1
|
Tang L, Chen X, Hou J, Wei X. CXCL14 in prostate cancer: complex interactions in the tumor microenvironment and future prospects. J Transl Med 2025; 23:9. [PMID: 39755616 DOI: 10.1186/s12967-024-06022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025] Open
Abstract
CXCL14 is a highly conserved chemokine expressed in various cell types, playing crucial roles in both physiological and pathological processes, including immune regulation and tumorigenesis. Recently, the role of CXCL14 in tumors has attracted considerable attention. However, previous pan-cancer studies have reported inconsistencies regarding the effects of CXCL14 on tumors, particularly concerning its expression levels in tumor tissues and its influence on various phenotypes of cancer cells. This variability is believed to stem from the context-dependent nature of CXCL14, as different sources of CXCL14 and its secretion within distinct tumor microenvironments may mediate diverse biological effects. Such phenomena have also been observed in prostate cancer research. Despite a foundational understanding of CXCL14 in prostate cancer, there remains a lack of comprehensive reviews summarizing the specific roles of this chemokine and systematically analyzing the reasons behind its complex effects. Therefore, this article aims to discuss the role of CXCL14 in the tumor microenvironment of prostate cancer and explore future research directions and potential applications.
Collapse
Affiliation(s)
- Lei Tang
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215006, People's Republic of China
| | - Xin Chen
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215006, People's Republic of China
- Department of Urology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, People's Republic of China
| | - Jianquan Hou
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215006, People's Republic of China.
- Department of Urology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, People's Republic of China.
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, 215006, People's Republic of China.
| |
Collapse
|
2
|
Roozitalab MR, Prekete N, Allen M, Grose RP, Louise Jones J. The Microenvironment in DCIS and Its Role in Disease Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:211-235. [PMID: 39821028 DOI: 10.1007/978-3-031-70875-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Ductal carcinoma in situ (DCIS) accounts for ~20% of all breast cancer diagnoses but whilst known to be a precursor of invasive breast cancer (IBC), evidence suggests only one in six patients will ever progress. A key challenge is to distinguish between those lesions that will progress and those that will remain indolent. Molecular analyses of neoplastic epithelial cells have not identified consistent differences between lesions that progressed and those that did not, and this has focused attention on the tumour microenvironment (ME).The DCIS ME is unique, complex and dynamic. Myoepithelial cells form the wall of the ductal-lobular tree and exhibit broad tumour suppressor functions. However, in DCIS they acquire phenotypic changes that bestow them with tumour promoter properties, an important evolution since they act as the primary barrier for invasion. Changes in the peri-ductal stromal environment also arise in DCIS, including transformation of fibroblasts into cancer-associated fibroblasts (CAFs). CAFs orchestrate other changes in the stroma, including the physical structure of the extracellular matrix (ECM) through altered protein synthesis, as well as release of a plethora of factors including proteases, cytokines and chemokines that remodel the ECM. CAFs can also modulate the immune ME as well as impact on tumour cell signalling pathways. The heterogeneity of CAFs, including recognition of anti-tumourigenic populations, is becoming evident, as well as heterogeneity of immune cells and the interplay between these and the adipocyte and vascular compartments. Knowledge of the impact of these changes is more advanced in IBC but evidence is starting to accumulate for a role in DCIS. Detailed in vitro, in vivo and tissue studies focusing on the interplay between DCIS epithelial cells and the ME should help to define features that can better predict DCIS behaviour.
Collapse
Affiliation(s)
- Mohammad Reza Roozitalab
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Niki Prekete
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Michael Allen
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, John Vane Science Centre, Charterhouse Square, Queen Mary University of London, London, UK.
| |
Collapse
|
3
|
Kaplan HG, Dowdell AK, Berry AB, Shimol RB, Robinson FL, Carney CA, Piening BD. Multi-omic profiling of simultaneous ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res Treat 2024; 205:451-464. [PMID: 38523186 PMCID: PMC11101558 DOI: 10.1007/s10549-024-07270-5] [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: 09/25/2023] [Accepted: 01/24/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE The progression of ductal carcinoma in situ (DCIS) to invasive breast carcinoma (IBC) in humans is highly variable. To better understand the relationship between them, we performed a multi-omic characterization of co-occurring DCIS and IBC lesions in a cohort of individuals. METHODS Formalin-fixed paraffin-embedded tissue samples from 50 patients with co-occurring DCIS and IBC lesions were subjected to DNA-seq and whole transcriptome RNA-seq. Paired DCIS and IBC multi-omics profiles were then interrogated for DNA mutations, gene expression profiles and pathway analysis. RESULTS Most small variants and copy number variations were shared between co-occurring DCIS and IBC lesions, with IBC exhibiting on average a higher degree of additional mutations. However, 36% of co-occurring lesions shared no common mutations and 49% shared no common copy number variations. The most frequent genomic variants in both DCIS and IBC were PIK3CA, TP53, KMT2C, MAP3K1, GATA3 and SF3B1, with KMT2C being more frequent in DCIS and TP53 and MAP3K1 more frequent in IBC, though the numbers are too small for definitive conclusions. The most frequent copy number variations were seen in MCL1, CKSB1 and ERBB2. ERBB2 changes were not seen in IBC unless present in the corresponding DCIS. Transcriptional profiles were highly distinct between DCIS and IBC, with DCIS exhibiting upregulation of immune-related signatures, while IBC showed significant overexpression in genes and pathways associated with cell division and proliferation. Interestingly, DCIS and IBC exhibited significant differential expression of different components of extracellular matrix (ECM) formation and regulation, with DCIS showing overexpression of ECM-membrane interaction components while IBC showed upregulation of genes associated with fibronectin and invadopodia. CONCLUSION While most co-occurring DCIS and IBC were mutationally similar and suggestive of a common clonal progenitor, transcriptionally the lesions are highly distinct, with IBC expressing key pathways that facilitate invasion and proliferation. These results are suggestive of additional levels of regulation, epigenetic or other, that facilitate the acquisition of invasive properties during tumor evolution.
Collapse
MESH Headings
- Humans
- Female
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Mutation
- DNA Copy Number Variations
- Gene Expression Profiling/methods
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/metabolism
- Biomarkers, Tumor/genetics
- Middle Aged
- Neoplasm Invasiveness
- Gene Expression Regulation, Neoplastic
- Transcriptome
- Aged
- Adult
- Genomics/methods
- Multiomics
Collapse
Affiliation(s)
- Henry G Kaplan
- Swedish Cancer Institute, 1221 Madison St., Suite 920, Seattle, WA, 98104, USA.
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Health, Portland, OR, 97213, USA
| | - Anna B Berry
- Swedish Cancer Institute, 1221 Madison St., Suite 920, Seattle, WA, 98104, USA
| | - Racheli Ben Shimol
- Earle A. Chiles Research Institute, Providence Health, Portland, OR, 97213, USA
| | - Fred L Robinson
- Earle A. Chiles Research Institute, Providence Health, Portland, OR, 97213, USA
| | | | - Brian D Piening
- Earle A. Chiles Research Institute, Providence Health, Portland, OR, 97213, USA
| |
Collapse
|
4
|
Rao J, Sinn M, Pelzer U, Riess H, Oettle H, Demir IE, Friess H, Jäger C, Steiger K, Muckenhuber A. KRT81 and HNF1A expression in pancreatic ductal adenocarcinoma: investigation of predictive and prognostic value of immunohistochemistry-based subtyping. J Pathol Clin Res 2024; 10:e12377. [PMID: 38750616 PMCID: PMC11096282 DOI: 10.1002/2056-4538.12377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/19/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
Even after decades of research, pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal disease and responses to conventional treatments remain mostly poor. Subclassification of PDAC into distinct biological subtypes has been proposed by various groups to further improve patient outcome and reduce unnecessary side effects. Recently, an immunohistochemistry (IHC)-based subtyping method using cytokeratin-81 (KRT81) and hepatocyte nuclear factor 1A (HNF1A) could recapitulate some of the previously established molecular subtyping methods, while providing significant prognostic and, to a limited degree, also predictive information. We refined the KRT81/HNF1A subtyping method to classify PDAC into three distinct biological subtypes. The prognostic value of the IHC-based method was investigated in two primary resected cohorts, which include 269 and 286 patients, respectively. In the second cohort, we also assessed the predictive effect for response to erlotinib + gemcitabine. In both PDAC cohorts, the new HNF1A-positive subtype was associated with the best survival, the KRT81-positive subtype with the worst, and the double-negative with an intermediate survival (p < 0.001 and p < 0.001, respectively) in univariate and multivariate analyses. In the second cohort (CONKO-005), the IHC-based subtype was additionally found to have a potential predictive value for the erlotinib-based treatment effect. The revised IHC-based subtyping using KRT81 and HNF1A has prognostic significance for PDAC patients and may be of value in predicting treatment response to specific therapeutic agents.
Collapse
Affiliation(s)
- Jia Rao
- Institute of PathologyTechnical University of MunichMunichGermany
| | - Marianne Sinn
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
- Department of Internal Medicine IIUniversity Medical Center of Hamburg‐EppendorfHamburgGermany
| | - Uwe Pelzer
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
| | - Hanno Riess
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
| | - Helmut Oettle
- Department of Haematology, Oncology and Tumour Immunology, CONKO‐Study‐GroupCharité – University Medicine BerlinBerlinGermany
| | - Ihsan E Demir
- Department of Surgery, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
- Else Kröner Clinician Scientist Professor for Translational Pancreatic SurgeryMunichGermany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
| | - Carsten Jäger
- Department of Surgery, Klinikum rechts der Isar, School of MedicineTechnical University of MunichMunichGermany
| | - Katja Steiger
- Institute of PathologyTechnical University of MunichMunichGermany
| | | |
Collapse
|
5
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Kang DS, Moriarty A, Wang YJ, Thomas A, Hao J, Unger BA, Klotz R, Ahmmed S, Amzaleg Y, Martin S, Vanapalli S, Xu K, Smith A, Shen K, Yu M. Ectopic Expression of a Truncated Isoform of Hair Keratin 81 in Breast Cancer Alters Biophysical Characteristics to Promote Metastatic Propensity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2300509. [PMID: 37949677 PMCID: PMC10837353 DOI: 10.1002/advs.202300509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/28/2023] [Indexed: 11/12/2023]
Abstract
Keratins are an integral part of cell structure and function. Here, it is shown that ectopic expression of a truncated isoform of keratin 81 (tKRT81) in breast cancer is upregulated in metastatic lesions compared to primary tumors and patient-derived circulating tumor cells, and is associated with more aggressive subtypes. tKRT81 physically interacts with keratin 18 (KRT18) and leads to changes in the cytosolic keratin intermediate filament network and desmosomal plaque formation. These structural changes are associated with a softer, more elastically deformable cancer cell with enhanced adhesion and clustering ability leading to greater in vivo lung metastatic burden. This work describes a novel biomechanical mechanism by which tKRT81 promotes metastasis, highlighting the importance of the biophysical characteristics of tumor cells.
Collapse
Affiliation(s)
- Diane S. Kang
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
| | - Aidan Moriarty
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Yiru Jess Wang
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Amal Thomas
- Department of Molecular and Computational BiologyUSC David and Dana Dornsife College of LettersArts and SciencesUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Jia Hao
- Department of Biomedical EngineeringViterbi School of EngineeringUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Bret A. Unger
- Department of ChemistryUniversity of California at BerkeleyBerkeleyCA94720USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Shamim Ahmmed
- Department of Chemical EngineeringTexas Tech UniversityLubbockTX79409USA
| | - Yonatan Amzaleg
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
| | - Stuart Martin
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| | - Siva Vanapalli
- Department of Chemical EngineeringTexas Tech UniversityLubbockTX79409USA
| | - Ke Xu
- Department of ChemistryUniversity of California at BerkeleyBerkeleyCA94720USA
| | - Andrew Smith
- Department of Molecular and Computational BiologyUSC David and Dana Dornsife College of LettersArts and SciencesUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Keyue Shen
- Department of Biomedical EngineeringViterbi School of EngineeringUniversity of Southern CaliforniaLos AngelesCA90089USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative MedicineKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- USC Norris Comprehensive Cancer CenterKeck School of Medicine of the University of Southern CaliforniaLos AngelesCA90033USA
- Department of PharmacologyUniversity of Maryland School of MedicineBaltimoreMD21201USA
- Marlene and Stewart Greenebaum Comprehensive Cancer CenterUniversity of Maryland School of MedicineBaltimoreMD21201USA
| |
Collapse
|
7
|
Peng J, Chan C, Zhang S, Sklavounos AA, Olson ME, Scott EY, Hu Y, Rajesh V, Li BB, Chamberlain MD, Zhang S, Peng H, Wheeler AR. All-in-One digital microfluidics pipeline for proteomic sample preparation and analysis. Chem Sci 2023; 14:2887-2900. [PMID: 36937585 PMCID: PMC10016607 DOI: 10.1039/d3sc00560g] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Highly sensitive and reproducible analysis of samples containing low amounts of protein is restricted by sample loss and the introduction of contaminants during processing. Here, we report an All-in-One digital microfluidic (DMF) pipeline for proteomic sample reduction, alkylation, digestion, isotopic labeling and analysis. The system features end-to-end automation, with integrated thermal control for digestion, optimized droplet additives for sample manipulation and analysis, and an automated interface to liquid chromatography with tandem mass spectrometry (HPLC-MS/MS). Dimethyl labeling was integrated into the pipeline to allow for relative quantification of the trace samples at the nanogram level, and the new pipeline was applied to evaluating cancer cell lines and cancer tissue samples. Several known proteins (including HSP90AB1, HSPB1, LDHA, ENO1, PGK1, KRT18, and AKR1C2) and pathways were observed between model breast cancer cell lines related to hormone response, cell metabolism, and cell morphology. Furthermore, differentially quantified proteins (such as PGS2, UGDH, ASPN, LUM, COEA1, and PRELP) were found in comparisons of healthy and cancer breast tissues, suggesting potential utility of the All-in-One pipeline for the emerging application of proteomic cancer sub-typing. In sum, the All-in-One pipeline represents a powerful new tool for automated proteome processing and analysis, with the potential to be useful for evaluating mass-limited samples for a wide range of applications.
Collapse
Affiliation(s)
- Jiaxi Peng
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - Calvin Chan
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
| | - Shuailong Zhang
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- School of Mechatronical Engineering, Beijing Institute of Technology Beijing 100081 China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology Beijing 100081 China
| | - Alexandros A Sklavounos
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
| | - Maxwell E Olson
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
| | - Erica Y Scott
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - Yechen Hu
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - Vigneshwar Rajesh
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
| | - Bingyu B Li
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| | - M Dean Chamberlain
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
- Saskatchewan Cancer Agency, University of Saskatchewan 107 Wiggins Road Saskatoon SK S7N 5E5 Canada
| | - Shen Zhang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital 600 University Avenue Toronto ON M5G 1X5 Canada
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA Changsha Hunan 410000 China
| | - Hui Peng
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- School of Environment, University of Toronto 33 Willcocks Street Toronto ON M5S 3E8 Canada
| | - Aaron R Wheeler
- Department of Chemistry, University of Toronto 80 St. George Street Toronto ON M5S 3H6 Canada +1-416-946-3865 +1-416-946-3866
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto 160 College Street Toronto ON M5S 3E1 Canada
- Institute of Biomedical Engineering, University of Toronto 164 College Street Toronto ON M5S 3G9 Canada
| |
Collapse
|
8
|
Corsi F, Albasini S, Ciciriello S, Villani L, Truffi M, Sevieri M, Sorrentino L. Extensive Intraductal Component in Breast Cancer: What Role in Disease-Free Survival? J Surg Res 2023; 283:233-240. [PMID: 36423471 DOI: 10.1016/j.jss.2022.10.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Extensive intraductal component (EIC) associated to early breast cancer could increase the risk locoregional recurrence, but its impact on distant metastases is still unclear. The aim of the present study was to assess the role of EIC on 5-year survival outcomes in patients affected by early breast cancer treated with breast-conserving surgery. METHODS A total of 414 consecutive patients with a minimum follow-up of 60 mo were collected from January 2007 to December 2015. Disease-free survival (DFS), distant metastasis-free survival (DMFS), and locoregional recurrence-free survival at 5 y were assessed considering the presence or absence of EIC and other clinical and pathological features. RESULTS Absence of EIC was independently associated with worse 5-year DFS (hazard ratio [HR] 1.68, P = 0.008) and 5-year DMFS (HR 1.93, P = 0.007), whereas 5-year locoregional recurrence-free survival was not affected (HR 1.50, P = 0.16). Five-year DFS was increased by EIC in T1 patients (P = 0.03) but not in T2 stage. Moreover, EIC was associated to better DFS in G2 (P = 0.03) and G3 patients (P = 0.01) but not in G1 cases. CONCLUSIONS Our results suggest that EIC is independently correlated with increased 5-year DFS and in particular with 5-year DMFS.
Collapse
Affiliation(s)
- Fabio Corsi
- Breast Unit, Department of Surgery, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy; Department of Biomedical and Clinical Sciences "Luigi Sacco", Università di Milano, Milan, Italy.
| | - Sara Albasini
- Breast Unit, Department of Surgery, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Simone Ciciriello
- Breast Unit, Department of Surgery, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Laura Villani
- Department of Pathology, Istituto Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Marta Truffi
- Nanomedicine and Molecular Imaging Lab, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Marta Sevieri
- Department of Biomedical and Clinical Sciences "Luigi Sacco", Università di Milano, Milan, Italy
| | - Luca Sorrentino
- Colorectal Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| |
Collapse
|
9
|
Sourouni M, Opitz C, Radke I, Kiesel L, Tio J, Götte M, von Wahlde M. Establishment of a
3D
co‐culture model to investigate the role of primary fibroblasts in ductal carcinoma in situ of the breast. Cancer Rep (Hoboken) 2022; 6:e1771. [PMID: 36534078 PMCID: PMC10075300 DOI: 10.1002/cnr2.1771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a precursor form of breast cancer. 13%-50% of these lesions will progress to invasive breast cancer, but the individual progression risk cannot be estimated. Therefore, all patients receive the same therapy, resulting in potential overtreatment of a large proportion of patients. AIMS The role of the tumor microenvironment (TME) and especially of fibroblasts appears to be critical in DCIS development and a better understanding of their role may aid individualized treatment. METHODS AND RESULTS Primary fibroblasts isolated from benign or malignant punch biopsies of the breast and MCF10DCIS.com cells were seeded in a 3D cell culture system. The fibroblasts were cultured in a type I collagen layer beneath a Matrigel layer with MCF10DCIS.com cells. Dye-quenched (DQ) fluorescent collagen I and IV were used in collagen and Matrigel layer respectively to demonstrate proteolysis. Confocal microscopy was performed on day 2, 7, and 14 to reveal morphological changes, which could indicate the transition to an invasive phenotype. MCF10DCIS.com cells form smooth, round spheroids in co-culture with non-cancer associated fibroblasts (NAFs). Spheroids in co-culture with tumor-associated fibroblasts (TAFs) appear irregularly shaped and with an uneven surface; similar to spheroids formed from invasive cells. Therefore, these morphological changes represent the progression of an in situ to an invasive phenotype. In addition, TAFs show a higher proteolytic activity compared to NAFs. The distance between DCIS cells and fibroblasts decreases over time. CONCLUSION The TAFs seem to play an important role in the progression of DCIS to invasive breast cancer. The better characterization of the TME could lead to the identification of DCIS lesions with high or low risk of progression. This could enable personalized oncological therapy, prevention of overtreatment and individualized hormone replacement therapy after DCIS.
Collapse
Affiliation(s)
- Marina Sourouni
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Carl Opitz
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Isabel Radke
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Ludwig Kiesel
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Joke Tio
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Martin Götte
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Marie‐Kristin von Wahlde
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| |
Collapse
|
10
|
Casasent AK, Almekinders MM, Mulder C, Bhattacharjee P, Collyar D, Thompson AM, Jonkers J, Lips EH, van Rheenen J, Hwang ES, Nik-Zainal S, Navin NE, Wesseling J. Learning to distinguish progressive and non-progressive ductal carcinoma in situ. Nat Rev Cancer 2022; 22:663-678. [PMID: 36261705 DOI: 10.1038/s41568-022-00512-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 02/07/2023]
Abstract
Ductal carcinoma in situ (DCIS) is a non-invasive breast neoplasia that accounts for 25% of all screen-detected breast cancers diagnosed annually. Neoplastic cells in DCIS are confined to the ductal system of the breast, although they can escape and progress to invasive breast cancer in a subset of patients. A key concern of DCIS is overtreatment, as most patients screened for DCIS and in whom DCIS is diagnosed will not go on to exhibit symptoms or die of breast cancer, even if left untreated. However, differentiating low-risk, indolent DCIS from potentially progressive DCIS remains challenging. In this Review, we summarize our current knowledge of DCIS and explore open questions about the basic biology of DCIS, including those regarding how genomic events in neoplastic cells and the surrounding microenvironment contribute to the progression of DCIS to invasive breast cancer. Further, we discuss what information will be needed to prevent overtreatment of indolent DCIS lesions without compromising adequate treatment for high-risk patients.
Collapse
Affiliation(s)
- Anna K Casasent
- Department of Genetics, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Charlotta Mulder
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | | | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Serena Nik-Zainal
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Nicholas E Navin
- Department of Genetics, MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioinformatics, MD Anderson Cancer Center, Houston, TX, USA
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands.
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
11
|
Rebbeck CA, Xian J, Bornelöv S, Geradts J, Hobeika A, Geiger H, Alvarez JF, Rozhkova E, Nicholls A, Robine N, Lyerly HK, Hannon GJ. Gene expression signatures of individual ductal carcinoma in situ lesions identify processes and biomarkers associated with progression towards invasive ductal carcinoma. Nat Commun 2022; 13:3399. [PMID: 35697697 PMCID: PMC9192778 DOI: 10.1038/s41467-022-30573-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/06/2022] [Indexed: 12/27/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is considered a non-invasive precursor to breast cancer, and although associated with an increased risk of developing invasive disease, many women with DCIS will never progress beyond their in situ diagnosis. The path from normal duct to invasive ductal carcinoma (IDC) is not well understood, and efforts to do so are hampered by the substantial heterogeneity that exists between patients, and even within patients. Here we show gene expression analysis from > 2,000 individually micro-dissected ductal lesions representing 145 patients. Combining all samples into one continuous trajectory we show there is a progressive loss in basal layer integrity heading towards IDC, coupled with two epithelial to mesenchymal transitions, one early and a second coinciding with the convergence of DCIS and IDC expression profiles. We identify early processes and potential biomarkers, including CAMK2N1, MNX1, ADCY5, HOXC11 and ANKRD22, whose reduced expression is associated with the progression of DCIS to invasive breast cancer.
Collapse
Affiliation(s)
- Clare A Rebbeck
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| | - Jian Xian
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Susanne Bornelöv
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joseph Geradts
- Department of Pathology & Laboratory Medicine, East Carolina University Brody School of Medicine, Greenville, NC, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - Jose Franco Alvarez
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Elena Rozhkova
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Ashley Nicholls
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Herbert K Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
12
|
Belpaire M, Taminiau A, Geerts D, Rezsohazy R. HOXA1, a breast cancer oncogene. Biochim Biophys Acta Rev Cancer 2022; 1877:188747. [PMID: 35675857 DOI: 10.1016/j.bbcan.2022.188747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
More than 25 years ago, the first literature records mentioned HOXA1 expression in human breast cancer. A few years later, HOXA1 was confirmed as a proper oncogene in mammary tissue. In the following two decades, molecular data about the mode of action of the HOXA1 protein, the factors contributing to activate and maintain HOXA1 gene expression and the identity of its target genes have accumulated and provide a wider view on the association of this transcription factor to breast oncogenesis. Large-scale transcriptomic data gathered from wide cohorts of patients further allowed refining the relationship between breast cancer type and HOXA1 expression. Several recent reports have reviewed the connection between cancer hallmarks and the biology of HOX genes in general. Here we take HOXA1 as a paradigm and propose an extensive overview of the molecular data centered on this oncoprotein, from what its expression modulators, to the interactors contributing to its oncogenic activities, and to the pathways and genes it controls. The data converge to an intricate picture that answers questions on the multi-modality of its oncogene activities, point towards better understanding of breast cancer aetiology and thereby provides an appraisal for treatment opportunities.
Collapse
Affiliation(s)
- Magali Belpaire
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Arnaud Taminiau
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Dirk Geerts
- Heart Failure Research Center, Amsterdam University Medical Center (AMC), Universiteit van Amsterdam, Amsterdam, the Netherlands.
| | - René Rezsohazy
- Animal Molecular and Cellular Biology Group (AMCB), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium.
| |
Collapse
|
13
|
Saleh M, Chandrashekar DS, Shahin S, Agarwal S, Kim HG, Behring M, Shaikh AJ, Moloo Z, Eltoum IEA, Yates C, Varambally S, Manne U. Comparative analysis of triple-negative breast cancer transcriptomics of Kenyan, African American and Caucasian Women. Transl Oncol 2021; 14:101086. [PMID: 33839593 PMCID: PMC8058567 DOI: 10.1016/j.tranon.2021.101086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Abstract
The current study determined the molecular fingerprints of TNBCs of women from kenya (KE) and compared them with those of African–American (AA) and Caucasian (CA) women. RNA sequencing analysis highlights the role of molecular alterations in TNBCs and the potential benefit of targeting pathways in this disease for the KE population as compared to AAs and CAs. The dysregulated genes and signaling pathways could contributes to the aggressive phenotypes of TNBCs of KE women.
Purpose : Triple-negative breast cancer (TNBC) patients of various ethnic groups often have discrete clinical presentations and outcomes. Women of African descent have a disproportionately higher chance of developing TNBCs. The aim of the current study was to establish the transcriptome of TNBCs from Kenyan (KE) women of Bantu origin and compare it to those TNBCs of African-Americans (AA) and Caucasians (CA) for identifying KE TNBC-specific molecular determinants of cancer progression and potential biomarkers of clinical outcomes. Patients and Methods : Pathology-confirmed TNBC tissues from Kenyan women of Bantu origin (n = 15) and age and stage range matched AA (n = 19) and CA (n = 23) TNBCs of patients from Alabama were included in this study. RNA was isolated from paraffin-embedded tissues, and expression was analyzed by RNA sequencing. Results : At clinical presentation, young KE TNBC patients have tumors of higher stages. Differential expression analysis identified 160 up-regulated and 178 down-regulated genes in KE TNBCs compared to AA and CA TNBCs. Validation analyses of the TCGA breast cancer data identified 45 KE TNBC-specific genes that are involved in the apoptosis (ACTC1, ERCC6 and CD14), cell proliferation (UHRF2, KDM4C, UHMK1, KCNH5, KRT18, CSF1R and S100A13), and Wnt signaling (BCL9L) pathways. Conclusions : In this study, we identified biomarkers that are specific for KE TNBC patients of Bantu origin. Further study with a larger sample size of matched tumors could confirm our findings. If biologically confirmed, these molecular determinants could have clinical and biological implications and serve as targets for development of personalized therapeutics for KE TNBC patients.
Collapse
Affiliation(s)
- Mansoor Saleh
- Department of Medicine, University of Alabama at Birmingham, Birmingham 35233, AL, United States; Department of Hematology-Oncology, the Aga Khan University, Nairobi, Kenya; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham 35233, AL, United States
| | | | - Sayed Shahin
- Department of Pathology, the Aga Khan University, Nairobi, Kenya
| | - Sumit Agarwal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hyung-Gyoon Kim
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Michael Behring
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Zahir Moloo
- Department of Pathology, the Aga Khan University, Nairobi, Kenya
| | - Isam-Eldin A Eltoum
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Clayton Yates
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States; Department of Biology & Center for Cancer Research, Tuskegee University, Tuskegee, AL, United States
| | - Sooryanarayana Varambally
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham 35233, AL, United States; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Upender Manne
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham 35233, AL, United States; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
14
|
Patterns of treatment and outcome of ductal carcinoma in situ in the Netherlands. Breast Cancer Res Treat 2021; 187:245-254. [PMID: 33385265 PMCID: PMC8062340 DOI: 10.1007/s10549-020-06055-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/07/2020] [Indexed: 01/03/2023]
Abstract
Purpose To spare DCIS patients from overtreatment, treatment de-escalated over the years. This study evaluates the influence of these developments on the patterns of care in the treatment of DCIS with particular interest in the use of breast conserving surgery (BCS), radiotherapy following BCS and the use and type of axillary staging. Methods In this large population-based cohort study all women, aged 50–74 years diagnosed with DCIS from January 1989 until January 2019, were analyzed per two-year cohort. Results A total of 30,417 women were diagnosed with DCIS. The proportion of patients undergoing BCS increased from 47.7% in 1995–1996 to 72.7% in 2017–2018 (p < 0.001). Adjuvant radiotherapy following BCS increased from 28.9% (1995–1996) to 89.6% (2011–2012) and subsequently decreased to 74.9% (2017–2018; p < 0.001). Since its introduction, the use of sentinel lymph node biopsy (SLNB) increased to 63.1% in 2013–2014 and subsequently decreased to 52.8% in 2017–2018 (p < 0.001). Axillary surgery is already omitted in 55.8% of the patients undergoing BCS nowadays. The five-year invasive relapse-free survival (iRFS) for BCS with adjuvant radiotherapy in the period 1989–2010, was 98.7% [CI 98.4% – 99.0%], compared to 95.0% [CI 94.1% –95.8%] for BCS only (p < 0.001). In 2011–2018, this was 99.3% [CI 99.1% – 99.5%] and 98.8% [CI 98.2% – 99.4%] respectively (p = 0.01). Conclusions This study shows a shift toward less extensive treatment. DCIS is increasingly treated with BCS and less often followed by additional radiotherapy. The absence of radiotherapy still results in excellent iRFS. Axillary surgery is increasingly omitted in DCIS patients.
Collapse
|
15
|
Zhang P, Wu X, Gardashova G, Yang Y, Zhang Y, Xu L, Zeng Y. Molecular and functional extracellular vesicle analysis using nanopatterned microchips monitors tumor progression and metastasis. Sci Transl Med 2020; 12:eaaz2878. [PMID: 32522804 PMCID: PMC8024111 DOI: 10.1126/scitranslmed.aaz2878] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/30/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Longitudinal cancer monitoring is crucial to clinical implementation of precision medicine. There is growing evidence indicating important functions of extracellular vesicles (EVs) in tumor progression and metastasis, including matrix remodeling via transporting matrix metalloproteases (MMPs). However, the clinical relevance of EVs remains largely undetermined, partially owing to challenges in EV analysis. Distinct from existing technologies mostly focused on characterizing molecular constituents of EVs, here we report a nanoengineered lab-on-a-chip system that enables integrative functional and molecular phenotyping of tumor-associated EVs. A generalized, high-resolution colloidal inkjet printing method was developed to allow robust and scalable manufacturing of three-dimensional (3D) nanopatterned devices. With this nanochip platform, we demonstrated integrative analysis of the expression and proteolytic activity of MMP14 on EVs to detect in vitro cell invasiveness and monitor in vivo tumor metastasis, using cancer cell lines and mouse models. Analysis of clinical plasma specimen showed that our technology could be used for cancer detection including accurate classification of age-matched controls and patients with ductal carcinoma in situ, invasive ductal carcinoma, or locally metastatic breast cancer in a training cohort (n = 30, 96.7% accuracy) and an independent validation cohort (n = 70, 92.9% accuracy). With clinical validation, our technology could provide a useful liquid biopsy tool to improve cancer diagnostics and real-time surveillance of tumor evolution in patients to inform personalized therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Xiaoqing Wu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Gulhumay Gardashova
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Yang Yang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Yaohua Zhang
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS 66045, USA
| | - Liang Xu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA.
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Yong Zeng
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA.
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS 66045, USA
- University of Kansas Cancer Center, Kansas City, KS 66160, USA
| |
Collapse
|
16
|
Westrich JA, Vermeer DW, Colbert PL, Spanos WC, Pyeon D. The multifarious roles of the chemokine CXCL14 in cancer progression and immune responses. Mol Carcinog 2020; 59:794-806. [PMID: 32212206 DOI: 10.1002/mc.23188] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Abstract
The chemokine CXCL14 is a highly conserved, homeostatic chemokine that is constitutively expressed in skin epithelia. Responsible for immune cell recruitment and maturation, as well as impacting epithelial cell motility, CXCL14 contributes to the establishment of immune surveillance within normal epithelial layers. Furthermore, CXCL14 is critical to upregulating major histocompatibility complex class I expression on tumor cells. Given these important roles, CXCL14 is often dysregulated in several types of carcinomas including cervical, colorectal, endometrial, and head and neck cancers. Its disruption has been shown to limit critical antitumor immune regulation and is correlated to poor patient prognosis. However, other studies have found that in certain cancers, namely pancreatic and some breast cancers, overexpression of stromal CXCL14 correlates with poor patient survival due to increased invasiveness. Contributing to the ambiguity CXCL14 plays in cancer is that the native CXCL14 receptor remains uncharacterized, although several candidate receptors have been proposed. Despite the complexity of CXCL14 functions, it remains clear that this chemokine is a key regulatory factor in cancer and represents a potential target for future cancer immunotherapies.
Collapse
Affiliation(s)
- Joseph A Westrich
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel W Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - Paul L Colbert
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - William C Spanos
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - Dohun Pyeon
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan
| |
Collapse
|
17
|
Kushner MH, Ory V, Graham GT, Sharif GM, Kietzman WB, Thevissen S, Yuan M, Schmidt MO, Wellstein A, Riegel AT. Loss of ANCO1 repression at AIB1/YAP targets drives breast cancer progression. EMBO Rep 2020; 21:e48741. [PMID: 31788936 PMCID: PMC6945057 DOI: 10.15252/embr.201948741] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/28/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Transcription factors critical for the transition of normal breast epithelium to ductal carcinoma in situ (DCIS) and invasive breast cancer are not clearly defined. Here, we report that the expression of a subset of YAP-activated and YAP-repressed genes in normal mammary and early-stage breast cancer cells is dependent on the nuclear co-activator AIB1. Gene expression, sequential ChIP, and ChIP-seq analyses show that AIB1 and YAP converge upon TEAD for transcriptional activation and repression. We find that AIB1-YAP repression of genes at the 1q21.3 locus is mediated by AIB1-dependent recruitment of ANCO1, a tumor suppressor whose expression is progressively lost during breast cancer progression. Reducing ANCO1 reverts AIB1-YAP-dependent repression, increases cell size, and enhances YAP-driven aberrant 3D growth. Loss of endogenous ANCO1 occurs during DCIS xenograft progression, a pattern associated with poor prognosis in human breast cancer. We conclude that increased expression of AIB1-YAP co-activated targets coupled with a loss of normal ANCO1 repression is critical to patterns of gene expression that mediate malignant progression of early-stage breast cancer.
Collapse
Affiliation(s)
- Max H Kushner
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Virginie Ory
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Garrett T Graham
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Ghada M Sharif
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - William B Kietzman
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Sophia Thevissen
- Department of Molecular MedicineGoethe UniversityFrankfurt am MainGermany
| | - Meng Yuan
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Marcel O Schmidt
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Anton Wellstein
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| | - Anna T Riegel
- Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDCUSA
| |
Collapse
|
18
|
Analysis of macrophages and neutrophils infiltrating murine mammary carcinoma sites within hours of tumor delivery. Cell Immunol 2019; 346:103929. [DOI: 10.1016/j.cellimm.2019.103929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/02/2019] [Indexed: 11/15/2022]
|
19
|
Yoon JH, Han K, Koh J, Kim GR, Kim HJ, Park YM, Youk JH, Chung J, Chae IH, Choi EJ, Moon HJ. Outcomes of Ductal Carcinoma In Situ According to Detection Modality: A Multicenter Study Comparing Recurrence Between Mammography and Breast US. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2623-2633. [PMID: 31351671 DOI: 10.1016/j.ultrasmedbio.2019.06.420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/17/2019] [Accepted: 06/27/2019] [Indexed: 06/10/2023]
Abstract
The purpose of this study was to determine whether disease recurrence and intrinsic characteristics of ductal carcinoma in situ (DCIS) are associated with the imaging method of detection in asymptomatic women. This multicenter, retrospective study included 844 women treated for asymptomatic DCIS who had pre-operative mammography and breast ultrasonography (US) studies available. Of the 844 women, 25 (3.0%) developed recurrences. Patients in the US group had significantly lower 5- and 10-y recurrence-free survival (RFS) rates compared with patients in the mammography group (p = 0.011). US-detected DCIS showed significantly lower 5-and 10-y RFS rates compared with mammography-detected DCIS in patients <50 y or with mammographically dense breasts (p = 0.002 and 0.002, respectively). US as the detection modality (hazard ratio [HR]: 4.451; 95% confidence interval [CI]: 1.530, 12.950; p = 0.006) and HER2 positivity (HR: 4.036; 95% CI: 1.438; 11.330; p = 0.008) were significantly associated with recurrence. We concluded that US as the detection modality and HER2 positivity were significantly associated with recurrence in patients treated for asymptomatic DCIS.
Collapse
Affiliation(s)
- Jung Hyun Yoon
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University, College of Medicine, Seoul, Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University, College of Medicine, Seoul, Korea
| | - Jieun Koh
- Department of Radiology, CHA Bundang Medical Center, CHA University, College of Medicine, Seongnam, Korea
| | - Ga Ram Kim
- Department of Radiology, Inha University, College of Medicine, Incheon, Korea
| | - Hye Jung Kim
- Department of Radiology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Young Mi Park
- Department of Diagnostic Radiology, Busan Paik Hospital, Inje University, College of Medicine, Busan, Korea
| | - Ji Hyun Youk
- Department of Radiology, Gangnam Severance Hospital, Yonsei University, College of Medicine, Seoul, Korea
| | - Jin Chung
- Department of Radiology, Ewha Womans University Mokdong Hospital, Ewha Womans University, College of Medicine, Seoul, Korea
| | - In Hye Chae
- Department of Radiology, National Cancer Center, Goyang, Korea
| | - Eun Jung Choi
- Department of Radiology, Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Chonbuk National University, Medical School, Jeollabuk-do, Korea
| | - Hee Jung Moon
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University, College of Medicine, Seoul, Korea.
| |
Collapse
|
20
|
Dessources K, Sebastiao APM, Pareja F, Weigelt B, Reis-Filho JS. How Did We Get There? The Progression from Ductal Carcinoma In Situ to Invasive Ductal Carcinoma. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00318-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
KRT18 is correlated with the malignant status and acts as an oncogene in colorectal cancer. Biosci Rep 2019; 39:BSR20190884. [PMID: 31345960 PMCID: PMC6692566 DOI: 10.1042/bsr20190884] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/09/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022] Open
Abstract
Keratin 18 (KRT18) has been suggested to be overexpressed in most types of human tumor, but the expression pattern of KRT18 in colorectal cancer (CRC) remained unknown. In our research, KRT18 protein expression was markedly increased in CRC cancer tissues and cell lines compared with adjacent normal colorectal tissues and normal colonic epithelial cell line, respectively. Meanwhile, we observed high KRT18 expression was associated with advanced clinical stage, deep tumor invasion, lymph node metastasis, distant metastasis, poor differentiation and unfavorable prognosis in CRC patients. Multivariate Cox regression analysis showed high expression of KRT18 was an unfavorable independent predictor for overall survival in CRC patients. The in vitro studies indicated down-regulation of KRT18 expression depressed CRC cell viability, migration and invasion. In conclusion, KRT18 serves as an oncogenic role in CRC progression and may be a therapeutic target for promoting CRC patients' prognosis.
Collapse
|
22
|
Goh CW, Wu J, Ding S, Lin C, Chen X, Huang O, Chen W, Li Y, Shen K, Zhu L. Invasive ductal carcinoma with coexisting ductal carcinoma in situ (IDC/DCIS) versus pure invasive ductal carcinoma (IDC): a comparison of clinicopathological characteristics, molecular subtypes, and clinical outcomes. J Cancer Res Clin Oncol 2019; 145:1877-1886. [PMID: 31089799 DOI: 10.1007/s00432-019-02930-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 05/06/2019] [Indexed: 12/26/2022]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) is widely recognized as the precursor of invasive ductal carcinoma (IDC). We aimed to analyze the clinicopathological characteristics and clinical outcomes of coexisting DCIS component in IDC and its clinical significance according to molecular subtypes. METHODS Data from 3001 patients with IDC (79.4%) and IDC/DCIS (20.6%) who underwent surgery from January 2009 to June 2016 were retrospectively assessed. The clinical outcomes of IDC with coexistent DCIS in different molecular subtypes were evaluated. RESULTS IDC/DCIS patients were more likely to be younger (P < 0.001), had low tumor grade (P = 0.001), had less lymph node involvement (P = 0.038) and received more mastectomy (P = 0.002) than IDC patients. In the comparison of molecular subtype prevalence, IDC/DCIS patients were more frequently presented with luminal B/HER2 positive (12.5% vs 11.0%, P < 0.001) and HER2 positive subtypes (20.9% vs 9.8%, P < 0.001). The 5-year disease-free survival (DFS, 90.9% vs 87.5%, P = 0.021) and 5-year overall survival (OS 96.1% vs 94.0%, P = 0.018) were significantly improved in IDC/DCIS patients compared to IDC patients. In multivariate analysis, the presence of coexisting DCIS (P = 0.048), tumor size (P < 0.001), lymph node status (P < 0.001), lymphovascular invasion (P = 0.007) and molecular subtypes (P < 0.001) were independent prognostic factors for DFS. Furthermore, coexistence of DCIS component in IDC significantly improved DFS in HER2 positive (94.8% vs 78.5%, P = 0.003), but had no association in luminal and triple negative subtypes. CONCLUSIONS IDC with coexisting DCIS was associated with improved prognosis. Patients with IDC/DCIS presented with more HER2 positive expression and might improve DFS in HER2 positive breast cancer.
Collapse
MESH Headings
- Breast Neoplasms/classification
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/classification
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/classification
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/mortality
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Female
- Humans
- Kaplan-Meier Estimate
- Lymphatic Metastasis
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Retrospective Studies
Collapse
Affiliation(s)
- Chih Wan Goh
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Jiayi Wu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Shuning Ding
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Caijin Lin
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Xiaosong Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Ou Huang
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Weiguo Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Yafen Li
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Li Zhu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
23
|
Agahozo MC, Sieuwerts AM, Doebar SC, Verhoef EI, Beaufort CM, Ruigrok-Ritstier K, de Weerd V, Sleddens HFBM, Dinjens WNM, Martens JWM, van Deurzen CHM. PIK3CA mutations in ductal carcinoma in situ and adjacent invasive breast cancer. Endocr Relat Cancer 2019; 26:471-482. [PMID: 30844755 DOI: 10.1530/erc-19-0019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
PIK3CA is one of the most frequently mutated genes in invasive breast cancer (IBC). These mutations are generally associated with hyper-activation of the phosphatidylinositol 3-kinase signaling pathway, which involves increased phosphorylation of AKT (p-AKT). This pathway is negatively regulated by the tumor suppressor PTEN. Data are limited regarding the variant allele frequency (VAF) of PIK3CA, PTEN and p-AKT expression during various stages of breast carcinogenesis. Therefore, the aim of this study was to gain insight into PIK3CA VAF and associated PTEN and p-AKT expression during the progression from ductal carcinoma in situ (DCIS) to IBC. We isolated DNA from DCIS tissue, synchronous IBC and metastasis when present. These samples were pre-screened for PIK3CA hotspot mutations using the SNaPshot assay and, if positive, validated and quantified by digital PCR. PTEN and p-AKT expression was evaluated by immunohistochemistry using the Histo-score (H-score). Differences in PIK3CA VAF, PTEN and p-AKT H-scores between DCIS and IBC were analyzed. PIK3CA mutations were detected in 17 out of 73 DCIS samples, 16 out of 73 IBC samples and 3 out of 23 lymph node metastasis. We detected a significantly higher VAF of PIK3CA in the DCIS component compared to the adjacent IBC component (P = 0.007). The expression of PTEN was significantly higher in DCIS compared to the IBC component in cases with a wild-type (WT) PIK3CA status (P = 0.007), while it remained similar in both components when PIK3CA was mutated. There was no difference in p-AKT expression between DCIS and the IBC component. In conclusion, our data suggest that PIK3CA mutations could be essential specifically in early stages of breast carcinogenesis. In addition, these mutations do not co-occur with PTEN expression during DCIS progression to IBC in the majority of patients. These results may contribute to further unraveling the process of breast carcinogenesis, and this could aid in the development of patient-specific treatment.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- 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
- Class I Phosphatidylinositol 3-Kinases/genetics
- Disease Progression
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Inflammatory Breast Neoplasms/genetics
- Inflammatory Breast Neoplasms/pathology
- Middle Aged
- Mutation
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Prognosis
Collapse
Affiliation(s)
| | - Anieta M Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - S Charlane Doebar
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Esther I Verhoef
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Corine M Beaufort
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Vanja de Weerd
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Hein F B M Sleddens
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Winand N M Dinjens
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | |
Collapse
|
24
|
Shee K, Muller KE, Marotti J, Miller TW, Wells WA, Tsongalis GJ. Ductal Carcinoma in Situ Biomarkers in a Precision Medicine Era: Current and Future Molecular-Based Testing. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:956-965. [PMID: 30385093 DOI: 10.1016/j.ajpath.2018.08.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/09/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
Historically, ductal carcinoma in situ (DCIS) of the breast has been managed aggressively with surgery and radiotherapy because of a risk of progression to invasive ductal carcinoma. However, this treatment paradigm has been challenged by overtreatment concerns and evidence that suggests that DCIS can be stratified according to risk of recurrence or risk of progression to invasive disease. Traditional methods of risk stratification include histologic grade and hormone receptor status. Recent technological advancements have enabled an era of precision medicine, where DCIS can be molecularly analyzed by tools, such as next-generation DNA and RNA sequencing, to identify molecular biomarkers for risk stratification. These findings have led to the development of tools such as the Oncotype DX Breast DCIS Score, a gene expression-based assay with the potential to prevent overtreatment in low-risk disease.
Collapse
Affiliation(s)
- Kevin Shee
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon
| | - Kristen E Muller
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Jonathan Marotti
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Todd W Miller
- Department of Molecular & Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon
| | - Wendy A Wells
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
25
|
Aburjania Z, Jang S, Whitt J, Jaskula-Stzul R, Chen H, Rose JB. The Role of Notch3 in Cancer. Oncologist 2018; 23:900-911. [PMID: 29622701 PMCID: PMC6156186 DOI: 10.1634/theoncologist.2017-0677] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 12/15/2022] Open
Abstract
The Notch family is a highly conserved gene group that regulates cell-cell interaction, embryogenesis, and tissue commitment. This review article focuses on the third Notch family subtype, Notch3. Regulation via Notch3 signaling was first implicated in vasculogenesis. However, more recent findings suggest that Notch3 signaling may play an important role in oncogenesis, tumor maintenance, and resistance to chemotherapy. Its role is mainly oncogenic, although in some cancers it appears to be tumor suppressive. Despite the wealth of published literature, it remains relatively underexplored and requires further research to shed more light on its role in cancer development, determine its tissue-specific function, and elaborate novel treatment strategies. Herein we summarize the role of Notch3 in cancer, possible mechanisms of its action, and current cancer treatment strategies targeting Notch3 signaling. IMPLICATIONS FOR PRACTICE The Notch family is a highly conserved gene group that regulates cell-cell interaction, embryogenesis, and tissue commitment. This review summarizes the existing data on the third subtype of the Notch family, Notch3. The role of Notch3 in different types of cancers is discussed, as well as implications of its modification and new strategies to affect Notch3 signaling activity.
Collapse
Affiliation(s)
- Zviadi Aburjania
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samuel Jang
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jason Whitt
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Renata Jaskula-Stzul
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J Bart Rose
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
26
|
Vermeulen MA, Doebar SC, van Deurzen CHM, Martens JWM, van Diest PJ, Moelans CB. Copy number profiling of oncogenes in ductal carcinoma in situ of the male breast. Endocr Relat Cancer 2018; 25:173-184. [PMID: 29203614 DOI: 10.1530/erc-17-0338] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023]
Abstract
Characterizing male breast cancer (BC) and unraveling male breast carcinogenesis is challenging because of the rarity of this disease. We investigated copy number status of 22 BC-related genes in 18 cases of pure ductal carcinoma in situ (DCIS) and in 49 cases of invasive carcinoma (IC) with adjacent DCIS (DCIS-AIC) in males using multiplex ligation-dependent probe amplification (MLPA). Results were compared to female BC and correlated with survival. Overall, copy number ratio and aberration frequency including all 22 genes showed no significant difference between the 3 groups. Individual unpaired analysis revealed a significantly higher MTDH copy number ratio in IC compared to DCIS-AIC and pure DCIS (P = 0.009 and P = 0.038, respectively). ADAM9 showed a significantly lower copy number aberration frequency in male BC, compared to female BC (P = 0.020). In DCIS-AIC, MTDH, CPD, CDC6 and TOP2A showed a lower frequency of copy number increase in males compared to females (P < 0.001 for all 4 genes). In IC, CPD gain and CCNE1 gain were independent predictors of poor overall survival. In conclusion, male DCIS and IC showed a similar copy number profile for 21 out of 22 interrogated BC-related genes, illustrating their clonal relation and the genetically advanced state of male DCIS. MTDH showed a higher copy number ratio in IC compared to adjacent and pure DCIS and may therefore play a role in male breast carcinogenesis. Differences were detected between male and female DCIS for 4 genes pointing to differences in breast carcinogenesis between the sexes.
Collapse
Affiliation(s)
- Marijn A Vermeulen
- Department of PathologyUniversity Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Shusma C Doebar
- Department of PathologyErasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Carolien H M van Deurzen
- Department of PathologyErasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- BOOG Study Center/Dutch Breast Cancer Research GroupAmsterdam, The Netherlands
| | - John W M Martens
- BOOG Study Center/Dutch Breast Cancer Research GroupAmsterdam, The Netherlands
- Department of Medical Oncology and Cancer Genomics NetherlandsErasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Paul J van Diest
- Department of PathologyUniversity Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Cathy B Moelans
- Department of PathologyUniversity Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|