1
|
Loudig O, Mitchell MI, Ben-Dov IZ, Liu C, Fineberg S. MiRNA expression deregulation correlates with the Oncotype DX ® DCIS score. BREAST CANCER RESEARCH : BCR 2022; 24:62. [PMID: 36096802 PMCID: PMC9469592 DOI: 10.1186/s13058-022-01558-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022]
Abstract
Background Current clinical criteria do not discriminate well between women who will or those who will not develop ipsilateral invasive breast cancer (IBC), or a DCIS recurrence after a ductal carcinoma in situ (DCIS) diagnosis. The 12-gene Oncotype DX® DCIS assay (RT qPCR gene-based scoring system) was established and shown to predict the risk of subsequent ipsilateral IBC or DCIS recurrence. Recent studies have shown that microRNA (miRNA) expression deregulation can contribute to the development of IBC, but very few have evaluated miRNA deregulation in DCIS lesions. In this study, we sought to determine whether specific miRNA expression changes may correlate with Oncotype DX® DCIS scores. Methods For this study, we used archived formalin-fixed, paraffin-embedded (FFPE) specimens from 41 women diagnosed with DCIS between 2012 and 2018. The DCIS lesions were stratified into low (n = 26), intermediate (n = 10), and high (n = 5) risk score groups using the Oncotype DX® DCIS assay. Total RNA was extracted from DCIS lesions by macro-dissection of unstained FFPE sections, and next-generation small-RNA sequencing was performed. We evaluated the correlation between miRNA expression data and Oncotype score, as well as patient age. RT-qPCR validations were performed to validate the topmost differentially expressed miRNAs identified between the different risk score groups. Results MiRNA sequencing of 32 FFPE DCIS specimens from the three different risk group scores identified a correlation between expression deregulation of 17 miRNAs and Oncotype scores. Our analyses also revealed a correlation between the expression deregulation of 9 miRNAs and the patient’s age. Based on these results, a total of 15 miRNAs were selected for RT-qPCR validation. Of these, miR-190b (p = 0.043), miR-135a (p = 0.05), miR-205 (p = 0.00056), miR-30c (p = 0.011), and miR-744 (p = 0.038) showed a decreased expression in the intermediate/high Oncotype group when compared to the low-risk score group. A composite risk score was established using these 5 miRNAs and indicated a significant association between miRNA expression deregulation and the Oncotype DX® DCIS Score (p < 0.0021), between high/intermediate and low risk groups. Conclusions Our analyses identified a subset of 5 miRNAs able to discriminate between Oncotype DX® DCIS score subgroups. Together, our data suggest that miRNA expression analysis may add value to the predictive and prognostic evaluation of DCIS lesions. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-022-01558-4.
Collapse
Affiliation(s)
- Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA.
| | - Megan I Mitchell
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Iddo Z Ben-Dov
- Department of Nephrology and Hypertension, Hadassah Medical Center, 91120, Jerusalem, Israel
| | - Christina Liu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Susan Fineberg
- Department of Pathology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| |
Collapse
|
2
|
Miripour ZS, Abbasvandi F, Aghaee P, Shojaeian F, Faramarzpour M, Mohaghegh P, Hoseinpour P, Namdar N, Hassanpour Amiri M, Ghafari H, Parniani M, Kaviani A, Alamdar S, NajafiKhoshnoo S, Sanati H, Mapar M, Sadeghian N, Akbari ME, Yunesian M, Abdolahad M. Human study on cancer diagnostic probe (CDP) for real-time excising of breast positive cavity side margins based on tracing hypoxia glycolysis; checking diagnostic accuracy in non-neoadjuvant cases. Cancer Med 2022; 11:1630-1645. [PMID: 35224879 PMCID: PMC8986141 DOI: 10.1002/cam4.4503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/25/2021] [Accepted: 11/18/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cancer diagnostic probe (CDP) had been developed to detect involved breast cavity side margins in real-time (Miripour et al. Bioeng Transl Med. e10236.). Here, we presented the results of the in vivo human model CDP studies on non-neoadjuvant cases. METHODS This study is a prospective, blind comparison to a gold standard, and the medical group recruited patients. CDP and frozen data were achieved before the permanent pathology experiment. The main outcome of the study is surgical margin status. From November 2018 to April 2020, 202 patients were registered, and 188 were assigned for the study. Breast-conserving surgery at any age or gender, re-surgery due to re-currency, or involved margins are acceptable. Patients must be non-neoadjuvant. The reliability of CDP scoring had been evaluated by the pathology of the scored IMs. Then, three models of the study were designed to compare CDP with the frozen sections. Receiver operating characteristic (ROC) curves and AUC were measured based on the permanent postoperative pathology gold standard. RESULTS A matched clinical diagnostic categorization between the pathological results of the tested IMs and response peaks of CDP on 113 cases, was reported (sensitivity = 97%, specificity = 89.3%, accuracy = 92%, positive predictive value (PPV) = 84.2%, and negative predictive value (NPV) = 98%). Study A showed the independent ability of CDP for IM scoring (sensitivity = 80%, specificity = 90%, accuracy = 90%, PPV = 22.2%, and NPV = 99.2%). Study B showed the complementary role of CDP to cover the missed lesions of frozen sections (sensitivity = 93.8%, specificity = 91%, accuracy = 91%, PPV = 55.6%, and NPV = 99.2%). Study C showed the ability of CDP in helping the pathologist to reduce his/her frozen miss judgment (specificity = 92%, accuracy = 93%, PPV = 42.1%, and NPV = 100%). Results were reported based on the post-surgical permanent pathology gold standard. CONCLUSION CDP scoring ability in intra-operative margin detection was verified on non-neoadjuvant breast cancer patients. Non-invasive real-time diagnosis of IMs with pathological values may make CDP a distinct tool with handheld equipment to increase the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Zohreh Sadat Miripour
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Fereshteh Abbasvandi
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
- Cancer Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Parisa Aghaee
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Fatemeh Shojaeian
- Cancer Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Mahsa Faramarzpour
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Pooneh Mohaghegh
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | | | - Naser Namdar
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Morteza Hassanpour Amiri
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Hadi Ghafari
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Mohammad Parniani
- Pathology DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
| | - Ahmad Kaviani
- Institute of CancerImam Khomeini HospitalTehran University of Medical SciencesTehranIran
| | - Sedigheh Alamdar
- Department of PathobiologyIran University of Medical SciencesShahid Hemmat HighwayTehranIran
| | - Sahar NajafiKhoshnoo
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Hassan Sanati
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
| | - Mahna Mapar
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
| | - Nastaran Sadeghian
- ATMP DepartmentBreast Cancer Research CenterMotamed Cancer InstituteACECRTehranIran
| | | | - Masud Yunesian
- Department of Environmental HealthSchool of Public HealthTehran University of Medical SciencesTehranIran
- Department of Research Methodology and Data AnalysisInstitute for Environmental ResearchTehran University of Medical SciencesTehranIran
| | - Mohammad Abdolahad
- Nano Bio Electronic Devices LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of ExcellenceThin Film and Nanoelectronics LabSchool of Electrical and Computer EngineeringCollege of EngineeringUniversity of TehranTehranIran
- Institute of CancerImam Khomeini HospitalTehran University of Medical SciencesTehranIran
- UT&TUMS Cancer Electronics Research CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
3
|
Miripour ZS, Abbasvandi F, Aghaee P, NajafiKhoshnoo S, Faramarzpour M, Mohaghegh P, Hoseinpour P, Namdar N, Amiri MH, Ghafari H, Zareie S, Shojaeian F, Sanati H, Mapar M, Sadeghian N, Akbari ME, Khayamian MA, Abdolahad M. Electrochemical tracing of hypoxia glycolysis by carbon nanotube sensors, a new hallmark for intraoperative detection of suspicious margins to breast neoplasia. Bioeng Transl Med 2022; 7:e10236. [PMID: 35079624 PMCID: PMC8780057 DOI: 10.1002/btm2.10236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/18/2021] [Indexed: 12/03/2022] Open
Abstract
For most people, the first step in treatment is to take out the tumor (surgery), so precise and fast diagnosis of any sign of high-risk and neoplastic cells, especially in surgical cavity margins, is significant. The frozen pathology method is the conventional standard of intraoperative diagnosis, but the low number of slides prepared from non-fixed tissues prevents us from achieving a perfect diagnosis. Although many improvements in intraoperative margin detection were achieved, still real-time detection of neoplastic lesions is crucial to improving diagnostic quality. Functionalized carbon nanotubes grown on the electrode needles lively and selectively determine the H2O2 released from cancer/atypical cells through reverse Warburg effect and hypoxia assisted glycolysis pathways in a quantitative electrochemical manner. The study was carried out on cell lines, 57 in vivo mice models with breast cancer, and 258 fresh in vitro samples of breast cancer tumors. A real-time electrotechnical system, named cancer diagnostic probe (CDP) (US Patent Pub. No.: US 2018/02991 A1, US 2021/0007638 A1, and US 2021/0022650 A1 [publications], and US 10,786,188 B1 [granted]), has been developed to find pre-neoplastic/neoplastic cells in vivo in a quantitative electrochemical manner by tracing hypoxia glycolysis byproducts. Matched pathological evaluations with response peaks of CDP were found based on the presence of neoplasia (from atypia to invasive carcinoma) in live breast tissues. The ability of CDP to find neoplastic lesions in mice models in vivo and fresh breast tumors in vitro was verified with sensitivity and specificity of 95% and 97%, respectively. The system may help a surgeon assistant system for usage in the operating room after passing many trials and standard examinations in the future.
Collapse
Affiliation(s)
- Zohreh Sadat Miripour
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Fereshteh Abbasvandi
- ATMP DepartmentBreast Cancer Research Center, Motamed Cancer Institute, ACECRTehranIran
| | - Parisa Aghaee
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Sahar NajafiKhoshnoo
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Mahsa Faramarzpour
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Pooneh Mohaghegh
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | | | - Naser Namdar
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Morteza Hassanpour Amiri
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Hadi Ghafari
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Sarah Zareie
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Fatemeh Shojaeian
- Cancer Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Hassan Sanati
- Integrative Oncology DepartmentBreast Cancer Research Center, Motamed Cancer Institute, ACECRTehranIran
| | - Mahna Mapar
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Nastaran Sadeghian
- ATMP DepartmentBreast Cancer Research Center, Motamed Cancer Institute, ACECRTehranIran
| | | | - Mohammad Ali Khayamian
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
| | - Mohammad Abdolahad
- Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Nano Electronic Center of Excellence, Thin Film and Nanoelectronics Lab, School of Electrical and Computer Engineering, College of EngineeringUniversity of TehranTehranIran
- Cancer Institute, Imam Khomeini HospitalTehran University of Medical SciencesTehranIran
- UT&TUMS Cancer Electrotechnique Research CenterYAS Hospital, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
4
|
Dabbagh N, Abbasvandi F, Miripour ZS, Hoseinpour P, Jahanbakhshi F, Moradi A, Riazi H, Moradian F, Zanjani FS, Parniani M, Akbari ME, Abdolahad M. Accuracy of cancer diagnostic probe for intra-surgical checking of cavity side margins in neoadjuvant breast cancer cases: A human model study. Int J Med Robot 2021; 18:e2335. [PMID: 34571582 DOI: 10.1002/rcs.2335] [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: 05/17/2021] [Revised: 09/07/2021] [Accepted: 09/22/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Background Recently, a real-time system, named cancer diagnostic probe (CDP), has been developed to diagnose the presence of pre-neoplastic/neoplastic cells in breast cavity side margins. Detecting mechanism is real-time determination of the ROS/H2 O2 released from cancer or atypical cells, through reverse Warburg effect and hypoxia glycolysis pathways. AIMS Here, we designed a human model study based on real-time checking of 387 internal margins (IM) from 39 neoadjuvant breast cancer cases by CDP. MATERIALS & METHODS: Each lesion was checked by entered needle sensor and electrical scores were recorded. The permanent pathology result of each tested lesion was our gold standard to evaluate CDP scoring. CDP results were compared with permanent pathology of tumour side margins (as a conventional margin evaluation procedure). RESULTS Results showed that the sensitivity of CDP in scoring the cavity side margins of those cases is 91%. A total of 18 involved IM which had been detected by CDP were declared as free margins in pathology section of tumour side samples. Just five involved IM were missed by CDP. DISCUSSIONS Such sensitivity revealed that metabolism based (here: hypoxia glycolysis) tracing of cancer cells show distinct electrochemical responses between clear and involved cavity side margin evaluation. CONCLUSION This human study showed the promising role of CDP to achieve clear margins after BCS of neoadjuvant cases.
Collapse
Affiliation(s)
- Najmeh Dabbagh
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran.,Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Abbasvandi
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran.,ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.,SEPAS Pathology Laboratory, Tehran, Iran
| | - Zohreh Sadat Miripour
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Parisa Hoseinpour
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran.,SEPAS Pathology Laboratory, Tehran, Iran
| | - Fahimeh Jahanbakhshi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Afshin Moradi
- Department of Pathology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hooman Riazi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Moradian
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Parniani
- SEPAS Pathology Laboratory, Tehran, Iran.,Pathology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Mohammad Abdolahad
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran.,School of Medicine, Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran.,UT-TUMS Cancer Electronics Research Center, Tehran, Iran
| |
Collapse
|
5
|
Li X, Wang H, Sun Z, Fan C, Jin F, Mao X. A retrospective observational study of intraductal breast papilloma and its coexisting lesions: A real-world experience. Cancer Med 2020; 9:7751-7762. [PMID: 32822113 PMCID: PMC7571817 DOI: 10.1002/cam4.3308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/09/2020] [Accepted: 06/20/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Breast intraductal papilloma is a heterogeneous group. The aim of the study is to investigate the intraductal breast papilloma and its coexisting lesions retrospectively in real-world practice. METHODS We retrospectively identified 4450 intraductal breast papilloma and its coexisting lesions. RESULTS About 18.36% of intraductal papilloma coexisted with malignant lesions of the breast, 37.33% coexisted with atypia hyperplasia (AH), 25.24% coexisted with benign lesions, and only 19.10% coexisted without concomitant lesions. In addition, 36.80% of intraductal breast papilloma had nipple discharge, 51.46% had a palpable breast mass, and 16.45% had both nipple discharge and a palpable breast mass. About 28.18% experienced discomfort or were asymptomatic. Furthermore, 98.99% had ultrasound abnormalities, and 53.06% had intraductal hypoechogenicity upon ultrasound. 31.89% had mammographic distortion, and 14.45% had microcalcification upon mammography. Intraductal breast papilloma with malignancy had significant correlations with clinical manifestations. CONCLUSION Coexisting malignancy was also related to ultrasound abnormality (BIRADS 4C and 5), mammographic distortion, and microcalcification upon mammography but was not related to the intraductal hypoechoic upon ultrasound. Coexisting atypical hyperplasia correlated with nipple discharge but not palpable mass, mammographic distortion, or intraductal hypoechoic upon ultrasound. The coexisting AH was also related to abnormality upon ultrasound or microcalcification compared with the benign lesions. The intraductal papilloma coexists with malignancy or AH accounted for more than 50%, and the clinical information on papilloma and its coexisting lesions is nonspecific. We recommended surgical treatment for benign intraductal papillary lesions.
Collapse
Affiliation(s)
- Xiaona Li
- Department of Breast SurgeryThe First Affiliated Hospital of China Medical University & School of Fundamental ScienceChina Medical UniversityShenyangPeople’s Republic of China
| | - Huan Wang
- Department of GynecologyThe First Affiliated Hospital of China Medical UniversityShenyangPeople’s Republic of China
| | - Zhe Sun
- Department of Breast SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangPeople’s Republic of China
| | - Chuifeng Fan
- Department of PathologyThe First Affiliated Hospital and College of Basic Medical Sciences of China Medical UniversityShenyangPeople’s Republic of China
| | - Feng Jin
- Department of Breast SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangPeople’s Republic of China
| | - Xiaoyun Mao
- Department of Breast SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangPeople’s Republic of China
| |
Collapse
|
6
|
Mahdavi R, Hosseinpour P, Abbasvandi F, Mehrvarz S, Yousefpour N, Ataee H, Parniani M, Mamdouh A, Ghafari H, Abdolahad M. Bioelectrical pathology of the breast; real-time diagnosis of malignancy by clinically calibrated impedance spectroscopy of freshly dissected tissue. Biosens Bioelectron 2020; 165:112421. [PMID: 32729539 DOI: 10.1016/j.bios.2020.112421] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 10/23/2022]
Abstract
In this paper, freshly (non-fixed) dissected tissues obtained from breast cancer surgery were impedimetrically and pathologically scanned, analyzed, and probable electro-pathological mutual matching was investigated. A new electrical model was proposed for pathological scores of breast lesions based on the theory of electric current dispersion by different types of biological tissues. This integrated handheld bioimpedance sensor named EPA would score the clearance or malignancy involvement of dissected tumor margins by introducing two crucial classification parameters named Z1kHz and IPS (impedance phase slope in the frequency ranges of 100-500 kHz). EPA benefits from a precise signal recording and analysis method which leads to the detection of the presence of even about 5% distribution of premalignant cells among healthy breast tissue. EPA can be clinically used by pathologists, as a complementary device, for real-time diagnosis of suspicious margins of dissected tumors to declare more precise intraoperative diagnosis by scanning all around the dissected tissues. Each data sampling and analysis covers 2 mm of the surface in less than 5 s. Measurements on about 313 human breast tumor margins showed more than 90% accuracy and near 93% specificity for EPA as an independent diagnostic tool.
Collapse
Affiliation(s)
- Reihane Mahdavi
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran
| | - Parisa Hosseinpour
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran; SEPAS Pathology Laboratory, P.O.Box: 1991945391, Tehran, Iran
| | - Fereshteh Abbasvandi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX 15179/64311, Tehran, Iran
| | - Sajad Mehrvarz
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran
| | - Narges Yousefpour
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran
| | - Hossein Ataee
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran
| | - Mohammad Parniani
- Department of Clinical Investigations, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX 15179/64311, Tehran, Iran
| | - Amir Mamdouh
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran
| | - Hadi Ghafari
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran
| | - Mohammad Abdolahad
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical AndComputer Engineering, Faculty of Engineering, University of Tehran, Tehran, P.O. Box 14395/515, Iran.
| |
Collapse
|
7
|
Chan PMY, Chotai N, Lai ES, Sin PY, Chen J, Lu SQ, Goh MH, Chong BK, Ho BCS, Tan EY. Majority of flat epithelial atypia diagnosed on biopsy do not require surgical excision. Breast 2018; 37:13-17. [DOI: 10.1016/j.breast.2017.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/14/2017] [Accepted: 10/10/2017] [Indexed: 11/16/2022] Open
|
8
|
|
9
|
Ozerdem U, Tavassoli FA. Distribution pattern of Ki67 immunoreactivity in ductal intraepithelial neoplasia: Correlation with lesion grade and potential utility. Pathol Res Pract 2016; 212:872-875. [DOI: 10.1016/j.prp.2016.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 07/07/2016] [Indexed: 11/26/2022]
|
10
|
Mao X, Qiao Z, Fan C, Guo A, Yu X, Jin F. Expression pattern and methylation of estrogen receptor α in breast intraductal proliferative lesions. Oncol Rep 2016; 36:1868-74. [PMID: 27498697 PMCID: PMC5022874 DOI: 10.3892/or.2016.4988] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/13/2016] [Indexed: 01/12/2023] Open
Abstract
Intraductal proliferative lesions of the breast including usual ductal hyperplasia (UDH), atypical ductal hyperplasia (ADH) and ductal carcinoma in situ (DCIS) are associated with increased risk, albeit of greatly different magnitudes, for the subsequent development of invasive carcinoma. Estrogen receptor α (ERα) has been widely accepted as a prognostic marker and a predictor for endocrine therapy response of breast cancer. To investigate the ERα expression and methylation in breast intraductal proliferative lesions, we analyzed ERα expression in breast intraductal proliferative lesions including pure UDH (N=98), ADH without DCIS (N=160), DCIS without invasive breast cancer (N=149) by immunohistochemistry. Furthermore, the methylation status of ERα by methylation-specific PCR (MSP) was defined in 217 cases of breast intraductal proliferative lesions. Immunohistochemistry showed that 98/98 (100%) of the UDH cases were positive for ERα expression. ERα protein expression in ADH (132/160) (92.5%) was higher than in DCIS (101/149) (67.8%). But the ERα expression pattern was different with histological diversity of breast intraductal proliferative lesions. The average percent cells staining positive for ERα was 35.33% in UDH, 87.75% in ADH and 71.45% in DCIS. ERα methylation in 32/60 (53.3%) UDH, 11/77 (10.2%) ADH and 32/80 (40.0%) DCIS. Our results demonstrated a strong negative correlation between the percent of cells staining positive for ERα and ERα methylation (r=−0.831, p<0.001). Taken together, our results underlined that ERα expression or methylation may be involved in the breast carcinogenesis and advancement, thus it is not parallel to breast cancer risk in breast intraductal proliferative lesions. No obvious watershed between ERα-positive and -negative breast carcinogenesis was established. Estrogen receptor (ER) methylation or expression is a reversible signal in breast carcinogenesis which affected biological behavior of cells.
Collapse
Affiliation(s)
- Xiaoyun Mao
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhen Qiao
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chuifeng Fan
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ayao Guo
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinmiao Yu
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Feng Jin
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
11
|
Management of flat epithelial atypia on breast core biopsy may be individualized based on correlation with imaging studies. Mod Pathol 2015; 28:670-6. [PMID: 25412845 DOI: 10.1038/modpathol.2014.159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 10/15/2014] [Accepted: 10/16/2014] [Indexed: 11/08/2022]
Abstract
Flat epithelial atypia of the breast commonly co-exists with atypical ductal hyperplasia, lobular neoplasia, and indolent forms of invasive carcinomas such as tubular carcinoma. Most patients with pure flat epithelial atypia on core biopsy undergo surgical excision to evaluate for carcinoma in the adjacent breast tissue. Studies to date have reported varying upgrade rates with most recommending follow-up excision. These studies have often lacked detailed radiographic correlation, central review by breast pathologists and information regarding the biology of the carcinomas identified upon excision. In this study, we report the frequency of upgrade to invasive carcinoma or ductal carcinoma in situ in excision specimens following a diagnosis of pure flat epithelial atypia on core biopsy. Radiographic correlation is performed for each case and grade/receptor status of detected carcinomas is reported. Seventy-three (73) core biopsies containing pure flat epithelial atypia were identified from our files, meeting inclusion criteria for the study. In the subsequent excision biopsies, five (7%) cases contained invasive carcinoma or ductal carcinoma in situ and seventeen (23%) contained atypical ductal hyperplasia or lobular neoplasia. All of the ductal carcinoma in situ cases with estrogen receptor results were estrogen receptor positive and intermediate grade. The invasive tumors were small (pT1a) hormone receptor-positive, HER2-negative, low-grade invasive ductal or tubular carcinomas with negative sentinel lymph-node biopsies. No upgrades were identified in the 14 patients who had all of their calcifications removed by the stereotactic core biopsy. Our rate of upgrade to carcinoma, once cases with discordant imaging are excluded, is at the lower end of the range reported in the literature. Given the low upgrade rate and indolent nature of the carcinomas associated with flat epithelial atypia, case management may be individualized based on clinical and radiographic findings. Excision may not be necessary for patients without remaining calcifications following core biopsy.
Collapse
|
12
|
Curigliano G, Disalvatore D, Esposito A, Pruneri G, Lazzeroni M, Guerrieri-Gonzaga A, Luini A, Orecchia R, Goldhirsch A, Rotmensz N, Bonanni B, Viale G. Risk of subsequentin situ and invasive breast cancer in human epidermal growth factor receptor 2-positive ductal carcinomain situ. Ann Oncol 2015; 26:682-687. [DOI: 10.1093/annonc/mdv013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
13
|
Narod SA, Rakovitch E. A comparison of the risks of in-breast recurrence after a diagnosis of dcis or early invasive breast cancer. ACTA ACUST UNITED AC 2014; 21:119-24. [PMID: 24940092 DOI: 10.3747/co.21.1892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND It is controversial whether ductal carcinoma in situ (dcis) is a preinvasive marker of breast cancer or if it is part of a spectrum of small cancers with malignant potential. Comparing clinical outcomes in women with invasive and noninvasive breast lesions might help to resolve the issue. METHODS From a database of 2641 patients with breast cancer, we selected women who had been treated with breast-conserving surgery for a cancer that was 2.0 cm or less in size, node-negative, and nonpalpable. No subject received chemotherapy. Cancers were categorized as noninvasive (stage 0, n = 172) or invasive (stage 1, n = 401) based on a review of the pathology records. We compared the actuarial risks of in-breast recurrence after invasive and noninvasive breast lesions before and after adjusting for tamoxifen and radiotherapy. RESULTS The 18-year cumulative risk of in-breast recurrence was 35.2% for patients with dcis and 12.8% for patients with small invasive cancers (hazard ratio: 2.4; 95% confidence interval: 1.5 to 3.8; p < 0.0003). After adjustment for radiotherapy and tamoxifen treatment, the difference was small and nonsignificant (hazard ratio: 1.4; 95% confidence interval: 0.9 to 2.4; p = 0.22). CONCLUSIONS For women with small, nonpalpable, node-negative breast cancers, the likelihood of experiencing an in-breast recurrence was associated with radiotherapy and with tamoxifen, but not with the presence of cancer cells invading beyond the basement membrane.
Collapse
Affiliation(s)
- S A Narod
- Women's College Research Institute, Women's College Hospital, and the Dalla Lana School of Public Health University of Toronto, Toronto, ON
| | - E Rakovitch
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON
| |
Collapse
|
14
|
Sun Y, Mao X, Fan C, Liu C, Guo A, Guan S, Jin Q, Li B, Yao F, Jin F. CXCL12-CXCR4 axis promotes the natural selection of breast cancer cell metastasis. Tumour Biol 2014; 35:7765-73. [PMID: 24810923 PMCID: PMC4158177 DOI: 10.1007/s13277-014-1816-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 03/04/2014] [Indexed: 12/14/2022] Open
Abstract
CXCR4 and its ligand CXCL12 can promote the proliferation, survival, and invasion of cancer cells. They have been shown to play an important role in regulating metastasis of breast cancer to specific organs. High CXCR4 expression was also correlated to poor clinical outcome. Previous study also showed that tumor cells express a high level of CXCR4 and that tumor metastasis target tissues (lung, liver, and bone) express high levels of the ligand CXCL12, allowing tumor cells to directionally migrate to target organs via a CXCL12-CXCR4 chemotactic gradient. However, the exact mechanisms of how CXCR4 and CXCL12 enhance metastasis and/or tumor growth and their full implications on breast cancer progression are unknown. Yet it is likely that chemokine receptor signaling may provide more than just a migrational advantage by also helping the metastasized cells establish and survive in secondary environments. In this study, we investigated CXCR4 and CXCL12 expression in breast cancer and analyzed its association with clinicopathological factors by immunohistochemistry first. Then, we detected the mRNA and protein expression of CXCR4 and CXCL12 in breast cancer cell lines by Western blot and RT-PCR. The MDA-MB-231 has CXCR4 expression and very weak CXCL12 expression. So, we constructed the functional CXCL12 expression in MDA-MB-231 using a gene transfection technique. Further experiments were conducted to evaluate the effect of CXCL12 transfection on the biological behaviors of MDA-MB-231. The cell proliferation of MDA-MB-231–CXCL12 was accessed by MTT assay; the apoptosis was analyzed by an AnnexinV-FITC/propidium iodide double staining of flow cytometry method; and the cell invasive ability was examined by Matrigel invasion assay. Immunohistochemical analysis showed the co-expression of CXCR4 and CXCL12 correlated with lymph node metastasis and TNM stage (p < 0.01). It suggested that the chemokine CXCL12 and its sole ligand CXCR4 play important role in the malignance of breast cancer. To gain a deeper insight into it, we picked CXCR4-expressing cells MDA-MB-231 to be transfected with CXCL12 stably. The decreased cellular proliferation, increased apoptosis, and invasive ability were found in MDA-MB-231 with successful CXCL12 transfection (p < 0.05). Our findings underlined the CXCL12-CXCR4 axis correlated tightly with breast cancer metastasis. CXCL12-CXCR4 axis can increase the invasion and apoptosis of MDA-MB-231 simultaneously. These data strongly support the hypothesis that CXCL12-CXCR4 axis promotes the natural selection of breast cancer cell metastasis. Our findings could have significant implications in terms of breast cancer aggressiveness and the effectiveness of targeting the receptors and downstream signaling pathways for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yanan Sun
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning, 110001, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bijker N, Donker M, Wesseling J, den Heeten GJ, Rutgers EJT. Is DCIS breast cancer, and how do I treat it? Curr Treat Options Oncol 2013; 14:75-87. [PMID: 23239193 DOI: 10.1007/s11864-012-0217-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ductal carcinoma in situ (DCIS) is a pre-invasive stage of breast cancer with a heterogeneous clinical behaviour. Since the introduction of mammographic screening programmes, the incidence of DCIS has shown a dramatic increase. Treatment should focus on the prevention of progression to invasive disease. If progression occurs, poorly differentiated DCIS frequently gives rise to grade III invasive breast cancer, whereas well differentiated DCIS more often recurs as grade I invasive disease. However, at present, validated diagnostic test are lacking to predict progression accurately. The majority of women with DCIS are suitable for breast conserving therapy. Obtaining clear surgical margins is the most important goal of a local excision. Radiotherapy is effective in reducing the risk of local recurrence with about 50 % in all subgroups of patients with DCIS. (Breast cancer specific) survival of women with DCIS is excellent, and radiotherapy does not further improve this. Future research should be directed in enabling to select women who have a high risk of--invasive--recurrence, so in which radiotherapy should be standard part of the breast conserving approach, and those women with a more indolent lesion, in which after surgery a watchful waiting approach can be followed.
Collapse
Affiliation(s)
- N Bijker
- Department of Radiation Oncology, Academic Medical Center, P.O. Box 22700, 1100DE, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
16
|
DCIS and LCIS are confusing and outdated terms. They should be abandoned in favor of ductal intraepithelial neoplasia (DIN) and lobular intraepithelial neoplasia (LIN). Breast 2013; 22:431-5. [PMID: 23643807 DOI: 10.1016/j.breast.2013.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/03/2013] [Indexed: 11/22/2022] Open
Abstract
The terms ductal and lobular intraepithelial neoplasia (DIN and LIN) were introduced by Tavossoli 15 years ago, who proposed they should replace, respectively, ductal and lobular carcinoma in situ (DCIS and LCIS). This proposal has been slowly gaining ground. We argue that DCIS and LCIS should now be definitively abandoned. Bringing together 'in situ' and other entities into the simpler and more logical DIN/LIN framework--as has been done with intraepithelial neoplasias of cervix, vagina, vulva, prostate, and pancreas--would eliminate the artificial and illogical distinctions between 'not cancers' (e.g. flat epithelial atypia, atypical ductal hyperplasia--now classified as low grade DIN) and 'cancers' (e.g. DCIS--now considered medium-high grade DIN). Elimination of the term 'carcinoma' from entities that cannot metastasize will reduce confusion among health professionals and patients, and contribute to reducing the risk of overtreatment, as well as reducing adverse psychological reactions in patients.
Collapse
|
17
|
Lazzeroni M, Guerrieri-Gonzaga A, Botteri E, Leonardi MC, Rotmensz N, Serrano D, Varricchio C, Disalvatore D, Del Castillo A, Bassi F, Pagani G, DeCensi A, Viale G, Bonanni B, Pruneri G. Tailoring treatment for ductal intraepithelial neoplasia of the breast according to Ki-67 and molecular phenotype. Br J Cancer 2013; 108:1593-601. [PMID: 23579208 PMCID: PMC3668474 DOI: 10.1038/bjc.2013.147] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: The post-surgical management of ductal intraepithelial neoplasia (DIN) of the breast is still a dilemma. Ki-67 labelling index (LI) has been proposed as an independent predictive and prognostic factor in early breast cancer. Methods: The prognostic and predictive roles of Ki-67 LI were evaluated with a multivariable Cox regression model in a cohort of 1171 consecutive patients operated for DIN in a single institution from 1997 to 2007. Results: Radiotherapy (RT) was protective in subjects with DIN with Ki-67 LI ≥14%, whereas no evidence of benefit was seen for Ki-67 LI <14%, irrespective of nuclear grade and presence of necrosis. Notably, the higher the Ki-67 LI, the stronger the effect of RT (P-interaction <0.01). Hormonal therapy (HT) was effective in both Luminal A (adjusted hazard ratio (HR)=0.56 (95% CI, 0.33–0.97)) and Luminal B/Her2neg DIN (HR 0.51 (95% CI, 0.27–0.95)). Conclusion: Our data suggest that Ki-67 LI may be a useful prognostic and predictive adjunct in DIN patients. The Ki-67 LI of 14% could be a potential cutoff for better categorising this population of women at increased risk for breast cancer and in which adjuvant treatment (RT, HT) should be differently addressed, independent of histological grade and presence of necrosis.
Collapse
Affiliation(s)
- M Lazzeroni
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ruddy KJ, Meyer ME, Giobbie-Hurder A, Emmons KM, Weeks JC, Winer EP, Partridge AH. Long-term risk perceptions of women with ductal carcinoma in situ. Oncologist 2013; 18:362-8. [PMID: 23568001 DOI: 10.1634/theoncologist.2012-0376] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Previous research has demonstrated that many women with ductal carcinoma in situ (DCIS) overestimate their risk for future breast cancer at the time of diagnosis and soon thereafter. This study aims to evaluate risk perceptions after 5 years. PATIENTS AND METHODS In a longitudinal cohort study, we mailed long-term follow-up surveys to 315 women who had previously responded to a survey 18 months after they were diagnosed with DCIS, excluding those who had experienced recurrence and those not treated at our institution. We evaluated risk perceptions with items used previously in the cohort. RESULTS One hundred ninety-three women (61%) responded. The median time since diagnosis was 5.9 years. We excluded 12 because of recurrence. Of the 181 remaining, 32% perceived at least a moderate 5-year risk for developing DCIS again, 43% perceived at least a moderate lifetime risk for developing DCIS again, 27% perceived at least a moderate 5-year risk for invasive breast cancer, 38% perceived at least a moderate lifetime risk for invasive breast cancer, and 24% perceived at least a moderate risk for DCIS spreading to other body parts. In a multivariate model, worse financial status and higher perceived risk in the previous survey were the only predictors of at least a moderate perception of risk for DCIS spreading. CONCLUSION Women with a history of DCIS continue to harbor inaccurate perceptions of their risk for future breast cancer events even 5 years after diagnosis.
Collapse
Affiliation(s)
- Kathryn J Ruddy
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Mao XY, Chen H, Wang H, Wei J, Liu C, Zheng HC, Yao F, Jin F. MTA1 expression correlates significantly with ER-alpha methylation in breast cancer. Tumour Biol 2012; 33:1565-72. [PMID: 22644675 DOI: 10.1007/s13277-012-0410-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 04/24/2012] [Indexed: 01/24/2023] Open
Abstract
Metastasis tumor antigen 1 (MTA1), a novel candidate metastasis-associated gene, is known to increase the migration and invasion of various tumor cells in vitro. It also plays an important role in tumorigenesis and tumor aggressiveness of breast cancer. Estrogen receptor alpha (ERα) plays an important role in the etiology of breast cancer and has been widely accepted as a prognostic marker for breast cancer and a response predictor for endocrine therapy. The ERα gene methylation has been linked to the lack of ERα expression in breast cancer. The aim of the study is to assess the correlation between the ERα methylation and MTA1 expression in breast cancer and further to investigate whether the repressed ERα methylation can downregulate the expression of MTA1 in vitro. In general, we found ERα methylation had significant correlation with the MTA1 expression (p < 0.05) in female patients of breast cancer (n = 102) by methylation-specific polymerase chain reaction and immunohistochemistry. To gain a deeper insight into the molecular mechanism underlying the relation between MTA1 and ERα methylation, we treated the invasive breast cancer cell lines with the demethylating agent, found the downregulation of MTA1 protein expression, and mRNA with the unmethylation of ERα (p < 0.05). And the invasive ability of breast cancer cells was significantly positively associated with MTA1 expression. These unique findings have greatly extended our current knowledge about the relation between ERα methylation and MTA1 expression. These data strongly support the hypothesis that methylation is involved in the relation between MTA1 and ERα in breast cancer.
Collapse
Affiliation(s)
- Xiao-yun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Ductal carcinoma in situ (DCIS) of the breast represents a complex, heterogeneous pathologic condition in which malignant epithelial cells are confined within the ducts of the breast without evidence of invasion. The increased use of screening mammography has led to a significant shift in the diagnosis of DCIS, accounting for approximately 27% of all newly diagnosed cases of breast cancer in 2011, with an overall increase in incidence. As the incidence of DCIS increases, the treatment options continue to evolve. Consistent pathologic evaluation is crucial in optimizing treatment recommendations. Surgical treatment options include breast-conserving surgery (BCS) and mastectomy. Postoperative radiation therapy in combination with breast-conserving surgery is considered the standard of care with demonstrated decrease in local recurrence with the addition of radiation therapy. The role of endocrine therapy is currently being evaluated. The optimization of diagnostic imaging, treatment with regard to pathological risk assessment, and the role of partial breast irradiation continue to evolve.
Collapse
|
21
|
Cole K, Tabernero M, Anderson KS. Biologic characteristics of premalignant breast disease. Cancer Biomark 2012; 9:177-92. [PMID: 22112476 DOI: 10.3233/cbm-2011-0187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer is the second leading cause of cancer death in women in the United States. While mammography and breast magnetic resonance imaging (MRI) improve detection of early disease, there remains an unmet need for biomarkers for risk stratification, early detection, prediction, and disease prognosis. A number of early breast lesions, from atypical hyperplasias to carcinomas in situ, are associated with an increased risk of developing subsequent invasive breast carcinoma. The recent development of genomic, epigenomic, and proteomic tools for tissue biomarker detection, including array CGH, RNA expression microarrays, and proteomic arrays have identified a number of potential biomarkers that both identify patients at increased risk, as well as provided insights into the pathology of early breast cancer development. This chapter focuses on the detection and application of tissue and serum biomarkers for the identification and risk stratification of early breast cancer lesions.
Collapse
Affiliation(s)
- Kimberly Cole
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, USA
| | | | | |
Collapse
|
22
|
Orsaria P, Granai AV, Venditti D, Petrella G, Buonomo O. Investigational Paradigms in Downscoring and Upscoring DCIS: Surgical Management Review. Int J Surg Oncol 2012; 2012:560493. [PMID: 22666571 PMCID: PMC3362033 DOI: 10.1155/2012/560493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 03/14/2012] [Indexed: 11/21/2022] Open
Abstract
Counseling patients with DCIS in a rational manner can be extremely difficult when the range of treatment criteria results in diverse and confusing clinical recommendations. Surgeons need tools that quantify measurable prognostic factors to be used in conjunction with clinical experience for the complex decision-making process. Combination of statistically significant tumor recurrence predictors and lesion parameters obtained after initial excision suggests that patients with DCIS can be stratified into specific subsets allowing a scientifically based discussion. The goal is to choose the treatment regimen that will significantly benefit each patient group without subjecting the patients to unnecessary risks. Exploring the effectiveness of complete excision may offer a starting place in a new way of reasoning and conceiving surgical modalities in terms of "downscoring" or "upscoring" patient risk, perhaps changing clinical approach. Reexcison may lower the specific subsets' score and improve local recurrence-free survival also by revealing a larger tumor size, a higher nuclear grade, or an involved margin and so suggesting the best management. It seems, that the key could be identifying significant relapse predictive factors, according to validated risk investigation models, whose value is modifiable by the surgical approach which avails of different diagnostic and therapeutic potentials to be optimal. Certainly DCIS clinical question cannot have a single curative mode due to heterogeneity of pathological lesions and histologic classification.
Collapse
Affiliation(s)
- P. Orsaria
- Division of Surgical Oncology, Department of Surgery, Tor Vergata University Hospital, 00133 Rome, Italy
| | - A. V. Granai
- Division of Surgical Oncology, Department of Surgery, Tor Vergata University Hospital, 00133 Rome, Italy
| | - D. Venditti
- Division of Surgical Oncology, Department of Surgery, Tor Vergata University Hospital, 00133 Rome, Italy
| | - G. Petrella
- Division of Surgical Oncology, Department of Surgery, Tor Vergata University Hospital, 00133 Rome, Italy
| | - O. Buonomo
- Division of Surgical Oncology, Department of Surgery, Tor Vergata University Hospital, 00133 Rome, Italy
| |
Collapse
|
23
|
Partridge AH, Elmore JG, Saslow D, McCaskill-Stevens W, Schnitt SJ. Challenges in ductal carcinoma in situ risk communication and decision-making: report from an American Cancer Society and National Cancer Institute workshop. CA Cancer J Clin 2012; 62:203-10. [PMID: 22488610 PMCID: PMC4112288 DOI: 10.3322/caac.21140] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In September 2010, the American Cancer Society and National Cancer Institute convened a conference to review current issues in ductal carcinoma in situ (DCIS) risk communication and decision-making and to identify directions for future research. Specific topics included patient and health care provider knowledge and attitudes about DCIS and its treatment, how to explain DCIS to patients given the heterogeneity of the disease, consideration of nomenclature changes, and the usefulness of decision tools/aids. This report describes the proceedings of the workshop in the context of the current literature and discusses future directions. Evidence suggests that there is a lack of clarity about the implications and risks of a diagnosis of DCIS among patients, providers, and researchers. Research is needed to understand better the biology and mechanisms of the progression of DCIS to invasive breast cancer and the factors that predict those subtypes of DCIS that do not progress, as well as efforts to improve the communication and informed decision-making surrounding DCIS.
Collapse
Affiliation(s)
- Ann H Partridge
- Department of Medicine, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
24
|
Wei J, Han B, Mao XY, Wei MJ, Yao F, Jin F. Promoter methylation status and expression of estrogen receptor alpha in familial breast cancer patients. Tumour Biol 2011; 33:413-20. [PMID: 21922275 DOI: 10.1007/s13277-011-0234-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/26/2011] [Indexed: 12/18/2022] Open
Abstract
The hypermethylation of estrogen receptor alpha (ERα) promoter is a common molecular alteration in sporadic breast cancer (BC), but its involvement in familial BC remains largely unknown. In the present study, we analyzed the methylation statuses of four regions (ER1, ER3, ER4, and ER5) of the ERα promoter and the ERα expression levels of 113 familial BC patients in a Han Chinese Population from northeastern China and evaluated the association between major clinicopathological features and the hypermethylation statuses of the ERα gene. Tumor samples were analyzed for ERα methylation status by the methylation-specific polymerase chain reaction for ERα, PR, p53, BRCA-1, and BRCA-2 by immunohistochemical (IHC) staining and for Her-2 status by IHC and fluorescence in situ hybridization (FISH). ERα methylation was observed in tumor tissues in 47/113 (41.6%) familial BC patients. There were no significant differences in the methylation statuses among ER1 (20.4%), ER3 (18.6%), ER4 (17.7%), and ER5 (19.5%; χ (2) = 3.89, p > 0.05). An association between ERα expression level and its promoter methylation level was found. In addition, ERα methylation was significantly correlated with tumor size, PR expression, p53 nuclear accumulation, and BRCA-1 and BRCA-2 statuses. In conclusion, in familial BC patients, the level of ERα gene promoter methylation correlates with ERα expression, PR, p53 nuclear accumulation, and BRCA-1 and BRCA-2 statuses. Epigenetic alteration of ERα gene may play an important role in the pathogenesis of familial BC.
Collapse
Affiliation(s)
- Jing Wei
- Departments of Breast Surgery and Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | | | | | | | | | | |
Collapse
|
25
|
Increased N-myc downstream-regulated gene 1 expression is associated with breast atypia-to-carcinoma progression. Tumour Biol 2011; 32:1271-6. [DOI: 10.1007/s13277-011-0232-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Accepted: 08/24/2011] [Indexed: 11/26/2022] Open
|
26
|
Mao XY, Fan CF, Zheng HC, Wei J, Yao F, Jin F. p53 nuclear accumulation and ERalpha expression in ductal hyperplasia of breast in a cohort of 215 Chinese women. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:112. [PMID: 20712900 PMCID: PMC2936335 DOI: 10.1186/1756-9966-29-112] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Accepted: 08/16/2010] [Indexed: 01/04/2023]
Abstract
Introduction Women with ductal hyperplasia including usual ductal hyperplasia (UDH) and atypical ductal hyperplasia (ADH) have an increased risk of developing invasive ductal carcinoma (IDC) of breast. The importance of several molecular markers in breast cancer has been of considerable interest during recent years such as p53 and estrogen receptor alpha (ERα). However, p53 nuclear accumulation and ERα expression have not been assessed in ductal hyperplasia co-existing with ductal carcinoma in situ (DCIS) or IDC versus pure ductal hyperplasia without DCIS or IDC. Materials and methods We investigated p53 nuclear accumulation and ERα expression in breast ductal hyperplasia in a cohort of 215 Chinese women by immunohistochemistry (IHC), which included 129 cases of pure ductal hyperplasia, 86 cases of ductal hyperplasia co-existing with DCIS (41 cases) or IDC (45 cases). Results Nuclear p53 accumulation was identified in 22.8% of ADH (31/136), 41.5% of DCIS (17/41) and 42.2% of IDC (19/45), and no case of UDH (0/79). No difference in nuclear p53 accumulation was observed between pure ADH and ADH co-existing with DCIS (ADH/DCIS) or IDC (ADH/IDC) (P > 0.05). The positive rate of ERα expression was lower in ADH (118/136, 86.8%) than that in UDH (79/79, 100%) (P < 0.001), but higher than that in DCIS (28/41, 68.3%) or IDC (26/45, 57.8%) respectively (P < 0.001). The frequency of ERα expression was lower in ADH/DCIS (23/29, 79.31%) and ADH/IDC (23/30, 76.67%) than that in pure ADH (72/77, 93.51%) respectively (P < 0.05). There was a negative weak correlation between p53 nuclear accumulation and ERα expression as for ADH (coefficient correlation -0.51; P < 0.001). Conclusions Different pathological types of ductal hyperplasia of breast are accompanied by diversity in patterns of nuclear p53 accumulation and ERα expression. At least some pure ADH is molecularly distinct from ADH/CIS or ADH/IDC which indicated the two types of ADH are molecularly distinct entities although they have the same morphological appearance.
Collapse
Affiliation(s)
- Xiao-yun Mao
- Department of Breast Surgery, Research Unit of General Surgery, the First Affiliated Hospital of China Medical University, and Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning Province, (110001), China
| | | | | | | | | | | |
Collapse
|
27
|
Lopez-Garcia MA, Geyer FC, Lacroix-Triki M, Marchió C, Reis-Filho JS. Breast cancer precursors revisited: molecular features and progression pathways. Histopathology 2010; 57:171-92. [PMID: 20500230 DOI: 10.1111/j.1365-2559.2010.03568.x] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increasingly more coherent data on the molecular characteristics of benign breast lesions and breast cancer precursors have led to the delineation of new multistep pathways of breast cancer progression through genotypic-phenotypic correlations. It has become apparent that oestrogen receptor (ER)-positive and -negative breast lesions are fundamentally distinct diseases. Within the ER-positive group, histological grade is strongly associated with the number and complexity of genetic abnormalities in breast cancer cells. Genomic analyses of high-grade ER-positive breast cancers have revealed that a substantial proportion of these tumours harbour the characteristic genetic aberrations found in low-grade ER-positive disease, suggesting that at least a subgroup of high-grade ER-positive breast cancers may originate from low-grade lesions. The ER-negative group is more complex and heterogeneous, comprising distinct molecular entities, including basal-like, HER2 and molecular apocrine lesions. Importantly, the type and pattern of genetic aberrations found in ER-negative cancers differ from those of ER-positive disease. Here, we review the available molecular data on breast cancer risk indicator and precursor lesions, the putative mechanisms of progression from in situ to invasive disease, and propose a revised model of breast cancer evolution based on the molecular characteristics of distinct subtypes of in situ and invasive breast cancers.
Collapse
Affiliation(s)
- Maria A Lopez-Garcia
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, 237 Fulham Road, London, UK
| | | | | | | | | |
Collapse
|
28
|
Abstract
Lobular and ductal intraepithelial neoplasias reflect proliferations of immunophenotypically variable, biologically and morphologically diverse cells with a potential, not always realized, for progression to carcinoma by breaking through the barriers of the myoepithelial cell layer and basement membrane, ultimately invading the stroma. Starting with the lobular and then the ductal proliferations, this review will address the evolution of our understanding of these lesions; the problems associated with the conventional terminology of ductal hyperplasia, atypical hyperplasia, and carcinoma in situ; and reasons for and advantages of the intraepithelial neoplasia terminology.
Collapse
MESH Headings
- Adult
- Biopsy
- Breast/pathology
- Carcinoma in Situ/pathology
- Carcinoma in Situ/surgery
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/surgery
- Cell Transformation, Neoplastic/pathology
- Diagnosis, Differential
- Disease Progression
- Female
- Humans
- Mastectomy
- Neoplasms, Multiple Primary/pathology
- Neoplasms, Multiple Primary/surgery
Collapse
Affiliation(s)
- F A Tavassoli
- Department of Pathology, Yale University School of Medicine, Lauder Hall 222, 310 Cedar Str., New Haven, CT 06510, USA.
| |
Collapse
|
29
|
Tavassoli FA, Lannin DR. Re: Precancerous lesions of the breast. Nat Rev Clin Oncol 2009; 6:E1. [PMID: 19247307 DOI: 10.1038/ncponc1344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Fattaneh A Tavassoli
- Department of Pathology and Surgery, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | |
Collapse
|
30
|
Kuerer HM, Albarracin CT, Yang WT, Cardiff RD, Brewster AM, Symmans WF, Hylton NM, Middleton LP, Krishnamurthy S, Perkins GH, Babiera G, Edgerton ME, Czerniecki BJ, Arun BK, Hortobagyi GN. Ductal Carcinoma in Situ: State of the Science and Roadmap to Advance the Field. J Clin Oncol 2009; 27:279-88. [DOI: 10.1200/jco.2008.18.3103] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Ductal carcinoma in situ (DCIS) is the fourth leading cancer for women in the United States. Understanding of the biology and clinical behavior of DCIS is imperfect. This article highlights the current knowledge base and the scientific roadmap needed to advance the field. Methods This article is based on work done by and consultations obtained from leading experts in the field over a 6-month period that culminated in a full-day symposium designed to systematically review the most pertinent MEDLINE published reports and develop a roadmap to elucidate the molecular steps of carcinogenesis, reduce the extent or prevent the need for therapies, eliminate recurrences, and reduce morbidity. Results Expression profiling of pure DCIS will help elucidate the molecular characteristics that distinguish high-risk lesions from clinically irrelevant lesions. The development of new methods of extracting RNA from processed tissues may provide opportunities for research. Mammography often underestimates the pathologic extent of DCIS; other imaging methods need to be investigated for detection and monitoring of disease stability or progression. Novel biologic agents are being delivered in neoadjuvant clinical trials, and alternative methods for breast irradiation are being studied. Future trials of treatment versus no treatment for biologically selected cases of DCIS should be developed. Conclusion There is a critical need for a concerted international effort among patients with DCIS, clinicians, and basic scientists to conduct the research necessary to improve fundamental understanding of the biology and clinical behavior of DCIS and prevent development of invasive breast cancer.
Collapse
Affiliation(s)
- Henry M. Kuerer
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Constance T. Albarracin
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Wei T. Yang
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Robert D. Cardiff
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Abenaa M. Brewster
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - W. Fraser Symmans
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Nola M. Hylton
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Lavinia P. Middleton
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Savitri Krishnamurthy
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - George H. Perkins
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Gildy Babiera
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Mary E. Edgerton
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Brian J. Czerniecki
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Banu K. Arun
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Gabriel N. Hortobagyi
- From the Departments of Surgical Oncology, Pathology, Diagnostic Radiology, Clinical Cancer Prevention, Radiation Oncology, and Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX; Department of Pathology and Laboratory Medicine, University of California, Davis; Department of Radiology, University of California, San Francisco, CA; and the Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
31
|
Croce S, Bretz-Grenier MF, Mathelin C. [Most common benign epithelial breast diseases: diagnosis, treatment and cancer risk]. ACTA ACUST UNITED AC 2008; 36:788-99. [PMID: 18650113 DOI: 10.1016/j.gyobfe.2008.02.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 02/26/2008] [Indexed: 11/28/2022]
Abstract
As a consequence of breast imaging development and increased interventional radiology, benign epithelial breast diseases (BEBD) represent a growing percentage of breast pathology diagnoses. BEBD include numerous entities such as cysts, fibrosis, adenosis, duct ectasia, which require neither surgery nor follow-up. Some BEBD have to be individualized (radial scars, papillomas, complex sclerosing adenosis, lobular intraepithelial neoplasia, flat epithelial atypia, atypical hyperplasia), being preinvasive lesions or markers of increased breast cancer risk, or being associated with suspect radiological aspect. BEBD should be managed in a pluridisciplinar way and correctly diagnosed by percutaneous biopsies or surgical specimens. The goals of surgery vary according to lesions. It always allows a complete surgical specimen analysis and therefore a search for atypical or cancerous cells. Surgery can also have a preventive role by reducing the risk of potential malignant transformation. Finally, it enables in some cases the excision of a radiologically suspect mass. So the aim of this review is to give a clinical and morphological description of most common BEBD, underlying their cancer risk, specific diagnosis, therapeutic, follow-up and psychological repercussions.
Collapse
Affiliation(s)
- S Croce
- Département de pathologie, hôpital de Hautepierre, CHRU de Strasbourg, avenue Molière, 67200 Strasbourg cedex, France.
| | | | | |
Collapse
|
32
|
Folgiero V, Avetrani P, Bon G, Di Carlo SE, Fabi A, Nisticò C, Vici P, Melucci E, Buglioni S, Perracchio L, Sperduti I, Rosanò L, Sacchi A, Mottolese M, Falcioni R. Induction of ErbB-3 expression by alpha6beta4 integrin contributes to tamoxifen resistance in ERbeta1-negative breast carcinomas. PLoS One 2008; 3:e1592. [PMID: 18270579 PMCID: PMC2220038 DOI: 10.1371/journal.pone.0001592] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 01/18/2008] [Indexed: 12/14/2022] Open
Abstract
Background Tamoxifen is still the most widely used drug in hormone therapy for the treatment of breast cancer. Its benefits in adjuvant treatment are well documented in controlled and randomized clinical studies, which have demonstrated an increase in disease-free intervals of patients with positive hormonal receptors. However, the mechanisms involved in endocrine resistance are not clear. Laboratory and clinical data now indicate that bi-directional molecular cross-talk between nuclear or membrane ER and growth factor receptor pathways may be involved in endocrine resistance. We recently found a functional interaction between α6β4 integrin and ErbB-3 receptor to maintain the PI3K/Akt survival pathway of mammary tumour cells. We sought to improve understanding of this process in order to provide the involvement of both receptors insight into mechanism of Tamoxifen resistance. Methods and Findings Using human breast cancer cell lines displaying different levels of α6β4 and ErbB-3 receptors and a series of 232 breast cancer biopsies from patients submitted to adjuvant Tamoxifen monotherapy for five years, we evaluated the functional interaction between both receptors in relationship to Tamoxifen responsiveness. In mammary carcinoma cells, we evidenced that the α6β4 integrin strongly influence Akt phosphorylation through ErbB-3 protein regulation. Moreover, the ErbB-3 inactivation inhibits Akt phosphorylation, induces apoptosis and inhibits in vitro invasion favouring Tamoxifen responsiveness. The analysis of human tumors revealed a significant relationship between α6β4 and ErbB-3 in P-Akt-positive and ERβ1-negative breast cancers derived from patients with lower disease free survival. Conclusions We provided evidence that a strong relationship occurs between α6β4 and ErbB-3 positivity in ERβ1-negative breast cancers. We also found that the association between ErbB-3 and P-Akt positivity mainly occurs in ERβ1-negative breast cancer derived from patients with lower DFS indicating that both receptors are clinically relevant in predicting the response to Tamoxifen.
Collapse
Affiliation(s)
- Valentina Folgiero
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Paolo Avetrani
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Giulia Bon
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Selene E. Di Carlo
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Alessandra Fabi
- Department of Medical Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Cecilia Nisticò
- Department of Medical Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Patrizia Vici
- Department of Medical Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Elisa Melucci
- Department of Pathology, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | - Laura Rosanò
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
| | - Ada Sacchi
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
| | | | - Rita Falcioni
- Department of Experimental Oncology, Regina Elena Cancer Institute, Rome, Italy
- *E-mail:
| |
Collapse
|
33
|
Dotto J, Kluk M, Geramizadeh B, Tavassoli FA. Frequency of Clinically Occult Intraepithelial and Invasive Neoplasia in Reduction Mammoplasty Specimens: A Study of 516 Cases. Int J Surg Pathol 2008; 16:25-30. [DOI: 10.1177/1066896907307176] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Reduction mammoplasty is a frequently performed procedure for the treatment of macromastia and for the achievement of symmetry in breast cancer patients following lumpectomy. Slides from 516 consecutive bilateral reduction mammoplasties performed for macromastia over 15 years were reviewed. Among these, 92 (18%) low-risk ductal intraepithelial neoplasia/intraductal hyperplasia, 28 (5%) ductal intraepithelial neoplasia 1 (1 low-grade ductal carcinoma in situ, 11 atypical intraductal hyperplasia, and 16 flat type), 17 (3%) lobular intraepithelial neoplasia, and 1 (0.2%) tubular carcinoma were identified. The patients were categorized into 3 age groups: <40 (n = 352), 40 to 50 (n = 107), and over 50 years (n = 57); the frequency of the lesions increased with age. These data confirm the low frequency of clinically occult malignancies identified in reduction mammoplasty specimens and provide substantial information about the frequency of a variety of intraepithelial proliferations. Preoperative mammography, specimen orientation, and inking of margins with 1 color are advised when reduction mammoplasty is scheduled for women ≥ 40 years of age.
Collapse
Affiliation(s)
- Jorge Dotto
- Department of Pathology, Yale New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| | - Michael Kluk
- Department of Pathology, Yale New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| | - Bita Geramizadeh
- Department of Pathology, Namazi Hospital, Shiraz University School of Medical Sciences, Shiraz, Iran
| | - Fattaneh A. Tavassoli
- Department of Pathology, Yale New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut,
| |
Collapse
|
34
|
Martel M, Barron-Rodriguez P, Tolgay Ocal I, Dotto J, Tavassoli FA. Flat DIN 1 (flat epithelial atypia) on core needle biopsy: 63 cases identified retrospectively among 1,751 core biopsies performed over an 8-year period (1992–1999). Virchows Arch 2007; 451:883-91. [PMID: 17786469 DOI: 10.1007/s00428-007-0499-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 07/26/2007] [Accepted: 08/03/2007] [Indexed: 11/24/2022]
Abstract
Uniform management of flat DIN 1 (flat epithelial atypia) on core needle biopsy (CNB) concerning surgical excision or clinical follow-up are lacking. In a retrospective review of 1,751 CNB over an 8-year period, we found 63 cases with flat DIN 1 as the most advanced lesion; follow-up was available in 55 cases. Of the 63 patients, 24 had a subsequent biopsy for 15 days to 10 years after the initial CNB, an infiltrating carcinoma was found in nine (14.3%) patients, seven (11.1%) in the ipsilateral, and two (3.2%) in the contralateral breast. Five underwent an excisional biopsy of the ipsilateral breast within less than 3 months of the initial CNB; none had either an invasive or intraepithelial carcinoma. Based on our findings, we consider flat DIN 1 a marker of slightly increased risk for subsequent development of invasive breast carcinoma. When flat DIN 1 is found on CNB as the most advanced lesion after mammographic correlation, an excisional biopsy is not mandatory; however, close follow-up is advised with repeat mammograms for early detection of any clinically occult carcinoma in the vicinity of flat DIN 1 that may have been missed by the CNB.
Collapse
Affiliation(s)
- Maritza Martel
- Department of Pathology, Yale University School of Medicine, P. O. Box 208070, New Haven, CT 06520-8070, USA.
| | | | | | | | | |
Collapse
|
35
|
Ghofrani M, Tapia B, Tavassoli FA. Discrepancies in the diagnosis of intraductal proliferative lesions of the breast and its management implications: results of a multinational survey. Virchows Arch 2006; 449:609-16. [PMID: 17058097 PMCID: PMC1888715 DOI: 10.1007/s00428-006-0245-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Accepted: 05/31/2006] [Indexed: 12/21/2022]
Abstract
To measure discrepancies in diagnoses and recommendations impacting management of proliferative lesions of the breast, a questionnaire of five problem scenarios was distributed among over 300 practicing pathologists. Of the 230 respondents, 56.5% considered a partial cribriform proliferation within a duct adjacent to unequivocal ductal carcinoma in situ (DCIS) as atypical ductal hyperplasia (ADH), 37.7% of whom recommended reexcision if it were at a resection margin. Of the 43.5% who diagnosed the partially involved duct as DCIS, 28.0% would not recommend reexcision if the lesion were at a margin. When only five ducts had a partial cribriform proliferation, 35.7% considered it as DCIS, while if >or=20 ducts were so involved, this figure rose to 60.4%. When one duct with a complete cribriform pattern measured 0.5, 1.5, or 4 mm, a diagnosis of DCIS was made by 22.6, 31.3, and 94.8%, respectively. When multiple ducts with flat epithelial atypia were at a margin, 20.9% recommended reexcision. Much of these discrepancies arise from the artificial separation of ADH and low-grade DCIS and emphasize the need for combining these two under the umbrella designation of ductal intraepithelial neoplasia grade 1 (DIN 1) to diminish the impact of different terminologies applied to biologically similar lesions.
Collapse
Affiliation(s)
- Mohiedean Ghofrani
- Department of Pathology, Yale University School of Medicine, Lauder Hall (LH) 222, 310 Cedar St., New Haven, CT 06510 USA
| | - Beatriz Tapia
- Department of Pathology, Yale University School of Medicine, Lauder Hall (LH) 222, 310 Cedar St., New Haven, CT 06510 USA
| | - Fattaneh A. Tavassoli
- Department of Pathology, Yale University School of Medicine, Lauder Hall (LH) 222, 310 Cedar St., New Haven, CT 06510 USA
| |
Collapse
|
36
|
Fritzsche FR, Dietel M, Kristiansen G. Flache epitheliale Atypie und andere Zylinderzellläsionen der Brust. DER PATHOLOGE 2006; 27:381-6. [PMID: 16858557 DOI: 10.1007/s00292-006-0852-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Columnar cell lesions are being increasingly identified in specimens excised for mammographically suspect microcalcifications. The entity of flat epithelial neoplasia remains a challenge for surgical pathologists due to its uncertain biological and clinical significance, inconsistent nomenclature, lack of prognostic data and the often unobtrusive and easily overlooked histological findings. This review aims to summarize our experience and the currently available literature on this topic, and will lead to a better understanding of this lesion. Because of its putative role as a precursor lesion and its many similarities to atypical ductal hyperplasia or ductal carcinoma in situ, differentiation from these lesions on the one hand and from columnar cell lesions without atypia on the other, is of importance and should result in different therapeutic recommendations depending on its presence in excisional or core needle biopsies.
Collapse
Affiliation(s)
- F R Fritzsche
- Institut für Pathologie, Charité - Universitätsmedizin, Campus Mitte, Schumannstrasse 20/21, 10117, Berlin, Germany
| | | | | |
Collapse
|
37
|
Bratthauer GL, Strauss BL, Tavassoli FA. STAT 5a expression in various lesions of the breast. Virchows Arch 2005; 448:165-71. [PMID: 16133357 DOI: 10.1007/s00428-005-0056-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Accepted: 07/27/2005] [Indexed: 12/21/2022]
Abstract
The seven signal transducer and activator of transcription (STAT) molecules are effectors of hormonal or cytokine stimulation through receptors. STAT 5a, isolated from prolactin-stimulated mammary cells, contributes to normal proliferation and is essential for mammary gland differentiation. Using a monoclonal antibody, we tested 100 formalin-fixed, paraffin-embedded breast tissues representing everything from simple hyperplasia to invasive carcinoma for the expression of STAT 5a in comparison to normal breast epithelial cells. Immunohistochemical analysis was performed following heat treatment in a pressure cooker. STAT 5a was found in endothelial cells, adipocytes, and leukocytes as well as in the cytoplasm and nucleus of normal epithelial cells, usual ductal hyperplasia, and benign lesions such as fibroadenoma. Myoepithelial cells and stromal fibroblasts failed to demonstrate any STAT 5a in addition to most atypical proliferations including in situ and invasive carcinomas. A few examples of lobular intraepithelial neoplasia and invasive carcinoma demonstrated some reactivity, albeit comparatively reduced. The absence of STAT 5a in the abnormal breast epithelial cells may indicate a defect contributory to the abnormal state.
Collapse
Affiliation(s)
- Gary L Bratthauer
- Department of Gynecologic and Breast Pathology, Armed Forces Institute of Pathology, Washington, DC 20306-6000, USA.
| | | | | |
Collapse
|