51
|
Buza N. HER2 Testing in Endometrial Serous Carcinoma: Time for Standardized Pathology Practice to Meet the Clinical Demand. Arch Pathol Lab Med 2021; 145:687-691. [PMID: 32649220 DOI: 10.5858/arpa.2020-0207-ra] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2020] [Indexed: 01/08/2023]
Abstract
CONTEXT.— Endometrial serous carcinoma is an aggressive subtype of endometrial cancer with the highest rate of recurrence and mortality among all histotypes. A recent clinical trial showed prolonged progression-free survival in advanced-stage and recurrent human epidermal growth factor receptor 2 (HER2)-positive endometrial serous carcinoma when trastuzumab was added to the standard chemotherapy regimen. This targeted therapeutic approach was recently endorsed by the National Comprehensive Cancer Network clinical guidelines. There is a growing interest among clinicians to obtain HER2 testing in endometrial serous carcinoma, and pathologists need to be prepared to recognize the unique characteristics of HER2 protein expression and gene amplification in these tumors and apply specific HER2 scoring criteria. OBJECTIVE.— To provide a historical overview of targeted HER2 therapy in endometrial serous carcinoma and to summarize key findings from recent studies on the specific features of HER2 protein expression and gene amplification relative to other tumor types. Endometrial carcinoma-specific HER2 testing criteria are proposed based on evidence in the existing literature. DATA SOURCES.— Sources comprise review of the literature and personal experience of the author. CONCLUSIONS.— HER2 protein overexpression and/or gene amplification is present in approximately 25% to 30% of endometrial serous carcinomas, providing an opportunity for targeted therapy. Pathologists play a key role in tumor HER2 testing and scoring to ensure appropriate patient selection and successful clinical outcome.
Collapse
Affiliation(s)
- Natalia Buza
- From the Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
52
|
Polónia A, Caramelo A. HER2 in situ hybridization test in breast cancer: quantifying margins of error and genetic heterogeneity. Mod Pathol 2021; 34:1478-1486. [PMID: 33980971 DOI: 10.1038/s41379-021-00813-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/27/2021] [Accepted: 03/29/2021] [Indexed: 11/09/2022]
Abstract
The aim of the present study was to evaluate the effect of counting increasing number of invasive cancer cells in the result of the HER2 in situ hybridization (ISH) test in breast cancer as well as to compare two different approaches of measuring genomic heterogeneity (single cell and population based). A cohort of 100 consecutive breast cancer cases (primary and metastatic) were evaluated for HER2 gene amplification with bright-field ISH. The evaluation of the samples included scoring 20 nuclei, in five different areas, measuring the margins of error for each case. Genomic heterogeneity (GH) was defined by the 2018 ASCO/CAP guideline as a discrete population of tumor cells with HER2 amplification. We also evaluated GH as single tumor cells with HER2 amplification. The stabilization of the coefficient of variation of HER2/CEP17 ratio requires about 60 invasive cancer cells. The average margin of error of HER2/CEP17 ratio and of HER2 copy number was 0.40 and 0.53, respectively, when counting 20 cells, decreasing to 0.20 and 0.26 when counting 100 cells. Population GH was observed in 1% of the cases, while single cell GH was observed in 27% of the cases, reaching its maximum value in cases near the thresholds of positivity. Therefore, margins of error in HER2 ISH test are high, and the minimal cell number recommended in current guidelines should be raised to at least 60 cells. Population GH is a rare event and single cell GH is maximal in cases near the thresholds.
Collapse
Affiliation(s)
- António Polónia
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal. .,I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
| | - Ana Caramelo
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal.,I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| |
Collapse
|
53
|
HER2 Testing Characteristics Can Predict Residual Cancer Burden following Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer. Int J Breast Cancer 2021; 2021:6684629. [PMID: 34123431 PMCID: PMC8166502 DOI: 10.1155/2021/6684629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/09/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives The response to HER2-targeted neoadjuvant chemotherapy (NAC) in HER2-positive (+) breast cancer can be quantified using residual cancer burden (RCB) pathologic evaluation to predict relapse free/overall survival. However, more information is needed to characterize the relationship between patterns of HER2 testing results and response to NAC. We evaluated clinicopathologic characteristics associated with RCB categories in HER2+ patients who underwent HER2-directed NAC. Methods A retrospective chart review was conducted with Stage I-III HER2+ breast cancer cases following NAC and surgical resection. HER2 immunohistochemistry (IHC) staining and fluorescence in situ hybridization (FISH), histologic/clinical characteristics, hormone receptor status, and RCB scores (RCB-0, RCB-I, RCB-II, and RCB-III) were evaluated. Results 64/151 (42.4%) patients with HER2+ disease had pathologic complete response (pCR). Tumors with suboptimal response (RCB-II and RCB-III) were more likely to demonstrate less than 100% HER2 IHC 3+ staining (p < 0.0001), lower HER2 FISH copies (p < 0.0001), and lower HER2/CEP17 ratios (p = 0.0015) compared to RCB-I and RCB-II responses. Estrogen receptor classification using ≥10% versus ≥1% staining showed greater association with higher RCB categories. Conclusions HER2+ characteristics show differing response to therapy despite all being categorized as positive; tumors with less than 100% IHC 3+ staining, lower HER2 FISH copies, and lower HER2/CEP17 ratios resulted in higher RCB scores.
Collapse
|
54
|
Clinical significance of quantitative categorization of HER2 fluorescent in situ hybridization results in invasive breast cancer patients treated with HER2-targeted agents. Mod Pathol 2021; 34:720-734. [PMID: 33479447 DOI: 10.1038/s41379-020-00728-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 11/08/2022]
Abstract
HER2 (ERBB2) gene status serves as a strong predictive marker of response to HER2-targeted agents in invasive breast cancers, albeit with heterogeneous response. Our aim was to determine the distribution and prognosis of HER2 groups by fluorescent in situ hybridization (FISH) using the updated 2018 American Society of Clinical Oncology-College of American Pathologist (ASCO-CAP) guidelines. We identified 226 cases of equivocal or positive HER2 FISH invasive breast cancer (interpreted by ASCO-CAP guidelines at the time of reporting) who received HER2-targeted agents from 2006 to 2017. We subcategorized Group 1 further into three subgroups: low amplified (HER2/CEP17 ratio ≥ 2.0-2.99, mean HER2/cell 4.0-5.9), amplified (HER2/CEP17 ratio ≥ 2.0-2.99, mean HER2/cell ≥ 6), and excessive amplification (HER2/CEP17 ratio ≥ 3.0, mean HER2/cell ≥ 4.0). Outcomes studied were recurrence, metastasis, second breast primary, disease-specific survival (DSS), and overall survival (OS). Univariate analysis showed that the five categories of HER2 FISH were significantly associated with OS (p < 0.01), specifically higher HER2 amplification was associated with fewer deaths. HER2 FISH status also statistically significantly relates to DFS (p < 0.01) and metastasis (p = 0.01) but not with recurrence or second breast primary in our study. Tumor type and HER2 ISH Groups are independent predictors for both OS and DFS in our cohort. The proposed Group 1 subcategories were significantly associated with OS (p < 0.01) and DFS (p < 0.01), excessive HER2 amplification was associated with longer median survival. The Cox regression models showed better survival outcomes for the excessive amplification subgroup than the low amplified subgroup, with OS (hazard ratio = 0.63, 95% CI 0.42-0.93) and DFS (HR = 0.55, 95% CI 0.37-0.83). We demonstrated that in HER2 FISH Group 1 patients, high HER2 amplification was significantly associated with longer OS and DFS; these patients seem to benefit more from HER2-targeted regimens. We recommend reporting these Group 1 subcategories when assessing HER2 FISH.
Collapse
|
55
|
Robinson CL, Harrison BT, Ligon AH, Dong F, Maffeis V, Matulonis U, Nucci MR, Kolin DL. Detection of ERBB2 amplification in uterine serous carcinoma by next-generation sequencing: an approach highly concordant with standard assays. Mod Pathol 2021; 34:603-612. [PMID: 33077919 DOI: 10.1038/s41379-020-00695-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 01/29/2023]
Abstract
Uterine serous carcinoma is an aggressive subtype of endometrial cancer that accounts for fewer than 10% of endometrial carcinomas but is responsible for about half of deaths. A subset of cases has HER2 overexpression secondary to ERBB2 gene amplification, and these patients may benefit from anti-HER2 therapies, such as trastuzumab. HER2 protein overexpression is currently assessed by immunohistochemistry (IHC) and ERBB2 gene amplification by fluorescence in situ hybridization (FISH). Targeted next-generation sequencing (NGS) is increasingly used to routinely identify predictive and prognostic molecular abnormalities in endometrial carcinoma. To investigate the ability of a targeted NGS panel to detect ERBB2 amplification, we identified cases of uterine serous carcinoma (n = 93) and compared HER2 expression by IHC and copy number assessed by FISH with copy number status assessed by NGS. ERBB2 copy number status using a combination of IHC and FISH was interpreted using the 2018 ASCO/CAP guidelines for breast carcinoma. ERBB2 amplification by NGS was determined by the relative number of reads mapping to ERBB2 in tumor DNA compared to control nonneoplastic DNA. Cases with copy number ≥6 were considered amplified and copy number <6 were non-amplified. By IHC, 70 specimens were classified as negative (0 or 1+), 19 were classified as equivocal (2+), and 4 were classified as positive (3+). Using combined IHC/FISH, ERBB2 amplification was observed in 8 of 93 cases (9%). NGS identified the same 8 cases with copy number ≥6; all 85 others had copy number <6. In this series, NGS had 100% concordance with combined IHC/FISH in identifying ERBB2 amplification. NGS is highly accurate in detecting ERBB2 amplification in uterine serous carcinoma and provides an alternative to measurement by IHC and FISH.
Collapse
Affiliation(s)
| | - Beth T Harrison
- Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Azra H Ligon
- Department of Pathology, Division of Clinical Cytogenetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Valeria Maffeis
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | | | - Marisa R Nucci
- Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David L Kolin
- Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
56
|
Predictive significance of HER2 intratumoral heterogeneity, determined by simultaneous gene and protein analysis, for resistance to trastuzumab-based treatments for HER2-positive breast cancer. Virchows Arch 2021; 479:13-21. [PMID: 33496805 DOI: 10.1007/s00428-021-03036-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/01/2021] [Accepted: 01/15/2021] [Indexed: 10/22/2022]
Abstract
Gene-protein assay (GPA), a combination of immunohistochemistry and dual in situ hybridization, allows simultaneous visualization of HER2 protein and gene on a single slide. We aimed to clarify the clinical significance of HER2 intratumoral heterogeneity (ITH) using GPA. We investigated the relationships between various HER2 ITH indicators and clinical course in 102 patients with HER2-positive breast cancer, treated with neoadjuvant trastuzumab and chemotherapy. Five representative microscopic images were captured from each GPA slide of pre-therapeutic biopsy materials. All evaluable cancer cells in the images were individually assessed for HER2 gene copy number and protein expression. Mean and coefficient of variation (CV) of both gene copy number and protein category were calculated, and each was divided into negative, equivocal, and positive. Based on their combined status, cancer cells were classified into nine types. Pathological complete response (pCR) to neoadjuvant treatments showed positive relationships to mean gene copy number (P < 0.001), mean protein category (P < 0.001), and proportion of gene- and protein-positive tumor cells (P < 0.001) and showed negative relationships to the CV of protein category (P < 0.001) and the proportion of gene-amplified but protein-negative tumor cells (P = 0.002). Two diagnostic models, created by combining clinicopathological factors and ITH indicators, showed excellent potential diagnostic ability for pCR (mean gene copy number and protein category CV; AUC = 0.837, proportion of gene- and protein-positive tumor cells; AUC = 0.831). HER2 ITH quantified by GPA is a potential predictive indicator for efficacy of HER2-targeted treatment.
Collapse
|
57
|
Akashi M, Yamaguchi R, Kusano H, Yamaguchi M, Akiba J, Kakuma T, Tanaka M, Akagi Y, Yano H. ER staining levels affect HER2 staining and heterogeneity. Breast Cancer 2021; 28:720-726. [PMID: 33423217 DOI: 10.1007/s12282-020-01208-7] [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: 10/31/2020] [Accepted: 12/20/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE This study explored the relationship of ER expression levels with HER2 staining properties and heterogeneity and discussed the differences in HER2 assessment caused by the 2018 ASCO/CAP guideline updates from that of the 2013 version. METHODS HER2-positive breast cancer was divided into three groups of the high hormone receptor expression (LH-high) group, low expression (LH-low) group, or negative (NLH) group to (1) compare differences in the percentage of the HER2 IHC test score of 2 + based on the 2013 ASCO/CAP guideline and in the intratumor heterogeneity of HER2 expression for breast cancer with an IHC score of 3 + among these groups, (2) compare the HER2/CEP17 ratio and the average HER2 copy number, and classified ISH groupings according to the 2018 ASCO/CAP guideline algorithm. RESULTS (1) Of 244 HER2-positive breast cancers, the cases with a HER2 IHC score of 2 + (n = 54, 22.1%) were significantly more common in the LH-high group (n = 45, P < 0.001). The frequency of heterogeneity was low (n = 25, 10.2%) for the HER2 score of 3 + (n = 190, 77.9%), and significantly higher in the LH-high group (n = 19, 76%, P = 0.002). (2) In a HER2 IHC score of 2 + , Group 2 which is deemed HER2 negative according to the revised 2018 ASCO/CAP guideline was observed in 17 (39.5%) out of 43 cases, of which 16 cases (94.1%) were in the LH-high group. CONCLUSIONS The LH-high group is a heterogeneous group largely consisting of heterogeneous cases with HER2 IHC scores of 2 + or 3 + . NLH, in contrast, is a homogenous group.
Collapse
Affiliation(s)
- Momoko Akashi
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan.,Department of Breast Surgery, National Hospital Organization Kyushu Medical Center, Fukuoka, Fukuoka, Japan
| | - Rin Yamaguchi
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan. .,Department of Pathology and Laboratory Medicine, Kurume University Medical Center, 155-1 Kokubu, Kurume, Fukuoka, 859-0863, Japan.
| | - Hironori Kusano
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Miki Yamaguchi
- Departmen of Surgery, Japan Community Healthcare Organization Kurume General Hospital, Kurume, Fukuoka, Japan
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Tatsuyuki Kakuma
- Biostatistics Center, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Maki Tanaka
- Departmen of Surgery, Japan Community Healthcare Organization Kurume General Hospital, Kurume, Fukuoka, Japan
| | - Yoshito Akagi
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
58
|
Xu Y, Wang C, Chen X, Li Y, Bian W, Yao C. San Huang Decoction Targets Aurora Kinase A to Inhibit Tumor Angiogenesis in Breast Cancer. Integr Cancer Ther 2020; 19:1534735420983463. [PMID: 33349071 PMCID: PMC7758657 DOI: 10.1177/1534735420983463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
San Huang Decoction (SHD), a Chinese herb formula, has been popularly prescribed in the clinical treatment of patients suffering from breast cancer. The aim of this study was to explore the anti-angiogenic effects of SHD in breast cancer and explain the underlying mechanism. Transwell and Matrigel assays showed that SHD reduced human umbilical vein endothelial cell migration and tubule formation and ELISA and qRT-PCR assays demonstrated its mediation of vascular endothelial growth factor (VEGF) expression. siRNA silencing of aurora kinase A (AURKA) produced results similar to those obtained by inhibition of AURKA with SHD. In addition, a chorioallantoic membrane assay was carried out to directly examine the effect of SHD on breast cancer anti-angiogenesis and immunofluorescence and immunohistochemical staining analysis showed that SHD reduced the expression of CD31, AURKA, and VEGF in a xenograft model. Furthermore, SHD regulated extracellular signal-regulated kinase expression in breast cancer cells, which was examined by western blotting. In conclusion, our findings indicated that SHD treatment mimicked the decrease in tumor neovascularization in breast cancer cells after the siRNA-mediated knockdown of AURKA. Thus, SHD may inhibit tumor angiogenesis in breast cancer by targeting AURKA and downregulating the ERK signaling pathway.
Collapse
Affiliation(s)
- Yanlei Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- Liyang Branch of Jiangsu Province Hospital of Chinese Medicine, Changzhou, China
| | - Cong Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Xiyan Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yongfei Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Weihe Bian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Chang Yao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- Chang Yao, Affiliated Hospital of Nanjing University of Chinese Medicine, Han Zhong Road 155, Nanjing, Jiangsu 210029, China. The First Clinical School of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
59
|
Ciesielski M, Szajewski M, Walczak J, Pęksa R, Lenckowski R, Supeł M, Zieliński J, Kruszewski WJ. Impact of chromosome 17 centromere copy number increase on patient survival and human epidermal growth factor receptor 2 expression in gastric adenocarcinoma. Oncol Lett 2020; 21:142. [PMID: 33552261 PMCID: PMC7798021 DOI: 10.3892/ol.2020.12403] [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: 05/19/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022] Open
Abstract
The accurate evaluation of human epidermal growth factor receptor 2 (HER2) status is essential for the appropriate use of targeted therapies. An increased number of chromosome 17 centromere enumeration probe (CEP17) signals may underrate fluorescence in situ hybridization (FISH) outcomes, resulting in false-negative or a false-equivocal HER2 status assessment. The aim of the present study was to assess the frequency of CEP17 copy number increase (CNI), its effects on HER2 protein expression (and the subsequent effects on tumor cells), and the survival outcomes of patients with gastric cancer. Archival primary tumor samples from 244 patients that underwent gastric resection for adenocarcinoma were retrieved for both HER2 protein expression analysis (using immunochemistry) and HER2 gene amplification (using FISH). The associations between HER2 status, CEP17 CNI and multiple clinicopathological parameters (including survival outcome), were assessed. The relationship between CEP17 CNI and HER2 protein upregulation was also investigated. CEP17 CNI was detected in 17.2% of cases, and a strong association between CEP17 CNI and HER2 upregulation was revealed. The impact of CEP17 CNI on survival did not reach statistical significance. Consequently, CEP17 CNI was discovered to be strongly associated with HER2 upregulation in tumor cells, which may characterize a critical issue in HER2 testing. Therefore, the eligibility for HER2-targeted agents in CEP17 CNI-positive patients warrants further recognition.
Collapse
Affiliation(s)
- Maciej Ciesielski
- Department of Oncological Surgery, Gdynia Centre of Oncology, Pomeranian Hospitals, Gdynia, Pomeranian Voivodship 81-519, Poland.,Division of Propedeutics of Oncology, Medical University of Gdańsk, Gdańsk, Pomeranian Voivodship 80-210, Poland
| | - Mariusz Szajewski
- Department of Oncological Surgery, Gdynia Centre of Oncology, Pomeranian Hospitals, Gdynia, Pomeranian Voivodship 81-519, Poland.,Division of Propedeutics of Oncology, Medical University of Gdańsk, Gdańsk, Pomeranian Voivodship 80-210, Poland
| | - Jakub Walczak
- Department of Oncological Surgery, Gdynia Centre of Oncology, Pomeranian Hospitals, Gdynia, Pomeranian Voivodship 81-519, Poland
| | - Rafał Pęksa
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Pomeranian Voivodship 80-210, Poland
| | - Radosław Lenckowski
- Department of Pathomorphology, Gdynia Centre of Oncology, Pomeranian Hospitals, Gdynia, Pomeranian Voivodship 81-519, Poland
| | - Małgorzata Supeł
- Department of Pathomorphology, Gdynia Centre of Oncology, Pomeranian Hospitals, Gdynia, Pomeranian Voivodship 81-519, Poland
| | - Jacek Zieliński
- Department of Oncological Surgery, Medical University of Gdańsk, Gdańsk, Pomeranian Voivodship 80-210, Poland
| | - Wiesław Janusz Kruszewski
- Department of Oncological Surgery, Gdynia Centre of Oncology, Pomeranian Hospitals, Gdynia, Pomeranian Voivodship 81-519, Poland.,Division of Propedeutics of Oncology, Medical University of Gdańsk, Gdańsk, Pomeranian Voivodship 80-210, Poland
| |
Collapse
|
60
|
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
|
61
|
Schmitt ML. Molecular Biomarkers: A Review of Multiple Applications in Clinical Care of Colorectal Cancer. Clin J Oncol Nurs 2020; 24:635-643. [PMID: 33216064 DOI: 10.1188/20.cjon.635-643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Advancements in tumor profiling have identified multiple molecular biomarkers that influence tumor growth and behavior. Molecular biomarkers provide clinical prognostic and predictive information, which guides treatment decisions and forms the backbone of precision oncology. OBJECTIVES This article identifies key predictive and prognostic molecular biomarkers used in the treatment of colorectal cancer and provides greater understanding of their biologic significance and usefulness in guiding treatment decisions. METHODS A review of the literature and professional guidelines was performed to evaluate approved molecular biomarkers, targeted agents, and tumor testing modalities used for the management and treatment of colorectal cancer, with an emphasis on treatment decision making. FINDINGS Genomic biomarkers are increasingly used for the prevention, diagnosis, prognostication, and management of colorectal cancer. The introduction of targeted agents and advancements in tumor profiling technologies have increased treatment opportunities and improved clinical outcomes for patients with colorectal cancer.
Collapse
|
62
|
Klocker EV, Suppan C. Biomarkers in Her2- Positive Disease. Breast Care (Basel) 2020; 15:586-593. [PMID: 33447232 DOI: 10.1159/000512283] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Background Breast cancer is a heterogeneous disease with well-known characteristics such as hormone receptor (HR) status and human epidermal growth factor (Her)2 status. Although Her2 represents an established treatment target, the development of resistance mechanisms during treatment, cardiotoxicity, and a worse response to standard therapies lead to worse outcomes. Summary Therefore, we investigated various biomarkers in breast cancer such as Her2 mutations, Her2 heterogeneity, HR, PIK3CA, PTEN, programmed death receptor ligand 1 (PD-L1), tumor-infiltrating lymphocytes (TIL), micro RNA (miRNA), and BRCA mutations with regard to their clinical impact in Her2-positive disease. HR status and Her2 status, such as the presence of PIK3CA mutations, already play a role in treatment decision-making processes, whereas other biomarkers like PD-L1 status or TIL represent promising future markers. The influence of BRCA mutations in Her2-positive disease, Her2 mutations, and the impact of miRNA is vague to date. Antibody-drug conjugates (ADC) such as T-DM have been established as important treatment strategies, especially in Her2-positive disease. Key Message However, up-to-date biomarkers appropriate for clinical practice are missing. Further studies are needed.
Collapse
Affiliation(s)
| | - Christoph Suppan
- Department of Oncology, Medical University of Graz, Graz, Austria
| |
Collapse
|
63
|
Hiraoka N, Nitta H, Ohba A, Yoshida H, Morizane C, Okusaka T, Nara S, Esaki M, Kishi Y, Shimada K. Details of human epidermal growth factor receptor 2 status in 454 cases of biliary tract cancer. Hum Pathol 2020; 105:9-19. [PMID: 32891647 DOI: 10.1016/j.humpath.2020.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2)-targeted therapy has improved clinical outcomes in patients with HER2-positive breast and gastric cancers, although ineffective or recurrent cases are present. One reason for this is the heterogeneity of HER2 expression in cancer cells. The aim of this study was to investigate the clinicopathological characteristics and HER2 status of patients with biliary tract cancers (BTCs). We examined HER2 protein expression by immunohistochemistry, HER2 gene amplification by fluorescence in situ hybridization, and both HER2 protein and gene levels simultaneously by gene-protein assay. Samples were collected from 454 patients who underwent surgical resection for BTCs (110 intrahepatic cholangiocarcinomas [ICC], 67 perihilar extrahepatic cholangiocarcinomas [ECC-Bp], 119 distal extrahepatic cholangiocarcinomas [ECC-Bd], 80 gallbladder carcinomas [GBC], and 79 ampullary carcinomas [AVC]). HER2 status was assessed according to the guidelines for HER2 testing in gastroesophageal adenocarcinoma. HER2-positive status was detected in 14.5% of BTCs (3.7% of ICC, 3.0% of ECC-Bp, 18.5% of ECC-Bd, 31.3% of GBC, and 16.4% of AVC). Furthermore, HER2-positivity tended to correlate with low histological grade, tumor histology, and macroscopic features in certain tumors. HER2 heterogeneity was common and highly frequent (83%) in BTC cases. Reduced HER2 protein expression in the deeper invasive areas with simultaneous dedifferentiation was frequently observed in HER2-positive cancer cells. The findings of this study suggest that a large subgroup of HER2-positive BTC cases can be considered for HER2-targeted therapy. Moreover, the HER2 status in BTCs should be determined carefully using a sensitive approach toward larger cancer tissues.
Collapse
Affiliation(s)
- Nobuyoshi Hiraoka
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, 104-0045, Japan.
| | | | - Akihiro Ohba
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Hiroshi Yoshida
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Satoshi Nara
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Minoru Esaki
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yoji Kishi
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Kazuaki Shimada
- Hepatobiliary and Pancreatic Surgery Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| |
Collapse
|
64
|
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
|
65
|
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: 1.5] [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
|
66
|
Moussa M, Badawy A, Helal N, Hegab F, Youssef M, Aboushousha T, Al Faruok L, Elwy D. Differential Expression of HER2 and SKP2 in Benign and Malignant Colorectal Lesions. Asian Pac J Cancer Prev 2020; 21:2357-2366. [PMID: 32856866 PMCID: PMC7771937 DOI: 10.31557/apjcp.2020.21.8.2357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Colorectal cancer (CRC) is the fourth most common cancer worldwide. Both HER2 and SKP2 have a carcinogenic role in CRC making them attractive targets for tailored treatment. This work aims to correlate HER2 and SKP2 protein expression as well as HER2 gene amplification with clinicopathological parameters aiming at identifying potential candidates for targeted therapy. Methods: This Study was conducted on 127 paraffin-embedded tissue samples of different colorectal lesions [controls, chronic colitis, ulcerative colitis (UC), hyperplastic polyps (HPs), adenomas and CRCs] to investigate HER2 and SKP2 expression by immunohistochemistry (IHC), Selected CRC cases [equivocal (2+) and positive (3+) by IHC] were further evaluated by ISH (CISH and SISH ) to assess HER2 gene amplification. Results: Chronic colitis, UC, HPs and adenomas were HER2-negative. HER2 positivity (scores 2+ and 3+) was found only in15% of CRCs. Both SISH and CISH showed the same results with high concordance as 66.7% of equivocal and 100% of positive cases showed amplification of HER2 gene. SKP2 positivity was detected in 26.7% and 45% of adenomas and CRCs respectively, while other studied groups were negative. A significant correlation was noted between HER2 and SKP2 expression. Conclusion: A small percent of CRCs exhibited HER2 gene amplification, which would be potential candidates for anti HER2 therapy whereas IHC could be a primary screening test for patient selection. A potential carcinogenic role of SKP2 was suggested by the findings that SKP2 expression was undetectable in normal colonic mucosa but significantly increases from adenoma to carcinoma, hoping adenoma patients to get benefit from targeted therapy.
Collapse
Affiliation(s)
- Mona Moussa
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Afkar Badawy
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Noha Helal
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Fatma Hegab
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Magdy Youssef
- Department of Gastroenterology and Hepatology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Lubna Al Faruok
- Department of Pathology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Dalal Elwy
- Department of Pathology, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
67
|
Mezei T, Hajdu M, Czigléczki G, Lotz G, Kocsis J, Kulka J, Horváth A. Sterile, abscess-like cerebral lesion during trastuzumab therapy after HER2 status switch in a triple negative breast cancer patient: a case report and literature review. BMC Cancer 2020; 20:615. [PMID: 32611325 PMCID: PMC7329406 DOI: 10.1186/s12885-020-07114-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 06/25/2020] [Indexed: 12/31/2022] Open
Abstract
Background Breast cancer is a global health problem – it is the most common malignancy among women. Triple negative breast cancers (TNBC) account for 10–20% of female breast cancer. Most TNBC cases confer poor prognosis. Brain metastasis appears in more than 15% in the triple negative breast cancer population, which causes serious decrease in survival. Changes of immunophenotype are not uncommon in breast cancer, offering new therapeutic options in cases where targetable proteins or pathways are being identified. Case presentation After five lines of chemotherapy and 82 months following the first diagnosis, our patient with brain metastatic triple negative breast cancer had human epidermal growth factor receptor 2 (HER2) genetic heterogeneity in the metastatic tissue sample interpreted as HER2 status conversion. After the removal of the metastasis, we started first line therapy for metastatic HER2 positive cancer with trastuzumab and paclitaxel. After the first cycle of trastuzumab, on day 8, she had a seizure, and neurosurgical examination showed an abscess-like lesion. The punctate proved to be sterile by microbiological and pathological examination, so we continued cytostatic therapy without the anti-HER2 antibody. 3 months later, we could not identify the previous abscess-like lesion in the control computer tomography (CT) scan, and our patient had no neurological deficits. Conclusion We emphasize the importance of regular tissue confirmation of predictive markers in progressive tumorous disease even if our presented case is not unequivocally a “conversion case”. Tumor subtype is determined according to algorithms and definitions published in guidelines, nevertheless, use of different guidelines may lead to controversial interpretation in cases where HER2 genetic heterogeneity is present. Furthermore, we suggest that seronegative, aseptic intracranial fluid effusion after the removal of a brain metastasis may possibly be a side effect of trastuzumab.
Collapse
Affiliation(s)
- Tamás Mezei
- Department of Neurosurgery, Semmelweis University, 57 Amerikai street, Budapest, Pest, 1145, Hungary. .,National Institute of Clinical Neurosciences, 57 Amerikai street, Budapest, Pest, 1145, Hungary.
| | - Melinda Hajdu
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 26 Üllői street, Budapest, Pest, 1085, Hungary
| | - Gábor Czigléczki
- Department of Neurosurgery, Semmelweis University, 57 Amerikai street, Budapest, Pest, 1145, Hungary.,National Institute of Clinical Neurosciences, 57 Amerikai street, Budapest, Pest, 1145, Hungary
| | - Gábor Lotz
- 2nd Department of Pathology, Semmelweis University, 93 Üllői street, Budapest, Pest, 1091, Hungary
| | - Judit Kocsis
- 3rd Department of Internal Medicine, Semmelweis University, 4 Kútvölgyi street, Budapest, Pest, 1125, Hungary
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University, 93 Üllői street, Budapest, Pest, 1091, Hungary
| | - Anna Horváth
- 3rd Department of Internal Medicine, Semmelweis University, 4 Kútvölgyi street, Budapest, Pest, 1125, Hungary
| |
Collapse
|
68
|
Comparison of Dako HercepTest and Ventana PATHWAY Anti-HER2 (4B5) Tests and Their Correlation With Fluorescent In Situ Hybridization in Breast Carcinoma. Appl Immunohistochem Mol Morphol 2020; 27:403-409. [PMID: 31233398 DOI: 10.1097/pai.0000000000000646] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We compared the performance of two Food and Drug Administration-approved HER2 immunohistochemistry (IHC) tests: HercepTest (Dako) and PATHWAY anti-HER2 (4B5) (Ventana). MATERIALS AND METHODS In total, 180 invasive breast carcinomas previously tested by both HercepTest and fluorescent in situ hybridization (FISH) were retested with 4B5. Three pathologists scored the HER2 IHC using the 2013 American Society of Clinical Oncology/College of American Pathologists guidelines. The HER2 IHC results were correlated with FISH. RESULTS Among 135 equivocal cases by HercepTest, 100 (74.1%) were negative by 4B5. Among 45 positive HercepTest cases 9 (20%) were equivocal by 4B5. Among 135 equivocal HercepTest results, 100 (74.1%) were nonamplified, 18 (13.3%) equivocal, and 17 (12.6%) amplified by FISH. Among the 45 positive results with HercepTest, 2 (4.5%) were nonamplified and 1 (2.2%) was equivocal by FISH. All 37 positive and 3 negative by 4B5 cases were amplified by FISH. The absolute interobserver agreement was high for both tests (Fleiss kappa=0.838 for HercepTest and 0.771 for 4B5). CONCLUSIONS PATHWAY anti-HER2 (4B5) significantly reduced the number of equivocal results that require additional testing. Although HercepTest was positive in a small number of HER2 nonamplified cases, 4B5 failed to detect 3 cases that were interpreted as positive by FISH, all with nonclassic or low levels of amplification.
Collapse
|
69
|
Lu H, Yin J. Texture Analysis of Breast DCE-MRI Based on Intratumoral Subregions for Predicting HER2 2+ Status. Front Oncol 2020; 10:543. [PMID: 32373531 PMCID: PMC7186477 DOI: 10.3389/fonc.2020.00543] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/26/2020] [Indexed: 01/04/2023] Open
Abstract
Background: Breast tumor heterogeneity is related to risk factors that lead to aggressive tumor growth; however, such heterogeneity has not been thoroughly investigated. Purpose: To evaluate the performance of texture features extracted from heterogeneity subregions on subtraction MRI images for identifying human epidermal growth factor receptor 2 (HER2) 2+ status of breast cancers. Materials and Methods: Seventy-six patients with HER2 2+ breast cancer who underwent dynamic contrast-enhanced magnetic resonance imaging were enrolled, including 42 HER2 positive and 34 negative cases confirmed by fluorescence in situ hybridization. The lesion area was delineated semi-automatically on the subtraction MRI images at the second, fourth, and sixth phases (P-1, P-2, and P-3). A regionalization method was used to segment the lesion area into three subregions (rapid, medium, and slow) according to peak arrival time of the contrast agent. We extracted 488 texture features from the whole lesion area and three subregions independently. Wrapper, least absolute shrinkage and selection operator (LASSO), and stepwise methods were used to identify the optimal feature subsets. Univariate analysis was performed as well as support vector machine (SVM) with a leave-one-out-based cross-validation method. The area under the receiver operating characteristic curve (AUC) was calculated to evaluate the performance of the classifiers. Results: In univariate analysis, the variance from medium subregion at P-2 was the best-performing feature for distinguishing HER2 2+ status (AUC = 0.836); for the whole lesion region, the variance at P-2 achieved the best performance (AUC = 0.798). There was no significant difference between the two methods (P = 0.271). In the machine learning with SVM, the best performance (AUC = 0.929) was achieved with LASSO from rapid subregion at P-2; for the whole region, the highest AUC value was 0.847 obtained at P-2 with LASSO. The difference was significant between the two methods (P = 0.021). Conclusion: The texture analysis of heterogeneity subregions based on intratumoral regionalization method showed potential value for recognizing HER2 2+ status in breast cancer.
Collapse
Affiliation(s)
- Hecheng Lu
- School of Medicine and Bioinformatics Engineering, Northeastern University, Shenyang, China.,Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, ShengJing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
70
|
Van Bockstal MR, Agahozo MC, van Marion R, Atmodimedjo PN, Sleddens HFBM, Dinjens WNM, Visser LL, Lips EH, Wesseling J, van Deurzen CHM. Somatic mutations and copy number variations in breast cancers with heterogeneous HER2 amplification. Mol Oncol 2020; 14:671-685. [PMID: 32058674 PMCID: PMC7138394 DOI: 10.1002/1878-0261.12650] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/13/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022] Open
Abstract
Intratumour heterogeneity fuels carcinogenesis and allows circumventing specific targeted therapies. HER2 gene amplification is associated with poor outcome in invasive breast cancer. Heterogeneous HER2 amplification has been described in 5-41% of breast cancers. Here, we investigated the genetic differences between HER2-positive and HER2-negative admixed breast cancer components. We performed an in-depth analysis to explore the potential heterogeneity in the somatic mutational landscape of each individual tumour component. Formalin-fixed, paraffin-embedded breast cancer tissue of ten patients with at least one HER2-negative and at least one HER2-positive component was microdissected. Targeted next-generation sequencing was performed using a customized 53-gene panel. Somatic mutations and copy number variations were analysed. Overall, the tumours showed a heterogeneous distribution of 12 deletions, 9 insertions, 32 missense variants and 7 nonsense variants in 26 different genes, which are (likely) pathogenic. Three splice site alterations were identified. One patient had an EGFR copy number gain restricted to a HER2-negative in situ component, resulting in EGFR protein overexpression. Two patients had FGFR1 copy number gains in at least one tumour component. Two patients had an 8q24 gain in at least one tumour component, resulting in a copy number increase in MYC and PVT1. One patient had a CCND1 copy number gain restricted to a HER2-negative tumour component. No common alternative drivers were identified in the HER2-negative tumour components. This series of 10 breast cancers with heterogeneous HER2 gene amplification illustrates that HER2 positivity is not an unconditional prerequisite for the maintenance of tumour growth. Many other molecular aberrations are likely to act as alternative or collaborative drivers. This study demonstrates that breast carcinogenesis is a dynamically evolving process characterized by a versatile somatic mutational profile, of which some genetic aberrations will be crucial for cancer progression, and others will be mere 'passenger' molecular anomalies.
Collapse
Affiliation(s)
| | | | - Ronald van Marion
- Department of PathologyErasmus MC Cancer Institute RotterdamThe Netherlands
| | | | | | | | - Lindy L. Visser
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Esther H. Lips
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jelle Wesseling
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | | |
Collapse
|
71
|
Van Bockstal MR, Dubois D, Talpe S, Galant C. Isolated CEP17 Copy Number Gain in Invasive Breast Cancer Results in a "Reverse" Amplification Status. Int J Surg Pathol 2020; 29:76-77. [PMID: 32131665 DOI: 10.1177/1066896920911421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Mieke R Van Bockstal
- Cliniques universitaires Saint-Luc, Brussels, Belgium
- Ghent University, Ghent, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Christine Galant
- Cliniques universitaires Saint-Luc, Brussels, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
72
|
Marchiò C, Annaratone L, Marques A, Casorzo L, Berrino E, Sapino A. Evolving concepts in HER2 evaluation in breast cancer: Heterogeneity, HER2-low carcinomas and beyond. Semin Cancer Biol 2020; 72:123-135. [PMID: 32112814 DOI: 10.1016/j.semcancer.2020.02.016] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/16/2020] [Accepted: 02/22/2020] [Indexed: 02/06/2023]
Abstract
The human epidermal growth factor receptor 2 (HER2) is a well-known negative prognostic factor in breast cancer and a target of the monoclonal antibody trastuzumab as well as of other anti-HER2 compounds. Pioneering works on HER2-positive breast cancer in the 90s' launched a new era in clinical research and oncology practice that has reshaped the natural history of this disease. In diagnostic pathology the HER2 status is routinely assessed by using a combination of immunohistochemistry (IHC, to evaluate HER2 protein expression levels) and in situ hybridization (ISH, to assess HER2 gene status). For this purpose, international recommendations have been developed by a consensus of experts in the field, which have changed over the years according to new experimental and clinical data. In this review article we will document the changes that have contributed to a better evaluation of the HER2 status in clinical practice, furthermore we will discuss HER2 heterogeneity defined by IHC and ISH as well as by transcriptomic analysis and we will critically describe the complexity of HER2 equivocal results. Finally, we will introduce the clinical impact of HER2 mutations and we will define the upcoming category of HER2-low breast cancer with respect to emerging clinical data on the efficacy of specific anti-HER2 agents in subgroups of breast carcinomas lacking the classical oncogene addition dictated by HER2 amplification.
Collapse
Affiliation(s)
- Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| | - Laura Annaratone
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Ana Marques
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Pathology Unit, Centro Hospitalar São João, Porto, Portugal
| | - Laura Casorzo
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Enrico Berrino
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Turin, Italy; Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
73
|
Woo JW, Lee K, Chung YR, Jang MH, Ahn S, Park SY. The updated 2018 American Society of Clinical Oncology/College of American Pathologists guideline on human epidermal growth factor receptor 2 interpretation in breast cancer: comparison with previous guidelines and clinical significance of the proposed in situ hybridization groups. Hum Pathol 2020; 98:10-21. [PMID: 32027910 DOI: 10.1016/j.humpath.2020.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/06/2020] [Accepted: 01/23/2020] [Indexed: 12/17/2022]
Abstract
The aim of the study was to evaluate the impact of the updated 2018 American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guideline on human epidermal growth factor receptor 2 (HER2) interpretation in breast cancer compared with that of the previous guidelines and also the significance of in situ hybridization (ISH) groups proposed by the updated guideline. HER2 ISH reports and immunohistochemistry (IHC) data from 1,348 invasive breast cancers diagnosed at a single institution were included in this study. HER2 IHC was reassessed using the 2018 guideline, and HER2 ISH status was determined by the 2007, 2013, and 2018 guidelines. When applying the updated guideline, most of the HER2 ISH-equivocal cases as per the previous guidelines were reclassified as ISH negative, and 0.8% of HER2 ISH-positive tumors as per the 2007 guideline and 2.5% of those as per the 2013 guideline were changed to ISH negative. Accordingly, the negative HER2 ISH results significantly increased in the 2018 guideline compared with the 2013 guideline. HER2 ISH-positive tumors in ISH group 3 (HER2/chromosome enumeration probe 17 [CEP17] ratio <2.0 and average HER2 copy number ≥6.0 per cell) were characterized by equivocal HER2 protein expression, CEP17 copy number gain, and low HER2 copy numbers compared with classic HER2 ISH-positive tumors in ISH group 1 (HER2/CEP17 ratio ≥2.0 and average HER2 copy number ≥4.0 per cell). HER2 ISH-negative tumors in ISH group 4 (HER2/CEP17 ratio <2.0 with average HER2 copy number ≥4.0 and < 6.0 per cell) revealed more aggressive clinicopathologic features and poorer clinical outcomes than those in ISH group 5 (HER2/CEP17 ratio <2.0 and average HER2 copy number <4.0 per cell), especially in the hormone receptor-positive subgroup. In conclusion, implementation of the updated 2018 ASCO/CAP guideline leads to a significant increase in HER2 ISH-negative results compared with the 2013 guideline, mainly via reclassification of the ISH-equivocal cases to ISH-negative ones. ISH groups proposed by the updated guideline provide additional information on the clinicopathologic characteristics of the tumors.
Collapse
Affiliation(s)
- Ji Won Woo
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, 13620, Republic of Korea
| | - Kyoungyul Lee
- Department of Pathology, Kangwon National University Hospital, Chuncheon, Kangwon, 24289, Republic of Korea
| | - Yul Ri Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, 13620, Republic of Korea
| | - Min Hye Jang
- Department of Pathology, Yeungnam University Medical Center, Daegu, 42415, Republic of Korea
| | - Soomin Ahn
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, 13620, Republic of Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Gyeonggi, 13620, Republic of Korea.
| |
Collapse
|
74
|
Hou Y, Nitta H, Parwani AV, Li Z. The assessment of HER2 status and its clinical implication in breast cancer. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.mpdhp.2019.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
75
|
Lee K, Kim HJ, Jang MH, Lee S, Ahn S, Park SY. Centromere 17 copy number gain reflects chromosomal instability in breast cancer. Sci Rep 2019; 9:17968. [PMID: 31784614 PMCID: PMC6884473 DOI: 10.1038/s41598-019-54471-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/15/2019] [Indexed: 12/23/2022] Open
Abstract
Chromosomal instability (CIN) is known to be associated with prognosis and treatment response in breast cancer. This study was conducted to determine whether copy number gain of centromere 17 (CEP17) reflects CIN, and to evaluate the prognostic and predictive value of CIN in breast cancer. CIN status was determined by summing copy number gains of four centromeric probes (CEP1, CEP8, CEP11, and CEP16) based on fluorescence in situ hybridization and CIN scores were calculated using next generation sequencing data. High CIN was associated with adverse clinicopatholgical parameters of breast cancer. Among them, positive HER2 status, high Ki-67 index and CEP17 copy number gain were found to be independent predictors of high CIN. High CIN was associated with poor clinical outcome of the patients in the whole group, as well as in luminal/HER2-negative and HER2-positive subtypes. CEP17 copy number was significantly higher in the high-CIN-score group than in the low-CIN-score group. A positive linear correlation between the mean CEP17 copy number and the CIN score was found. In conclusion, CEP17 copy number was confirmed as a useful predictor for CIN in breast cancer, and high CIN was revealed as an indicator of poor prognosis in breast cancer.
Collapse
Affiliation(s)
- Kyoungyul Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Kangwon National University Hospital, Chuncheon, Kangwon, Republic of Korea
| | - Hyun Jeong Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea
| | - Min Hye Jang
- Department of Pathology, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Sejoon Lee
- Precision Medicine Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea
| | - Soomin Ahn
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea.
| |
Collapse
|
76
|
Ahn S, Woo JW, Lee K, Park SY. HER2 status in breast cancer: changes in guidelines and complicating factors for interpretation. J Pathol Transl Med 2019; 54:34-44. [PMID: 31693827 PMCID: PMC6986968 DOI: 10.4132/jptm.2019.11.03] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/03/2019] [Indexed: 12/16/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) protein overexpression and/or HER2 gene amplification is found in about 20% of invasive breast cancers. It is a sole predictive marker for treatment benefits from HER2 targeted therapy and thus, HER2 testing is a routine practice for newly diagnosed breast cancer in pathology. Currently, HER2 immunohistochemistry (IHC) is used for a screening test, and in situ hybridization is used as a confirmation test for HER2 IHC equivocal cases. Since the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines on HER2 testing was first released in 2007, it has been updated to provide clear instructions for HER2 testing and accurate determination of HER2 status in breast cancer. During HER2 interpretation, some pitfalls such as intratumoral HER2 heterogeneity and increase in chromosome enumeration probe 17 signals may lead to inaccurate assessment of HER2 status. Moreover, HER2 status can be altered after neoadjuvant chemotherapy or during metastatic progression, due to biologic or methodologic issues. This review addresses recent updates of ASCO/CAP guidelines and factors complicating in the interpretation of HER2 status in breast cancers.
Collapse
Affiliation(s)
- Soomin Ahn
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Won Woo
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoungyul Lee
- Department of Pathology, Kangwon National University Hospital, Chuncheon, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
77
|
Godoy-Ortiz A, Sanchez-Muñoz A, Chica Parrado MR, Álvarez M, Ribelles N, Rueda Dominguez A, Alba E. Deciphering HER2 Breast Cancer Disease: Biological and Clinical Implications. Front Oncol 2019; 9:1124. [PMID: 31737566 PMCID: PMC6828840 DOI: 10.3389/fonc.2019.01124] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
The main obstacle for designing effective treatment approaches in breast cancer is the extensive and the characteristic heterogeneity of this tumor. The vast majority of critical genomic changes occurs during breast cancer progression, creating a significant variability within primary tumors as well as between the primary breast cancer and their metastases, a hypothesis have already demonstrated in retrospective studies (1). A clear example of this is the HER2-positive breast cancer. In these tumors, we can find all of the transcriptional subtypes of breast cancer, even the basal like or luminal A subtypes. Although the HER2-enriched is the most representative transcriptional subtype in the HER2-positive breast cancer, we can find it too in breast cancers with HER2-negative status. This intrinsic subtype shows a high expression of the HER2 and is associated with proliferation-related genes clusters, among other features. Therefore, two hypotheses can be suggested. First, the HER2 amplification can be a well-defined driver event present in all of the intrinsic subtypes, and not a subtype marker isolated. Secondly, HER2-enriched subtype can have a distinctive transcriptional landscape independent of HER2 amplification. In this review, we present an extensive revision about the last highlights and advances in clinical and genomic settings of the HER2-positive breast cancer and the HER2-enriched subtype, in an attempt to improving the knowledge of the underlying biology of both entities and to explaining the intrinsic heterogeneity of HER2-positive breast cancers.
Collapse
Affiliation(s)
- Ana Godoy-Ortiz
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Alfonso Sanchez-Muñoz
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Maria Rosario Chica Parrado
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Martina Álvarez
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Nuria Ribelles
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Antonio Rueda Dominguez
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
| | - Emilio Alba
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
- Laboratorio de Biología Molecular del Centro de Investigaciones Médico-Sanitarias de Málaga (CIMES), Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga (UMA), Málaga, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| |
Collapse
|
78
|
Patil Okaly GV, Panwar D, Lingappa KB, Kumari P, Anand A, Kumar P, Chikkalingaiah MH, Kumar RV. FISH and HER2/ neu equivocal immunohistochemistry in breast carcinoma. Indian J Cancer 2019; 56:119-123. [PMID: 31062729 DOI: 10.4103/ijc.ijc_333_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIM The aim of this study was to validate the role of fluorescence in situ hybridization (FISH) in investigating HER2/neu gene amplification (human epidermal growth factor receptor 2) in patients with HER2/neu equivocal breast cancer diagnosed on immunohistochemistry (IHC). MATERIALS AND METHODS This was a retrospective study conducted from January 2013 to October 2017. A total of 134 patients diagnosed with invasive breast carcinoma and HER2/neu equivocal status on IHC were analyzed. Also, the cases for the years 2016 and 2017 formed a subgroup that was analyzed further to study the impact of pre-analytical factors on IHC and FISH results. RESULTS A total of 134 women with HER2/neu IHC equivocal breast cancer were included in the study with a median age of 50 years (range 25-81). HER2/neu amplification by FISH was noted in 72 (54%) cases, whereas it was non-amplified in 52 (39%) cases. Ten cases were reported as equivocal even on FISH (ASCO/CAP 2013 guidelines). Polysomy 17 was noted in 55 cases (41%), of which 26 patients were≤50 years and 29 patients were >50 years of age. Twenty (36%) of these 55 cases showed HER2/neu amplification, whereas 26 (48%) cases were non-amplified and 9 (16%) cases were reported as equivocal on FISH. Also, more than half of the polysomy cases were hormone receptor negative. CONCLUSION IHC is a good screening tool for negative and positive results. Any patient targeted for trastuzumab therapy should undergo confirmation of HER2/neu equivocal status by FISH analysis. We also suggest that if a non-classical FISH pattern is seen, the test should be repeated with a non-centromeric chromosome 17 reference locus probe for better treatment planning.
Collapse
Affiliation(s)
| | - Dipti Panwar
- Department of Pathology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | | | - Prasanna Kumari
- Department of Pathology, Cytogenetics Unit, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - Abhishek Anand
- Department of Medical Oncology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | - Prashantha Kumar
- Department of Pathology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| | | | - Rekha Vijay Kumar
- Department of Pathology, Kidwai Cancer Institute, Bengaluru, Karnataka, India
| |
Collapse
|
79
|
Wei Q, Zhang Y, Gao J, Li J, Li J, Li Y, Zhou J, Lu M, Gong J, Zhang X, Shen L, Sun Y, Chang L, Wang X. Clinicopathologic Characteristics of HER2-positive Metastatic Colorectal Cancer and Detection of HER2 in Plasma Circulating Tumor DNA. Clin Colorectal Cancer 2019; 18:175-182. [DOI: 10.1016/j.clcc.2019.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/17/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
|
80
|
Baroš IV, Tanasković N, Pellas U, Eri Ž, Tadić Latinović L, Tot T. Internodal HER2 heterogeneity of axillary lymph node metastases in breast cancer patients. Bosn J Basic Med Sci 2019; 19:242-248. [PMID: 30957723 DOI: 10.17305/bjbms.2019.3970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/11/2018] [Indexed: 11/16/2022] Open
Abstract
Determination of human epidermal growth factor receptor 2 (HER2) status is important for adequate treatment of breast cancer (BC) patients. The novel HER2 gene protein assay (GPA) is particularly convenient, as it allows the simultaneous assessment of HER2 protein expression and gene amplification at individual cell level. Here we investigated the frequency of internodal HER2 heterogeneity in axillary lymph node macrometastases of BC patients and compared HER2 status between primary breast tumor and its metastases. We included a total of 41 female patients operated between 2014 and 2015 for primary BC with axillary lymph node macrometastases. Representative paraffin blocks of metastatic lymph nodes were sectioned and the slides were stained using the GPA in 38 BC cases. GPA results were assessed according to the ASCO/CAP 2013 criteria. We analyzed 12586 individual tumor cells, 120 cells per section of each metastatic lymph node. HER2 status differed between the primary tumor and its metastases in 5/38 cases (13.2%). In patients with at least two metastatic nodes, the HER2 status of lymph node metastases was only slightly different in 4/23 cases (17.4%). Our results indicate rare but substantial differences in HER2 status between primary breast tumor and its axillary lymph node metastases that may direct the choice and outcomes of targeted therapy in BC patients. The impact of the rare and subtle internodal HER2 heterogeneity evidenced in this study remains uncertain. Determining the HER2 status of lymph node metastases in BC seems to be rational, but assessing a limited number of metastatic nodes may be sufficient.
Collapse
Affiliation(s)
- Ilija Vladimir Baroš
- Department of Clinical Pathology, University Clinical Centre of the Republic of Srpska, Banja Luka, Bosnia and Herzegovina.
| | | | | | | | | | | |
Collapse
|
81
|
Halilovic A, Verweij DI, Simons A, Stevens-Kroef MJPL, Vermeulen S, Elsink J, Tops BBJ, Otte-Höller I, van der Laak JAWM, van de Water C, Boelens OBA, Schlooz-Vries MS, Dijkstra JR, Nagtegaal ID, Tol J, van Cleef PHJ, Span PN, Bult P. HER2, chromosome 17 polysomy and DNA ploidy status in breast cancer; a translational study. Sci Rep 2019; 9:11679. [PMID: 31406196 PMCID: PMC6690925 DOI: 10.1038/s41598-019-48212-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 07/24/2019] [Indexed: 01/19/2023] Open
Abstract
Breast cancer treatment depends on human epidermal growth factor receptor-2 (HER2) status, which is often determined using dual probe fluorescence in situ hybridisation (FISH). Hereby, also loss and gain of the centromere of chromosome 17 (CEP17) can be observed (HER2 is located on chromosome 17). CEP17 gain can lead to difficulty in interpretation of HER2 status, since this might represent true polysomy. With this study we investigated whether isolated polysomy is present and how this effects HER2 status in six breast cancer cell lines and 97 breast cancer cases, using HER2 FISH and immunohistochemistry, DNA ploidy assessment and multiplex ligation dependent probe amplification. We observed no isolated polysomy of chromosome 17 in any cell line. However, FISH analysis did show CEP17 gain in five of six cell lines, which reflected gains of the whole chromosome in metaphase spreads and aneuploidy with gain of multiple chromosomes in all these cases. In patients' samples, gain of CEP17 indeed correlated with aneuploidy of the tumour (91.1%; p < 0.001). Our results indicate that CEP17 gain is not due to isolated polysomy, but rather due to widespread aneuploidy with gain of multiple chromosomes. As aneuploidy is associated with poor clinical outcome, irrespective of tumour grade, this could improve future therapeutic decision making.
Collapse
Affiliation(s)
- Altuna Halilovic
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands. .,Department of Tumor Immunology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands.
| | - Dagmar I Verweij
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Annet Simons
- Department of Human Genetics, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | | | - Susan Vermeulen
- Department of Human Genetics, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Janet Elsink
- Department of Human Genetics, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Bastiaan B J Tops
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Irene Otte-Höller
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | | | - Carlijn van de Water
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | | | | | - Jeroen R Dijkstra
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Jolien Tol
- Department of Medical Oncology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands.,Department of Medical Oncology, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Patricia H J van Cleef
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy & OncoImmunology laboratory, Department of Radiation Oncology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| | - Peter Bult
- Department of Pathology, Radboud university medical center (Radboudumc), Nijmegen, The Netherlands
| |
Collapse
|
82
|
Comparison of Dual-ISH (DISH) With Fluorescence In Situ Hybridization (FISH) and Correlation With Immunohistochemical Findings for HER2/Neu Status in Breast Carcinoma. Appl Immunohistochem Mol Morphol 2019; 25:231-236. [PMID: 26766122 DOI: 10.1097/pai.0000000000000304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION The most widely used methods for determination of HER2/neu status in breast carcinoma are immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). Both techniques are associated with technical and interpretive difficulties. Alternative methods exist including quantitative PCR and the newly developed chromogenic dual in situ hybridization (DISH). METHODS We evaluated HER2 DISH as an alternative to FISH and report our findings from 101 cases. In addition, we correlated HER2 DISH and FISH results with HercepTest and 4B5 immunohistochemistry. RESULTS Eight cases failed FISH analysis and none failed DISH analysis. A 95% (88/93) concordance was found between DISH and FISH for all cases in the series. When only 2+ IHC cases were evaluated, the concordance was 94% for DISH and FISH. Using the 2013 ASCO/CAP recommendations, none of the tested cases were equivocal by FISH or DISH despite 66% of cases being 2+ by HercepTest and 32% by the 4B5 antibody. COMMENT Our study, which utilizes a majority of IHC equivocal cases, demonstrates that HER2 FISH and DISH are concordant methodologies. HER2 DISH is therefore an acceptable alternative to FISH.
Collapse
|
83
|
Masuda S, Nitta H, Kelly BD, Zhang W, Farrell M, Dennis E. Intratumoral Estrogen Receptor Heterogeneity of Expression in Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer as Evaluated by a Brightfield Multiplex Assay. J Histochem Cytochem 2019; 67:563-574. [PMID: 31184528 DOI: 10.1369/0022155419856862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Breast cancer (BC) is a heterogeneous disease with evolving genetic alterations and expressions of receptor proteins. Intratumoral heterogeneity (ITH) is considered to be a resistance factor in response to targeted therapies. The current single-slide, single-marker immunohistochemistry techniques cannot accurately assess ITH at the individual cancer cell level. In this study, we develop a novel brightfield multiplex assay to simultaneously assess estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) protein markers, together with the HER2 gene and the centromere of chromosome 17 (CEP17) copy numbers, using a single tissue section. The data presented herein demonstrate heterogeneous cancer cell subpopulations in 11 HER2-positive/ER-positive (HER2+/ER+) tumors among 33 BCs analyzed immunohistochemically (HER2 score of 2+ or 3+). The predominant cancer cell subpopulation was HER2+/ER- (50.18%), followed by HER2+/ER+ (39.05%), HER2-/ER+ (4.26%), ER- with HER2 microheterogeneity cancer cells (3.58%), and ER+ with HER2 microheterogeneity cancer cells (2.93%). The three other tumor subtypes, namely, HER2-/ER+, HER2+/ER-, and HER2-/ER-, were more homogeneous, representing 82.59%, 99.22%, and 100% of cancer cells, respectively. This novel assay revealed that HER2+ cancer cells were more predominant than ER+ cancer cells in HER2+/ER+ tumors and provided new insights toward our understanding of BC carcinogenesis.
Collapse
Affiliation(s)
- Shinobu Masuda
- Division of Oncologic Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Hiroaki Nitta
- Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona
| | - Brian D Kelly
- Tissue Research and Early Development, Roche Tissue Diagnostics, Tucson, Arizona
| | - Wenjun Zhang
- Tissue Research and Early Development, Roche Tissue Diagnostics, Tucson, Arizona
| | - Michael Farrell
- Tissue Research and Early Development, Roche Tissue Diagnostics, Tucson, Arizona
| | - Eslie Dennis
- Medical & Scientific Affairs, Roche Tissue Diagnostics, Tucson, Arizona
| |
Collapse
|
84
|
Muller KE, Marotti JD, Tafe LJ. Pathologic Features and Clinical Implications of Breast Cancer With HER2 Intratumoral Genetic Heterogeneity. Am J Clin Pathol 2019; 152:7-16. [PMID: 30892594 DOI: 10.1093/ajcp/aqz010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To review breast cancer with human epidermal growth factor receptor 2 (HER2) genetic heterogeneity (GH), with a focus on clinician interpretation of the results and subsequent management. METHODS We retrospectively searched all consecutive invasive breast cancers that underwent HER2 fluorescent in situ hybridization testing and identified cases with HER2 GH. RESULTS Eleven (0.4%) cases of primary breast tumors with reported HER2 GH were identified over 76 months. HER2-positive tumor cells comprised a mean ± SD of 20% ± 15% of the overall tumor population (range, 5%-50%) and exhibited high levels of amplification (HER2/CEP17 = 5.4 ± 2.9). Three cases had morphologic heterogeneity. Of the four patients who completed neoadjuvant chemotherapy, two had trastuzumab; all showed a partial pathologic response. We identified two cases in which there appeared to be clinical misinterpretation of the HER2 results. CONCLUSIONS HER2 GH is an infrequent event, may contain morphologic heterogeneity, could infer incomplete response to neoadjuvant therapy, and requires adequate communication with treating clinicians.
Collapse
Affiliation(s)
- Kristen E Muller
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Laura J Tafe
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| |
Collapse
|
85
|
Kurihara N, Imai K, Nanjo H, Nakamura R, Wakamatsu Y, Akagami K, Terata K, Wakita A, Sato Y, Motoyama S, Akagami Y, Minamiya Y. Practical application of non-contact alternating current electric field mixing for reagent-saving in situ hybridisation of HER2. J Clin Pathol 2019; 72:603-608. [PMID: 31129615 DOI: 10.1136/jclinpath-2019-205830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/24/2019] [Accepted: 05/01/2019] [Indexed: 11/03/2022]
Abstract
AIMS Human epidermal growth factor receptor 2 (HER2)-targeted agents are effective against HER2-positive breast cancers. However, their lack of survival benefit in HER2-negative patients as well as their toxic effects and high cost highlight the need for accurate assessment of HER2 status. Our aim was to evaluate the clinical utility of a reagent-saving in situ hybridisation (Saving ISH) that facilitates hybridisation and saves HER2/chromosome enumeration probe by taking advantage of the non-contact mixing effect of an alternating current (AC) electric field. METHODS With a new device, we apply a high-voltage, low-frequency AC electric field to the tissue sections, which mixes the probe within microdroplets as the voltage is switched on and off. Specimens (n=113) from patients with breast cancers identified immunohistochemically as HER2 0/1(+), (2+) or (3+) were used. The specimens were all tested using conventional dual ISH (DISH), DISH with an automated slide stainer (ASS) and Saving ISH (1:1-1:3 dilution). RESULTS The Saving ISH with 1:2 probe dilution produced stable results with less non-specific staining while using smaller amounts of probe. The accuracy of HER2 status with Saving ISH was equal to standard. We found 96.4% agreement between DISH using ASS and Saving ISH (kappa coefficient=0.912). CONCLUSIONS These results suggest reagent-saving HER2 ISH could be used as a clinical tool for accurate and stable HER2 assessment, even when reagent concentrations vary.
Collapse
Affiliation(s)
- Nobuyasu Kurihara
- 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
- Division of Clinical Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | | | - Yuki Wakamatsu
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Koji Akagami
- 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
| | - Akiyuki Wakita
- 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
| | | | - Yoshihiro Minamiya
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
86
|
Chromogenic and Silver in Situ Hybridization for Identification of HER 2 Overexpression in Breast Cancer Patients: A Systematic Review and Meta-Analysis. Appl Immunohistochem Mol Morphol 2019; 28:411-421. [DOI: 10.1097/pai.0000000000000773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
87
|
High-content, cell-by-cell assessment of HER2 overexpression and amplification: a tool for intratumoral heterogeneity detection in breast cancer. J Transl Med 2019; 99:722-732. [PMID: 30659272 PMCID: PMC6522386 DOI: 10.1038/s41374-018-0172-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 01/25/2023] Open
Abstract
Immunohistochemistry and fluorescence in situ hybridization are the two standard methods for human epidermal growth factor receptor 2 (HER2) assessment. However, they have severe limitations to assess quantitatively intratumoral heterogeneity (ITH) when multiple subclones of tumor cells co-exist. We develop here a high-content, quantitative analysis of breast cancer tissues based on microfluidic experimentation and image processing, to characterize both HER2 protein overexpression and HER2 gene amplification at the cellular level. The technique consists of performing sequential steps on the same tissue slide: an immunofluorescence (IF) assay using a microfluidic protocol, an elution step for removing the IF staining agents, a standard FISH staining protocol, followed by automated quantitative cell-by-cell image processing. Moreover, ITH is accurately detected in both cluster and mosaic form using an analysis of spatial association and a mathematical model that allows discriminating true heterogeneity from artifacts due to the use of thin tissue sections. This study paves the way to evaluate ITH with high accuracy and content while requiring standard staining methods.
Collapse
|
88
|
Sawada R, Maehara R, Oshikiri T, Nakamura T, Itoh T, Kodama Y, Kakeji Y, Zen Y. MDM2 copy number increase: a poor prognostic, molecular event in esophageal squamous cell carcinoma. Hum Pathol 2019; 89:1-9. [PMID: 31004651 DOI: 10.1016/j.humpath.2019.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 12/09/2022]
Abstract
The present study aimed to elucidate the clinicopathological significance of molecular alterations in MDM2 in esophageal squamous cell carcinoma (ESCC). A total of 399 resected cases of ESCC were examined by dual-color in situ hybridization for MDM2 and immunohistochemistry for p53 using tissue microarrays. Clinicopathological features were correlated with the MDM2 status. Among 362 cases with a successful dual-color in situ hybridization analysis, 19 (5%) and 13 (4%) had MDM2 amplification and chromosome 12 polysomy, respectively, and these were examined as an MDM2-positive group. A comparison between amplified and polysomic cases revealed that the latter were more strongly associated with preoperative chemotherapy than the former. Sixteen (50%) of 32 MDM2-positive cases had positive results in all tissue cores examined, indicating diffuse MDM2 alterations. Cases with the diffuse alteration of MDM2 were characterized by an advanced pT stage and extensive vascular infiltration. The relationship between MDM2 copy number increases and p53 mutations was weak, with the overexpression of p53 being similarly detected in MDM2-positive and MDM2-negative cases (59% versus 49%; P = .267). Overall survival was shorter in patients with MDM2-positive ESCC than in those without MDM2 alterations (P = .033). The poor prognostic value of MDM2 alterations became more obvious when only diffusely altered cases were counted (P = .005). In conclusion, the present study revealed that MDM2 copy number increases occurred in 9% of ESCC cases, and MDM2 alterations, particularly diffuse abnormalities, were associated with a poor prognosis. MDM2-altered ESCC may achieve beneficial effects from MDM2-targeted therapy.
Collapse
Affiliation(s)
- Ryuichiro Sawada
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Ritsuko Maehara
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Taro Oshikiri
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Tetsu Nakamura
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Tomoo Itoh
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Yuzo Kodama
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Yoshihiro Kakeji
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Yoh Zen
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Institute of Liver Studies, King's College Hospital & King's College London, London SE5 9RS, United Kingdom.
| |
Collapse
|
89
|
Impact of the updated 2018 ASCO/CAP guidelines on HER2 FISH testing in invasive breast cancer: a retrospective study of HER2 fish results of 2233 cases. Breast Cancer Res Treat 2019; 175:51-57. [DOI: 10.1007/s10549-019-05148-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/22/2019] [Indexed: 12/18/2022]
|
90
|
Gibbons-Fideler IS, Nitta H, Murillo A, Tozbikian G, Banks P, Parwani AV, Li Z. Identification of HER2 Immunohistochemistry-Negative, FISH-Amplified Breast Cancers and Their Response to Anti-HER2 Neoadjuvant Chemotherapy. Am J Clin Pathol 2019; 151:176-184. [PMID: 30339245 DOI: 10.1093/ajcp/aqy136] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Objectives Either immunohistochemistry (IHC) or in situ hybridization (ISH) can be used to determine human epidermal growth factor receptor 2 (HER2) status. Breast cancers (BCs) with HER2 IHC-negative (IHC-) and ISH-amplified (ISH+) results have been rarely reported but not well studied. We investigated the frequency of HER2 IHC-/ISH+ BCs and their response to anti-HER2 neoadjuvant chemotherapy (NAC). Methods Seventeen BCs with HER2 IHC-/ISH+ results were identified from 1,107 consecutive invasive BCs (1.5%, 17/1,107). Results Gene protein assay confirmed the original HER2 IHC and ISH results. Increased HER2 RNA level was detected in HER2 IHC-/ISH+ cases compared with HER2 IHC-/ISH- cases. Eight patients had anti-HER2 NAC; three had pathologic complete response, and five had residual tumors. Conclusions A small percentage of patients (1.5%) showed discordant HER2 IHC and ISH results (IHC-/ISH+) and would have lost the opportunity for potentially beneficial anti-HER2-targeted therapy if only HER2 IHC testing had been used.
Collapse
Affiliation(s)
| | | | | | - Gary Tozbikian
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH
| | | | - Anil V Parwani
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH
| | - Zaibo Li
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH
| |
Collapse
|
91
|
Comparison between digital image analysis and visual assessment of immunohistochemical HER2 expression in breast cancer. Pathol Res Pract 2018; 214:2087-2092. [DOI: 10.1016/j.prp.2018.10.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/20/2018] [Accepted: 10/19/2018] [Indexed: 11/20/2022]
|
92
|
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: 576] [Impact Index Per Article: 96.0] [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
|
93
|
Shu S, Iimori M, Nakanishi R, Jogo T, Saeki H, Oki E, Maehara Y. Changes in HER2 Expression and Amplification Status Following Preoperative Chemotherapy for Gastric Cancer. In Vivo 2018; 32:1491-1498. [PMID: 30348707 PMCID: PMC6365720 DOI: 10.21873/invivo.11405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND It is essential to establish a strategy for second-line treatment for human epidermal growth factor receptor 2 (HER2)-positive gastric cancer; however, HER2 expression status after chemotherapy treatment is not routinely determined. MATERIALS AND METHODS We analyzed 25 cases of gastric cancer that received preoperative chemotherapy and selected the six pre-treatment samples that were HER2-positive. Pre- and post-treatment tumor samples were examined for HER2 expression, and for HER2, epidermal growth factor receptor (EGFR), and hepatocyte growth factor receptor (MET) gene amplification. RESULTS Three patients had been treated with trastuzumab plus chemotherapy, and three patients with cytotoxic chemotherapy alone. Only one case that had an initial HER2 score of 3+ and had received trastuzumab plus chemotherapy remained HER2-positive after treatment. Decrease or loss of HER2 expression and amplification was observed in the other five patients. Amplification of EGFR or MET was not observed in any pre- or post-treatment specimens. CONCLUSION Our data suggest that trastuzumab plus chemotherapy or chemotherapy alone may induce loss of HER2 positivity.
Collapse
Affiliation(s)
- Sei Shu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
- Product Research Department, Medical Affairs Division, Chugai Pharmaceutical Co., Ltd., Kamakura, Japan
| | - Makoto Iimori
- Department of Molecular Cancer Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
| | - Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
| | - Tomoko Jogo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
| | - Hiroshi Saeki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Kyushu University, Fukuoka, Japan
- Kyushu Central Hospital of the Mutual Aid Association of Public School Teachers, Fukuoka, Japan
| |
Collapse
|
94
|
Khandelwal S, Boylan M, Spallholz JE, Gollahon L. Cytotoxicity of Selenium Immunoconjugates against Triple Negative Breast Cancer Cells. Int J Mol Sci 2018; 19:E3352. [PMID: 30373175 PMCID: PMC6274915 DOI: 10.3390/ijms19113352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/19/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Within the subtypes of breast cancer, those identified as triple negative for expression of estrogen receptor α (ESR1), progesterone receptor (PR) and human epidermal growth factor 2 (HER2), account for 10⁻20% of breast cancers, yet result in 30% of global breast cancer-associated deaths. Thus, it is critical to develop more targeted and efficacious therapies that also demonstrate less side effects. Selenium, an essential dietary supplement, is incorporated as selenocysteine (Sec) in vivo into human selenoproteins, some of which exist as anti-oxidant enzymes and are of importance to human health. Studies have also shown that selenium compounds hinder cancer cell growth and induce apoptosis in cancer cell culture models. The focus of this study was to investigate whether selenium-antibody conjugates could be effective against triple negative breast cancer cell lines using clinically relevant, antibody therapies targeted for high expressing breast cancers and whether selenium cytotoxicity was attenuated in normal breast epithelial cells. To that end, the humanized monoclonal IgG1 antibodies, Bevacizumab and Trastuzumab were conjugated with redox selenium to form Selenobevacizumab and Selenotrastuzumab and tested against the triple negative breast cancer (TNBC) cell lines MDA-MB-468 and MDA-MB-231 as well as a normal, immortalized, human mammary epithelial cell line, HME50-5E. VEGF and HER2 protein expression were assessed by Western. Although expression levels of HER2 were low or absent in all test cells, our results showed that Selenobevacizumab and Selenotrastuzumab produced superoxide (O2•-) anions in the presence of glutathione (GSH) and this was confirmed by a dihydroethidium (DHE) assay. Interestingly, superoxide was not elevated within HME50-5E cells assessed by DHE. The cytotoxicity of selenite and the selenium immunoconjugates towards triple negative cells compared to HME-50E cells was performed in a time and dose-dependent manner as measured by Trypan Blue exclusion, MTT assay and Annexin V assays. Selenobevacizumab and Selenotrastuzumab were shown to arrest the cancer cell growth but not the HME50-5E cells. These results suggest that selenium-induced toxicity may be effective in treating TNBC cells by exploiting different immunotherapeutic approaches potentially reducing the debilitating side effects associated with current TNBC anticancer drugs. Thus, clinically relevant, targeting antibody therapies may be repurposed for TNBC treatment by attachment of redox selenium.
Collapse
Affiliation(s)
- Soni Khandelwal
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| | - Mallory Boylan
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| | - Julian E Spallholz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| | - Lauren Gollahon
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA.
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
95
|
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
|
96
|
Koopman T, van der Vegt B, Dijkstra M, Bart J, Duiker E, Wisman GBA, de Bock GH, Hollema H. HER2 immunohistochemistry in endometrial and ovarian clear cell carcinoma: discordance between antibodies and with in-situ hybridisation. Histopathology 2018; 73:852-863. [PMID: 29989198 DOI: 10.1111/his.13704] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 07/09/2018] [Indexed: 11/26/2022]
Abstract
AIMS Treatment with anti-HER2 therapy could be beneficial for patients with HER2-positive endometrial and ovarian clear cell carcinoma (CCC). We studied HER2 overexpression by immunohistochemistry (IHC) using three different antibodies, including concordance with amplification by in-situ hybridisation (ISH). METHODS AND RESULTS IHC and ISH were performed on tissue microarrays of 101 tumours: 58 endometrial pure CCC, 19 endometrial mixed carcinomas with a CCC component and 24 ovarian pure CCC. IHC was performed using SP3, 4B5 and HercepTest antibodies, and was scored by two independent observers. ISH was performed using dual-colour silver ISH. Using IHC, agreement was poor between SP3/4B5 (61.4%), poor between SP3/HercepTest (68.3%) and reasonable between 4B5/HercepTest (75.2%). Interobserver agreement was substantial to almost perfect for all antibodies (SP3: linear weighted κ = 0.89, 4B5: κ = 0.90, HercepTest: κ = 0.76). HER2-positivity by ISH was 17.8% (endometrial pure CCC: 24.1%, endometrial mixed: 0%, ovarian pure CCC: 16.7%). IHC/ISH concordance was poor, with a high false-negative rate of all three IHC antibodies: sensitivity (38.9-50.0%) and positive predictive value (PPV) (37.5-58.3%) were poor; specificity (81.9-94.0%) and negative predictive value (NPV) (87.1-88.3%) were reasonable. When excluding 2+ cases, sensitivity declined (26.7-43.8%) but PPV (80.0-87.5%) and specificity (98.6-98.7%) improved. CONCLUSIONS In ovarian and endometrial CCC, there is considerable difference in HER2 overexpression by different IHC antibodies and marked discordance with ISH. As such, no single antibody can be considered conclusive for determining HER2 status in CCC. Based on these results, the lack of predictive value of different HER2 testing methods, as used in other studies, could be explained.
Collapse
Affiliation(s)
- Timco Koopman
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marcel Dijkstra
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Joost Bart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Evelien Duiker
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - G Bea A Wisman
- Department of Gynaecologic Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Harry Hollema
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
97
|
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: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
98
|
Reappraisal of HER2 status in the spectrum of advanced urothelial carcinoma: a need of guidelines for treatment eligibility. Mod Pathol 2018; 31:1270-1281. [PMID: 29467478 DOI: 10.1038/s41379-018-0023-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 01/05/2023]
Abstract
Although human epidermal growth factor receptor 2 (HER2) may represent a therapeutic target, its evaluation in urothelial carcinoma of the bladder does not rely on a standardized scoring system by immunohistochemistry or fluorescent in situ hybridization (FISH), as reflected by various methodology in the literature and clinical trials. Our aim was to improve and standardize HER2 amplification detection in bladder cancer. We assessed immunohistochemical criteria derived from 2013 American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAPs) guidelines for breast cancer and investigated intratumoral heterogeneity in a retrospective multicentric cohort of 188 patients with locally advanced urothelial carcinoma of the bladder. Immunohistochemistry was performed on 178 primary tumors and 126 lymph node metastases, eligible cases (moderate/strong, complete/incomplete membrane staining) were assessed by FISH. HER2 overexpression was more frequent with 2013 ASCO/CAP than 2007 ASCO/CAP guidelines (p < 0.0001). The rate of positive HER2 FISH was similar between primary tumor and lymph node metastases (8%). Among positive FISH cases, 48% were associated with moderate/strong incomplete membrane staining that were not scored eligible for FISH by 2007 ASCO/CAP criteria. Among 3+ immunohistochemistry score cases, 67% were associated with HER2-positive FISH. Concordance between primary tumors and matched lymph node metastases was moderate for immunohistochemistry (κ = 0.54 (CI 95%, 0.41-0.67)) and FISH (κ = 0.50 (CI 95%, 0.20-0.79)). HER2-positive FISH was more frequent in micropapillary carcinomas (12%) and carcinoma with squamous differentiation (11%) than in pure conventional carcinoma (6%). Intratumoral heterogeneity for HER2 immunohistochemistry was observed in 7% primary tumor and 6% lymph node metastases; 24% positive HER2 FISH presented intratumoral heterogeneity. Our study suggests that HER2 evaluation should include an immunohistochemistry screening step with eligibility for FISH including incomplete/complete and moderate/strong membrane staining. Spatial or temporal intratumoral heterogeneity prompts to perform evaluation on both tumor and lymph node, and for each histological variant observed.
Collapse
|
99
|
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: 1353] [Impact Index Per Article: 225.5] [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
|
100
|
Colomer R, Aranda-López I, Albanell J, García-Caballero T, Ciruelos E, López-García MÁ, Cortés J, Rojo F, Martín M, Palacios-Calvo J. Biomarkers in breast cancer: A consensus statement by the Spanish Society of Medical Oncology and the Spanish Society of Pathology. Clin Transl Oncol 2018; 20:815-826. [PMID: 29273958 PMCID: PMC5996012 DOI: 10.1007/s12094-017-1800-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 11/05/2022]
Abstract
This consensus statement revises and updates the recommendations for biomarkers use in the diagnosis and treatment of breast cancer, and is a joint initiative of the Spanish Society of Medical Oncology and the Spanish Society of Pathology. This expert group recommends determining in all cases of breast cancer the histologic grade and the alpha-estrogen receptor (ER), progesterone receptor, Ki-67 and HER2 status, in order to assist prognosis and establish therapeutic options, including hormone therapy, chemotherapy and anti-HER2 therapy. One of the four available genetic prognostic platforms (MammaPrint®, Oncotype DX®, Prosigna® or EndoPredict®) may be used in node-negative ER-positive patients to establish a prognostic category and decide with the patient whether adjuvant treatment may be limited to hormonal therapy. Newer technologies including next-generation sequencing, liquid biopsy, tumour-infiltrating lymphocytes or PD-1 determination are at this point investigational.
Collapse
Affiliation(s)
- R Colomer
- Departamento de Oncología Médica, Hospital Universitario La Princesa, C/Diego de León, 62, 28006, Madrid, Spain.
| | - I Aranda-López
- Pathology Department, General University Hospital of Alicante, Alicante, Spain
| | - J Albanell
- Medical Oncology Department, Mar University Hospital, Hospital del Mar Medical Research Institute (IMIM), Pompeu Fabra University, CIBERONC, Barcelona, Spain
| | - T García-Caballero
- Pathology Department, University Hospital Complex of Santiago, Santiago de Compostela, Spain
| | - E Ciruelos
- Medical Oncology Department, Doce de Octubre University Hospital, Madrid, Spain
| | - M Á López-García
- Pathology Department, Virgen del Rocio University Hospital, CIBERONC, Seville, Spain
| | - J Cortés
- Medical Oncology Department, Ramón y Cajal University Hospital, Madrid, Spain
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Baselga Institute of Oncology (IOB), Madrid, Barcelona, Spain
| | - F Rojo
- Pathology Department, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - M Martín
- Medical Oncology Department, Gregorio Marañón University Hospital, CIBERONC, GEICAM, Madrid, Spain
| | - J Palacios-Calvo
- Pathology Department, Ramón y Cajal University Hospital, CIBERONC, IRYCIS and University of Alcalá, Madrid, Spain.
| |
Collapse
|