1
|
Zhou Y, Wang H, Yang J, Wang F, Dong D, Zhao X, Wang L, He R, Ruan Z, Yang J. Comparison of the prognostic effect of pyrotinib plus trastuzumab and chemotherapy different lines therapy in HER2-positive advanced breast cancer. J Chemother 2025; 37:135-145. [PMID: 38557437 DOI: 10.1080/1120009x.2024.2335714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/24/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
This study aimed to compare the efficacy of pyrotinib, trastuzumab combined with chemotherapy with different lines therapy in human epidermal growth factor receptor 2- (HER2-) positive advanced breast cancer (ABC) and analyze the factors affecting the prognosis. A total of 84 patients with median age of 49 year-old. The mPFS of patients receiving first-line pyrotinib plus trastuzumab and chemotherapy was the longest (11 months) compared with second- and third line patients (p = 0.106). The objective response rate (ORR) and disease control rate (DCR) of the total population were 33.3% and 82.1% respectively. Subgroup analysis suggested that using pyrotinib plus trastuzumab and Albumin-bound paclitaxel was not inferior to combine with Vinorelbine in regards of PFS. Histological grade (OR: 0.233[0.069 ∼ 0.781], p = 0.018) and tumor location (OR: 0.286[0.087 ∼ 0.942], p = 0.040) were independent factors influencing the ORR. Multivariate cox analysis showed that Ki-67 was independently associated with increased risk of progression (HR: 1.843[1.044-3.254], p = 0.035). The most common adverse events were diarrhea (17.9%) and neutropenia (11.9%). In the first-, second- and third-line treatment, pyrotinib plus trastuzumab and chemotherapy is effective and safe. Pyrotinib and trastuzumab combined with Albumin-bound paclitaxel may be a potential ideal treatment plan for HER2-positive advanced breast cancer.
Collapse
Affiliation(s)
- Yangqingqing Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiao Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fan Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Danfeng Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoai Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Le Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruiyuan He
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiping Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
2
|
Wilcock D, Sirohi D, Coleman JF, Adelhardt P, Kim JT, Albertson D, Affolter K, Beech C, Jedrzkiewicz J, Ruano AL, Cleary AS, Mahlow J, Balatico M, Gulbahce HE. HER2 fluorescence in situ hybridization groups 2-4 breast cancers classified as positive after targeted recounts following equivocal (2+) immunohistochemistry. Am J Clin Pathol 2025:aqaf006. [PMID: 39937121 DOI: 10.1093/ajcp/aqaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
OBJECTIVES To investigate the correlation between the extent of (percentage of tumor cells) immunohistochemistry (IHC) staining and final human epidermal growth factor receptor 2 (HER2)-positive result in fluorescence in situ hybridization (FISH) groups 2 to 4 with equivocal (2+) IHC requiring second, blinded FISH evaluation. METHODS Breast cancer cases submitted for HER2 FISH testing with group 2 to 4 results were included. RESULTS Of the 2548 cases with HER2 FISH groups 2 to 4 that had HER2 IHC performed, 1104 (43.3%) (76/182 [41.8%] of group 2, 94/161 [58.4%] of group 3, 934/2205 [42.4%] of group 4) had equivocal (2+) IHC. After second blinded, IHC-guided recounts, 217 of 1104 (19.7%) (17/76 [22.4%], 75/94 [79.8%], 125/934 [13.4%] of FISH groups 2, 3, 4 with IHC 2+, respectively) had final HER2-positive status. Only 13 of 217 (6%) of the cases with HER2-positive status had more than 50% circumferential staining of the tumor targeted for rescoring. CONCLUSIONS In over 90% of HER2 FISH group 2 to 4 breast cancers with equivocal (2+) IHC followed by targeted, blinded second FISH evaluation and final HER2-positive result, the amplified population of tumor cells was limited (<50%). Current guidelines recommend cancers having 10% to 50% of the subpopulation with amplified cells classified as having genetic heterogeneity (GH), which have a poor response to targeted therapies. Identifying these tumors as having GH and/or repeat testing may be recommended.
Collapse
Affiliation(s)
| | - Deepika Sirohi
- Department of Pathology, University of California-San Francisco, San Francisco, California, US
| | - Joshua F Coleman
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Parisa Adelhardt
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Jong Taek Kim
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Daniel Albertson
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Kajsa Affolter
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Cameron Beech
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Jolanta Jedrzkiewicz
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Ana L Ruano
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Allison S Cleary
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Jonathan Mahlow
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - Michael Balatico
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| | - H Evin Gulbahce
- ARUP Laboratories, Salt Lake City, Utah, US
- Department of Pathology and Laboratory Medicine, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, Utah, US
| |
Collapse
|
3
|
Li Z, Hu Y, Jones D, Zhao W, Tozbikian G, Parwani AV. Clinicopathologic Characteristics and Follow-Up Outcomes of Invasive Breast Carcinoma With Different Positive HER2 Fluorescence In Situ Hybridization Patterns: Experience From a Single Academic Institution. Mod Pathol 2025; 38:100637. [PMID: 39490739 DOI: 10.1016/j.modpat.2024.100637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/03/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast carcinoma (BC) encompasses a spectrum of molecular subtypes, characterized by varying HER2/CEP17 ratios and HER2 copy numbers, influencing responses to anti-HER2 therapy. This study stratified HER2 fluorescence in situ hybridization (FISH)-positive patients into 3 distinct groups-group 1 with high copy number (G1-HC: ratio ≥2, copy number ≥6), group 1 with low copy number (G1-LC: ratio ≥2, copy number ≥4 and <6), and group 3 (G3: ratio <2.0, copy number ≥6.0)-and evaluated their clinicopathologic features, response to anti-HER2 therapy, and outcomes. In a cohort of 2702 continuous primary BCs, G1-HC BCs accounted for 304 cases (11.3%), G1-LC for 37 cases (1.4%), and G3 for 75 cases (2.8%). G1-HC BCs were associated with younger age, higher tumor grade, and estrogen receptor negativity compared with G1-LC BCs. Furthermore, G1-HC BCs exhibited increased progesterone receptor negativity and HER2 immunohistochemistry 3+ compared with G1-LC and G3 BCs. Analysis of the subgroup of HER2 immunohistochemistry 2+-only cases (n = 166) showed similar results. Notably, G1-HC patients exhibited significantly enhanced responses to anti-HER2 neoadjuvant chemotherapy compared with G1-LC and G3 patients. Conversely, G1-LC patients displayed a lower likelihood of disease-free status compared with G1-HC and G3 patients, albeit with no significant differences in overall survival, distant metastasis, or local recurrence among the groups. These findings offer valuable clinicopathologic insights into different HER2 FISH positive subgroups, potentially informing future criteria for interpreting HER2 FISH results.
Collapse
Affiliation(s)
- Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| | - Yan Hu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Dan Jones
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Weiqiang Zhao
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Gary Tozbikian
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
4
|
Li AC, Hammond S, Crosby D, Li Z, Parwani AV. Clinicopathologic Features and Digital Imaging Analysis of HER2 Protein in Breast Carcinomas With Different HER2 Fluorescence in Situ Hybridization Patterns. Clin Breast Cancer 2025; 25:38-45. [PMID: 39500658 DOI: 10.1016/j.clbc.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/21/2024] [Accepted: 10/04/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND HER2-targeted therapies have significantly improved outcomes for patients with HER2-positive breast cancer (BC), which represents 15% to 20% of all BC cases. HER2 status is assessed via immunohistochemistry (IHC) and/or in situ hybridization (ISH), dividing BCs into five groups (G1-G5). PATIENTS AND METHODS In a study of 2,702 primary BC cases, comprising 12.7% G1, 0.2% G2, 2.8% G3, 8.5% G4, and 75.9% G5, we analyzed clinicopathologic features and HER2 protein expression digitally for each ISH group. RESULTS Notably, G5 cases had a higher proportion of lobular carcinoma (13.9%) compared to other groups. G3 cases showed the highest percentage of grade 3 tumors (56.9%), while G5 cases had the lowest (21.4%). Additionally, G5 cases had the highest rate of estrogen receptor (ER) positivity (84.6%), while G1-HC (high copy number) cases had the lowest (70.4%). Most G1-HC cases were HER2 IHC 3+ (76.1%), while most G5 cases were IHC 0/1+ (75.7%). IHC 2+ was most common in G1-LC (low copy number) and G3 cases (83.8% and 90.7%, respectively), with G4 cases predominantly IHC 2+ (56.3%) and IHC 1+ (30.1%). Discordant HER2 IHC and ISH results were observed in 12 cases (0.4%), including 7 G1-HC (2.3%), 4 G1-LC (10.8%), and 1 G5 case (0.1%). Digital quantification of HER2 IHC levels in all groups except G5 revealed that G1-HC tumors had the highest HER2 protein expression, followed by G3, with G4 showing the lowest. CONCLUSION These findings offer valuable insights into the clinicopathologic characteristics and future management for different HER2 ISH groups.
Collapse
Affiliation(s)
- Aidan C Li
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO
| | - Scott Hammond
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Debra Crosby
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH.
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH.
| |
Collapse
|
5
|
Laokulrath N, Gudi M, Salahuddin SA, Chong APY, Ding C, Iqbal J, Leow WQ, Tan BY, Tse G, Rakha E, Tan PH. Human epidermal growth factor receptor 2 (HER2) status in breast cancer: practice points and challenges. Histopathology 2024; 85:371-382. [PMID: 38845396 DOI: 10.1111/his.15213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 08/09/2024]
Abstract
Human epidermal growth factor receptor 2 (HER2)-enriched breast cancer benefits significantly from anti-HER2 targeted therapies. This highlights the critical need for precise HER2 immunohistochemistry (IHC) interpretation serving as a triage tool for selecting patients for anti-HER2 regimens. Recently, the emerging eligibility of patients with HER2-low breast cancers for a novel HER2-targeted antibody-drug conjugate (T-DXd) adds challenges to HER2 IHC scoring interpretation, notably in the 0-1+ range, which shows high interobserver and interlaboratory staining platform variability. In this review, we navigate evolving challenges and suggest practical recommendations for HER2 IHC interpretation.
Collapse
Affiliation(s)
- Natthawadee Laokulrath
- Department of Pathology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | - Mihir Gudi
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | | | | | - Cristine Ding
- Division of Anatomical Pathology, Changi General Hospital, Singapore
| | - Jabed Iqbal
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | | | - Gary Tse
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Emad Rakha
- Cellular Pathology Department, School of Medicine, University of Nottingham, Nottingham, UK
| | - Puay Hoon Tan
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
- Luma Medical Centre, Singapore
| |
Collapse
|
6
|
Ajabnoor R, Zhang G, Hu Y, Gao Y, Finkelman BS, Turner BM, Yi S, Dhakal A, Audeh W, Li Z, Li X, Hicks DG, Zhang H. Breast Cancer With HER2 Immunohistochemical Score 2 and Average HER2 Signals/Cell 6 or More and HER2/CEP17 Ratio Less Than 2 ('ISH Group 3'): A Multi-Institutional Cohort Analysis Emphasizing Outcome and Molecular Subtype. Mod Pathol 2024; 37:100530. [PMID: 38810729 DOI: 10.1016/j.modpat.2024.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
Breast cancer (BC) with average human epidermal growth factor receptor 2 (HER2) signals/cell ≥6 and HER2/chromosome enumeration probe 17 (CEP17) ratio <2 (in situ hybridization [ISH] group 3) is very rare, accounting for 0.4% to 3.0% of cases sent for the dual-probe ISH assay. Although such patients are currently eligible for treatment with HER2-targeted therapy, their characteristics and outcomes remain poorly understood. Sixty-two BCs with equivocal HER2 immunohistochemical score (2+) and reflex ISH group 3 results were identified across 4 institutions. Available clinicopathologic characteristics, MammaPrint and BluePrint molecular results, and follow-up information were retrospectively analyzed. Most BCs with HER2 equivocal immunohistochemical and ISH group 3 results were histologic grade 2 or 3 (100%), estrogen receptor (ER) positive (90.3%), with an average HER2 signals/cell of 7.3. Molecular profiles revealed that 80% (16/20) of tumors were luminal subtypes, and HER2 molecular subtype was identified in 10% of tumors (2/20). Twelve (19.4%) out of 62 patients developed local recurrence and/or distant metastasis with a median follow-up of 50 months. One (10%) of 10 patients achieved pathologic complete response after neoadjuvant chemotherapy. Forty-nine (79%) out of 62 patients completed anti-HER2 agents, and exploratory analysis showed no statistically significant difference in disease outcomes between patients who completed anti-HER2 treatment and those who did not. Univariate analysis revealed advanced clinical stage, and ER/progesterone receptor negativity was associated with unfavorable disease outcomes, and exploratory multivariate analysis demonstrated that clinical stage was the most significant factor associated with disease outcomes in the studied population. These findings increase our understanding of this rare, but clinically important HER2 category. Large-scale prospective randomized studies are needed to further evaluate the role of perioperative HER2-targeted therapy in this patient population.
Collapse
Affiliation(s)
- Rana Ajabnoor
- Department of Pathology, University of Rochester Medical Center, Rochester, New York; Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Gloria Zhang
- Department of Pathology and Laboratory Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Yan Hu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yuan Gao
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia
| | - Brian S Finkelman
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Sha Yi
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia
| | - Ajay Dhakal
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - William Audeh
- Department of Medical Affairs, Agendia Inc, Irvine, California
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia
| | - David G Hicks
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
7
|
Ayana G, Lee E, Choe SW. Vision Transformers for Breast Cancer Human Epidermal Growth Factor Receptor 2 Expression Staging without Immunohistochemical Staining. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:402-414. [PMID: 38096984 DOI: 10.1016/j.ajpath.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/10/2023] [Accepted: 11/20/2023] [Indexed: 12/31/2023]
Abstract
Accurate staging of human epidermal growth factor receptor 2 (HER2) expression is vital for evaluating breast cancer treatment efficacy. However, it typically involves costly and complex immunohistochemical staining, along with hematoxylin and eosin staining. This work presents customized vision transformers for staging HER2 expression in breast cancer using only hematoxylin and eosin-stained images. The proposed algorithm comprised three modules: a localization module for weakly localizing critical image features using spatial transformers, an attention module for global learning via vision transformers, and a loss module to determine proximity to a HER2 expression level based on input images by calculating ordinal loss. Results, reported with 95% CIs, reveal the proposed approach's success in HER2 expression staging: area under the receiver operating characteristic curve, 0.9202 ± 0.01; precision, 0.922 ± 0.01; sensitivity, 0.876 ± 0.01; and specificity, 0.959 ± 0.02 over fivefold cross-validation. Comparatively, this approach significantly outperformed conventional vision transformer models and state-of-the-art convolutional neural network models (P < 0.001). Furthermore, it surpassed existing methods when evaluated on an independent test data set. This work holds great importance, aiding HER2 expression staging in breast cancer treatment while circumventing the costly and time-consuming immunohistochemical staining procedure, thereby addressing diagnostic disparities in low-resource settings and low-income countries.
Collapse
Affiliation(s)
- Gelan Ayana
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea; School of Biomedical Engineering, Jimma University, Jimma, Ethiopia
| | - Eonjin Lee
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea
| | - Se-Woon Choe
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea; Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea.
| |
Collapse
|
8
|
Bai Q, Lv H, Bao L, Yang Y, Zhang X, Chang H, Xue T, Ren M, Zhu X, Zhou X, Yang W. Invasive Breast Cancer with HER2 ≥4.0 and <6.0: Risk Classification and Molecular Typing by a 21-Gene Expression Assay and MammaPrint Plus BluePrint Testing. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:563-575. [PMID: 37554155 PMCID: PMC10406110 DOI: 10.2147/bctt.s420738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023]
Abstract
PURPOSE To investigate the HER2 status and clinicopathological features in invasive breast cancer with HER2 ≥4.0 and <6.0, which has always been controversial. METHODS Forty breast cancer cases with HER2 ≥4.0 and <6.0 by fluorescence in situ hybridization (FISH) were collected and classified into two groups based on the HRE2/CEP17 ratio (Group A: ≥2.0, n=22; Group B: <2.0, n=18). Clinicopathological characteristics, HER2 status, risk classification, and molecular typing were further analyzed and compared by 21-Gene expression assay and MammaPrint plus BluePrint test. RESULTS The majority of cases in both groups were invasive carcinoma (NOS), with histological grade II, HR+, Ki-67 ≥20%, HER2 2+, and a high risk of recurrence, although younger patients and lymph node metastases were more common in Group A. Surprisingly, all HR+ breast cancers in both groups were classified as luminal-type, HR- cases were all basal-type or unknown, and the index of HER2 in all cases was <0.000 using the BluePrint test, which indicated that HER2 status should be negative. Furthermore, the level of HER2 mRNA expression in all cases of both groups was <10.7, which was defined as HER2 negative by the 21-Gene expression assay. In addition, 10 patients of Group A received anti-HER2 neoadjuvant therapy; only one patient with HR- achieved Grade 5 based on the Miller-Payne system, whereas none of the patients achieved pathological complete response (pCR) based on the Residual Cancer Burden system. CONCLUSION Group A breast cancer, which has always been unquestionably diagnosed as HER2 amplification, was more likely to be HER2 negative and derived less benefit from anti-HER2 neoadjuvant chemotherapy. Group A breast cancer should be distinguished from classical HER2-positive breast cancers when assessing HER2 FISH, and a larger cohort of Group A patients should be included in further studies.
Collapse
Affiliation(s)
- Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Longlong Bao
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Yu Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xin Zhang
- Department of Pathology, Fudan University Zhongshan Hospital, Shanghai, 200032, People’s Republic of China
| | - Heng Chang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Tian Xue
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Min Ren
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiaoli Zhu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Institute of Pathology, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
9
|
Zheng L, Zhang Y, Wang Z, Wang H, Hao C, Li C, Zhao Y, Lyu Z, Song F, Chen K, Huang Y, Song F. Comparisons of clinical characteristics, prognosis, epidemiological factors, and genetic susceptibility between HER2-low and HER2-zero breast cancer among Chinese females. Cancer Med 2023; 12:14937-14948. [PMID: 37387469 PMCID: PMC10417066 DOI: 10.1002/cam4.6129] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Traditional human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC) is recommended to be divided into HER2-low and HER2-zero subtypes due to different prognosis. However, few studies investigated their differences in clinical characteristics and prognosis among Chinese HER2-negative BC and their stratified differences by hormone receptor (HR), while fewer studies investigated their differences in epidemiological factors and genetic susceptibility. METHODS A total of 11,911 HER2-negative BC were included to compare the clinical characteristics and prognosis between HER2-zero and HER2-low BC, and 4227 of the 11,911 HER2-negative BC were further compared to 5653 controls to investigate subtype-specific epidemiological factors and single nucleotide polymorphisms(SNPs). RESULTS Overall, 64.2% of HER2-negative BC were HER2-low BC, and the stratified proportions of HER2-low BC were 61.9% and 75.2% for HR-positive and HR-negative BC, respectively. Compared to HER2-zero BC, HER2-low BC among HR-positive BC showed younger age at diagnosis, later stage, poorer differentiation, and higher Ki-67, while elder age at diagnosis and lower mortality were observed for HER2-low BC among HR-negative BC (all p values <0.05). Compared to healthy controls, both HER2-low and HER2-zero BC are associated with similar epidemiological factors and SNPs. However, stronger interaction between epidemiological factors and polygenic risk scores were observed for HER2-zero BC than HER2-low BC among either HR-positive [odds ratios: 10.71 (7.55-15.17) and 8.84 (6.19-12.62) for the highest risk group compared to the lowest risk group] or HR-negative BC [7.00 (3.14-15.63) and 5.70 (3.26-9.98)]. CONCLUSIONS HER2-low BC should deserve more attention than HER2-zero BC, especially in HR-negative BC, due to larger proportion, less clinical heterogeneity, better prognosis, and less susceptibility to risk factors.
Collapse
Affiliation(s)
- Lu Zheng
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Yunmeng Zhang
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Zhipeng Wang
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Huan Wang
- Department of Infectious Disease Control and PreventionHeping Centers for Disease Control and Prevention of TianjinTianjinChina
| | - Chunfang Hao
- Department of Breast Cancer, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Chenyang Li
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Yanrui Zhao
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Zhangyan Lyu
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Yubei Huang
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Tianjin's Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for CancerTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjinChina
| |
Collapse
|
10
|
Zhou S, Lv H, Li A, Li M, Zhong S, Lu H, Zhou X, Bai Q, Yang W. A clinicopathological study and survival analysis of 99 breast cancers with HER2/CEP17 ratio ≥ 2.0 and an average HER2 copy number < 4.0 per cell in China. BMC Cancer 2023; 23:84. [PMID: 36698078 PMCID: PMC9875391 DOI: 10.1186/s12885-023-10531-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Breast cancer patients of American Society of Clinical Oncology and the College of American Pathologists (ASCO/CAP) Group 2 were all HER2-negative according to the 2018 guideline, not HER2-positive as defined in the 2013 guideline. METHODS We aims to elucidate the unique clinicopathological features of ASCO/CAP Group 2 patients by comparing with classic HER2-nonamplified cancers, and reveal the efficacy of the former to anti-HER2 therapy. The clinicopathological features, treatment and prognosis information of 99 patients between 2014 and 2018 were collected. HER2 status was re-defined using the updated recommendations. RESULTS Of the 99 ASCO/CAP Group 2 tumors, 25.5% (25/99) tumors were immunohistochemical (IHC) 0/1+ and 74.7% (74/99) tumors were IHC 2+. According to the updated 2018 guideline, all of them were HER2 negative. When compared to ASCO/CAP Group 5, patients of ASCO/CAP Group 2 displayed higher ratio of histological grade 3 (P = .03), high Ki67 proliferation index (P = .03) and pN3 (more than 9 lymph nodes metastasis, P = .02), and lower estrogen receptor (ER) positivity (P = .04). There was no statistical difference in the survival of patients received anti-HER2 therapy and patients not received anti-HER2 therapy. CONCLUSIONS Patients of ASCO/CAP Group 2 did not received apparent benefit from anti-HER2 treatment. Although according to the updated guidelines and latest reports, HER2 is negative, but when compared with classic HER2-nonamplified cancers, patients of this group seemed to be more aggressive. We suggest that this group still be regarded as an independent category, in order to accumulate more cases in the future to expand the scope of research.
Collapse
Affiliation(s)
- Shuling Zhou
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Hong Lv
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Anqi Li
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Ming Li
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Siyuan Zhong
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Hongfen Lu
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Xiaoyan Zhou
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Qianming Bai
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| | - Wentao Yang
- grid.452404.30000 0004 1808 0942Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong’an Road, Shanghai, 200032 P. R. China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong’an Road, Shanghai, 200032 P. R. China
| |
Collapse
|
11
|
Comparison of HercepTest™ mAb pharmDx (Dako Omnis, GE001) with Ventana PATHWAY anti-HER-2/neu (4B5) in breast cancer: correlation with HER2 amplification and HER2 low status. Virchows Arch 2022; 481:685-694. [PMID: 35970977 PMCID: PMC9636083 DOI: 10.1007/s00428-022-03378-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/27/2022]
Abstract
Performance of the new CE-IVD-marked HercepTest™ mAb pharmDx (Dako Omnis) assay (HercepTest (mAb)) was compared against the PATHWAY® anti-HER-2/neu (4B5) (PATHWAY 4B5) assay using 119 pre-selected breast cancer samples covering the entire range of HER2 immunohistochemistry (IHC) expression scores (0, 1 + , 2 + , 3 +). The sensitivity and specificity of both assays were assessed based on consensus IHC scores and amplification status, as determined by fluorescence in situ hybridization (FISH) according to 2018 ASCO/CAP testing guidelines. There was a high concordance between results from the HercepTest (mAb) and PATHWAY 4B5 assays for HER2-negative (IHC 0, 1 + , 2 + and FISH negative) and HER2-positive (IHC 3 + , 2 + and FISH positive) breast carcinomas (98.2%). Regarding individual IHC scores, complete agreement was achieved in 69.7% (83/119) of cases, and all but one of the discordant cases were due to higher HER2-status scoring using the HercepTest (mAb). Thus, more tumors were overscored as IHC 2 + by HercepTest (mAb) (27 versus 15) as evidenced by their lower FISH positivity rate (48.1% versus 80%). However, two amplified tumors identified as IHC 2 + by HercepTest (mAb) were missed by PATHWAY 4B5 (IHC 1 +). Four additional cases identified as IHC 2 + by HercepTest (mAb), with FISH ratio < 2 but elevated gene counts (≥ 4 to < 6), were recorded negative by PATHWAY 4B5. The HercepTest (mAb) detects HER2 expression with higher sensitivity in tumors with gene amplification (ISH group 1) and increased gene counts (ISH group 4) as well as in HER2-low tumors (HER2 IHC2 + /FISH negative or IHC 1 +). Future studies will demonstrate whether this translates into improved patient selection especially for new HER2-directed therapies.
Collapse
|
12
|
Yoder A, Inge LJ, Chen CC, Marati VR, Nguyen TK, Zuiderveld K, Martin J, Gladden S, Miri MS, Venugopal R, Lopez B, Ranger-Moore J, Guetter C. Computer-Aided Scoring Of () Gene Amplification Status In Breast Cancer. J Pathol Inform 2022; 13:100116. [PMID: 36268099 PMCID: PMC9577051 DOI: 10.1016/j.jpi.2022.100116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/05/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Background Identification of HER2 protein overexpression and/or amplification of the HER2 gene are required to qualify breast cancer patients for HER2 targeted therapies. In situ hybridization (ISH) assays that identify HER2 gene amplification function as a stand-alone test for determination of HER2 status and rely on the manual quantification of the number of HER2 genes and copies of chromosome 17 to determine HER2 amplification. Methods To assist pathologists, we have developed the uPath HER2 Dual ISH Image Analysis for Breast (uPath HER2 DISH IA) algorithm, as an adjunctive aid in the determination of HER2 gene status in breast cancer specimens. The objective of this study was to compare uPath HER2 DISH image analysis vs manual read scoring of VENTANA HER2 DISH-stained breast carcinoma specimens with ground truth (GT) gene status as the reference. Three reader pathologists reviewed 220, formalin-fixed, paraffin-embedded (FFPE) breast cancer cases by both manual and uPath HER2 DISH IA methods. Scoring results from manual read (MR) and computer-assisted scores (image analysis, IA) were compared against the GT gene status generated by consensus of a panel of pathologists. The differences in agreement rates of HER2 gene status between manual, computer-assisted, and GT gene status were determined. Results The positive percent agreement (PPA) and negative percent agreement (NPA) rates for image analysis (IA) vs GT were 97.2% (95% confidence interval [CI]: 95.0, 99.3) and 94.3% (95% CI: 90.8, 97.3) respectively. Comparison of agreement rates showed that the lower bounds of the 95% CIs for the difference of PPA and NPA for IA vs MR were –0.9% and –6.2%, respectively. Further, inter- and intra-reader agreement rates in the IA method were observed with point estimates of at least 96.7%. Conclusions Overall, our data show that the uPath HER2 DISH IA is non-inferior to manual scoring and supports its use as an aid for pathologists in routine diagnosis of breast cancer. Image analysis algorithm for HER2 amplification using Bright-field ISH in Breast. Automated tumor cell selection and quantitation within pathologist defined ROI. The image analysis algorithm is non-inferior to manual scoring. Integrated solution to support pathologists in determining HER2 gene status.
Collapse
|
13
|
Chen X, Lin Y, Jiang Z, Li Y, Zhang Y, Wang Y, Yu F, Guo W, Chen L, Chen M, Zhang W, Wang C, Fu F. HER2 copy number quantification in primary tumor and cell-free DNA provides additional prognostic information in HER2 positive early breast cancer. Breast 2022; 62:114-122. [PMID: 35158152 PMCID: PMC8850316 DOI: 10.1016/j.breast.2022.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 11/07/2021] [Accepted: 02/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The quantitative relationship between HER2 copy number and prognosis in HER2 positive adjuvant setting remain controversial, and few studies have focused on adjuvant setting to illustrate the potential clinical relevance of HER2 in cfDNA. Our study aim to develop a novel method in HER2 quantification and explore the relationship between HER2 copy number in primary tumors or cfDNA and prognosis in HER2 positive early breast cancer. METHODS Two hundred and two patients with early breast cancer were prospectively included in a study where primary tumors, matching non-cancer breast tissue, corresponding plasma, and the plasma from 20 healthy volunteers were collected. Cox proportional hazard analysis was employed to determine the prognostic value of HER2 gene copy number in tissue and cfDNA. Tissue based nomograms and time-dependent decision curve analysis were used to evaluate the practicality of HER2 copy number stratification. RESULTS HER2 amplification by CNVplex demonstrated a robust concordance with FISH (concordance 89.2%). A three-tiered system of tissue and a two-tiered system of cfDNA classification were shown to be independent prognostic factors. A tissue copy number-based nomogram was fitted and further evaluation revealed a good performance in discrimination (c statistic 0.801) and calibration. CONCLUSIONS We first report CNVplex as a viable alternative for HER2 detection. Quantitative evaluation of HER2 presents tremendous potential for use in risk stratification. We also uncover the potential for using HER2 copy number in cfDNA as a biomarker for prognosis in a HER2 positive adjuvant setting.
Collapse
Affiliation(s)
- Xiaobin Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yuxiang Lin
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | | | - Yan Li
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yihua Zhang
- Department of Dermatology, Fujian Medical University First Affiliated Hospital, Fuzhou, Fujian Province, 350000, China
| | - Ying Wang
- Genesky Biotechnologies Inc., Shanghai, 201315, China
| | - Feng Yu
- Genesky Biotechnologies Inc., Shanghai, 201315, China
| | - Wenhui Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lili Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Minyan Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wenzhe Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
14
|
Hwang HW, Hong SA, Nam SJ, Kim SW, Lee JE, Yu JH, Lee SK, Cho SY, Cho EY. Histologic analysis according to HER2 gene status in HER2 2 + invasive breast cancer: a study of 280 cases comparing ASCO/CAP 2013 and 2018 guideline recommendations. Virchows Arch 2022; 480:749-758. [PMID: 35138452 DOI: 10.1007/s00428-022-03280-0] [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: 09/02/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
Abstract
The American Society of Clinical Oncology and College of American Pathologists guidelines for HER2 testing in breast cancer (BC) have been updated with more stringent criteria regarding immunohistochemistry (IHC) 2 + interpretation. The aim of our study was to determine HER2 status in IHC 2 + cases based on 2013 and 2018 guidelines and to investigate specific histologic characteristics that might predict HER2 status in tumors with equivocal IHC staining. Two hundred eighty BC cases reported as IHC 2 + and 24 cases reported as non-IHC 2 + were reviewed with 12 histologic characteristics. Of the IHC 2 + cases based on 2013 guideline, 21% were reclassified to IHC 1 + when applying the 2018 guidelines. Consequently, it led to an 8% increase of HER2 amplification rate in 2018 IHC 2 + group. Seven characteristics were significantly associated with prediction of HER2 amplification in IHC 2 + BCs, including high tumor-infiltrating lymphocytes (TILs), distinct cellular membrane, no apical snout, large nuclear size, nuclear size variation, high nuclear grade, and tubule formation < 10%. Using these criteria, the presence of four or more characteristics significantly indicates HER2 amplification. Moreover, four characteristics among them, including high TILs, distinct cellular membrane, nuclear size variation, and high nuclear grade, were also associated with HER2 amplification in non-IHC 2 + cases, demonstrating their predictive value as complements to IHC. In conclusion, we provide specific morphologic features that will improve pathologist performance in identifying more HER2-positive BCs. We further suggest an algorithm for trastuzumab therapy decisions using a combination of histomorphologic evaluation and the updated 2018 guidelines.
Collapse
Affiliation(s)
- Hye Won Hwang
- Department of Pathology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Seok Jin Nam
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok Won Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Eon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Han Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Kyung Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo Youn Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Eun Yoon Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
15
|
Mohanty SK, Mishra SK, Tiwari A, Sharma S, Bhardwaj M, Pattnaik N, Jaiswal S, Baisakh MR, Das S, Pradhan MR, Swain TR, Satpathy K, Williamson SR, Parwani AV. Reappraisal of HER2 Amplification in High-Grade Urothelial Carcinoma Based on 2018 ASCO/CAP Clinical Practice Guidelines. Am J Clin Pathol 2021; 156:1130-1141. [PMID: 34124742 DOI: 10.1093/ajcp/aqab083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES To examine and compare human epidermal growth factor receptor 2 (HER2) amplification status in high-grade urothelial carcinoma (HGUCa), using both 2013 and 2018 HER2 reporting guidelines for breast carcinoma from the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP). METHODS HER2 status by fluorescence in situ hybridization (FISH) assay in 78 cases of HGUCa was compared using 2013 and 2018 HER2 reporting guidelines. RESULTS HER2 amplification was observed in 22 (28.2%) of 78 tumors, of which 17 were in group 1, 1 in group 2, and 2 each in groups 3 and 4 (FISH assay, 2018). The remaining 14 HER2-amplified tumors (FISH assay, 2013) became negative, falling into group 2 (FISH assay, 2018) and were either negative or equivocal on immunohistochemistry (IHC, 2018). All FISH-negative tumors (n = 37) using 2013 criteria remained negative (group 5, 2018). FISH-equivocal tumors (2013) were further categorized into HER2 amplified (n = 1) and HER2 negative (n = 4) (2018). Overall, 20 (25.6%) tumors had discordant HER2 FISH results (2018 vs 2013). CONCLUSIONS Implementing 2018 guidelines, HER2 amplification decreased from 36 to 22 cases. The group with a HER2/CEP17 ratio of 2 or more and average HER2 copy number less than 4 (group 2) were predominantly negative by IHC, suggesting a biologically distinct group of HGUCa that is different from HER2-amplified tumors, which may not respond to HER2-targeted therapy.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Sourav K Mishra
- Department of Medical Oncology, Advanced Medical Research Institute, Bhubaneswar, India
| | - Ankit Tiwari
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
| | - Shivani Sharma
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Mohit Bhardwaj
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Niharika Pattnaik
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
| | - Sunil Jaiswal
- Department of Surgical Oncology, Advanced Medical Research Institute, Bhubaneswar, India
| | - Manas R Baisakh
- Department of Pathology and Laboratory Medicine, Prolife Diagnostics, Bhubaneswar, India
| | - Subodh Das
- Department of Urology, Advanced Medical Research Institute, Bhubaneswar, India
| | - Manas R Pradhan
- Department of Urology, Advanced Medical Research Institute, Bhubaneswar, India
| | - Tapas R Swain
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India
| | | | | | - Anil V Parwani
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| |
Collapse
|
16
|
Nie Z, Wang J, Ji XC. Retracted: Microcalcification-associated breast cancer: HER2-enriched molecular subtype is associated with mammographic features. Br J Radiol 2021:20170942. [PMID: 29927639 DOI: 10.1259/bjr.20170942] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To investigate whether the mammographic features were different between breast cancer HER2-enriched molecular subtype and non-HER2-enriched molecular subtype. METHODS 283 microcalcification-associated breast cancers were identified (HER2-enriched: n = 57; non-HER2-enriched: n = 226). Mammographic tumor mass and calcification features in relation to HER2 molecular subtype were analyzed. RESULTS On univariate analysis, HER2-enriched molecular subtype rates were significantly higher (a) in tumor size ≤2 cm [33 of 57 (57.9%)] than in tumor size >2 cm lesions [22 of 226 (9.7%)] (p = 0.007), (b) in non-spiculated mass [39 of 57 (68.4%)] than in spiculated mass lesions [18 of 226 (7.9%)] (p = 0.034),(c) in calcifications extent >2 cm [41 of 57 (71.9%)] lesions than in calcifications extent ≤2 cm lesions [16 of 226 (7.1%)] (p < 0.001) and (d) in calcification density >20 cm-2 [44 of 57 (71.2%)] lesions than in calcification density ≤20 cm-2 lesions [13 of 226 (5.8%)] (p = 0.034).On multivariate analysis, three mammographic features [tumor size >2 cm vs size ≤2 cm odds ratio (OR): 0.415 95% confidence interval (CI) (0.215 to 0.802), p = 0.009, spiculated mass vs non-spiculated mass OR: 0.226 95% CI (0.114 to 0.446), p < 0.001 and calcifications extent >2 cm vs calcifications extent ≤2 cm OR: 7.754, 95% CI (3.100 to 19.339) p< 0.001] were independent predictors. Our results indicated that small tumor size, non-spiculated mass and calcification extent >2 cm are more likely to be HER2 molecular subtype. The discrimination of this model, as quantified by the area under the curve, was 0.751 [95% CI (0.701 to 0.854)]. CONCLUSION Our study presents a prediction model that incorporates the mammographic features of tumor size, non-spiculated mass and calcification extent, which can potentially be used to preoperative predict breast cancer HER2-enriched subtype. ADVANCES IN KNOWLEDGE Mammographic features can noninvasively visualize breast tumor phenotype characteristics.
Collapse
Affiliation(s)
- Zhong Nie
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jian Wang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiao-Chun Ji
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
17
|
Watanabe SN, Imai K, Nanjo H, Wakamatsu Y, Kimura Y, Katayose Y, Kamata S, Terata K, Takahashi E, Ibonai A, Yamaguchi A, Konno H, Yatsuyanagi M, Kudo C, Takashima S, Akagami Y, Nakamura R, Sato Y, Motoyama S, Nomura K, Minamiya Y. Rapid HER2 cytologic fluorescence in situ hybridization for breast cancer using noncontact alternating current electric field mixing. Cancer Med 2020; 10:586-594. [PMID: 33280268 PMCID: PMC7877363 DOI: 10.1002/cam4.3626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/07/2020] [Accepted: 11/10/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2-in situ hybridization (HER2-ISH) is widely approved for diagnostic, prognostic biomarker testing of formalin-fixed paraffin-embedded tissue blocks. However, cytologic ISH analysis has a potential advantage in tumor samples such as pleural effusion and ascites that are difficult to obtain the histological specimens. Our aim was to evaluate the clinical reliability of a novel rapid cytologic HER2 fluorescence ISH protocol (rapid-CytoFISH). MATERIALS AND METHODS Using a new device, we applied a high-voltage/frequency, noncontact alternating current electric field to tissue imprints and needle rinses, which mixed the probe within microdroplets as the voltage was switched on and off (AC mixing). Cytologic samples (n = 143) were collected from patients with immunohistochemically identified HER2 breast cancers. The specimens were then tested using standard dual-color ISH using formalin-fixed paraffin-embedded tissue (FFPE-tissue DISH) for HER2-targeted therapies, CytoFISH, and rapid-CytoFISH (completed within 4 h). RESULTS All 143 collected cytologic specimens (50 imprinted cytology specimens from resected tumors and 93 liquid-based cytology specimens from needle rinses) were suitable for FISH analysis. The HER2/chromosome enumeration probe (CEP) 17 ratios did not significantly differ between FFPE-tissue DISH and either CytoFISH protocol. Based on HER2 scoring criteria, we found 95.1% agreement between FFPE-tissue DISH and CytoFISH (Cohen's kappa coefficient = 0.771 and 95% confidence interval (CI): 0.614-0.927). CONCLUSION CytoFISH could potentially serve as a clinical tool for prompt determination of HER2 status in breast cancer cytology. Rapid-CytoFISH with AC mixing will enable cancer diagnoses and HER2 status to be determined on the same day a patient comes to a clinic or hospital.
Collapse
Affiliation(s)
- Shin-Nosuke Watanabe
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhiro Imai
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuki Wakamatsu
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | | | - Kaori Terata
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Eriko Takahashi
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Ayano Ibonai
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Ayuko Yamaguchi
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Hikari Konno
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Misako Yatsuyanagi
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Chiaki Kudo
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Shinogu Takashima
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | - Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Satoru Motoyama
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Kyoko Nomura
- Department of Environmental Health Science and Public Health, Akita University Graduate School of Medicine, Akita, Japan
| | - Yoshihiro Minamiya
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
18
|
Geiersbach KB, Sill DR, Meyer RG, Yuhas JA, Sukov WR, Mounajjed T, Carter JM, Jenkins RB, Chen B. HER2 Testing for Breast Cancer in the Genomics Laboratory: A Sea Change for Fluorescence In Situ Hybridization. Arch Pathol Lab Med 2020; 145:883-886. [PMID: 33112955 DOI: 10.5858/arpa.2020-0273-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Guidelines for HER2 testing in breast cancer have changed over time, from the US Food and Drug Administration guideline to the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines published in 2007, 2013, and 2018. OBJECTIVE.— To investigate the change in assignment of HER2 status in breast cancers with equivocal (2+) immunohistochemistry (IHC) results by fluorescence in situ hybridization (FISH) following implementation of the ASCO/CAP 2018 guideline. DESIGN.— The study included 3556 invasive breast cancers that were HER2 equivocal (2+) by IHC and were submitted to our FISH laboratory after July 2018. Reflex testing (with repeat IHC staining) was performed on certain categories of FISH results known as groups 2, 3, and 4. Concomitant review of IHC and FISH was performed on these reflex cases per 2018 guideline recommendations. The FISH data were analyzed to compare US Food and Drug Administration and ASCO/CAP 2007, 2013, and 2018 interpretations. RESULTS.— Of 3548 invasive breast cancers with complete data available, the percentage agreement for FISH according to different guidelines was highest for ASCO/CAP 2018 versus US Food and Drug Administration (96.5%), followed by ASCO/CAP 2018 versus 2007 (93.8%), and lowest with ASCO/CAP 2018 versus 2013 (83.7%). Per the 2018 guideline, reflex IHC testing was performed on 633 breast cancers (17.8%); the majority of reflex testing results were negative (541 of 633; 85.5%). The overall distribution of HER2 FISH results (per the 2018 guideline) was 88.5% negative and 11.5% positive. CONCLUSIONS.— By eliminating the equivocal FISH category, the 2018 ASCO/CAP guideline significantly reduced the HER2 FISH-positive rate in tumors with equivocal (2+) IHC results.
Collapse
Affiliation(s)
- Katherine B Geiersbach
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Daniel R Sill
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Reid G Meyer
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jason A Yuhas
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - William R Sukov
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Taofic Mounajjed
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jodi M Carter
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Robert B Jenkins
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Beiyun Chen
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
19
|
Li A, Bai Q, Kong H, Zhou S, Lv H, Zhong S, Li M, Bi R, Zhou X, Yang W. Impact of the Updated 2018 American Society of Clinical Oncology/College of American Pathologists Guideline for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer. Arch Pathol Lab Med 2020; 144:1097-1107. [PMID: 32142367 DOI: 10.5858/arpa.2019-0369-oa] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2019] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The updated American Society of Clinical Oncology/College of American Pathologists guideline for human epidermal growth factor receptor 2 (HER2) testing in breast cancer requires pathologists to re-evaluate HER2 status. OBJECTIVE.— To define HER2 status of breast cancer using immunohistochemistry and fluorescence in situ hybridization. DESIGN.— Diagnostic reports of invasive breast cancers made between 2014 and 2018 with HER2 immunohistochemistry and fluorescence in situ hybridization results were retrieved. HER2 status was re-defined using the updated recommendations. RESULTS.— Of 2514 tumors, 89.7% (2254 of 2514) suggested for fluorescence in situ hybridization assay were HER2 immunohistochemistry 2+. Approximately 8.9% (225 of 2514) and 1.4% (35 of 2514) of tumors were of immunohistochemistry 0/1+ and 3+, respectively. Based on the average HER2 copy number and HER2:CEP17 ratio, tumors were assigned into 5 groups, including 13.1% (330 of 2514) group 1 tumors, 2.1% (52 of 2514) group 2 tumors, 1.1% (27 of 2514) group 3 tumors, 7.0% (175 of 2514) group 4 tumors, and 76.8% (1930 of 2514) group 5 tumors. In combination with immunohistochemistry, all tumors in group 2 and group 4 changed HER2 status, from positive and equivocal into negative, respectively, while group 3 tumors remained positive. Comparative analyses of clinicopathologic features of tumors in different groups revealed that group 2 and 4 tumors displayed worse clinicopathologic features than those of group 5, while group 3 tumors shared similar clinicopathologic features to those of group 1. CONCLUSIONS.— Following the updated guideline, HER2 status is clearly designated. Significant differences regarding clinical features were observed between tumors in different groups but they share the same HER2 status, suggesting further validation of the accuracy of this diagnostic approach is warranted.
Collapse
Affiliation(s)
- Anqi Li
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Qianming Bai
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Hui Kong
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Shuling Zhou
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Hong Lv
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Siyuan Zhong
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Ming Li
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Rui Bi
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Xiaoyan Zhou
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| | - Wentao Yang
- From the Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Pathology, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Blanton KC, Deal AM, Kaiser-Rogers KA, Anders CK, O'Connor SM, Hertel JD, Calhoun BC. Clinicopathologic features of breast cancer reclassified as HER2-amplified by fluorescence in situ hybridization with alternative chromosome 17 probes. Ann Diagn Pathol 2020; 48:151576. [PMID: 32805517 DOI: 10.1016/j.anndiagpath.2020.151576] [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: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Dual probe fluorescence in situ hybridization (FISH) assays for determination of human epidermal growth factor receptor 2 (HER2) gene amplification in breast cancer provide a ratio of HER2 to chromosome 17. The ratio may be skewed by copy number alterations (CNA) in the control locus for chromosome 17 (CEP17). We analyzed the impact of alternative chromosome 17 control probes on HER2 status in a series of breast cancers with an emphasis on patients reclassified as amplified. METHODS Breast cancer patients with equivocal HER2 immunohistochemistry (2+) and equivocal FISH with CEP17 were included. Reclassification of HER2 status was assessed with alternative chromosome 17 control probes (LIS1 and RARA). RESULTS A total of 40 unique patients with 46 specimens reflexed to alternative chromosome 17 probe testing were identified. The majority (>80%) of patients had pT1-2, hormone receptor-positive tumors with an intermediate or high combined histologic grade. There were 34/46 (73.9%) specimens reclassified as amplified with alternative probes, corresponding to 29/40 (72.5%) patients. Of the patients reclassified as amplified with alternative probes, 34.5% (10/29) received HER2-targeted therapy. CONCLUSION In this series, the majority of breast cancers tested with alternative chromosome 17 control probes under the 2013 ASCO/CAP Guidelines were converted to HER2-amplified. The treatment data and the clinicopathologic profile of the tumors suggest that most of these patients will neither receive nor benefit from HER2-targeted therapy. The findings support the recommendation in the 2018 ASCO/CAP HER2 Guidelines to discontinue the use of alternative chromosome 17 probes.
Collapse
Affiliation(s)
- Kristen C Blanton
- School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Allison M Deal
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kathleen A Kaiser-Rogers
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Carey K Anders
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Durham, NC, USA
| | - Siobhan M O'Connor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Johann D Hertel
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Benjamin C Calhoun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
21
|
Wang X, Teng X, Ding W, Sun K, Wang B. A clinicopathological study of 30 breast cancer cases with a HER2/CEP17 ratio of ≥2.0 but an average HER2 copy number of <4.0 signals per cell. Mod Pathol 2020; 33:1557-1562. [PMID: 32203091 DOI: 10.1038/s41379-020-0519-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 11/09/2022]
Abstract
The American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) have recently issued updated guidelines on human epidermal growth factor receptor 2 (HER2) testing by fluorescence in situ hybridization (FISH) in invasive breast cancers. Cases with a HER2/chromosome enumeration probe 17 (CEP17) ratio of ≥2.0 but an average HER2 copy number of <4.0 signals per cell (ISH group 2) are no longer automatically classified as ISH positive. Herein, 30 cases in ISH group 2 were collected. Another 100 patients with a HER2/CEP17 ratio <2.0 and <4.0 HER2 signals per cell (ISH group 5) and 100 patients with a HER2/CEP17 ratio of ≥2.0 and an average HER2 copy number of ≥4.0 signals per cell (ISH group 1) were also recruited for comparison. According to the 2018 ASCO/CAP guidelines, all the cases in ISH group 2 were categorized as HER2 negative. The clinicopathological characteristics of the patients in ISH group 2 were intermediate between ISH group 1 and group 5. Survival analyses revealed that there was no significant disease-free survival (DFS) and overall survival (OS) difference between patients with or without targeted therapy in ISH group 2, as well as between patients with targeted therapy in ISH group 1 and patients in ISH group 2. Patients without targeted therapy in ISH group 2 had a significantly worse OS than patients with targeted therapy in ISH group 1 and patients in ISH group 5. In conclusion, patients in ISH group 2 represent a biologically heterogeneous subset, which are different from those in ISH group 1 and 5. A larger cohort of patients in ISH group 2 should be included for future researches to define the efficacy of HER2-targeted therapy.
Collapse
Affiliation(s)
- Xiaoling Wang
- Department of Pathology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaodong Teng
- Department of Pathology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Ding
- Department of Pathology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Ke Sun
- Department of Pathology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Wang
- Department of Pathology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
22
|
Zhang H, Moisini I, Ajabnoor RM, Turner BM, Hicks DG. Applying the New Guidelines of HER2 Testing in Breast Cancer. Curr Oncol Rep 2020; 22:51. [PMID: 32346807 DOI: 10.1007/s11912-020-0901-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The human epidermal growth factor receptor 2 (HER2) is an important prognostic and predictive biomarker in the breast cancer. The American Society of Clinical Oncology/College of American Pathology (ASCO/CAP) has published HER2 testing guidelines in breast cancer. We herein reviewed the HER2 testing guidelines in breast cancer with a focus on the application of the current guidelines. RECENT FINDINGS The continual investigation of HER2 testing in breast cancer has resulted in updates in the HER2 testing guidelines. The current guidelines focus on the uncommon clinical scenarios and emphasize the coordination between immunohistochemistry and in situ hybridization results, in an effort to improve clarity and accuracy. The ASCO/CAP guidelines provide valuable recommendations to ensure the accurate evaluation of HER2 status in breast cancer patients through standardization. Additional studies, particularly those with long-term outcome data are still needed to validate the guideline recommendations, especially the uncommon cases.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ioana Moisini
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Rana M Ajabnoor
- Department of Pathology, Faculty of medicine, King Abdulaziz University, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - David G Hicks
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, NY, 14642, USA.
| |
Collapse
|
23
|
Pasricha S, Asthana S, Labani S, Kailash U, Srivastav A, Gupta G, Mehta A, Kamboj M. Impact of 2013 ASCO/CAP guidelines on various HER2 reporting categories in breast cancer by fluorescentin-situhybridization and Immunohistochemistry: A meta-analysis with systematic review. ACTA ACUST UNITED AC 2020. [DOI: 10.25259/ijmio_17_2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objective:The ASCO/CAP guidelines for reporting HER2 in breast cancer, first released in 2007, aimed to standardize the reporting protocol, and were updated in 2013 and 2018, to ensure right treatment. Several studies have analyzed the changes attributed to 2013 updated guidelines, and majority of them found increase in positive and equivocal cases. However, the precise implication of these updated guidelines is still contentious, in spite of the latest update (2018 guidelines) addressing some of the issues. We conducted systematic review and meta- analysis to see the impact of 2013 guidelines on various HER2 reporting categories by both FISH and IHC.Materials and Methods:After extensively searching the pertinent literature, 16 studies were included for the systematic review. We divided our approach in three strategies: (1) Studies in which breast cancer cases were scored for HER2 by FISH or IHC as a primary test concurrently by both 2007 and 2013 guidelines, (2) Studies in which HER2 results were equivocal by IHC and were followed by reflex-FISH test by both 2007 and 2013 guidelines, and (3) Studies in which trends of HER2 reporting were compared in the two periods before and after implementation of updated 2013 guidelines. All the paired data in these respective categories was pooled and analyzed statistically to see the overall impact of the updated guidelines.Results:In the first category, by pooled analysis of primary FISH testing there has been a significant increase in the equivocal cases (P< 0.001) and positive cases (P= 0.037). We also found 8.3% and 0.8% of all the negative cases from 2007 guidelines shifted to equivocal and positive categories, respectively. Similarly by primary IHC testing there has been a significant increase in both equivocal cases (P< 0.001) and positive cases (P= 0.02). In the second category of reflex-FISH testing there was a substantial increase in the equivocal cases (P< 0.0001); however there is insignificant decrease (10% to 9.7%;P= 0.66) in the amplified cases. In the third approach for evaluating the trend, with the implementation of 2013 guidelines, there was increase in the equivocal category (P= 0.025) and positive category (P= 0.0088) by IHC. By FISH test also there was significant increase in the equivocal category (P< 0.001) while the increase in the positive category was non-significant (P= 0.159).Conclusions:The updated 2013 guidelines has significantly increased the positive and equivocal cases using primary FISH or IHC test and with further reflex testing, thereby increasing the double equivocal cases and increasing the cost and delaying the decision for definite management. However, whether the additional patients becoming eligible for HDT will derive treatment benefit needs to be answered by further large clinical trials.
Collapse
Affiliation(s)
- Sunil Pasricha
- Department of Pathology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India,
| | - Smita Asthana
- Division of Epidemiology and Biostatistics, ICMR-National Institute of Cancer Prevention and Research, Noida, Uttar Pradesh, India,
| | - Satyanarayana Labani
- Division of Epidemiology and Biostatistics, ICMR-National Institute of Cancer Prevention and Research, Noida, Uttar Pradesh, India,
| | - Uma Kailash
- Division of Epidemiology and Biostatistics, ICMR-National Institute of Cancer Prevention and Research, Noida, Uttar Pradesh, India,
| | - Abhinav Srivastav
- Division of Epidemiology and Biostatistics, ICMR-National Institute of Cancer Prevention and Research, Noida, Uttar Pradesh, India,
| | - Gurudutt Gupta
- Department of Pathology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India,
| | - Anurag Mehta
- Department of Pathology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India,
| | - Meenakshi Kamboj
- Department of Pathology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India,
| |
Collapse
|
24
|
Geiersbach KB, Meyer RG, Sill DR, Mounajjed T, Chen B, Sukov WR, Jenkins RB. Working Up Group 4 Equivocal HER2 Samples Tested by Fluorescence in Situ Hybridization in a Reference Laboratory Setting: Past, Present, and Future. J Clin Oncol 2020; 38:175-176. [DOI: 10.1200/jco.19.02482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Katherine B. Geiersbach
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Reid G. Meyer
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Daniel R. Sill
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Taofic Mounajjed
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Beiyun Chen
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - William R. Sukov
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Robert B. Jenkins
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| |
Collapse
|
25
|
Perrier A, Boelle PY, Chrétien Y, Gligorov J, Lotz JP, Brault D, Comperat E, Lefèvre G, Boissan M. An updated evaluation of serum sHER2, CA15.3, and CEA levels as biomarkers for the response of patients with metastatic breast cancer to trastuzumab-based therapies. PLoS One 2020; 15:e0227356. [PMID: 31910438 PMCID: PMC6946590 DOI: 10.1371/journal.pone.0227356] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 12/17/2019] [Indexed: 11/29/2022] Open
Abstract
Background The transmembrane receptor tyrosine kinase HER2 is overexpressed in approximately 15% of breast tumors and correlates with poor clinical prognosis. Several treatments that target HER2 are approved for treatment of HER2-positive metastatic breast cancer. The serum biomarkers most widely used to monitor anti-HER2 therapies in patients with HER2-positive metastatic breast cancer currently are CA15.3 and CEA. Nevertheless, their clinical utility in patients with breast cancer remains a subject of discussion and controversy; thus, additional markers may prove useful in monitoring the therapeutic responses of these patients. The extracellular domain of HER2 can be shed by proteolytic cleavage into the circulation and this shed form, sHER2, is reported to be augmented during metastasis of HER2-positive breast tumors. Here, we studied the clinical usefulness of sHER2, CA15.3, and CEA for monitoring treatment for breast cancer. Methods We measured prospectively pretreatment and post-treatment serum levels (day 1, 30, 60 and 90) of these three biomarkers in 47 HER2-positive, metastatic breast cancer patients treated with trastuzumab in combination with paclitaxel. Evaluation of the disease was performed according to the Response Evaluation Criteria in Solid Tumor (RECIST) at day 90. Results Patients with progressive disease at day 90 had smaller relative changes between day 1 and day 30 than those with complete, partial or stable responses at day 90: -9% versus -38% for sHER2 (P = 0.02), +23% versus -17% for CA15.3 (P = 0.005) and +29% versus -26% for CEA (P = 0.02). Patients with progressive disease at day 90 were less likely than the other patients to have a relative decrease of > 20% in their biomarker levels at day 30: 6% vs 33% for sHER2 (P = 0.03), 0% vs 27% for CA15.3 (P = 0.03), 4% vs 29% for CEA (P = 0.04). No patient with progressive disease at day 90 had > 20% reduction of the average combined biomarker levels at day 30 whereas 63% of the other patients had (P = 0.003). Moreover, when we analyzed a > 10% reduction of the average biomarker levels no patient with progressive disease at day 90 had a decrease > 10% at day 30 whereas 78% of other patients had (P<0.001, Se = 100%, Sp = 78%). Conclusion We show that regular measurement of sHER2, CA15.3, and CEA levels is useful for predicting the therapeutic response and for monitoring HER2-targeted therapy in patients with HER2-positive metastatic breast cancer. The average decrease of the three biomarkers with a threshold of > 10% appears to be the best parameter to distinguish patients who go on to have progressive disease from those who will have a complete, partial or stable response.
Collapse
Affiliation(s)
- Alexandre Perrier
- Laboratoire de Biochimie et Hormonologie, Hôpital Tenon, Groupe Hospitalier Est Parisien, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Pierre-Yves Boelle
- Sorbonne Université, INSERM, Institut Pierre Louis d’Epidémiologie et de Santé Publique (IPLESP), Assistance Publique–Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Yves Chrétien
- Centre de Recherche Saint-Antoine, INSERM, Sorbonne Université, Paris, France
| | - Joseph Gligorov
- Service d’Oncologie Médicale, Institut Universitaire de Cancérologie APHP–Sorbonne Université, Paris, France
| | - Jean-Pierre Lotz
- Service d’Oncologie Médicale, Institut Universitaire de Cancérologie APHP–Sorbonne Université, Paris, France
| | - Didier Brault
- Laboratoire de Biochimie et Hormonologie, Hôpital Tenon, Groupe Hospitalier Est Parisien, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Eva Comperat
- Department of Pathology, Hôpital Tenon, Groupe Hospitalier Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Guillaume Lefèvre
- Laboratoire de Biochimie et Hormonologie, Hôpital Tenon, Groupe Hospitalier Est Parisien, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Mathieu Boissan
- Laboratoire de Biochimie et Hormonologie, Hôpital Tenon, Groupe Hospitalier Est Parisien, Assistance Publique–Hôpitaux de Paris, Paris, France
- Centre de Recherche Saint-Antoine, INSERM, Sorbonne Université, Paris, France
- * E-mail:
| |
Collapse
|
26
|
Press MF, Seoane JA, Curtis C, Quinaux E, Guzman R, Sauter G, Eiermann W, Mackey JR, Robert N, Pienkowski T, Crown J, Martin M, Valero V, Bee V, Ma Y, Villalobos I, Slamon DJ. Assessment of ERBB2/HER2 Status in HER2-Equivocal Breast Cancers by FISH and 2013/2014 ASCO-CAP Guidelines. JAMA Oncol 2019; 5:366-375. [PMID: 30520947 PMCID: PMC6439848 DOI: 10.1001/jamaoncol.2018.6012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance The 2013/2014 American Society of Clinical Oncology and College of American Pathologists (ASCO-CAP) guidelines for HER2 testing by fluorescence in situ hybridization (FISH) designated an "equivocal" category (average HER2 copies per tumor cell ≥4-6 with HER2/CEP17 ratio <2.0) to be resolved as negative or positive by assessments with alternative control probes. Approximately 4% to 12% of all invasive breast cancers are characterized as HER2-equivocal based on FISH. Objective To evaluate the following hypotheses: (1) genetic loci used as alternative controls are heterozygously deleted in a substantial proportion of breast cancers; (2) use of these loci for assessment of HER2 by FISH leads to false-positive assessments; and (3) these HER2 false-positive breast cancer patients have outcomes that do not differ from clinical outcomes for patients with HER2-negative breast cancer. Design, Setting, and Participants We retrospectively assessed the use of chromosome 17 p-arm and q-arm alternative control genomic sites (TP53, D17S122, SMS, RARA, TOP2A), as recommended by the 2013/2014 ASCO-CAP guidelines for HER2 testing, in patients whose data were available through Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) and whose tissues were available through the Breast Cancer International Research Group clinical trials. We used data from an international cohort database of invasive breast cancers (1980 participants) and international clinical trial of adjuvant chemotherapy in invasive, node-positive breast cancer patients. Main Outcomes and Measures The primary objectives were to (1) assess frequency of heterozygous deletions in chromosome 17 genomic sites used as FISH internal controls for evaluation of HER2 status among HER2-equivocal cancers; (2) characterize impact of using deleted sites for determination of HER2-to-internal-control-gene ratios; (3) assess HER2 protein expression in each subgroup; and (4) compare clinical outcomes for each subgroup. Results Of the 1980 patients in METABRIC,1915 patients were fully evaluated. In addition, 100 HER2-equivocal breast cancers by FISH and 100 comparator FISH-negative breast cancers from the BCIRG-005 trial were analyzed. Heterozygous deletions, particularly in specific p-arm sites, were common in both HER2-amplified and HER2-not-amplified breast cancers. Use of alternative control probes from these regions to assess HER2 by FISH in HER2-equivocal as well as HER2-not-amplified breast cancers resulted in high rates of false-positive ratios (HER2-to-alternative control ratio ≥2.0) owing to heterozygous deletions of control p-arm genomic sites used in ratio denominators. Misclassification of HER2 status was observed not only in breast cancers with ASCO-CAP equivocal status but also in breast cancers with an average of fewer than 4.0 HER2 copies per tumor cell when using alternative control probes. Conclusions and Relevance The indiscriminate use of alternative control probes to calculate HER2 FISH ratios in HER2-equivocal breast cancers may lead to false-positive interpretations of HER2 status resulting from unrecognized heterozygous deletions in 1 or more of these alternative control genomic sites and incorrect HER2 ratio determinations.
Collapse
Affiliation(s)
- Michael F Press
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles
| | - Jose A Seoane
- Departments of Medicine & Genetics, Stanford University, Stanford, California
| | - Christina Curtis
- Departments of Medicine & Genetics, Stanford University, Stanford, California
| | - Emmanuel Quinaux
- International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Roberta Guzman
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles
| | | | | | - John R Mackey
- Department of Oncology, University of Alberta, Edmonton, Canada
| | - Nicholas Robert
- Virginia Cancer Specialists/US Oncology Research Network, Fairfax, Virginia
| | | | - John Crown
- Irish Cooperative Oncology Research Group, St Vincent's University Hospital, Dublin, Ireland
| | - Miguel Martin
- Instituto de Investigación Sanitaria Gregorio Marañón, CIBERONC, GEICAM, Universidad Complutense, Madrid, Spain
| | - Vicente Valero
- The University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Valerie Bee
- Cancer International Research Group/Translational Research in Oncology, Paris, France
| | - Yanling Ma
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles
| | - Ivonne Villalobos
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles
| | - Dennis J Slamon
- Department of Medicine, Geffen School of Medicine at University of California Los Angeles, Los Angeles
| |
Collapse
|
27
|
Suryavanshi M, Jaipuria J, Mehta A, Kumar D, Panigrahi MK, Verma H, Saifi M, Sharma S, Tandon S, Doval DC, Das BC. Droplet digital polymerase chain reaction offers an improvisation over conventional immunohistochemistry and fluorescent in situ hybridization for ascertaining Her2 status of breast cancer. South Asian J Cancer 2019; 8:203-210. [PMID: 31807475 PMCID: PMC6852626 DOI: 10.4103/sajc.sajc_344_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background Droplet digital polymerase chain reaction (DDPCR) is a recent modality for detecting Her2 expression which is quantitative, cheaper, easier to standardize, and free from interobserver variation. Purpose The purpose of this study is to incorporate DDPCR in the current diagnostic paradigm with clinical benefit. Materials and Methods Fifty-four consecutive patients were tested by immunohistochemistry (IHC), fluorescent in situ hybridization (FISH), and DDPCR. With FISH result as gold standard, receiver operating characteristic curves for DDPCR ratio were analyzed to label Her2-negative, equivocal, and positive cases as DDPCR score 1, 2, and 3, respectively. Proportion of patients labeled unequivocally as Her2 positive or negative was defined to have "clinically benefitted" from the test. Drawing parallel to inter-relationships between DDPCR, IHC, and FISH in the test cohort, four diagnostic pathways were defined - (1) initial IHC followed by FISH, (2) initial DDPCR followed by FISH, (3) initial IHC followed by DDPCR followed by FISH, and (4) initial DDPCR followed by IHC followed by FISH. Results Clinical benefit of DDPCR as an initial test in the test cohort was 57%, while it was 65% if used as a second-line test among those with an initial inconclusive IHC result. Sensitivity analysis in the simulation cohort revealed that if DDPCR cost was ≤0.6 times the cost of IHC, then a three-step pathway with DDPCR upfront would near certainly prove most cost beneficial. If DDPCR cost was >0.6 but ≤2 times the cost of IHC, then a three-step pathway with DDPCR as second-line test had a higher probability to prove most cost beneficial. If DDPCR cost was >2 times the cost of IHC, then conventional pathway had a higher probability to prove most cost-effective. Conclusion Incorporating DDPCR in the current clinical diagnostic paradigm has the potential to improve its cost-effectiveness and benefit.
Collapse
Affiliation(s)
| | - Jiten Jaipuria
- Department of Urogynaeoncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Anurag Mehta
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Dushyant Kumar
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | | | - Haristuti Verma
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Mumtaz Saifi
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Sanjeev Sharma
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | | | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
28
|
Yang L, Chen M, Pu T, Wu S, Wei B, Yang J, Bu H, Zhang Z. The differences of clinicopathologic characteristics among subgroups of reclassified HER2 fluorescence in situ hybridization (FISH) according to the ASCO/CAP 2018 breast cancer HER2 testing guidelines. J Clin Pathol 2019; 73:283-290. [DOI: 10.1136/jclinpath-2019-206222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022]
Abstract
AimsThe aim of this study is to analyse differences in clinicopathologic features among reclassified human epidermal growth factor receptor-2 (HER2) fluorescence in situ hybridization (FISH) results in breast cancers according to 2018 guidelines.MethodsAccording to different ratios of HER2 copy numbers to chromosome 17 centromere numbers (HER2/CEP17) and average HER2 copy numbers, 3795 invasive breast cancers were classified into six groups. Clinicopathologic features were collected and compared among different FISH groups.ResultsThere were no statistically significant differences about HER2 positive rate between 2013 and 2018 guidelines (p=0.518). After re-evaluating these cases according to 2018 guidelines, the cases that converted to a HER2 positive status had clinicopathologic features similar to samples in group 1 (ratio ≥2.0, HER2 ≥4.0). Compared with group 5 (ratio <2.0, HER2 <4.0), the cases in groups 1 had higher histological grade, more frequent occurrence of negative oestrogen receptor and progesterone receptor status and a higher Ki67 index. The samples in group 4 (ratio <2.0, 4.0≤HER2<6.0) showed a higher histological grade and higher Ki67 index than did the samples in group 5 but had a lower histological grade and lower Ki67 index than did the samples in group 1a (ratio ≥2.0, HER2 ≥6.0).ConclusionDifferent categories of HER2 FISH test results have significant differences in clinicopathologic features. With no equivocal cases in 2018 HER2 guidelines, the clear division of HER2 status is helpful for making treatment recommendations about HER2 targeted therapy.
Collapse
|
29
|
Wang B, Ding W, Sun K, Wang X, Xu L, Teng X. Impact of the 2018 ASCO/CAP guidelines on HER2 fluorescence in situ hybridization interpretation in invasive breast cancers with immunohistochemically equivocal results. Sci Rep 2019; 9:16726. [PMID: 31723206 PMCID: PMC6854277 DOI: 10.1038/s41598-019-53003-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/25/2019] [Indexed: 11/09/2022] Open
Abstract
The American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) recently issued updated guidelines on human epidermal growth factor receptor 2 (HER2) testing by fluorescence in situ hybridization (FISH) in invasive breast cancers. In this study, we aimed to investigate the impact of the new recommendations on HER2 FISH interpretation in invasive breast cancers with immunohistochemically (IHC) equivocal results. 1810 breast cancer cases with IHC equivocal results were enrolled in this study between January 2012 and May 2019. Concomitant IHC was performed on the same tissue blocks detected by FISH testing. According to the 2018 guidelines, all the cases in ISH group 2 were categorized as HER2 negative; three of four cases in ISH group 3 were considered as HER2 positive, while the one scored IHC 1+ was reclassified as HER2 negative; Fifty-three previously ISH equivocal cases were redistributed into ten HER2-positive cases and forty-three HER2-negative cases. In conclusion, the utility of 2018 ASCO/CAP guidelines resulted in a slight decrease in HER2 positive rate, due to the reclassification of cases in ISH group 2 and group 4. The implementation of the new guidelines can reduce reflex FISH test and make the diagnosis of HER2 gene status more definitive.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pathology, the first affiliated hospital of Zhejiang University, Qingchun Road 79#, Hangzhou, China
| | - Wei Ding
- Department of Pathology, the first affiliated hospital of Zhejiang University, Qingchun Road 79#, Hangzhou, China
| | - Ke Sun
- Department of Pathology, the first affiliated hospital of Zhejiang University, Qingchun Road 79#, Hangzhou, China
| | - Xiaoling Wang
- Department of Pathology, the first affiliated hospital of Zhejiang University, Qingchun Road 79#, Hangzhou, China
| | - Liming Xu
- Department of Pathology, the first affiliated hospital of Zhejiang University, Qingchun Road 79#, Hangzhou, China
| | - Xiaodong Teng
- Department of Pathology, the first affiliated hospital of Zhejiang University, Qingchun Road 79#, Hangzhou, China.
| |
Collapse
|
30
|
Zare S, Rong J, Daehne S, Roma A, Hasteh F, Dell'Aquila M, Fadare O. Implementation of the 2018 American Society of Clinical Oncology/College of American Pathologists Guidelines on HER2/neu Assessment by FISH in breast cancers: predicted impact in a single institutional cohort. Mod Pathol 2019; 32:1566-1573. [PMID: 31190000 DOI: 10.1038/s41379-019-0295-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/21/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
Abstract
The 2018 American Society of Clinical Oncology (ASCO) and College of American Pathologists (CAP) update modified the interpretation guidelines for human epidermal growth factor receptor 2 (HER2) testing by incorporating immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) results in a subset of cases. Importantly, the new guidelines eliminate "equivocal" results, as well as the use of alternative chromosome 17 probes as the primary strategy for resolving the indeterminate FISH results. Herein, we investigate the predicted impact of implementing the 2018 ASCO/CAP guidelines on HER2 assessment by FISH in breast cancers, using data from a single institution. We compared the HER2 status of 1542 consecutive cases of breast carcinoma, interpreted by 2013 and 2018 ASCO/CAP guidelines. In total, 10.7% (165/1542) of the cases had a different final interpretation by 2018 guidelines compared with 2013 guidelines, including 70 previously HER2-positive cases reclassified as negative, four previously negative cases reclassified as positive, and 91 previously equivocal cases reclassified as negative. Overall, the number of HER2-positive cancers was reduced by 66 cases (4.3% reduction in the HER2 positivity rate). The newly HER2-negative cases were mostly estrogen receptor positive (90%), progesterone receptor positive (80%), stage 1 (60.9%), and grade 1-2 (59.4%) cancers; 70% of them had been designated as HER2 positive only after the use of an alternative chromosome 17 FISH probe after an intially equivocal result from the standard CEP17 probe. Overall, implementing the revised 2018 HER2 guidelines is predicted to change the HER2 results of 10.7% of breast cancers, mainly by reclassifying previously equivocal to negative results.
Collapse
Affiliation(s)
- Somaye Zare
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Juan Rong
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Svenja Daehne
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Andres Roma
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Farnaz Hasteh
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Marie Dell'Aquila
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Oluwole Fadare
- Department of Pathology, Anatomic Pathology Division, University of California San Diego Health, La Jolla, CA, 92037, USA.
| |
Collapse
|
31
|
Lin L, Sirohi D, Coleman JF, Gulbahce HE. American Society of Clinical Oncology/College of American Pathologists 2018 Focused Update of Breast Cancer HER2 FISH Testing GuidelinesResults From a National Reference Laboratory. Am J Clin Pathol 2019; 152:479-485. [PMID: 31172196 DOI: 10.1093/ajcp/aqz061] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To review impact of the ASCO/CAP 2018 update on HER2 testing. METHODS HER2 fluorescence in situ hybridization (FISH) test requests from primary and metastatic breast cancers between August 2018 and January 2019 were included. FISH results requiring a changed algorithm under the new guidelines (groups 2, 3, and 4) were identified and HER2:CEN17 ratios, average HER2, CEN17 signals/cell, and HER2 immunohistochemistry (IHC) results were recorded. RESULTS Of the HER2 FISH cases 176/812(21.7%) fell within groups 2, 3, or 4; 0/12, 1/12, and 2/152 cases were positive (3+) by IHC, and 1/12, 2/12, and 6/152 cases were positive after targeted scoring from groups 2, 3, and 4, respectively. Following 2018 updates, 8.3%, 25%, and 5.3% of the groups 2, 3, and 4 were positive, respectively. CONCLUSIONS Groups 2, 3, and 4 constituted over 20% of HER2 FISH tests in a reference laboratory. The 2018 ASCO/CAP update significantly decreased the HER2 positivity rate.
Collapse
Affiliation(s)
- Leo Lin
- Department of Pathology, University of Utah, Salt Lake City
| | - Deepika Sirohi
- Department of Pathology, University of Utah, Salt Lake City
| | | | | |
Collapse
|
32
|
Gupta S, Neumeister V, McGuire J, Song YS, Acs B, Ho K, Weidler J, Wong W, Rhees B, Bates M, Rimm DL, Bossuyt V. Quantitative assessments and clinical outcomes in HER2 equivocal 2018 ASCO/CAP ISH group 4 breast cancer. NPJ Breast Cancer 2019; 5:28. [PMID: 31482108 PMCID: PMC6715641 DOI: 10.1038/s41523-019-0122-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/29/2019] [Indexed: 12/17/2022] Open
Abstract
We quantified human epidermal growth factor receptor 2 (HER2) RNA and protein expression in 2018 American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) in situ hybridization (ISH) group 4 (HER2/centromeric probe 17 (CEP17) ratio <2.0, average HER2 copy number ≥4.0 and <6.0, and 2013 ASCO/CAP ISH equivocal) breast cancers. Breast cancers in 2018 ASCO/CAP ISH group 4 between 2014 and 2017 were identified from the Yale archives. Sixty-three patients (34 with HER2 immunohistochemistry (IHC) 0/1+ and 29 with HER2 IHC 2+) were included. We compared patient characteristics, systemic treatments, and outcomes. We assessed HER2 by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and quantitative immunofluorescence (QIF). Among ISH group 4 cancers, higher HER2 mRNA (P < 0.0001) but similar HER2 protein levels were observed in IHC 2+ compared to IHC 0/1+ cancers. The distribution of RT-qPCR and QIF scores were independent of fluorescence in situ hybridization (FISH) ratio/copy number. Concordance between HER2 RT-qPCR and QIF was 69.8% (r = 0.52). Among 29 patients with IHC2+ results, 16 were HER2 positive by RT-qPCR and 12 were HER2 positive by QIF. Systemic treatment, recurrence, and survival outcomes were comparable among ISH group 4 cancers regardless of IHC 0/1+ or 2+ results. ISH group 4 cancers appear to form a distinct group with intermediate levels of RNA/protein expression, close to positive/negative cut points. Therefore, adjudication into positive or negative categories may not be meaningful. Our results support the 2018 ASCO/CAP recommendation to refrain from routine additional testing of these samples. Additional outcome information after trastuzumab treatment for patients in this special group might help to guide treatment decisions in these patients.
Collapse
Affiliation(s)
- Swati Gupta
- Department of Pathology, Yale University School of Medicine, New Haven, CT USA
| | - Veronique Neumeister
- Department of Pathology, Yale University School of Medicine, New Haven, CT USA
- Indivumed Inc, Frederick, MD USA
| | - John McGuire
- Department of Pathology, Yale University School of Medicine, New Haven, CT USA
| | - Yan S. Song
- Department of Pathology, Yale University School of Medicine, New Haven, CT USA
| | - Balazs Acs
- Department of Pathology, Yale University School of Medicine, New Haven, CT USA
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Kenneth Ho
- Division of Oncology Research and Development, Cepheid, Sunnyvale, CA USA
| | - Jodi Weidler
- Medical and Scientific Affairs and Strategy, Oncology, Cepheid, Sunnyvale, CA USA
| | - Wendy Wong
- Division of Oncology Research and Development, Cepheid, Sunnyvale, CA USA
| | - Brian Rhees
- Division of Oncology Research and Development, Cepheid, Sunnyvale, CA USA
| | - Michael Bates
- Medical and Scientific Affairs and Strategy, Oncology, Cepheid, Sunnyvale, CA USA
| | - David L. Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT USA
| | | |
Collapse
|
33
|
The Dilemma of HER2 Double-equivocal Breast Carcinomas: Genomic Profiling and Implications for Treatment. Am J Surg Pathol 2019; 42:1190-1200. [PMID: 29975246 PMCID: PMC6110371 DOI: 10.1097/pas.0000000000001100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Supplemental Digital Content is available in the text. The American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) 2013 guidelines for HER2 assessment have increased the number of HER2 equivocal breast carcinomas following in situ hybridization reflex testing, that is, HER2 “double equivocal” (equivocal protein expression and equivocal gene copy number). Forty-five double-equivocal carcinomas were subjected to Prosigna analysis. Twenty-seven cases were investigated for the expression of genes found to be differentially expressed between estrogen receptor (ER)-positive/HER2-positive (N=22) and ER-positive/HER2-negative (N=22) control cases. Twenty-nine of the 45 cases were also analyzed by targeted sequencing using a panel of 14 genes. We then explored the pathologic complete response rates in an independent series of double-equivocal carcinoma patients treated with trastuzumab-containing chemotherapy. All cases were ER-positive, with a mean Ki67 of 28%. Double-equivocal carcinomas were predominantly luminal B (76%); 9 cases (20%) were luminal A, and 2 cases (4%) HER2-enriched. The majority (73%) showed a high risk of recurrence by Prosigna, even when the carcinomas were small (<2 cm), node-negative/micrometastatic, and/or grade 2. Double-equivocal carcinomas showed TP53 (6/29, 20%), PIK3CA (3/29, 10%), HER2 (1/29, 3%), and MAP2K4 (1/29, 3%) mutations. Compared with grade-matched ER-positive/HER2-negative breast carcinomas from METABRIC, double-equivocal carcinomas harbored more frequently TP53 mutations and less frequently PIK3CA mutations (P<0.05). No significant differences were observed with grade-matched ER-positive/HER2-positive carcinomas. Lower pathologic complete response rates were observed in double-equivocal compared with HER2-positive patients (10% vs. 60%, P=0.009). Double-equivocal carcinomas are preferentially luminal B and show a high risk of recurrence. A subset of these tumors can be labeled as HER2-enriched by transcriptomic analysis. HER2 mutations can be identified in HER2 double-equivocal cases.
Collapse
|
34
|
Mahtani R, Holmes FA, Badve S, Caldera H, Coleman R, Mamounas E, Kalinsky K, Kittaneh M, Lower E, Pegram M, Press MF, Rugo HS, Schwartzberg L, Vogel C. A Roundtable Discussion of the Breast Cancer Therapy Expert Group (BCTEG): Clinical Developments and Practice Guidance on Human Epidermal Growth Factor Receptor 2 (HER2)-positive Breast Cancer. Clin Breast Cancer 2019; 20:e251-e260. [PMID: 32139271 DOI: 10.1016/j.clbc.2019.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/17/2019] [Accepted: 08/12/2019] [Indexed: 11/26/2022]
Abstract
Expression of human epidermal growth factor receptor 2 (HER2) in breast cancer defines a subset of patients (∼15%-20%) who are candidates for anti-HER2 therapies, most notably, trastuzumab, pertuzumab, antibody drug conjugates (eg, T-DM1), and tyrosine kinase inhibitor (TKI) drugs (eg, lapatinib and neratinib), all of which have dramatically changed the prognosis for this aggressive subtype of breast cancer. A roundtable meeting of the Breast Cancer Therapy Expert Group (BCTEG) was convened in March 2018 in an effort to discuss and clarify, from the perspective of the practicing community oncologist, recent developments in the diagnosis and treatment of HER2-positive (HER2+) breast cancer. Members of the group selected 4 key topics for discussion prior to the meeting, including diagnosis of HER2+ disease, and its treatment in the neoadjuvant, adjuvant, and metastatic settings. Approved testing methods, such as immunohistochemistry and fluorescence in situ hybridization, are used to demonstrate overexpression and/or amplification of HER2 in breast tumors, and established clinical guidelines are used to appropriately define treatment plans for patients with HER2+ disease. The panel acknowledges a range of treatment options now available for treatment of HER2+ breast cancer in the neoadjuvant, adjuvant, and advanced/metastatic settings, although it is noted that many controversies remain, including the optimal sequence of therapies, the most appropriate treatment(s) for subsets of patients with HER2+ disease (eg, hormone receptor-negative or -positive/HER2+), and uncertainties surrounding the diagnosis and definition of HER2+ disease. The current report summarizes the discussion of the BCTEG panel on this topic.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Hope S Rugo
- University of California San Francisco, San Francisco, CA
| | | | - Charles Vogel
- University of Miami Health System, Deerfield Beach, FL
| | | |
Collapse
|
35
|
Assessing the impact of the 2018 American Society of Clinical Oncology/College of American Pathologists recommendations on human epidermal growth factor receptor 2 testing by fluorescence in situ hybridization in breast carcinoma. Virchows Arch 2019; 476:367-372. [PMID: 31375912 DOI: 10.1007/s00428-019-02636-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/13/2019] [Accepted: 07/24/2019] [Indexed: 10/26/2022]
Abstract
The American Society of Clinical Oncology/College of American Pathologists recently updated their recommendations on human epidermal growth factor receptor 2 (HER2) testing by fluorescence in situ hybridization (FISH) in invasive breast cancer, with a focus on the clarification of less common test patterns of ISH. We assessed the impact of the updated ASCO/CAP guidelines on 1044 FISH tested tumors by comparing categorization according to the 2007, 2013, and 2018 ISH classification criteria. The 2013 guidelines increased the number of positive cases (17.4% vs 10.7%) identifying 70 (6.7%) additional patients who met the eligibility criteria for consideration for HER2-targeted therapy compared with the 2007 guidelines. There was a reduction in equivocal tumors (7.7%) with tumors classified as equivocal by the 2007 guidelines (n = 136) redistributed into positive (74, 54.4%) and negative (49, 36.0%) groups. The 2018 guidelines reclassified 10.8% of tumors in our series with a reduction in the number of positive tumors (7.1%). While the proportion of positive tumors (10.2%) was similar to that in 2007 (10.7%), the composition of this group was significantly altered. HER2 equivocal cases, a group which under the 2013 guidelines caused diagnostic and treatment difficulties, were largely eliminated. Our findings suggest that the 2018 update represents a potentially significant change in therapeutic options for a substantial proportion of patients with 2.9% of FISH-positive tumors according to the 2007 and 2013 guidelines now categorized as HER2 negative and, thus, ineligible for HER2-targeted therapy.
Collapse
|
36
|
Gao X, Wang M, Zhang Y, Xu Z, Ding J, Tang J. MicroRNA-16 sensitizes drug-resistant breast cancer cells to Adriamycin by targeting Wip1 and Bcl-2. Oncol Lett 2019; 18:2897-2906. [PMID: 31452770 PMCID: PMC6676408 DOI: 10.3892/ol.2019.10637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 06/12/2019] [Indexed: 12/14/2022] Open
Abstract
Clinical evidence indicates that drug resistance is a major obstacle in the treatment of breast cancer (BC). Drug resistance results in the disease being uncontrollable, and leads to high mortality rates. The aim of the present study was to investigate the chemosensitizing effect of microRNA (miR)-16 on Adriamycin (ADM)-resistant BC cells and the associated mechanisms. BC tumors from 40 patients were collected and reverse transcription-quantitative PCR was used to examine the expression of miR-16. ADM-sensitive (MCF-7/S) and -resistant (MCF-7/A) BC cell lines were used to determine the expression of miR-16 prior to and following transfection with miR-16 mimics or inhibitor. The effects of increased and decreased miR-16 expression on the chemosensitivity of BC cells to ADM was analyzed using MTT, colony survival and flow cytometry assays. miR-16 was found to regulate wild-type p53-induced phosphatase 1 (Wip1) and Bcl-2 expression, as confirmed by western blotting, immunofluorescence staining and luciferase reporter assays. miR-16 expression in drug-resistant tumor tissues and cells was decreased, compared with that the drug-sensitive equivalents. Overexpression of miR-16 in MCF-7/A was associated with a sharp downregulation of Wip1 and Bcl-2 expression, leading to increased ADM-induced cell apoptosis and sensitization of MCF-7/A cells to ADM treatment. Taken together, the results demonstrate that miR-16 may serve as an effective chemosensitizing target to enhance the effects of chemotherapy in drug-resistant BC cells with high Wip1 and Bcl-2 expression. This provides a novel approach to improving the chemotherapeutic efficacy in drug-resistant BC via regulation of miRs.
Collapse
Affiliation(s)
- Xitao Gao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Lianyungang Clinical College of Nanjing Medical University, The First People Hospital of Lianyungang City, Lianyungang, Jiangsu 222001, P.R. China
| | - Mei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yanyan Zhang
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Zhi Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiaji Ding
- Department of Burns and Plastic Surgery, Xuzhou No. 1 People's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
37
|
Curado M, Caramelo AS, Eloy C, Polónia A. What to expect from the 2018 ASCO/CAP HER2 guideline in the reflex in situ hybridization test of immunohistochemically equivocal 2+ cases? Virchows Arch 2019; 475:303-311. [PMID: 30953146 DOI: 10.1007/s00428-019-02567-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 01/12/2023]
Abstract
To evaluate the effect of the 2018 ASCO/CAP guideline in the identification of HER2-positive breast carcinomas (BC) in reflex in situ hybridization (ISH) test. A total of 592 primary invasive BC cases from before and after the publication of the updated ASCO/CAP guideline were evaluated for HER2 amplification by silver ISH according to the 2013 and 2018 guidelines. Cases were mostly (95%) HER2 equivocal by immunohistochemistry (IHC), not centrally reviewed. Other reasons for referring cases were IHC confirmation, IHC discordancy (either between needle-core-biopsy (NCB) and surgical excision specimen (SES) or between different laboratories) and IHC result unexpected for histopathologic features. Cases evaluated with the 2013 guideline (1st cohort) were 14.6% HER2-positive, decreasing significantly after the reclassification with the 2018 guideline due to the exclusion of group 2 cases without HER2 protein overexpression. Cases studied after the implementation of the 2018 guideline (2nd cohort) were 8.7% HER2-positive, a frequency that was not significantly different from the reclassification of the 1st cohort with the 2018 guideline. All cases referred for IHC confirmation had the expected ISH result. Cases with IHC discordancy between NCB and SES were ISH concordant. Only one out of 14 cases with an IHC score 3+ and classified as histological grade 1 or with a Ki67 below 10% was classified as ISH HER2-positive. The 2018 ASCO/CAP guideline resulted in a decrease of HER2-positive cases in reflex ISH test, selecting less patients for anti-HER2-targeted therapy.
Collapse
Affiliation(s)
- Mónica Curado
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal
| | - Ana Sofia Caramelo
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal
| | - Catarina Eloy
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.,Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - António Polónia
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal. .,I3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.
| |
Collapse
|
38
|
Wang X, He Y, Fan Z, Wang T, Xie Y, Li J, Ouyang T. Effect of Trastuzumab among HER2-Positive Breast Cancer Patients that Achieved Pathologic Complete Response after Neoadjuvant Chemotherapy. Breast Care (Basel) 2019; 14:388-393. [PMID: 31933585 DOI: 10.1159/000495186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background We sought to investigate the incremental benefit of trastuzumab in patients with HER2-positive breast cancer who achieved a pathologic complete response (pCR) after neoadjuvant chemotherapy (NACT). Methods The data of HER2-positive invasive breast cancer patients treated with NACT and achieving pCR were obtained from the institutional database. Patients were categorized according to trastuzumab administration. The Kaplan-Meier method and log-rank estimates were used to test the association between trastuzumab administration and survival. Univariate and multivariate Cox regressions were used to obtain hazard ratios. Results Of 223 patients, 83 (37.2%) were treated with NACT without trastuzumab and 140 (62.8%) were treated with NACT plus trastuzumab for 1 year. After a median follow-up of 67 months, the trastuzumab group showed improved relapse-free survival compared with the no-trastuzumab group (95.7 vs. 87.8%, hazard ratio = 0.31, p = 0.028). No significant difference in distant disease-free survival or overall survival was observed (p = 0.250 and 0.432, respectively). Multivariate analysis identified endocrine therapy and trastuzumab administration to be associated with decreased risk of relapse (p = 0.018 and 0.030, respectively). Conclusion The administration of trastuzumab should be considered standard treatment for HER2-positive patients who have achieved pCR after NACT alone.
Collapse
Affiliation(s)
- Xinguang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yingjian He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhaoqing Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Tianfeng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yuntao Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jinfeng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| | - Tao Ouyang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
39
|
Xu B, Shen J, Guo W, Zhao W, Zhuang Y, Wang L. Impact of the 2018 ASCO/CAP HER2 guidelines update for HER2 testing by FISH in breast cancer. Pathol Res Pract 2019; 215:251-255. [DOI: 10.1016/j.prp.2018.10.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022]
|
40
|
Martin V, Valera A, De Joffrey M, Banfi S, Mazzucchelli L. Implementation of the 2018 Human Epidermal Growth Factor Receptor 2 Guideline by American Society of Clinical Oncology/College of American Pathologists Will Reduce False-Positive Tests. Arch Pathol Lab Med 2019; 143:411-412. [PMID: 30605369 DOI: 10.5858/arpa.2018-0904-le] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | | | - Sara Banfi
- Institute of Pathology, Locarno, Switzerland
| | | |
Collapse
|
41
|
Zare SY, Lin L, Alghamdi AG, Daehne S, Roma AA, Hasteh F, Dell'Aquila M, Fadare O. Breast cancers with a HER2/CEP17 ratio of 2.0 or greater and an average HER2 copy number of less than 4.0 per cell: frequency, immunohistochemical correlation, and clinicopathological features. Hum Pathol 2019; 83:7-13. [PMID: 30121371 DOI: 10.1016/j.humpath.2018.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 11/28/2022]
Abstract
The 2013 American Society of Clinical Oncology and College of American Pathologists (ASCO/CAP) guidelines classified breast cancers with a fluorescence in situ hybridization dual-probe HER2/CEP17 ratio of 2 or greater as "amplified," inclusive of cases with a HER2 copy number less than 4. The 2018 ASCO/CAP update assigns HER2/neu status for the latter group in a fashion that is highly dependent on the associated immunohistochemical findings. Herein, the authors define the frequency, immunohistochemical correlates, and other clinicopathological features of breast cancers with HER2/CEP17 ratio of 2 or greater and HER2/neu copy number less than 4 (group A), based on an analysis of an institutional cohort assessed for HER2/neu status by both florescence in situ hybridization and immunohistochemistry and scored using 2013 ASCO/CAP criteria. Group A cases were compared with a group B of HER2/neu-amplified breast cancers with a HER2/neu copy number of 4 or greater regarding a variety of clinicopathological features. One hundred sixty-nine (14%) of 1201 cases were HER2/neu amplified, 18 (10.7%) in group A and 151 (89.3%) in group B. By immunohistochemistry, 61.1% of group A cases were HER2/neu negative, 7 (38.9%) were equivocal, and none were positive. In contrast, 66.9% of group B cases were HER2 positive (3+). We could not demonstrate statistically significant differences between the 2 groups regarding standard clinicopathological variables. In summary, our group A cases account for 1.5% of breast cancers, and 10.7% of all HER2/neu-amplified cancers classified as such based on 2013 ASCO/CAP criteria. They are predominantly HER2/neu negative by immunohistochemistry, which suggests that they are biologically different from classically HER2/neu-amplified cases and which validates the 2018 ASCO/CAP guideline against automatically classifying such cases as HER2/neu amplified.
Collapse
Affiliation(s)
- Somaye Y Zare
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Leo Lin
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Abrar G Alghamdi
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Svenja Daehne
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Andres A Roma
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Farnaz Hasteh
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Marie Dell'Aquila
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA
| | - Oluwole Fadare
- Department of Pathology, University of California San Diego, San Diego, CA, 92037 USA.
| |
Collapse
|
42
|
Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, Bilous M, Ellis IO, Fitzgibbons P, Hanna W, Jenkins RB, Press MF, Spears PA, Vance GH, Viale G, McShane LM, Dowsett M. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. Arch Pathol Lab Med 2018; 142:1364-1382. [PMID: 29846104 DOI: 10.5858/arpa.2018-0902-sa] [Citation(s) in RCA: 651] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE.— To update key recommendations of the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) human epidermal growth factor receptor 2 (HER2) testing in breast cancer guideline. METHODS.— Based on the signals approach, an Expert Panel reviewed published literature and research survey results on the observed frequency of less common in situ hybridization (ISH) patterns to update the recommendations. RECOMMENDATIONS.— Two recommendations addressed via correspondence in 2015 are included. First, immunohistochemistry (IHC) 2+ is defined as invasive breast cancer with weak to moderate complete membrane staining observed in >10% of tumor cells. Second, if the initial HER2 test result in a core needle biopsy specimen of a primary breast cancer is negative, a new HER2 test may (not "must") be ordered on the excision specimen based on specific clinical criteria. The HER2 testing algorithm for breast cancer is updated to address the recommended workup for less common clinical scenarios (approximately 5% of cases) observed when using a dual-probe ISH assay. These scenarios are described as ISH group 2 ( HER2/chromosome enumeration probe 17 [CEP17] ratio ≥2.0; average HER2 copy number <4.0 signals per cell), ISH group 3 ( HER2/CEP17 ratio <2.0; average HER2 copy number ≥6.0 signals per cell), and ISH group 4 ( HER2/CEP17 ratio <2.0; average HER2 copy number ≥4.0 and <6.0 signals per cell). The diagnostic approach includes more rigorous interpretation criteria for ISH and requires concomitant IHC review for dual-probe ISH groups 2 to 4 to arrive at the most accurate HER2 status designation (positive or negative) based on combined interpretation of the ISH and IHC assays. The Expert Panel recommends that laboratories using single-probe ISH assays include concomitant IHC review as part of the interpretation of all single-probe ISH assay results.
Collapse
Affiliation(s)
- Antonio C Wolff
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - M Elizabeth Hale Hammond
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Kimberly H Allison
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Brittany E Harvey
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Pamela B Mangu
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - John M S Bartlett
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Michael Bilous
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Ian O Ellis
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Patrick Fitzgibbons
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Wedad Hanna
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Robert B Jenkins
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Michael F Press
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Patricia A Spears
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Gail H Vance
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Giuseppe Viale
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Lisa M McShane
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Mitchell Dowsett
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland; Lisa M. McShane, National Cancer Institute, Bethesda, Maryland; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City; Kimberly H. Allison, Stanford University School of Medicine, Stanford, California; Patrick Fitzgibbons, St Jude Medical Center, Fullerton, California; Michael F. Press, University of Southern California, Los Angeles; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, Virginia; John M.S. Bartlett, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, Australia; Ian O. Ellis, The University of Nottingham, Nottingham, United Kingdom; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, Minnesota; Patricia A. Spears, Cancer Information and Support Network, Raleigh, North Carolina; Gail H. Vance, Indiana University School of Medicine, Indianapolis; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| |
Collapse
|
43
|
Martin V, Valera A, De Joffrey M, Banfi S, Mazzucchelli L. Implementation of the 2018 Human Epidermal Growth Factor Receptor 2 Guidelines by ASCO/College of American Pathologists Will Reduce False-Positive Tests. J Clin Oncol 2018; 36:JCO1800809. [PMID: 30346902 DOI: 10.1200/jco.18.00809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Vittoria Martin
- Vittoria Martin, Alexandra Valera, Melissa De Joffrey, Sara Banfi, and Luca Mazzucchelli, Institute of Pathology, Locarno, Switzerland
| | - Alexandra Valera
- Vittoria Martin, Alexandra Valera, Melissa De Joffrey, Sara Banfi, and Luca Mazzucchelli, Institute of Pathology, Locarno, Switzerland
| | - Melissa De Joffrey
- Vittoria Martin, Alexandra Valera, Melissa De Joffrey, Sara Banfi, and Luca Mazzucchelli, Institute of Pathology, Locarno, Switzerland
| | - Sara Banfi
- Vittoria Martin, Alexandra Valera, Melissa De Joffrey, Sara Banfi, and Luca Mazzucchelli, Institute of Pathology, Locarno, Switzerland
| | - Luca Mazzucchelli
- Vittoria Martin, Alexandra Valera, Melissa De Joffrey, Sara Banfi, and Luca Mazzucchelli, Institute of Pathology, Locarno, Switzerland
| |
Collapse
|
44
|
Allison KH. Ancillary Prognostic and Predictive Testing in Breast Cancer: Focus on Discordant, Unusual, and Borderline Results. Surg Pathol Clin 2018; 11:147-176. [PMID: 29413654 DOI: 10.1016/j.path.2017.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ancillary testing in breast cancer has become standard of care to determine what therapies may be most effective for individual patients with breast cancer. Single-marker tests are required on all newly diagnosed and newly metastatic breast cancers. Markers of proliferation are also used, and include both single-marker tests like Ki67 as well as panel-based gene expression tests, which have made more recent contributions to prognostic and predictive testing in breast cancers. This review focuses on pathologist interpretation of these ancillary test results, with a focus on expected versus unexpected results and troubleshooting borderline, unusual, or discordant results.
Collapse
Affiliation(s)
- Kimberly H Allison
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Lane 235, Stanford, CA 94305, USA.
| |
Collapse
|
45
|
Comparative Pathologic Analysis of Breast Cancers Classified as HER2/neu-Amplified by FISH Using a Standard HER2/CEP17 Dual Probe and an Alternative Chromosome 17 Control Probe. Am J Surg Pathol 2018; 42:1208-1215. [DOI: 10.1097/pas.0000000000001106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Desai NV, Torous V, Parker J, Auman JT, Rosson GB, Cruz C, Perou CM, Schnitt SJ, Tung N. Intrinsic molecular subtypes of breast cancers categorized as HER2-positive using an alternative chromosome 17 probe assay. Breast Cancer Res 2018; 20:75. [PMID: 29996866 PMCID: PMC6042281 DOI: 10.1186/s13058-018-1005-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The 2013 update of the American Society of Clinical Oncology-College of American Pathologists (ASCO-CAP) human epidermal growth factor receptor 2 (HER2) testing guidelines recommend using an alternative chromosome 17 probe assay to resolve HER2 results determined to be equivocal by immunohistochemistry (IHC) or fluorescence in-situ hybridization (FISH). However, it is unclear if cases considered HER2-positive (HER2+) by the alternative probe method are similar to those classified as HER2+ by traditional IHC and FISH criteria and benefit the same from HER2-targeted therapies. We studied the clinical and pathologic features of all 31 breast cancers classified as HER2+ by the alternative probe method at our institution since 2013 and determined their PAM50 intrinsic molecular subtypes. For comparison, we analyzed 19 consecutive cases that were classified as HER2+ by traditional FISH criteria during the same time period. Thirty (97%) cancers in the alternative probe cohort were estrogen receptor (ER)-positive (ER+), while only 9/19 (47%) of traditional HER2 controls were ER+ (p = 0.0002). Sufficient tissue for intrinsic subtype analysis was available for 20/31 cancers in the alternative probe cohort and 9/19 in the traditional HER2+ group. None (0%) of the 20 alternative probe-positive cases were of the HER2-enriched intrinsic subtype, while 8/9 (89%) of those HER2+ by traditional FISH criteria were HER2-enriched (p = 0.0001). These findings suggest that breast cancers classified as HER2+ only by the alternative probe method are biologically distinct from those classified as HER2+ by traditional criteria, and raises questions as to whether or not they derive the same benefit from HER2-targeted therapies.
Collapse
Affiliation(s)
- Neelam V Desai
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Shapiro 9, Boston, MA, 02215, USA.
| | - Vanda Torous
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Joel Parker
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - James T Auman
- Department of Pathology and Laboratory Medicine and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Gary B Rosson
- Department of Pathology and Laboratory Medicine and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Cassandra Cruz
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Shapiro 9, Boston, MA, 02215, USA
| | - Charles M Perou
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stuart J Schnitt
- Department of Pathology, Dana-Farber/Brigham and Women's Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Nadine Tung
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Ave, Shapiro 9, Boston, MA, 02215, USA
| |
Collapse
|
47
|
Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, Bilous M, Ellis IO, Fitzgibbons P, Hanna W, Jenkins RB, Press MF, Spears PA, Vance GH, Viale G, McShane LM, Dowsett M. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol 2018; 36:2105-2122. [PMID: 29846122 DOI: 10.1200/jco.2018.77.8738] [Citation(s) in RCA: 1527] [Impact Index Per Article: 218.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Purpose To update key recommendations of the American Society of Clinical Oncology/College of American Pathologists human epidermal growth factor receptor 2 (HER2) testing in breast cancer guideline. Methods Based on the signals approach, an Expert Panel reviewed published literature and research survey results on the observed frequency of less common in situ hybridization (ISH) patterns to update the recommendations. Recommendations Two recommendations addressed via correspondence in 2015 are included. First, immunohistochemistry (IHC) 2+ is defined as invasive breast cancer with weak to moderate complete membrane staining observed in > 10% of tumor cells. Second, if the initial HER2 test result in a core needle biopsy specimen of a primary breast cancer is negative, a new HER2 test may (not "must") be ordered on the excision specimen based on specific clinical criteria. The HER2 testing algorithm for breast cancer is updated to address the recommended work-up for less common clinical scenarios (approximately 5% of cases) observed when using a dual-probe ISH assay. These scenarios are described as ISH group 2 ( HER2/chromosome enumeration probe 17 [CEP17] ratio ≥ 2.0; average HER2 copy number < 4.0 signals per cell), ISH group 3 ( HER2/CEP17 ratio < 2.0; average HER2 copy number ≥ 6.0 signals per cell), and ISH group 4 ( HER2/CEP17 ratio < 2.0; average HER2 copy number ≥ 4.0 and < 6.0 signals per cell). The diagnostic approach includes more rigorous interpretation criteria for ISH and requires concomitant IHC review for dual-probe ISH groups 2 to 4 to arrive at the most accurate HER2 status designation (positive or negative) based on combined interpretation of the ISH and IHC assays. The Expert Panel recommends that laboratories using single-probe ISH assays include concomitant IHC review as part of the interpretation of all single-probe ISH assay results. Find additional information at www.asco.org/breast-cancer-guidelines .
Collapse
Affiliation(s)
- Antonio C Wolff
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - M Elizabeth Hale Hammond
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Kimberly H Allison
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Brittany E Harvey
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Pamela B Mangu
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - John M S Bartlett
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Michael Bilous
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Ian O Ellis
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Patrick Fitzgibbons
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Wedad Hanna
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Robert B Jenkins
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Michael F Press
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Patricia A Spears
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Gail H Vance
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Giuseppe Viale
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Lisa M McShane
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| | - Mitchell Dowsett
- Antonio C. Wolff, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth Hale Hammond, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Kimberly H. Allison, Stanford University School of Medicine, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Brittany E. Harvey and Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Centre and Women's College Hospital, Toronto, Ontario, Canada; Michael Bilous, Western Sydney University and Australian Clinical Laboratories, Sydney, New South Wales, Australia; Ian O. Ellis, The University of Nottingham, Nottingham; Mitchell Dowsett, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Patricia A. Spears, Cancer Information and Support Network, Raleigh, NC; Gail H. Vance, Indiana University School of Medicine, Indianapolis, IN; and Giuseppe Viale, University of Milan and Istituto Europeo di Oncologia, Milan, Italy
| |
Collapse
|
48
|
Wolff AC, Hammond MEH, Allison KH, Harvey BE, McShane LM, Dowsett M. HER2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update Summary. J Oncol Pract 2018; 14:437-441. [PMID: 29920138 DOI: 10.1200/jop.18.00206] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Antonio C Wolff
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Stanford University School of Medicine, Stanford, CA; American Society of Clinical Oncology, Alexandria, VA; National Cancer Institute, Bethesda, MD; and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - M Elizabeth Hale Hammond
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Stanford University School of Medicine, Stanford, CA; American Society of Clinical Oncology, Alexandria, VA; National Cancer Institute, Bethesda, MD; and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Kimberly H Allison
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Stanford University School of Medicine, Stanford, CA; American Society of Clinical Oncology, Alexandria, VA; National Cancer Institute, Bethesda, MD; and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Brittany E Harvey
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Stanford University School of Medicine, Stanford, CA; American Society of Clinical Oncology, Alexandria, VA; National Cancer Institute, Bethesda, MD; and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Lisa M McShane
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Stanford University School of Medicine, Stanford, CA; American Society of Clinical Oncology, Alexandria, VA; National Cancer Institute, Bethesda, MD; and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Mitchell Dowsett
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City, UT; Stanford University School of Medicine, Stanford, CA; American Society of Clinical Oncology, Alexandria, VA; National Cancer Institute, Bethesda, MD; and The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
49
|
The extracellular domain of Her2 in serum as a biomarker of breast cancer. J Transl Med 2018; 98:696-707. [PMID: 29491426 DOI: 10.1038/s41374-018-0033-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/13/2017] [Accepted: 12/20/2017] [Indexed: 11/08/2022] Open
Abstract
Breast cancer is a major health problem worldwide. In ~15% of breast cancers, the epidermal growth factor receptor HER2, a transmembrane protein, is overexpressed. This HER2 overexpression is associated with an aggressive form of the disease and a poor clinical prognosis. The extracellular domain (ECD) of HER2 is released into the blood by a proteolytic mechanism known as "ECD shedding". This proteolytic shedding leaves a constitutively active truncated receptor in the membrane that is 10-100-fold more oncogenic than the full-length receptor and promotes the growth and survival of cancer cells. Shedding of the HER2 ECD is increased during metastasis: whereas 15% of primary breast cancer patients have elevated levels of serum HER2 ECD (sHER2 ECD), the levels reach 45% in patients with metastatic disease. Thus, sHER2 ECD has been proposed as a promising biomarker for cancer recurrence and for monitoring the disease status of patients overexpressing HER2. Nevertheless, in 2016, the American Society of Clinical Oncology advises clinicians not to use soluble HER2 levels to guide their choice of adjuvant therapy for patients with HER2-positive breast cancer, because the evidence was considered not strong enough. Currently, biomarkers such as carcinoembryonic antigen and cancer antigen 15-3 are widely used to monitor metastatic breast cancer disease even if the level of evidence of clinical impact of this monitoring is poor. In this article, we review the evidence that sHER2 ECD might be used in some situations as a biomarker for breast cancer. Although this serum biomarker will not replace the direct measurement of tumor HER2 status for diagnosis of early-stage tumors; it might be especially useful in metastatic disease for prognosis, as an indicator of cancer progression and of therapy response, particularly to anti-HER2 therapies. Owing to these data, sHER2 ECD should be considered as a promising biomarker to detect cancer recurrence and metastasis.
Collapse
|
50
|
Geiersbach KB, Bridge JA, Dolan M, Jennings LJ, Persons DL, Souers RJ, Tsuchiya KD, Vasalos PH, Moncur JT. Comparative Performance of Breast Cancer Human Epidermal Growth Factor Receptor 2 Fluorescence In Situ Hybridization and Brightfield In Situ Hybridization on College of American Pathologists Proficiency Tests. Arch Pathol Lab Med 2018; 142:1254-1259. [PMID: 29733681 DOI: 10.5858/arpa.2017-0457-cp] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Fluorescence in situ hybridization (FISH) and brightfield in situ hybridization (ISH) are 2 clinically approved laboratory methods for detecting ERBB2 (HER2) amplification in breast cancer. OBJECTIVE.— To compare the performance of FISH and brightfield ISH on proficiency testing administered by the College of American Pathologists Laboratory Accreditation Program. DESIGN.— Retrospective review was performed on 70 tissue core samples in 7 separate proficiency testing surveys conducted between 2009 and 2013. RESULTS.— The samples included 13 consensus-amplified tissue cores, 53 consensus-nonamplified cores, and 4 cores that did not reach consensus for FISH and/or brightfield ISH. There were 2552 individual responses for FISH and 1871 individual responses for brightfield ISH. Consensus response rates were comparable for FISH (2474 of 2524; 98.0%) and brightfield ISH (2135 of 2189; 97.5%). The FISH analysis yielded an average HER2 copy number per cell that was significantly higher (by 2.86; P = .02) compared with brightfield ISH for amplified cores. For nonamplified cores, FISH yielded slightly, but not significantly, higher (by 0.17; P = .10) HER2 copy numbers per cell. There was no significant difference in the average HER2 to control ratio for either consensus-amplified or consensus-nonamplified cores. Participants reported "unable to analyze" more frequently for brightfield ISH (244 of 2453; 9.9%) than they did for FISH (160 of 2684; 6.0%). CONCLUSIONS.— Our study indicates a high concordance rate in proficiency testing surveys, with some significant differences noted in the technical performance of these assays. In borderline cases, updated American Society of Clinical Oncology/College of American Pathologists cutoff thresholds that place greater emphasis on HER2 copy number per cell could accentuate those differences between FISH and brightfield ISH.
Collapse
Affiliation(s)
- Katherine B Geiersbach
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Julia A Bridge
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Michelle Dolan
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Lawrence J Jennings
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Diane L Persons
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Rhona J Souers
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Karen D Tsuchiya
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Patricia H Vasalos
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| | - Joel T Moncur
- From the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Geiersbach); the Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha (Dr Bridge); the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Dolan); the Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Illinois (Dr Jennings); the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Dr Persons); the Departments of Biostatistics (Ms Souers) and Proficiency Testing (Ms Vasalos), College of American Pathologists, Northfield, Illinois; the Department of Laboratories, Seattle Children's Hospital, Seattle, Washington (Dr Tsuchiya); and the Department of Pathology, Walter Reed National Military Medical Center, Bethesda, Maryland (Dr Moncur)
| |
Collapse
|