histopathological features and age of patients. Diagn Pathol 2014; 9:227. [PMID: 25471940 PMCID: PMC4260240 DOI: 10.1186/s13000-014-0227-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/20/2014] [Indexed: 12/21/2022] Open
Abstract
Background The histopathological subtype, nuclear grade and presence or absence of comedonecrosis are established as critical elements in the reporting of ductal carcinoma in situ (DCIS) of the breast. The aims of this study were to determine the frequencies of morphological subtypes of DCIS, nuclear grade and comedonecrosis; to compare the age of patients with the histopathological characteristics of DCIS, and to assess the agreement of grade between in situ and invasive components in DCIS cases that were associated with invasive carcinoma. Methods We evaluated a series of 403 cases of DCIS, pure or associated with invasive mammary carcinoma, consecutively identified from the histopathology files of the Breast Pathology Laboratory, Federal University of Minas Gerais, Brazil, from 2003 to 2008. Results DCIS displayed a single growth pattern in most cases (55.1%) and the solid subtype was the most common morphology (42.2% of the total). High-grade DCIS was identified in 293/403 cases (72.7%) and comedonecrosis was present in 222/403 cases (55%). Among DCIS with a single architectural pattern, high grade was more common in the solid subtype (151/168 cases, 89.9%; p < 0.001). Only 32% of tumours with a cribriform pattern had high nuclear grade. Comedonecrosis was more common in the solid morphology than in the cribriform, papillary and micropapillary subtypes (p < 0.001). Patients with high-grade DCIS were younger in relation to patients with low-grade DCIS (p = 0.027) and patients with tumours with comedonecrosis were also younger in comparison to patients with tumours without comedonecrosis (p = 0.003). Fair agreement was observed between in situ and invasive components with regard to grade (weighted kappa = 0.23). Conclusions The high nuclear grade and the presence of comedonecrosis were identified more frequently in younger patients and more often correlated with the solid pattern of DCIS. Virtual Slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/13000_2014_227
Collapse
|
44
|
Toesca A, Botteri E, Lazzeroni M, Vila J, Manika A, Ballardini B, Bettarini F, Guerrieri-Gonzaga A, Bonanni B, Rotmensz N, Viale G, Veronesi P, Luini A, Veronesi U, Gentilini O. Breast conservative surgery for well-differentiated ductal intraepithelial neoplasia: Risk factors for ipsilateral breast tumor recurrence. Breast 2014; 23:829-35. [DOI: 10.1016/j.breast.2014.08.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 08/11/2014] [Accepted: 08/31/2014] [Indexed: 11/17/2022] Open
|
45
|
Shigematsu H, Kadoya T, Masumoto N, Matsuura K, Emi A, Kajitani K, Amioka A, Okada M. Role of FDG-PET/CT in prediction of underestimation of invasive breast cancer in cases of ductal carcinoma in situ diagnosed at needle biopsy. Clin Breast Cancer 2014; 14:358-64. [PMID: 24962555 DOI: 10.1016/j.clbc.2014.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 03/28/2014] [Accepted: 04/23/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the significance of FDG-PET/CT for predicting the underestimation of invasive breast cancer in cases of DCIS at needle biopsy. PATIENTS AND METHODS Of 83 consecutive cases with diagnoses of DCIS at primary needle biopsy who underwent curative surgery between 2010 and 2013, the association between the SUVmax on FDG-PET/CT before excision and the underestimation of invasive breast cancer was examined. RESULTS There were 29 (34.9%) cases diagnosed to have invasive breast cancer at excision. Receiver operating characteristics curve analysis showed the cutoff value of the SUVmax to predict underestimation of invasive breast cancer was 1.6. The rates of underestimation were 61.5% for patients with a tumor of SUVmax > 1.6 and 11.4% for patients with a tumor of SUVmax ≤ 1.6 (P < .001). A high value of SUVmax was significantly associated with symptomatic presentation (P < .001), palpability (P < .001), mass formation (P = .013), high Breast Imaging Reporting and Data System category (P = .011), and core needle biopsy (P = .007). In multivariate analysis, high SUVmax was only a significant predictive factor of underestimation of invasive breast cancer (hazard ratio, 11.7; 95% confidence interval, 3.70-37.0; P < .001). CONCLUSION SUVmax on FDG-PET/CT is useful for predicting the underestimation of invasive breast cancer in cases of DCIS at needle biopsy.
Collapse
Affiliation(s)
- Hideo Shigematsu
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takayuki Kadoya
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Norio Masumoto
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kazuo Matsuura
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Akiko Emi
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Keiko Kajitani
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ai Amioka
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
46
|
Abstract
We have previously indicated that the ideal animal tumor model should mimic the human disease. This means that the investigator should be able to ascertain the influence of host factors on the initiation of tumorigenesis, mimic the susceptibility of tumor response based on age and reproductive history, and determine the response of the tumors induced to chemotherapy. The utilization of experimental models of mammary carcinogenesis in risk assessment requires that the influence of ovarian, pituitary, and placental hormones, among others, as well as overall reproductive events are taken into consideration, since they are important modifiers of the susceptibility of the organ to neoplastic development. Several species, such as rodents, dogs, cats, and monkeys, have been evaluated for these purposes; however, none of them fulfills all the criteria specified previously. Rodents, however, are the most widely used models; therefore, this work will concentrate on discussing the rat rodent model of mammary carcinogenesis.
Collapse
Affiliation(s)
- Jose Russo
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Omranipour R, Alipour S, Hadji M, Bagheri K. Two Decades of Experience with Ductal Carcinoma in Situ of the Breast in the Cancer Institute of Tehran, Iran. Asian Pac J Cancer Prev 2014; 15:2771-6. [DOI: 10.7314/apjcp.2014.15.6.2771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
48
|
Lim YJ, Kim K, Chie EK, Han W, Noh DY, Ha SW. Treatment outcome of ductal carcinoma in situ patients treated with postoperative radiation therapy. Radiat Oncol J 2014; 32:1-6. [PMID: 24724045 PMCID: PMC3977126 DOI: 10.3857/roj.2014.32.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 11/22/2022] Open
Abstract
Purpose To evaluate the outcome of ductal carcinoma in situ (DCIS) patients who underwent surgery followed by radiation therapy (RT). Materials and Methods We retrospectively reviewed 106 DCIS patients who underwent surgery followed by postoperative RT between 1994 and 2006. Ninety-four patients underwent breast-conserving surgery, and mastectomy was performed in 12 patients due to extensive DCIS. Postoperative RT was delivered to whole breast with 50.4 Gy/28 fx. Tumor bed boost was offered to 7 patients (6.6%). Patients with hormonal receptor-positive tumors were treated with hormonal therapy. Results The median follow-up duration was 83.4 months (range, 33.4 to 191.5 months) and the median age was 47.8 years. Ten patients (9.4%) had resection margin <1 mm and high-grade and estrogen receptor-negative tumors were observed in 39 (36.8%) and 20 (18.9%) patients, respectively. The 7-year ipsilateral breast tumor recurrence (IBTR)-free survival rate was 95.3%. Resection margin (<1 or ≥1 mm) was the significant prognostic factor for IBTR in univariate and multivariate analyses (p < 0.001 and p = 0.016, respectively). Conclusion Postoperative RT for DCIS can achieve favorable treatment outcome. Resection margin was the important prognostic factor for IBTR in the DCIS patients who underwent postoperative RT.
Collapse
Affiliation(s)
- Yu Jin Lim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyubo Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | - Eui Kyu Chie
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea. ; Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Young Noh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Sung W Ha
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea. ; Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
49
|
Rewiring cell polarity signaling in cancer. Oncogene 2014; 34:939-50. [PMID: 24632617 DOI: 10.1038/onc.2014.59] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 02/08/2023]
Abstract
Disrupted cell polarity is a feature of epithelial cancers. The Crumbs, Par and Scribble polarity complexes function to specify and maintain apical and basolateral membrane domains, which are essential to organize intracellular signaling pathways that maintain epithelial homeostasis. Disruption of apical-basal polarity proteins facilitates rewiring of oncogene and tumor suppressor signaling pathways to deregulate proliferation, apoptosis, invasion and metastasis. Moreover, apical-basal polarity integrates intracellular signaling with the microenvironment by regulating metabolic signaling, extracellular matrix remodeling and tissue level organization. In this review, we discuss recent advances in our understanding of how polarity proteins regulate diverse signaling pathways throughout cancer progression from initiation to metastasis.
Collapse
|
50
|
Caballero OL, Shousha S, Zhao Q, Simpson AJG, Coombes RC, Neville AM. Expression of Cancer/Testis genes in ductal carcinoma in situ and benign lesions of the breast. Oncoscience 2013; 1:14-20. [PMID: 25593980 PMCID: PMC4295763 DOI: 10.18632/oncoscience.4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 12/10/2013] [Indexed: 01/22/2023] Open
Abstract
Cancer/testis (CT) genes represent a unique class of genes, which are expressed by germ cells, normally silenced in somatic cells, but activated in various cancers. CT proteins can elicit spontaneous immune responses in cancer patients and this feature makes them attractive targets for immunotherapy-based approaches. We have previously reported that CTs are relatively commonly expressed in estrogen receptor (ER) negative, high risk carcinomas. In this study, we examined the expression of selected CT genes in ductal carcinoma in situ (DCIS), lobular carcinoma in situ (LCIS) and benign proliferative lesions of the breast. ER negative DCIS were found to be associated with significant CT gene expression together with HER2 positivity and a marked stromal immune response.
Collapse
Affiliation(s)
- Otavia L Caballero
- Ludwig Collaborative Laboratory, Ludwig Institute for Cancer Research, Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Sami Shousha
- Imperial College Healthcare NHS Trust & Imperial College, London, Charing Cross Hospital
| | - Qi Zhao
- Ludwig Collaborative Laboratory, Ludwig Institute for Cancer Research, Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Andrew J G Simpson
- Ludwig Institute for Cancer Research, New York, NY., USA.,Current affiliation: Orygen Biotecnologia, São Paulo, Brazil
| | - R Charles Coombes
- Imperial College Healthcare NHS Trust & Imperial College, London, Charing Cross Hospital
| | | |
Collapse
|