1
|
Lee ES. Recent Advances in Malignant Phyllodes Tumors of the Breast. J Breast Cancer 2025; 28:51-60. [PMID: 40313121 PMCID: PMC12046350 DOI: 10.4048/jbc.2024.0284] [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: 11/18/2024] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Phyllodes tumors (PTs) represent an uncommon category of fibroepithelial neoplasms found in the breast tissue and are classified as benign, borderline, or malignant based on their histopathological features. Histological assessment remains the cornerstone of a PT diagnosis. However, recent genomic advancements have revealed the biological mechanisms underlying PTs, particularly malignant phyllodes tumors (MPTs). This comprehensive review integrates current genomic and molecular investigations that have elucidated the distinguishing features of PTs. Mutations in the MED12 gene represent early tumorigenic phenomena that are often observed in benign lesions, whereas modifications in the TERT promoter, alterations in TP53, and amplification of EGFR are associated with malignant transformation. Moreover, novel transcriptomic investigations have characterized discrete molecular subtypes exhibiting epithelial and fibrous attributes, thereby enriching our understanding of MPT heterogeneity. Actionable genomic modifications, including dysregulation of the PI3K/Akt/mTOR, MET, and IGF1R signaling cascades, represent promising directions for targeted therapeutic strategies; however, clinical validation is still insufficient. Advances in patient-derived models have created functional platforms conducive to drug screening and preclinical evaluation. Molecular examination has expanded our understanding of PTs, revealing the genomic components linked to malignant progression and identifying prospective therapeutic targets. Nevertheless, obstacles remain in the practical application of these discoveries, primarily owing to the intratumoral heterogeneity and rarity of MPTs. Future investigations should prioritize the integration of diverse omics methodologies, enhancement of preclinical testing frameworks, and establishment of global data-sharing initiatives to promote biomarker-driven treatment strategies.
Collapse
Affiliation(s)
- Eun-Shin Lee
- Department of Surgery, Korea University Hospital, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
2
|
Chen J, Liu L, Yang Y, Luo J, Liu S. Patient-derived organoid models of malignant phyllodes tumours for drug sensitivity testing and identification of targeted therapeutic strategies. J Drug Target 2025:1-11. [PMID: 40059613 DOI: 10.1080/1061186x.2025.2473010] [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: 10/18/2024] [Revised: 01/16/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Malignant phyllodes tumours (MPT) of the breast are rare fibroepithelial neoplasms. It exhibits rapid growth, large size, and a high local recurrence rate. METHODS In this study, we established novel patient-derived organoid (PDO) models from two primary MPT samples and conducted comprehensive genetic profiling and drug screening. RESULTS The PDO models faithfully recapped the histopathological and molecular features of the primary tumours, including stromal overgrowth, leaf-like projections, and the expression of key diagnostic markers. Drug testing revealed significant heterogeneity in response profiles to chemotherapeutic reagents between the two MPT-derived organoids, implying the importance of personalised drug testing. Next-generation sequencing analysis identified recurrent mutations in TP53, RB1, EGFR, ATM, and RECQL4, which correlated with the drug sensitivity profiles observed in the organoid models. Targeted therapeutic drugs, such as Abemaciclib (targeting the RB1 pathway) with an IC50 value of 1.744 µM, and Alflutinib Mesylate (targeting the EGFR pathway) with an IC50 value of 0.9150 µM, exhibited significant cytotoxic effects in the MPT2 organoid models. CONCLUSIONS This study highlights the novel application of PDOs for studying the molecular landscape of MPTs and identifying effective therapeutic targets, offering a promising platform for guiding personalised treatment strategies for this rare and challenging cancer.
Collapse
Affiliation(s)
- Jie Chen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Liangquan Liu
- Department of Breast Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunxu Yang
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, China
| | - Jing Luo
- Department of Breast Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Yan Y, Liu Y, Yao J, Sui L, Chen C, Jiang T, Liu X, Wang Y, Ou D, Chen J, Wang H, Feng L, Pan Q, Su Y, Wang Y, Wang L, Zhou L, Xu D. Deep learning-assisted distinguishing breast phyllodes tumours from fibroadenomas based on ultrasound images: a diagnostic study. Br J Radiol 2024; 97:1816-1825. [PMID: 39288312 DOI: 10.1093/bjr/tqae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/25/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES To evaluate the performance of ultrasound-based deep learning (DL) models in distinguishing breast phyllodes tumours (PTs) from fibroadenomas (FAs) and their clinical utility in assisting radiologists with varying diagnostic experiences. METHODS We retrospectively collected 1180 ultrasound images from 539 patients (247 PTs and 292 FAs). Five DL network models with different structures were trained and validated using nodule regions annotated by radiologists on breast ultrasound images. DL models were trained using the methods of transfer learning and 3-fold cross-validation. The model demonstrated the best evaluation index in the 3-fold cross-validation was selected for comparison with radiologists' diagnostic decisions. Two-round reader studies were conducted to investigate the value of DL model in assisting 6 radiologists with different levels of experience. RESULTS Upon testing, Xception model demonstrated the best diagnostic performance (area under the receiver-operating characteristic curve: 0.87; 95% CI, 0.81-0.92), outperforming all radiologists (all P < .05). Additionally, the DL model enhanced the diagnostic performance of radiologists. Accuracy demonstrated improvements of 4%, 4%, and 3% for senior, intermediate, and junior radiologists, respectively. CONCLUSIONS The DL models showed superior predictive abilities compared to experienced radiologists in distinguishing breast PTs from FAs. Utilizing the model led to improved efficiency and diagnostic performance for radiologists with different levels of experience (6-25 years of work). ADVANCES IN KNOWLEDGE We developed and validated a DL model based on the largest available dataset to assist in diagnosing PTs. This model has the potential to allow radiologists to discriminate 2 types of breast tumours which are challenging to identify with precision and accuracy, and subsequently to make more informed decisions about surgical plans.
Collapse
Affiliation(s)
- Yuqi Yan
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, Zhejiang 310022, China
| | - Yuanzhen Liu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Jincao Yao
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Lin Sui
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, Zhejiang 310022, China
| | - Chen Chen
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Tian Jiang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Xiaofang Liu
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Yifan Wang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Di Ou
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Jing Chen
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Hui Wang
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Lina Feng
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Qianmeng Pan
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Ying Su
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yukai Wang
- Zunyi Medical University, Zunyi 563000, China
| | - Liping Wang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Lingyan Zhou
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Dong Xu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| |
Collapse
|
4
|
Waitzberg ÂFL, Ferreira ENE, Pinilla M, Pineda P, Malinverni ACDM, Soares FA, Carraro DM. Are both distinct epithelial and stromal cells molecular analysis from phyllodes tumors versus fibroadenoma components affected in breast fibroepithelial progression? Acta Cir Bras 2023; 38:e386823. [PMID: 38055384 DOI: 10.1590/acb386823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/19/2023] [Indexed: 12/08/2023] Open
Abstract
PURPOSE To determine molecular events involved in the tumorigenesis of phyllodes tumors (PT) and the role of each stromal (SC) and epithelial (EC) cell. METHODS Frozen breast samples enriched with epithelial and stromal cells from three fibroadenomas and 14 PT were retrieved and laser microdissected. Sanger and polymerase chain reaction-based sequencing of exon 2 MED12 and TERT promoter hotspot mutations were performed; 44K microarray platform was used to analyze gene expression. RESULTS All three fibroadenomas (FAs) presented mutations in MED12, but not in TERT, whose mutation was observed in five of the 14 PTs. EC and SC of each affected tumor displayed identical alterations. Of the total differentially expressed genes (DEG) (EC = 1,543 and SC = 850), 984 were EC-eDEGs and 291 were SC-eDEGs. We found a high similarity of diseases and functions enriched by both cell types, but dissimilarity in the number of enriched canonical pathways. Three signaling canonical pathways overlapping with EC and SC were predicted to be activated in one cell type and inactivated in the other, while no overlap in eDEGs was assigned to them. We also identified 13 EC-eDEGs and five SC-eDEGs enriched networks, in which the SC-eDEGs were able to segregate FA from PT samples. CONCLUSIONS Identical TERT mutations from both SC and ES origins might affect the PTs tumorigenesis. Gene expression differences suggest coordinated molecular processes between these components with determinant differences acquired by SC, able to fully distinguish PTs from FAs lesions.
Collapse
Affiliation(s)
| | - Elisa Napolitano E Ferreira
- Universidade Federal de São Paulo - Paulista School of Medicine - Department of Pathology - São Paulo (SP), Brazil
| | - Mabel Pinilla
- Universidad de Concepción - Facultad de Medicina - Department of Medical Technology - Concepción, Chile
| | - Paulo Pineda
- Hospital A C Camargo - Genomics and Molecular Biology Group - São Paulo (SP), Brazil
| | - Andréa Cristina de Moraes Malinverni
- Universidade Federal de São Paulo - Paulista School of Medicine - Department of Pathology - São Paulo (SP), Brazil
- Universidade Federal de São Paulo - Laboratory of Molecular and Experimental Pathology I - São Paulo (SP), Brazil
| | | | - Dirce Maria Carraro
- Hospital A C Camargo - Genomics and Molecular Biology Group - São Paulo (SP), Brazil
| |
Collapse
|
5
|
Zhang M, Arjmandi FK, Porembka JH, Seiler SJ, Goudreau SH, Merchant K, Hwang H, Hayes JC. Imaging and Management of Fibroepithelial Lesions of the Breast: Radiologic-Pathologic Correlation. Radiographics 2023; 43:e230051. [PMID: 37856317 DOI: 10.1148/rg.230051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Fibroepithelial lesions (FELs) are among the most common breast masses encountered by breast radiologists and pathologists. They encompass a spectrum of benign and malignant lesions, including fibroadenomas (FAs) and phyllodes tumors (PTs). FAs are typically seen in young premenopausal women, with a peak incidence at 20-30 years of age, and have imaging features of oval circumscribed hypoechoic masses. Although some FA variants are especially sensitive to hormonal influences and can exhibit rapid growth (eg, juvenile FA and lactational adenomas), most simple FAs are slow growing and involute after menopause. PTs can be benign, borderline, or malignant and are more common in older women aged 40-50 years. PTs usually manifest as enlarging palpable masses and are associated with a larger size and sometimes with an irregular shape at imaging compared with FAs. Although FA and FA variants are typically managed conservatively unless large and symptomatic, PTs are surgically excised because of the risk of undersampling at percutaneous biopsy and the malignant potential of borderline and malignant PTs. As a result of the overlap in imaging and histologic appearances, FELs can present a diagnostic challenge for the radiologist and pathologist. Radiologists can facilitate accurate diagnosis by supplying adequate tissue sampling and including critical information for the pathologist at the time of biopsy. Understanding the spectrum of FELs can facilitate and guide appropriate radiologic-pathologic correlation and timely diagnosis and management of PTs. Published under a CC BY 4.0 license. Online supplemental material is available for this article. Quiz questions for this article are available through the Online Learning Center.
Collapse
Affiliation(s)
- Meng Zhang
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Firouzeh K Arjmandi
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Jessica H Porembka
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Stephen J Seiler
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Sally H Goudreau
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Kanwal Merchant
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Helena Hwang
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| | - Jody C Hayes
- From the Departments of Radiology (M.Z., F.K.A., J.H.P., S.J.S., S.H.G., K.M., J.C.H.) and Pathology (H.H.), University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, MC 8896, Dallas, TX 75390-8896
| |
Collapse
|
6
|
Tan PH. Refining the classification of breast phyllodes tumours. Pathology 2023; 55:437-448. [PMID: 37085395 DOI: 10.1016/j.pathol.2023.02.001] [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: 11/20/2022] [Revised: 01/17/2023] [Accepted: 02/05/2023] [Indexed: 04/23/2023]
Abstract
Phyllodes tumours of the breast are uncommon fibroepithelial neoplasms that pose recurrent classification challenges, in large part due to the multiple histological parameters of stromal hypercellularity and atypia, stromal mitotic count, stromal overgrowth and tumour borders, that are used for grading. While the World Health Organization (WHO) Classification of Breast Tumours provides recommendations on diagnostic features, defining criteria are not always applied in routine practice. Lack of concordance among pathologists in typing and grading further underscores the classification difficulties, especially in the borderline category. Although there has been significant molecular information on phyllodes tumours in recent years which has been diagnostically helpful, it has not been translated into daily clinical practice. In order to refine the classification of phyllodes tumours into one that is simple yet comprehensive, reproducible and prognostically precise, a multipronged approach is needed that leverages on global contributions of the International Fibroepithelial Consortium, support by the International Collaboration on Cancer Classification and Research (IC3 R) in amalgamating evidence translation, and guidance from the International Collaboration on Cancer Reporting (ICCR) for standardised reporting. It is hoped that the evidence generated can be used towards refining the classification of phyllodes tumours for the future.
Collapse
Affiliation(s)
- Puay Hoon Tan
- Luma Medical Centre, Singapore; KK Women's and Children's Hospital, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Pathology, University of Western Sydney, Sydney, NSW, Australia.
| |
Collapse
|
7
|
Nourieh M, Vibert R, Saint-Ghislain M, Cyrta J, Vincent-Salomon A. Next-generation sequencing in breast pathology: real impact on routine practice over a decade since its introduction. Histopathology 2023; 82:162-169. [PMID: 36482269 PMCID: PMC10108312 DOI: 10.1111/his.14794] [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: 08/14/2022] [Revised: 09/08/2022] [Accepted: 09/11/2022] [Indexed: 12/13/2022]
Abstract
The diagnosis, histomolecular classes of breast cancers (luminal A, luminal B, HER2-enriched, and basal-like), and accurate prediction of prognosis are commonly determined using morphological and phenotypical analyses in clinical practice worldwide. Therapeutic strategies are mostly based on the disease stage and molecular subclasses of breast cancer. Targeted therapies, such as anti-HER2s, poly-ADP ribose polymerase inhibitors or, to a lesser extent, phosphatidylinositol 3 kinase inhibitors, have substantially improved breast cancer patient prognosis over the past decades. Human epidermal growth factor receptor 2 (HER2) overexpression is widely determined based on immunohistochemistry, while next-generation sequencing (NGS) is currently employed to assess the presence of molecular alterations, including breast cancer gene 1 (BRCA1) and 2 or phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations, which are targets of these new approved therapies. In addition, next-generation sequencing (NGS) can aid the pathologist in challenging situations, such as a diagnostic workup for a metastatic carcinoma in lymph nodes of unknown origin, differential diagnosis of spindle cell tumourtumor in the breast between metaplastic carcinoma, malignant PT and sarcoma, o, as well as determining relatedness between primary breast cancers and recurrences. NGS offers a powerful tool that enables the pathologist to combine morphological analyses together with molecular alterations in challenging diagnostic situations.
Collapse
Affiliation(s)
- Maya Nourieh
- Department of Diagnostic and Theranostic Medicine, Versailles Saint Quentin University UVSQ, Institut CURIE, Saint-Cloud, France
| | - Roseline Vibert
- Department of Diagnostic and Theranostic Medicine, Paris Sciences Lettres University PSL, Institut CURIE, Paris, France
| | - Mathilde Saint-Ghislain
- Department of Medical Oncology, Paris Sciences Lettres University PSL, Institut CURIE, Paris, France
| | - Joanna Cyrta
- Department of Diagnostic and Theranostic Medicine, Paris Sciences Lettres University PSL, Institut CURIE, Paris, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theranostic Medicine, Paris Sciences Lettres University PSL, Institut CURIE, Paris, France
| |
Collapse
|
8
|
Tan BY, Fox SB, Lakhani SR, Tan PH. Survey of recurrent diagnostic challenges in breast phyllodes tumours. Histopathology 2023; 82:95-105. [PMID: 36468287 DOI: 10.1111/his.14730] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/15/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND Breast phyllodes tumours (PTs) are graded as benign, borderline, or malignant by analysis of multiple histological features. PT grading is often inconsistent, likely due to variation in the weighting of grading criteria by pathologists. DESIGN The hierarchy of use of diagnostic criteria was identified using a 20-question survey. RESULTS In all, 213 pathologists from 29 countries responded. 54% reported 10-50 PT cases per year. Criteria considered key to PT diagnosis were: increased stromal cellularity (84.3%), stromal overgrowth (76.6%), increased stromal mitoses (67.8%), stromal atypia (61.5%), stromal fronding (59.0%), periductal stromal condensation (58.0%), irregular tumour borders (46.3%), and/or lesional heterogeneity (33.7%). The importance of grading parameters were: mitotic activity (55.5%), stromal overgrowth (54.0%), stromal atypia (51.9%), increased stromal cellularity (41.7%), and nature of the tumour border (38.9%). 49% would diagnose malignant PT without a full array of adverse features. 89% used the term "cellular fibroepithelial lesion (FEL)" for difficult cases; 45% would diagnose an FEL with stromal fronding (but lacking other PT features) as fibroadenoma (FA), 35% FEL, and 17% PT. 59% deemed clinico-radiological findings diagnostically significant; 68% considered age (≥40 years) important in determining if an FEL was a FA or PT. In FELs from young patients, increased stromal cellularity (83%), fronding (52%), and mitoses (41%) were more common. 34% regarded differentiating cellular FA from PT as a specific challenge; 54% had issues assigning a borderline PT grade. CONCLUSION Criteria for grading PT lie on a spectrum, leading to interpretive variability. The survey highlights the criteria most used by pathologists, which do not completely align with WHO recommendations.
Collapse
Affiliation(s)
| | - Stephen B Fox
- Peter MacCallum Cancer Centre and University of Melbourne, Australia
| | - Sunil R Lakhani
- The University of Queensland and Pathology Queensland, Australia
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| |
Collapse
|
9
|
Seow DY, Tay TK, Tan PH. FIBROEPITHELIAL LESIONS OF THE BREAST: A REVIEW OF RECURRING DIAGNOSTIC ISSUES. Semin Diagn Pathol 2022; 39:333-343. [DOI: 10.1053/j.semdp.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 11/11/2022]
|
10
|
Cheng CL, Md Nasir ND, Ng GJZ, Chua KWJ, Li Y, Rodrigues J, Thike AA, Heng SY, Koh VCY, Lim JX, Hiew VJN, Shi R, Tan BY, Tay TKY, Ravi S, Ng KH, Oh KSL, Tan PH. Artificial intelligence modelling in differentiating core biopsies of fibroadenoma from phyllodes tumor. J Transl Med 2022; 102:245-252. [PMID: 34819630 DOI: 10.1038/s41374-021-00689-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 02/06/2023] Open
Abstract
Breast fibroepithelial lesions (FEL) are biphasic tumors which consist of benign fibroadenomas (FAs) and the rarer phyllodes tumors (PTs). FAs and PTs have overlapping features, but have different clinical management, which makes correct core biopsy diagnosis important. This study used whole-slide images (WSIs) of 187 FA and 100 PT core biopsies, to investigate the potential role of artificial intelligence (AI) in FEL diagnosis. A total of 9228 FA patches and 6443 PT patches was generated from WSIs of the training subset, with each patch being 224 × 224 pixel in size. Our model employed a two-stage architecture comprising a convolutional neural network (CNN) component for feature extraction from the patches, and a recurrent neural network (RNN) component for whole-slide classification using activation values from the global average pooling layer in the CNN model. It achieved an overall slide-level accuracy of 87.5%, with accuracies of 80% and 95% for FA and PT slides respectively. This affirms the potential role of AI in diagnostic discrimination between FA and PT on core biopsies which may be further refined for use in routine practice.
Collapse
Affiliation(s)
- Chee Leong Cheng
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Nur Diyana Md Nasir
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | | | | | - Yier Li
- AI Singapore, Singapore, Singapore
| | | | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Seow Ye Heng
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Valerie Cui Yun Koh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | | | - Venice Jing Ning Hiew
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Ruoyu Shi
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | | | | | | | | | | | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore. .,Division of Pathology, Singapore General Hospital, Singapore, Singapore.
| |
Collapse
|
11
|
Ng CCY, Md Nasir ND, Loke BN, Tay TKY, Thike AA, Rajasegaran V, Liu W, Lee JY, Guan P, Lim AH, Chang KTE, Gudi MA, Madhukumar P, Tan BKT, Tan VKM, Wong CY, Yong WS, Ho GH, Ong KW, Yip GWC, Bay BH, Tan P, Teh BT, Tan PH. Genetic differences between benign phyllodes tumors and fibroadenomas revealed through targeted next generation sequencing. Mod Pathol 2021; 34:1320-1332. [PMID: 33727697 DOI: 10.1038/s41379-021-00787-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/14/2022]
Abstract
Breast fibroepithelial lesions are biphasic tumors which comprise the common benign fibroadenomas (FAs) and the rarer phyllodes tumors (PTs). This study analyzed 262 (42%) conventional FAs, 45 (7%) cellular FAs, and 321 (51%) benign PTs contributed by the International Fibroepithelial Consortium, using a previously curated 16 gene panel. Benign PTs were found to possess a higher number of mutations, and higher rates of cancer driver gene alterations than both groups of FAs, in particular MED12, TERT promoter, RARA, FLNA, SETD2, RB1, and EGFR. Cases with MED12 mutations were also more likely to have TERT promoter, RARA, SETD2, and EGFR. There were no significant differences detected between conventional FAs and cellular FAs, except for PIK3CA and MAP3K1. TERT promoter alterations were most optimal in discriminating between FAs and benign PTs. Our study affirms the role of sequencing and key mutations that may assist in refining diagnoses of these lesions.
Collapse
Affiliation(s)
- Cedric Chuan Young Ng
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Nur Diyana Md Nasir
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Benjamin Nathanael Loke
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | | | - Wei Liu
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Jing Yi Lee
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Peiyong Guan
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Quantitative Biology and Medicine Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Abner Herbert Lim
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mihir Ananta Gudi
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Preetha Madhukumar
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
| | - Benita Kiat Tee Tan
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore.,Department of Surgery, Sengkang General Hospital, Singapore, Singapore
| | - Veronique Kiak Mien Tan
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
| | - Chow Yin Wong
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
| | - Wei Sean Yong
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
| | - Gay Hui Ho
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Kong Wee Ong
- Division of Surgery and Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | | | - George Wai Cheong Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Patrick Tan
- Duke-NUS Medical School, Singapore, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore. .,Duke-NUS Medical School, Singapore, Singapore.
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore. .,Duke-NUS Medical School, Singapore, Singapore. .,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Division of Pathology, Singapore General Hospital, Singapore, Singapore.
| |
Collapse
|
12
|
Ahmed SS, Lim JCT, Thike AA, Iqbal J, Tan PH. Epithelial-mesenchymal transition and cancer stem cell interactions in breast phyllodes tumours: immunohistochemical evaluation of EZH2, EZR, HMGA2, CD24 and CD44 in correlation with outcome analysis. J Clin Pathol 2021; 75:316-323. [PMID: 33627375 DOI: 10.1136/jclinpath-2020-207068] [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: 08/28/2020] [Revised: 12/21/2020] [Accepted: 01/23/2021] [Indexed: 11/04/2022]
Abstract
AIM Phyllodes tumours (PTs) categorised as benign, borderline and malignant, account for 1% of all breast tumours. Histological assessment does not always predict tumour behaviour, hindering determination of the clinical course and management.Epithelial-mesenchymal transition (EMT) is an important process during embryogenesis. Dysregulation of EMT causes loss of cell polarity, decreased intercellular adhesion, increased motility and invasiveness, promoting tumour progression. Similarly, cancer stem cells (CSCs) promote tumour growth, resistance and recurrence. The aim of this study is to evaluate expression of CSC markers; enhancer of zeste homolog 2 (EZH2), CD24 and CD44 and EMT associated proteins; ezrin (EZR) and high-mobility group AT-hook 2 (HMGA2) in PTs. METHOD Uing tissue microarray sections, immunohistochemistry was performed on 360 PTs. Epithelial and stromal expressions of EZH2, EZR, HMGA2, CD24 and CD44 were evaluated to assess their impact on disease progression and behaviour in correlation with clinicopathological parameters. RESULTS Stromal expression of EZH2, EZR and HMGA2 was observed in 73 (20.3%), 53 (14.7%) and 28 (7.8%) of tumours, epithelial expression in 121 (35.9%), 3 (0.8%) and 351 (97.5%) tumours, respectively. CD24 and CD44 staining was absent in both components. CONCLUSION Expression of biomarkers correlated significantly with aggressive tumour traits such as stromal hypercellularity, atypia, mitoses and permeative tumour borders.Stromal expression of EZH2 and EZR shortened disease-free survival and overall survival; HMGA2 expression did not alter patient survival. EZH2 and EZR may thus be useful in predicting PT behaviour.
Collapse
Affiliation(s)
| | | | - Aye Aye Thike
- Anatomical Pathology, Singapore General Hospital, Division of Pathology, Singapore
| | - Jabed Iqbal
- Anatomical Pathology, Singapore General Hospital, Division of Pathology, Singapore
| | - Puay Hoon Tan
- Pathology, Singapore General Hospital, Division of Pathology, Singapore
| |
Collapse
|
13
|
Tan PH. Fibroepithelial lesions revisited: implications for diagnosis and management. Mod Pathol 2021; 34:15-37. [PMID: 32461622 DOI: 10.1038/s41379-020-0583-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
Fibroepithelial lesions of the breast, comprising the fibroadenoma and phyllodes tumour, are a unique group of neoplasms that share histological characteristics but possess different clinical behaviour. The fibroadenoma is the commonest benign breast tumour in women, while the phyllodes tumour is rare and may be associated with recurrences, grade progression and even metastasis. The diagnosis of fibroadenoma is usually straightforward, with recognised histological variants such as the cellular, complex, juvenile and myxoid forms. The phyllodes tumour comprises benign, borderline and malignant varieties, graded using a constellation of histological parameters based on stromal characteristics of hypercellularity, atypia, mitoses, overgrowth and the nature of tumour borders. While phyllodes tumour grade correlates with clinical behaviour, interobserver variability in assessing multiple parameters that are potentially of different biological weightage leads to significant challenges in accurate grade determination and consequently therapy. Differential diagnostic considerations along the spectrum of fibroepithelial tumours can be problematic in routine practice. Recent discoveries of the molecular underpinnings of these tumours may have diagnostic, prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Academia, Diagnostics Tower Level 7, 20 College Road, Singapore, 169856, Singapore.
| |
Collapse
|
14
|
Fortarezza F, Pezzuto F, Cazzato G, Punzo C, d’Amati A, Lettini T, Gentile M, Buonadonna AL, Mariano M, Pezzolla A, Serio G. Bilateral Phyllodes Giant Tumor. A Case Report Analyzed by Array-CGH. Diagnostics (Basel) 2020; 10:825. [PMID: 33076253 PMCID: PMC7602371 DOI: 10.3390/diagnostics10100825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 02/05/2023] Open
Abstract
The breast phyllodes tumor is a biphasic tumor that accounts for less than of 1% of all breast neoplasms. It is classified as benign, borderline, or malignant, and can mimic benign masses. Some recurrent alterations have been identified. However, a precise molecular classification of these tumors has not yet been established. Herein, we describe a case of a 43-year-old woman that was admitted to the emergency room for a significant bleeding from the breast skin. A voluminous ulcerative mass of the left breast and multiple nodules with micro-calcifications on the right side were detected at a physical examination. A left total mastectomy and a nodulectomy of the right breast was performed. The histological diagnosis of the surgical specimens reported a bilateral giant phyllodes tumor, showing malignant features on the left and borderline characteristics associated with a fibroadenoma on the right. A further molecular analysis was carried out by an array-Comparative Genomic Hybridization (CGH) to characterize copy-number alterations. Many losses were detected in the malignant mass, involving several tumor suppressor genes. These findings could explain the malignant growth and the metastatic risk. In our study, genomic profiling by an array-CGH revealed a greater chromosomal instability in the borderline mass (40 total defects) than in the malignant (19 total defects) giant phyllodes tumor, reflecting the tumor heterogeneity. Should our results be confirmed with more sensitive and specific molecular tests (DNA sequencing and FISH analysis), they could allow a better selection of patients with adverse pathological features, thus optimizing and improving patient's management.
Collapse
Affiliation(s)
- Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Pathology Unit, University of Padova, 35121 Padova, Italy; (F.F.); (F.P.)
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Pathology Unit, University of Padova, 35121 Padova, Italy; (F.F.); (F.P.)
| | - Gerardo Cazzato
- Department of Emergency and Organ Transplantation (DETO), Pathology Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (G.C.); (A.d.); (T.L.); (M.M.)
| | - Clelia Punzo
- Department of Emergency and Organ Transplantation (DETO), Surgery Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (C.P.); (A.P.)
| | - Antonio d’Amati
- Department of Emergency and Organ Transplantation (DETO), Pathology Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (G.C.); (A.d.); (T.L.); (M.M.)
| | - Teresa Lettini
- Department of Emergency and Organ Transplantation (DETO), Pathology Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (G.C.); (A.d.); (T.L.); (M.M.)
| | - Mattia Gentile
- Medical Genetics, “Di Venere” Hospital, 70131 Carbonara (Bari), Italy; (M.G.); (A.L.B.)
| | | | - Marta Mariano
- Department of Emergency and Organ Transplantation (DETO), Pathology Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (G.C.); (A.d.); (T.L.); (M.M.)
| | - Angela Pezzolla
- Department of Emergency and Organ Transplantation (DETO), Surgery Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (C.P.); (A.P.)
| | - Gabriella Serio
- Department of Emergency and Organ Transplantation (DETO), Pathology Section, Breast Unit Care, University of Bari, Medical School, 70124 Bari, Italy; (G.C.); (A.d.); (T.L.); (M.M.)
| |
Collapse
|