1
|
Gamrani S, Akhouayri L, Boukansa S, Karkouri M, El Fatemi H. The Clinicopathological Features and Prognostic Significance of HER2-Low in Early Breast Tumors Patients Prognostic Comparison of HER-Low and HER2-Negative Breast Cancer Stratified by Hormone Receptor Status. Breast J 2023; 2023:6621409. [PMID: 38075551 PMCID: PMC10703525 DOI: 10.1155/2023/6621409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023]
Abstract
Introduction There has been increased interest in HER2-low breast tumors recently, as these tumors may have distinct clinical and molecular characteristics compared to HER2-negative and HER2-positive tumors. A new nomenclature has been proposed for HER2 1+ and HER2 2+ tumors that are confirmed negative according to fluorescence in situ hybridization (FISH). These tumors are now referred to as HER2-low, and it is thought that they may represent a distinct subtype of breast cancer that warrants further investigation. In this study, we aimed to evaluate the clinicopathological characteristics and prognostic impact of this particular subtype in a North-African context where HER2-low breast cancer is a relatively understudied subtype, particularly in non-Western populations. Methods We conducted a retrospective cohort study on 1955 breast tumors in Moroccan patients over 10 years, collected at the Pathology Department of Ibn Rochd University Hospital in Casablanca and at the pathology department of Hassan II University Hospital in Fes. We elaborated on their complete immunohistochemical profile based on the main breast cancer biomarkers: Ki-67, HER2, estrogen, and progesterone receptors. Their overall survival and disease free survival data were also retrieved from their respective records. Results Out of 1955 BC patients, 49.3% were classified as HER2-low; of which 80.7% and 19.2% were hormone receptors positive and negative, respectively. The clinicopathologic features indicate that HER2-low subtype tumors behave much more like HER2-positive than HER2-negative tumors. The survival analysis showed that the HER2-low subtype-belonging patients present significantly the poorest prognosis in disease-free survival (p = 0.003) in comparison with HER2-negative ones. When considering the hormonal status, hormonal-dependent tumors show a significant difference according to HER2 subtypes in disease-free survival (p < 0.001). Yet no significant difference was shown among hormonal negative tumors. Moreover, patients with hormonal positive tumors and simultaneously belonging to the HER2-low subgroup present a significantly good prognosis in overall survival compared to the ones with hormonal negative tumors (p = 0.008). Conclusion Our study has shown that the HER2-low phenotype is common among hormone-positive patients. The clinicopathological features and prognostic data indicate that the hormonal receptors effect and HER2 heterogeneity are crucial factors to consider. It is important to note that this particular subgroup is different from the HER2-negative one and should not be treated in the same way. Therefore, this study offers a new perspective in the management of HER2-low patients and can serve as a basis for future prospective analyses.
Collapse
Affiliation(s)
- Sanaa Gamrani
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Laila Akhouayri
- Dipartimento di Scienze Cliniche e Biologiche, Università Degli Studi Di Torino, Via Giuseppe Verdi, Torino, Italy
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Rue Tariq Ibn Ziad, Casablanca, Morocco
| | - Sara Boukansa
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mehdi Karkouri
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Rue Tariq Ibn Ziad, Casablanca, Morocco
- Department of Pathology, Ibn Rochd University Hospital, Rue des Hôpitaux, Casablanca, Morocco
| | - Hinde El Fatemi
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
2
|
Wu Y, Zhong R, Ma F. HER2-low breast cancer: Novel detections and treatment advances. Crit Rev Oncol Hematol 2023; 181:103883. [PMID: 36427769 DOI: 10.1016/j.critrevonc.2022.103883] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/20/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Breast cancer (BC), which has the highest cancer incidence in women, seriously threatens women's health. Since human epidermal growth factor receptor-2 (HER2) characterization, breast cancer treatment has entered an era of individualized targeted therapy. With the emergence of anti-HER2 targeting agents, monoclonal antibodies (mAbs) and tyrosine kinase inhibitors have considerably improved the prognosis of HER2-positive BC. However, HER2-low BC, accounting for 45-55% of BC patients, is less likely to benefit from conventional HER2-targeting mAbs. The growing success of the new generation of drugs, especially promising HER2-directed antibody-drug conjugates, has changed the treatment landscape for patients with HER2-low BC, leading to a research boom. HER-2-low BC is a heterogeneous entity, and there many areas remain to be explored. In this article, we review the literature on HER2-low BC, mainly focusing on its detection assays, clinicopathological profiles and treatment landscape, and hopefully provide insight into future perspectives.
Collapse
Affiliation(s)
- Yun Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruiqi Zhong
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Morsberger L, Pallavajjala A, Long P, Hardy M, Park R, Parish R, Nozari A, Zou YS. HER2 amplification by next-generation sequencing to identify HER2-positive invasive breast cancer with negative HER2 immunohistochemistry. Cancer Cell Int 2022; 22:350. [PMCID: PMC9664724 DOI: 10.1186/s12935-022-02761-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Human epidermal growth factor receptor 2 (HER2) positive breast carcinomas due to HER2 amplification are associated with aggressive behavior and a poor prognosis. Anti-HER2-targeted therapies are widely used to treat HER2-positive breast carcinomas with excellent outcomes. Accurate identification of HER2 amplification status in breast carcinomas is of important diagnostic and treatment value. Currently, HER2 amplification status is routinely determined by immunohistochemistry (IHC) and/or fluorescence in situ hybridization (FISH) testing. This study will review our past HER2 data to determine and characterize discordant results between HER2 IHC and FISH. It will also determine a potential impact of HER2 amplification status by next-generation sequencing (NGS) on these patients.
Methods
We reviewed a total of 4884 breast carcinomas with coexisting HER2 IHC and HER2 FISH performed at our institution from 2010 to 2022. 57 cases also had a Next-Generation-Sequencing-based (NGS) gene panel performed. Given the advances in biostatic analysis pipelines, NGS methods were utilized to provide results on HER2 amplification status along with somatic mutations.
Results
While the majority (ranging from 98.5% with IHC score of 0 and 93.1% with IHC score of 1 +) of 4884 breast carcinomas had concordant results from HER2 IHC and HER2 FISH testing, a small percentage of patients (ranging from 1.5% in those with IHC score of 0, to 6.9% with IHC score of 1 +) had discordant results, with negative HER2 IHC and positive HER2 FISH results. These patients could be reported as HER2-negative breast carcinomas if only HER2 IHC testing has been performed according to a current cost-effective HER2 test strategy. 57 patients had HER2 amplification status determined by NGS, and all patients had concordant results between HER2 NGS and FISH tests. A HER2-amplified breast carcinoma by NGS had a negative IHC and a positive HER2 FISH result. This case was classified as a HER2-positive breast carcinoma, had anti-HER2-targeted therapy, and achieved a complete clinical response.
Conclusions
A small percentage of HER2-positive breast carcinomas are unidentified because of a negative HER2 IHC based on our current cost-effective HER2 test strategy. It is not feasible and affordable in routine clinical practice to perform HER2 FISH for the cases with negative HER2 IHC (IHC score 0 and 1 +). Therefore, NGS assays capable of simultaneously detecting both somatic mutations and HER2 amplification could provide a more comprehensive genetic profiling for breast carcinomas in a clinical setting. Identification of HER2 amplification by NGS in HER2-positive breast carcinomas with negative HER2 IHC results is important since these cases are concealed by our current cost-effective HER2 test strategy with IHC first (for all cases) and FISH reflex (only for cases with IHC score of 2 +), and would offer the opportunity for potentially beneficial anti-HER2-targeted therapies for these patients.
Collapse
|
4
|
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
|
5
|
Smith LR, Kok HJ, Zhang B, Chung D, Spradlin RA, Rakoczy KD, Lei H, Boesze-Battaglia K, Barton ER. Matrix Metalloproteinase 13 from Satellite Cells is Required for Efficient Muscle Growth and Regeneration. Cell Physiol Biochem 2020; 54:333-353. [PMID: 32275813 DOI: 10.33594/000000223] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIMS Cell migration and extracellular matrix remodeling underlie normal mammalian development and growth as well as pathologic tumor invasion. Skeletal muscle is no exception, where satellite cell migration replenishes nuclear content in damaged tissue and extracellular matrix reforms during regeneration. A key set of enzymes that regulate these processes are matrix metalloproteinases (MMP)s. The collagenase MMP-13 is transiently upregulated during muscle regeneration, but its contribution to damage resolution is unknown. The purpose of this work was to examine the importance of MMP-13 in muscle regeneration and growth in vivo and to delineate a satellite cell specific role for this collagenase. METHODS Mice with total and satellite cell specific Mmp13 deletion were utilized to determine the importance of MMP-13 for postnatal growth, regeneration after acute injury, and in chronic injury from a genetic cross with dystrophic (mdx) mice. We also evaluated insulin-like growth factor 1 (IGF-1) mediated hypertrophy in the presence and absence of MMP-13. We employed live-cell imaging and 3D migration measurements on primary myoblasts obtained from these animals. Outcome measures included muscle morphology and function. RESULTS Under basal conditions, Mmp13-/- mice did not exhibit histological or functional deficits in muscle. However, following acute injury, regeneration was impaired at 11 and 14 days post injury. Muscle hypertrophy caused by increased IGF-1 was blunted with minimal satellite cell incorporation in the absence of MMP-13. Mmp13-/- primary myoblasts displayed reduced migratory capacity in 2D and 3D, while maintaining normal proliferation and differentiation. Satellite cell specific deletion of MMP-13 recapitulated the effects of global MMP-13 ablation on muscle regeneration, growth and myoblast movement. CONCLUSION These results show that satellite cells provide an essential autocrine source of MMP-13, which not only regulates their migration, but also supports postnatal growth and resolution of acute damage.
Collapse
Affiliation(s)
- Lucas R Smith
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Neurobiology, Physiology & Behavior, Physical Medicine & Rehabilitation, University of California, Davis, CA, USA
| | - Hui Jean Kok
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Boshi Zhang
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Du Chung
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ray A Spradlin
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Kyla D Rakoczy
- Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Hanqin Lei
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | | | - Elisabeth R Barton
- Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA, .,Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Xu Z, Xu P, Fan W, Huang B, Cheng Q, Zhang Z, Wang P, Yu M. The effect of an alternative chromosome 17 probe on fluorescence in situ hybridization for the assessment of HER2 amplification in invasive breast cancer. Exp Ther Med 2019; 18:2095-2103. [PMID: 31410164 PMCID: PMC6676089 DOI: 10.3892/etm.2019.7756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 05/09/2019] [Indexed: 12/16/2022] Open
Abstract
Fluorescent in situ hybridization (FISH) is commonly used to determine the ratio of human epidermal growth factor receptor 2 (HER2) to centromere enumeration probe for chromosome 17 (CEP17), which further determines HER2 gene status in breast cancer. However, due to copy number alteration in CEP17, inaccurate diagnoses can occur. The current study was performed to investigate the diagnostic value of an alternative CEP17 reference probe for HER2 status in invasive breast cancer. A higher-order repeat in the centromeric region of chromosome 17 was identified and an alternative probe (SCEP17) was subsequently prepared. Karyotype analysis of peripheral blood was used to detect SCEP17 probe specificity. Using a HER2/CEP17 probe, karyotype analysis revealed two strong green signals at the centromere of chromosome 17 and one weaker signal at the other centromere. However, two strong hybridization signals at the centromere of chromosome 17 were observed when the HER2/SCEP17 probe was used. In the 425 patients with invasive breast cancer, no statistical difference was observed between HER2/SCEP17 and HER2/CEP17 when detecting HER2 gene amplification (P=0.157). However, in terms of copy number, the SCEP17 probe exhibited a reduced number compared with the conventional CEP17 probe (P<0.001). In conclusion, the HER2/SCEP17 probe may lead to increased accuracy HER2 status assessment in invasive breast cancer. However, a further large-scale and prospective clinical trial is required for confirmation of the potential benefits of using the HER2/SCEP17 probe.
Collapse
Affiliation(s)
- Zhigao Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Peipei Xu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China.,Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450072, P.R. China
| | - Wei Fan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ben Huang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qingyuan Cheng
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zheng Zhang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ping Wang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Mingxia Yu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
9
|
Reference Size Matching, Whole-Genome Amplification, and Fluorescent Labeling as a Method for Chromosomal Microarray Analysis of Clinically Actionable Copy Number Alterations in Formalin-Fixed, Paraffin-Embedded Tumor Tissue. J Mol Diagn 2018; 20:279-288. [DOI: 10.1016/j.jmoldx.2018.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 01/01/2018] [Accepted: 01/09/2018] [Indexed: 12/14/2022] Open
|
10
|
Gray PN, Vuong H, Tsai P, Lu HM, Mu W, Hsuan V, Hoo J, Shah S, Uyeda L, Fox S, Patel H, Janicek M, Brown S, Dobrea L, Wagman L, Plimack E, Mehra R, Golemis EA, Bilusic M, Zibelman M, Elliott A. TumorNext: A comprehensive tumor profiling assay that incorporates high resolution copy number analysis and germline status to improve testing accuracy. Oncotarget 2018; 7:68206-68228. [PMID: 27626691 PMCID: PMC5356550 DOI: 10.18632/oncotarget.11910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023] Open
Abstract
The development of targeted therapies for both germline and somatic DNA mutations has increased the need for molecular profiling assays to determine the mutational status of specific genes. Moreover, the potential of off-label prescription of targeted therapies favors classifying tumors based on DNA alterations rather than traditional tissue pathology. Here we describe the analytical validation of a custom probe-based NGS tumor panel, TumorNext, which can detect single nucleotide variants, small insertions and deletions in 142 genes that are frequently mutated in somatic and/or germline cancers. TumorNext also detects gene fusions and structural variants, such as tandem duplications and inversions, in 15 frequently disrupted oncogenes and tumor suppressors. The assay uses a matched control and custom bioinformatics pipeline to differentiate between somatic and germline mutations, allowing precise variant classification. We tested 170 previously characterized samples, of which > 95% were formalin-fixed paraffin embedded tissue from 8 different cancer types, and highlight examples where lack of germline status may have led to the inappropriate prescription of therapy. We also describe the validation of the Affymetrix OncoScan platform, an array technology for high resolution copy number variant detection for use in parallel with the NGS panel that can detect single copy amplifications and hemizygous deletions. We analyzed 80 previously characterized formalin-fixed paraffin-embedded specimens and provide examples of hemizygous deletion detection in samples with known pathogenic germline mutations. Thus, the TumorNext combined approach of NGS and OncoScan potentially allows for the identification of the “second hit” in hereditary cancer patients.
Collapse
Affiliation(s)
| | - Huy Vuong
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | - Pei Tsai
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | | | - Wenbo Mu
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | | | - Jayne Hoo
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | - Swati Shah
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | - Lisa Uyeda
- Ambry Genetics, Aliso Viejo, CA, 92656, USA
| | | | | | | | | | | | | | | | - Ranee Mehra
- Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | | | | | | | | |
Collapse
|
11
|
Christgen M, van Luttikhuizen JL, Raap M, Braubach P, Schmidt L, Jonigk D, Feuerhake F, Lehmann U, Schlegelberger B, Kreipe HH, Steinemann D. Precise ERBB2 copy number assessment in breast cancer by means of molecular inversion probe array analysis. Oncotarget 2018; 7:82733-82740. [PMID: 27716627 PMCID: PMC5347728 DOI: 10.18632/oncotarget.12421] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 09/19/2016] [Indexed: 01/01/2023] Open
Abstract
HER2/ERBB2 amplification/overexpression determines the eligibility of breast cancer patients to HER2-targeted therapy. This study evaluates the agreement between ERBB2 copy number assessment by fluorescence in situ hybridization, a standard method recommended by the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP), and newly available DNA extraction-based methods. A series of n=29 formalin-fixed paraffin-embedded breast cancers were subjected to ERBB2 copy number assessment by fluorescence in situ hybridization (FISH, Vysis, Abbott). Following macrodissection of invasive breast cancer tissue and DNA extraction, ERBB2 copy number was also determined by molecular inversion probe array analysis (MIP, OncoScan, Affymetrix) and next generation sequencing combined with normalized amplicon coverage analysis (NGS/NAC, AmpliSeq, Ion Torrent). ERBB2 copy number values obtained by MIP or NGS/NAC were tightly correlated with ERBB2 copy number values obtained by conventional FISH (rs = 0.940 and rs = 0.894, P < 0.001). Using ASCO/CAP guideline-conform thresholds for categorization of breast cancers as HER2-negative, equivocal or positive, nearly perfect concordance was observed for HER2 classification by FISH and MIP (93% concordant classifications, κ = 0.87). Substantial concordance was observed for FISH and NGS/NAC (83% concordant classifications, κ = 0.62). In conclusion, MIP facilitates precise ERBB2 copy number detection and should be considered as an ancillary method for clinical HER2 testing.
Collapse
Affiliation(s)
| | | | - Mieke Raap
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Peter Braubach
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Lars Schmidt
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | | - Ulrich Lehmann
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | | - Hans H Kreipe
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Doris Steinemann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Hu X, Li Y, Yuan D, Li R, Kong L, Li H, Yang Z, Yu Q. Retrospective analysis of the association between human epidermal growth factor receptor 2 amplification and chromosome enumeration probe 17 status in patients with breast cancer. Oncol Lett 2017; 14:5265-5270. [PMID: 29113162 PMCID: PMC5656029 DOI: 10.3892/ol.2017.6897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/11/2017] [Indexed: 01/28/2023] Open
Abstract
The aim of the present study was to identify potential human epidermal growth factor receptor 2 (HER2) amplification, according to American Society of Clinical Oncology and the College of American Pathologists (ASCO/CAP) 2013 HER2 testing guidelines, in patients previously determined not to possess HER2 amplification, in accordance with previous 2007 guidelines. Potential discrepancies may arise from chromosome enumeration probe 17 (CEP17) amplification, deletion, polysomyor monosomy. HER2, CEP17, tumor protein p53 (TP53) and retinoic acid receptor α (RARA) genes from 67 patient specimens with suspected amplification, polysomy or monosomy of CEP17 were analyzed using fluorescence in situ hybridization. HER2 status was interpreted using 2007 and 2013 ASCO HER2 test guidelines as well as the reference genes TP53 and RARA. According to ASCO/CAP2007 HER2 guidelines, 20 patients exhibited HER2 amplification (29.85%), 41 were without HER2 amplification (including 25 with polysomy, 15 with monosomy and 1 with suspected monosomy plus co-amplification of HER2 and CEP17) and the remaining 6 patients were equivocal. Using ASCO/CAP 2013 HER2 guidelines, 49 patients exhibited HER2 gene amplification (73.1%). The 29-patient increase included 6 originally at equivocal levels but now demonstrating amplification, 22 originally with polysomy but now revealing co-amplification, and 1 with suspected monosomy plus co-amplification of HER2 and CEP17. According to TP53 and RARA, HER2 was amplified in 43 patients (64.1%). Using the revised guidelines, HER2, originally identified as amplified in 6 patients, was not amplified following the introduction of TP53 and RARA control genes. Among these 6, 4 possessed normal TP53 and RARA. The incidence of co-amplification of HER2 and CEP17 was 1.4% (21/1,518). RARA and TP53 are suitable control genes to evaluate HER2 status.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Molecular Medical Laboratory, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Yanan Li
- Molecular Medical Laboratory, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Dong Yuan
- Molecular Medical Laboratory, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Ruohan Li
- Molecular Medical Laboratory, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Lingquan Kong
- Endocrine Breast Surgery, The First Affiliated Hospital, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Hongyuan Li
- Endocrine Breast Surgery, The First Affiliated Hospital, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Zhu Yang
- Molecular Medical Laboratory, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Qiubo Yu
- Molecular Medical Laboratory, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| |
Collapse
|
13
|
Lee K, Jang MH, Chung YR, Lee Y, Kang E, Kim SW, Kim YJ, Kim JH, Kim IA, Park SY. Prognostic significance of centromere 17 copy number gain in breast cancer depends on breast cancer subtype. Hum Pathol 2016; 61:111-120. [PMID: 27989787 DOI: 10.1016/j.humpath.2016.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/25/2016] [Accepted: 12/01/2016] [Indexed: 02/07/2023]
Abstract
Increased copy number of chromosome enumeration probe (CEP) targeting centromere 17 is frequently encountered during HER2 in situ hybridization (ISH) in breast cancer. The aim of this study was to clarify the clinicopathologic significance of CEP17 copy number gain in a relatively large series of breast cancer patients. We analyzed 945 cases of invasive breast cancers whose HER2 fluorescence ISH reports were available from 2004 to 2011 at a single institution and evaluated the association of CEP17 copy number gain with clinicopathologic features of tumors and patient survival. We detected 186 (19.7%) cases of CEP17 copy number gain (CEP17≥3.0) among 945 invasive breast cancers. In survival analysis, CEP17 copy number gain was not associated with disease-free survival of the patients in the whole group. Nonetheless, it was found to be an independent adverse prognostic factor in the HER2-negative group but not in the HER2-positive group. In further subgroup analyses, CEP17 copy number gain was revealed as an independent poor prognostic factor in HER2-negative and hormone receptor-positive breast cancers, and it was associated with aggressive histologic variables including high T stage, high histologic grade, lymphovascular invasion, p53 overexpression, and high Ki-67 proliferative index. In conclusion, we found that elevated CEP17 count can serve as a prognostic marker in luminal/HER2-negative subtype of invasive breast cancer. We advocate the use of the dual-colored fluorescence ISH using CEP17 rather than the single-colored one because it gives additional valuable information on CEP17 copy number alterations.
Collapse
Affiliation(s)
- Kyuongyul Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Pathology, Kangwon National University Hospital, Chuncheon, Kangwon 24289, Republic of Korea
| | - Min Hye Jang
- Department of Pathology, Yeungnam University Medical Center, Daegu 42415, Republic of Korea
| | - Yul Ri Chung
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yangkyu Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Eunyoung Kang
- Breast Care Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Sung-Won Kim
- Breast Care Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Yu Jung Kim
- Breast Care Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Jee Hyun Kim
- Breast Care Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea
| | - In Ah Kim
- Breast Care Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea; Breast Care Center, Seoul National University Bundang Hospital, Seongnam, Gyeonggi 13620, Republic of Korea.
| |
Collapse
|
14
|
Sneige N, Hess KR, Multani AS, Gong Y, Ibrahim NK. Prognostic significance of equivocal human epidermal growth factor receptor 2 results and clinical utility of alternative chromosome 17 genes in patients with invasive breast cancer: A cohort study. Cancer 2016; 123:1115-1123. [PMID: 27893937 DOI: 10.1002/cncr.30460] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 09/30/2016] [Accepted: 10/31/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND The 2013 testing guidelines for determining the human epidermal growth factor receptor 2 (HER2) status include new cutoff points for the HER2/chromosome enumeration probe 17 (CEP17) ratio and the average HER2 copy number per cell, and they recommend using a reflex test with alternative chromosome 17 probes (Ch17Ps) to resolve equivocal HER2 results. This study sought to determine the clinical utility of alternative Ch17Ps in equivocal cases and the effects of equivocal results and/or a change in the HER2 status on patients' outcomes. METHODS The University of Texas MD Anderson Cancer Center database of HER2 dual-probe fluorescence in situ hybridization results from 2000 to 2010 was searched for cases of invasive breast cancer with HER2/CEP17 ratios < 2 and average HER2 copy numbers < 6 per cell. Cases with HER2 copy numbers of 4 to < 6 (the definition of equivocal HER2 results) were analyzed with alternative Ch17Ps for Smith-Magenis syndrome and retinoic acid receptor α genes. Disease-free survival (DFS) and overall survival (OS) were evaluated with respect to the HER2 copy number with multivariate Cox proportional hazards regression. RESULTS Among the 3630 patients meeting the inclusion criteria, 137 (4%) had equivocal HER2 results. With alternative Ch17Ps, 35 of 57 equivocal HER2 cases (61%) were upgraded to a positive HER2 status, and 22 cases (39%) remained unchanged. The 5-year DFS and OS adjusted hazard ratios (HRs) for copy numbers of 4 to < 6 versus < 4 were 0.6 (95% confidence interval [CI], 0.3-1.2) and 0.5 (95% CI, 0.2-1.0) with P values of .16 and .66, respectively. In comparison with HER2-negative cases, these CIs indicated that equivocal HER2 results were associated with either a protective effect (HR, < 0.5) or no effect (HR, 1.0). CONCLUSIONS These findings rule out a significant deleterious effect of equivocal HER2 results. Alternative Ch17Ps may erroneously upgrade the HER2 status; therefore, they cannot be considered reliable in clinical practice. Cancer 2017;123:1115-1123. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Nour Sneige
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kenneth R Hess
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Asha S Multani
- Department of Genetics, The University of Texas MD Anderson Cancer, Houston, Texas
| | - Yun Gong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nuhad K Ibrahim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Willmore-Payne C, Damjanovich-Colmenares K, Pasi AV, Werner TL, Gulbahce HE, Downs-Kelly E, Geiersbach KB. Inconsistent Results With Different Secondary Reflex Assays for Resolving HER2 Status. Am J Clin Pathol 2016; 146:618-626. [PMID: 27773872 DOI: 10.1093/ajcp/aqw177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Guidelines suggest that secondary reflex testing may be useful for resolving HER2 status in breast cancers with equivocal results by both immunohistochemistry (IHC) and in situ hybridization (ISH). We compared two reflex ISH assays and a polymerase chain reaction (PCR) assay for this application. METHODS Twenty-nine breast cancers with equivocal IHC and ISH results were retested two ways: (1) ISH using differentially labeled probes targeting ERBB2 ( HER2 , 17q12) and either RAI1 (17p11.2) or ORC4 (2q22.3-2q23.1) in two separate assays and (2) real-time quantitative PCR amplification of ERBB2 and a control locus ( EIF5B , 2q11.2). RESULTS Results of the HER2 / RAI1 and HER2 / ORC4 ISH assays were concordant for 21 (72%) cases, and results of all three secondary reflex assays were concordant for only 18 (62%) cases. Result discrepancies between the two ISH readers were observed for cases close to the cutoff threshold. CONCLUSIONS Use of different control loci for ISH is associated with discordant results, and PCR is more likely to classify cases as nonamplified, possibly due to contamination with nontumor cells. While resolution of HER2 -equivocal results is desirable from a clinical perspective, different secondary reflex assays yield different results, and the correlation of these results with clinical outcomes is unknown.
Collapse
Affiliation(s)
| | | | - Alexandra V Pasi
- ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, UT
| | | | | | | | | |
Collapse
|
16
|
Quantum dot nanoparticle for optimization of breast cancer diagnostics and therapy in a clinical setting. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1581-92. [DOI: 10.1016/j.nano.2016.02.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 01/30/2023]
|
17
|
Gogas H, Kotoula V, Alexopoulou Z, Christodoulou C, Kostopoulos I, Bobos M, Raptou G, Charalambous E, Tsolaki E, Xanthakis I, Pentheroudakis G, Koutras A, Bafaloukos D, Papakostas P, Aravantinos G, Psyrri A, Petraki K, Kalogeras KT, Pectasides D, Fountzilas G. MYC copy gain, chromosomal instability and PI3K activation as potential markers of unfavourable outcome in trastuzumab-treated patients with metastatic breast cancer. J Transl Med 2016; 14:136. [PMID: 27184134 PMCID: PMC4869295 DOI: 10.1186/s12967-016-0883-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 04/28/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND There is an unmet need for more efficient patient stratification for receiving trastuzumab in the metastatic breast cancer (mBC) setting, since only part of such patients benefit from the addition of this agent to chemotherapy. The aim of this study was to investigate the prognostic value of biomarkers including MYC and MET in mBC patients treated with trastuzumab-based regimens. METHODS mBC patients, locally tested as HER2-positive, treated with trastuzumab and chemotherapy between 1998 and 2010 were evaluated. Paraffin tumors (n = 229) were retrospectively centrally assessed by immunohistochemistry (IHC) for HER2, ER, PgR and Ki67; fluorescence in situ hybridization (FISH) for HER2, TOP2A and centromere (CEN) 17, MYC and CEN8, MET and CEN7; qPCR for MYC, MET copy number (CN); and, for PI3K activation (PIK3CA mutations; PTEN and phospho-mTOR protein expression). Increased CEN CN was assessed based on normal cut-offs. Time to progression (TTP) and survival were evaluated from the initiation of trastuzumab as first line treatment. RESULTS Among all tumors, 90 were HER2-negative upon central testing (ambiguous HER2) and the rest were true HER2-positive. Further, 156 patients presented with mBC upon relapse of pre-treated disease (R-mBC) and 65 were diagnosed at stage IV (de novo mBC). Concordance between FISH and qPCR on gene CN status was fair for MYC (Kappa = 0.458) and absent for MET. The presence of MYC CN gain with qPCR and the absence of PI3K activation were infrequent events (7 and 8 % of evaluable tumors, respectively), while 41 % of tumors had increased CEN CN in one or more chromosomes, indicative of chromosomal instability. The most consistent finding in the entire cohort and in the above patient subgroups with respect to outcome was the unfavourable effect of MYC CN gain, which was retained upon multivariable analysis (e.g., survival in the entire cohort, HR 6.02; 95 % CI 2.67-13.6; p < 0.001). Further unfavourable prognosticators were increased CEN CN in one chromosome in R-mBC but not in de novo mBC (multivariable interaction p = 0.048), PI3K activation in R-mBC (multivariable p = 0.004) and increased Ki67 for patient TTP. CONCLUSIONS MYC gene copies, centromere status and PI3K activation may adversely impact trastuzumab treated mBC patient outcome and seem worthy validating in larger series.
Collapse
Affiliation(s)
- Helen Gogas
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Vassiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece.,Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Zoi Alexopoulou
- Department of Biostatistics, Health Data Specialists Ltd, Athens, Greece
| | | | - Ioannis Kostopoulos
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Mattheos Bobos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Georgia Raptou
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Elpida Charalambous
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Eleftheria Tsolaki
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - Ioannis Xanthakis
- Department of Medical Oncology, Papageorgiou Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | | | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | | | | | - Gerasimos Aravantinos
- Second Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| | - Amanda Psyrri
- Division of Oncology, Second Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | | | - Konstantine T Kalogeras
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece.,Translational Research Section, Hellenic Cooperative Oncology Group, Data Office, Athens, Greece
| | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece.,Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
18
|
Koudelakova V, Trojanec R, Vrbkova J, Donevska S, Bouchalova K, Kolar Z, Varanasi L, Hajduch M. Frequency of chromosome 17 polysomy in relation to CEP17 copy number in a large breast cancer cohort. Genes Chromosomes Cancer 2016; 55:409-17. [PMID: 26847577 DOI: 10.1002/gcc.22337] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/30/2022] Open
Abstract
Eligibility to anti-HER2 therapy for breast tumors strictly depends on demonstrating HER2 overexpression (by immunohistochemistry) or HER2 gene amplification by in situ hybridization (ISH), usually defined by the ratio of HER2 gene to chromosome 17 centromere (CEP17) copies. However, the CEP17 copy number increase (CNI) has been proven responsible for misleading HER2 FISH results and recent small cohort studies suggest that chromosome 17 polysomy is actually very rare. Here we investigated by FISH the frequency of true chromosome 17 polysomy in a consecutive cohort of 5,477 invasive breast cancer patients. We evaluated and selected the LSI 17p11.2 probe for chromosome 17 enumeration on a training cohort of 67 breast cancer samples (CEP17 ≥ 2.5). LSI 17p11.2 was used in the 297/5,477 patients from the validation cohort displaying CEP17 CNI (CEP17 ≥ 3.0). Using HER2/17p11.2 scoring criteria, 37.3%/1.5% patients initially classified as equivocal/non-amplified were reclassified as amplified. For a more accurate assessment of chromosome 17 and ploidy in the samples, we tested six markers located on chromosome 17 and centromeric regions of chromosome 8 (CEP8) and 11 (CEP11) in 67 patients with CEP17 and LSI 17p11.2 CNI. True polysomy (hyperdiploidy) according to these markers was found in 0.48% of cases (24/5,020). CEP8 and CEP11 CNI (≥3.0) was more frequent in the hyperdiploid than CEP17 non-polysomic group (55.6% vs. 6.1% and 25% vs. 2.3%, respectively). Our results suggest that chromosome 17 polysomy is a rare event found in <1% breast cancer cases and that polysomy of other chromosomes frequently occurs with chromosome 17 polysomy.
Collapse
Affiliation(s)
- Vladimira Koudelakova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| | - Radek Trojanec
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| | - Jana Vrbkova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| | - Sandra Donevska
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| | - Katerina Bouchalova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| | - Zdenek Kolar
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 3, 775 15 Olomouc, the Czech Republic
| | - Lakshman Varanasi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Hnevotinska 5, 775 15 Olomouc, the Czech Republic
| |
Collapse
|
19
|
Polónia A, Leitão D, Schmitt F. Application of the 2013 ASCO/CAP guideline and the SISH technique for HER2 testing of breast cancer selects more patients for anti-HER2 treatment. Virchows Arch 2016; 468:417-23. [PMID: 26754674 DOI: 10.1007/s00428-016-1903-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/05/2015] [Accepted: 01/03/2016] [Indexed: 01/05/2023]
Abstract
The aim of this study is to assess the impact of changes of the 2013 ASCO/CAP guideline on the results of HER2 testing in breast cancer. A series of 916 primary invasive breast cancer cases, assessed as HER2 2+ by IHC in part using the 2007 and in part the 2013 ASCO/CAP criteria, was evaluated for HER2 amplification status by SISH and classified according to both 2007 and 2013 ASCO/CAP ISH guideline criteria. We observed a significant increase of HER2-positive cases (12.4 to 16.8%) and a decrease of HER2-equivocal cases (3.6 to 0.7%). Of the cases studied, 52.1% fulfilled both criteria of HER2/CEP17 ratio and average HER2 copy number per cell to be classified as HER2-positive. Reclassification of the cases from before the introduction of the new ASCO/CAP guideline with the 2013 ISH criteria resulted in an increase of cases with a HER2-positive status (12.4 to 14.2%) and in a decrease of HER2-equivocal cases (3.6 to 1.6%). The 2013 ASCO/CAP guideline selects more patients for anti-HER2 targeted therapy, mostly based on the modifications of criteria to evaluate ISH-HER2.
Collapse
Affiliation(s)
- António Polónia
- Department of Pathology, Ipatimup Diagnostics, Ipatimup, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Dina Leitão
- Department of Pathology, Ipatimup Diagnostics, Ipatimup, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fernando Schmitt
- Department of Pathology, Ipatimup Diagnostics, Ipatimup, Porto, Portugal.
- Faculty of Medicine, University of Porto, Porto, Portugal.
- Laboratoire national de santé, 1, rue Louis Rech, L-3555, Dudelange, Luxembourg.
| |
Collapse
|
20
|
Impact of updated HER2 testing guidelines in breast cancer--re-evaluation of HERA trial fluorescence in situ hybridization data. Mod Pathol 2015; 28:1528-34. [PMID: 26403781 DOI: 10.1038/modpathol.2015.112] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/03/2015] [Indexed: 11/08/2022]
Abstract
Recently the American Society of Clinical Oncology and the College of American Pathologists have updated their clinical practice guidelines for HER2 testing in breast cancer. In order to evaluate these new recommendations, we have re-assessed the HER2 status of 6018 breast cancer cases of the screening population for the HERceptin adjuvant (HERA) trial that were originally centrally tested by fluorescence in situ hybridization based on the FDA-released test guidelines. According to the most recent 2013 ASCO/CAP recommendations, 3380 (56.2%) cases were classified as HER2 positive compared with 3359 (55.8%) applying the HERA/FDA scheme and 3339 (55.5%) applying the 2007 ASCO/CAP guidelines. Twenty-one cases switched from negative (HERA/FDA scheme) to positive (2013 ASCO/CAP guidelines). This group is characterized by a mean HER2 gene copy number of ≥6.0, polysomy or co-amplification of CEP17 with an average CEP17 count of 5, and with HER2 receptor overexpression in 75% of cases. On the basis of the HER2 gene copy number alone, we observe 494 cases (8.2%) that are in the equivocal range. Most of these cases (>80%) were also nondecisive by immunohistochemistry (score 2+) irrespective of whether ratio was <2.0>. The number of equivocal cases that would require HER2 reflex testing decreases to 113 (1.9%) if in addition to the HER2 gene copy number also the ratio of HER2 and CEP17 copy numbers is considered via dual-color in situ hybridization. The combination of applying the HER2 mean gene copy number as well as the HER2/CEP17 ratio to define equivocal test decisions by fluorescence in situ hybridization as proposed by the current ASCO/CAP guidelines appears to be a more optimum approach to adopt in order to avoid or minimize reporting of false negative results. Using the mean HER2 gene copy number alone for decision making results in a significant increase of equivocal cases.
Collapse
|
21
|
Long TH, Lawce H, Durum C, Moore SR, Olson SB, Gatter K, Troxell ML. The New Equivocal: Changes to HER2 FISH Results When Applying the 2013 ASCO/CAP Guidelines. Am J Clin Pathol 2015; 144:253-62. [PMID: 26185310 DOI: 10.1309/ajcp3q9wfoqtkuvv] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVES Human epidermal growth factor receptor 2 (HER2, ERBB2) testing is an important prognostic/predictive marker in breast cancer management, especially in selecting HER2-targeted treatment. American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines address HER2 status and were recently revised in 2013, replacing the 2007 version. For in situ hybridization interpretation, 2013 guidelines return to the prior threshold of a HER2/CEP17 ratio of 2.0 or greater for positive and eliminate 1.8 to 2.2 as the equivocal range. Also, the HER2 signal/nucleus ratio is accounted for, with 6.0 or greater for positive and 4.0 to less than 6.0 for equivocal, even in cases with a HER2/CEP17 ratio less than 2.0. METHODS With institutional review board approval, we reviewed our 2006 to 2012 HER2 fluorescence in situ hybridization (FISH) results and classified them according to both the 2007 and 2013 guidelines as negative, positive, or equivocal. RESULTS Of 717 HER2 FISH results, 55 (7.7%) changed category when reassessed by 2013 guidelines. Nineteen of 25 results in the 2007 equivocal category were reassigned as positive (n = 13) or negative (n = 6). Thirty-five previously negative cases became equivocal in the 2013 scheme, 12 of these with 1+ immunohistochemistry. The positive category increased from 71 to 85. CONCLUSIONS The 2013 ASCO/CAP guidelines increased the number of HER2 FISH positive and equivocal results. The equivocal group is substantially different, posing a dilemma for clinical management.
Collapse
Affiliation(s)
- Thomas H. Long
- Department of Pathology, Oregon Health & Science University, Portland
| | - Helen Lawce
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Connie Durum
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Stephen R. Moore
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland
| | - Susan B. Olson
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland
| | - Ken Gatter
- Department of Pathology, Oregon Health & Science University, Portland
| | - Megan L. Troxell
- Department of Pathology, Oregon Health & Science University, Portland
- Knight Cancer Institute, Oregon Health & Science University, Portland
| |
Collapse
|
22
|
Jang MH, Kim EJ, Kim HJ, Chung YR, Park SY. Assessment of HER2 status in invasive breast cancers with increased centromere 17 copy number. Breast Cancer Res Treat 2015. [DOI: 10.1007/s10549-015-3522-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
23
|
Not All Next Generation Sequencing Diagnostics are Created Equal: Understanding the Nuances of Solid Tumor Assay Design for Somatic Mutation Detection. Cancers (Basel) 2015; 7:1313-32. [PMID: 26193321 PMCID: PMC4586770 DOI: 10.3390/cancers7030837] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 01/15/2023] Open
Abstract
The molecular characterization of tumors using next generation sequencing (NGS) is an emerging diagnostic tool that is quickly becoming an integral part of clinical decision making. Cancer genomic profiling involves significant challenges including DNA quality and quantity, tumor heterogeneity, and the need to detect a wide variety of complex genetic mutations. Most available comprehensive diagnostic tests rely on primer based amplification or probe based capture methods coupled with NGS to detect hotspot mutation sites or whole regions implicated in disease. These tumor panels utilize highly customized bioinformatics pipelines to perform the difficult task of accurately calling cancer relevant alterations such as single nucleotide variations, small indels or large genomic alterations from the NGS data. In this review, we will discuss the challenges of solid tumor assay design/analysis and report a case study that highlights the need to include complementary technologies (i.e., arrays) and germline analysis in tumor testing to reliably identify copy number alterations and actionable variants.
Collapse
|
24
|
The Clinicopathologic and Prognostic Value of Altered Chromosome 17 Centromere Copy Number in HER2 Fish Equivocal Breast Carcinomas. PLoS One 2015; 10:e0132824. [PMID: 26161550 PMCID: PMC4498752 DOI: 10.1371/journal.pone.0132824] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 06/19/2015] [Indexed: 11/19/2022] Open
Abstract
Chromosome 17 centromere (CEP17) gain is frequently observed in breast cancer by fluorescence in situ hybridization (FISH). To address the biologic characteristics and clinical significance of CEP17 gain in a large population of breast cancer patients, we performed FISH on a series of 770 breast cancer tissues to evaluate the status of human epidermal growth factor receptor 2 (HER2) gene and CEP17 by immunohistochemistry (IHC) and FISH. Among the 770 specimens, 184 cases showed CEP17 gain (23.9%). Histological grade, nodal status, HER2 by IHC, Ki 67 index, and p53 expression were significantly different between CEP17 gain tumors and HER2-positive tumors. In contrast with HER2-negative tumors, CEP17 gain tumors showed higher histological grade, higher HER2 score by IHC, and higher Ki 67 index. The patients with CEP17 gain tumors had an intermediate survival between HER2-negative and HER2-positive patients. By comparison to HER2-negative and HER2-positive patients, survival in luminal B patients with CEP17 gain tumors also fell in between. In conclusion, CEP17 gain tumors show specific differences compared with HER2-negative and HER2-positive tumors in clinical parameters and prognosis.
Collapse
|
25
|
Brunelli M, Nottegar A, Bogina G, Caliò A, Cima L, Eccher A, Vicentini C, Marcolini L, Scarpa A, Pedron S, Brunello E, Knuutila S, Sapino A, Marchiò C, Bria E, Molino A, Carbognin L, Tortora G, Jasani B, Miller K, Merdol I, Zanatta L, Laurino L, Wirtanen T, Zamboni G, Marconi M, Chilosi M, Manfrin E, Martignoni G, Bonetti F. Monosomy of chromosome 17 in breast cancer during interpretation of HER2 gene amplification. Am J Cancer Res 2015; 5:2212-2221. [PMID: 26328251 PMCID: PMC4548332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 06/11/2015] [Indexed: 06/04/2023] Open
Abstract
Monosomy of chromosome 17 may affect the assessment of HER2 amplification. Notably, the prevalence ranges from 1% up to 49% due to lack of consensus in recognition. We sought to investigate the impact of monosomy of chromosome 17 to interpretation of HER2 gene status. 201 breast carcinoma were reviewed for HER2 gene amplification and chromosome 17 status. FISH analysis was performed by using double probes (LSI/CEP). Absolute gene copy number was also scored per each probe. HER2 FISH test was repeated on serial tissue sections, ranging in thickness from 3 to 20 µm. Ratio was scored and subsequently corrected by monosomy after gold control test using the aCGH method to overcome false interpretation due to artefactual nuclear truncation. HER2 immunotests was performed on all cases. 26/201 cases were amplified (13%). Single signals per CEP17 were revealed in 7/201 (3.5%) cases. Five out of 7 cases appeared monosomic with aCGH (overall, 5/201, 2.5%) and evidenced single signals in >60% of nuclei after second-look on FISH when matching both techniques. Among 5, one case showed amplification with a pattern 7/1 (HER2/CEP17>2) of copies (3+ at immunotest); three cases revealed single signals per both probes (LSI/CEP=1) and one case revealed a 3:1 ratio; all last 4 cases showed 0/1+ immunoscore. We concluded that: 1) monosomy of chromosome 17 may be observed in 2.5% of breast carcinoma; 2) monosomy of chromosome 17 due to biological reasons rather than nuclear truncation was observed when using the cut-off of 60% of nuclei harboring single signals; 3) the skewing of the ratio due to single centromeric 17 probe may lead to false positive evaluation; 4) breast carcinomas showing a 3:1 ratio (HER2/CEP17) usually show negative 0/1+ immunoscore and <6 gene copy number at FISH.
Collapse
Affiliation(s)
- Matteo Brunelli
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Alessia Nottegar
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Giuseppe Bogina
- Anatomic Pathology, Sacro Cuore Don Calabria HospitalNegrar, Italy
| | - Anna Caliò
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Luca Cima
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Albino Eccher
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | | | - Lisa Marcolini
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Aldo Scarpa
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
- ARC-NET Applied Research Centre, University of VeronaItaly
| | - Serena Pedron
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Eleonora Brunello
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Sakari Knuutila
- Department of Pathology, Laboratory of Molecular Cytogenetic, University of HelsinkiFinland
| | - Anna Sapino
- Department of Medical Sciences, Anatomic Pathology, University and Hospital TrustTurin, Italy
| | - Caterina Marchiò
- Department of Medical Sciences, Anatomic Pathology, University and Hospital TrustTurin, Italy
| | - Emilio Bria
- Medical Oncology, University and Hospital TrustVerona, Italy
| | | | - Luisa Carbognin
- Medical Oncology, University and Hospital TrustVerona, Italy
| | | | - Bharat Jasani
- Institute of Cancer & Genetics, Pathology, Cardiff UniversityUnited Kingdom
| | - Keith Miller
- UK NEQAS, University College of LondonUnited Kingdom
| | | | - Lucia Zanatta
- Anatomic Pathology, S. Maria di Ca’ Foncello HospitalTreviso, Italy
| | - Licia Laurino
- Anatomic Pathology, S. Maria di Ca’ Foncello HospitalTreviso, Italy
| | - Tiina Wirtanen
- Department of Pathology, Laboratory of Molecular Cytogenetic, University of HelsinkiFinland
- Department of Pathology, HUSLab, University Central HospitalHelsinki, Finland
| | - Giuseppe Zamboni
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
- Anatomic Pathology, Sacro Cuore Don Calabria HospitalNegrar, Italy
| | - Marcella Marconi
- Anatomic Pathology, Sacro Cuore Don Calabria HospitalNegrar, Italy
| | - Marco Chilosi
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Erminia Manfrin
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Guido Martignoni
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| | - Franco Bonetti
- Department of Pathology and Diagnostic, Anatomic Pathology, University and Hospital TrustVerona, Italy
| |
Collapse
|
26
|
Pala EE, Bayol Ü, Özgüzer A, Küçük Ü, Akdeniz ÇY, Sezer Ö. Problems In Determining Her2 Status In Breast Carcinoma. THE JOURNAL OF BREAST HEALTH 2015; 11:10-16. [PMID: 28331683 DOI: 10.5152/tjbh.2014.2103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/14/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Human epidermal growth factor receptor 2 (HER2) oncoprotein is overexpressed in 15-25% of breast carcinomas and associated with poor outcome. Assessment of HER2 status accurately is important to select patients who will benefit from targeted therapy. MATERIALS AND METHODS In this study immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) were used to determine the HER2 status in 308 breast carcinoma cases of which 129 were consultation. The major problems in determining HER2 status and the reasons of discordant results between methods were discussed. RESULTS HER2 expression was (-) in 124, (+) in 29, (++) in 92, (+++) in 63 cases. 25 of 76 cases consulted as (++) were evaluated as (++) and 15 of 35 cases consulted as (+++) were evaluated as (+++). HER2 amplification was found in 88 (28.6%) of 308 cases by FISH. 3 of 124 (-), 1 of 29 (+), 22 of 92 (++), 62 of 63 (+++) cases were amplified by FISH. The relation between HER2 expression and amplification was statistically significant (p<0.001). Centromere 17 (CEN 17) region amplification was noted in 11 cases of which 2 were (+++), 9 were (++). 6 of the 11 cases showed focal low level, 1 of them showed diffuse high level amplification. CONCLUSION The concordance rate between IHC (+++) cases and FISH was 95.4% for consultation cases, 100% for our cases. The final concordance rate for both case groups was 98.4%. The possible reasons of discrepancy were triple negativity, preanalytical and analytical procedures of consultation cases and trucut samples.
Collapse
Affiliation(s)
- Emel Ebru Pala
- Clinic of Pathology, İzmir Tepecik Training and Research Hospital, İzmir, Turkey
| | - Ümit Bayol
- Clinic of Pathology, İzmir Tepecik Training and Research Hospital, İzmir, Turkey
| | - Alp Özgüzer
- Clinic of Pathology, İzmir Tepecik Training and Research Hospital, İzmir, Turkey
| | - Ülkü Küçük
- Clinic of Pathology, İzmir Tepecik Training and Research Hospital, İzmir, Turkey
| | | | - Özlem Sezer
- Clinic of Pathology, İzmir Tepecik Training and Research Hospital, İzmir, Turkey
| |
Collapse
|
27
|
Hanna WM, Barnes PJ, Chang MC, Gilks CB, Magliocco AM, Rees H, Quenneville L, Robertson SJ, SenGupta SK, Nofech-Mozes S. Human epidermal growth factor receptor 2 testing in primary breast cancer in the era of standardized testing: a Canadian prospective study. J Clin Oncol 2014; 32:3967-73. [PMID: 25385731 DOI: 10.1200/jco.2014.55.6092] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Therapies that target overexpression of human epidermal growth factor receptor 2 (HER2) rely on accurate and timely assessment of all patients with new diagnoses. This study examines HER2 testing of primary breast cancer tissue when performed with immunohistochemistry (IHC) and additional in situ hybridization (ISH) for negative cases (IHC 0/1+). The analysis focuses on the rate of false-negative HER2 tests, defined as IHC 0/1+ with an ISH ratio ≥ 2.0, in eight pathology centers across Canada. PATIENTS AND METHODS Whole sections of surgical resections or tissue microarrays (TMAs) from invasive breast carcinoma tissue were tested by both IHC and ISH using standardized local methods. Samples were scored by the local breast pathologist, and consecutive HER2-negative IHC results (IHC 0/1+) were compared with the corresponding fluorescence or silver ISH result. RESULTS Overall, 711 surgical excisions of primary breast cancer were analyzed by IHC and ISH; HER2 and chromosome 17 centromere (CEP17) counts were available in all cases. The overall rate of false-negative samples was 0.84% (six of 711 samples). Interpretable IHC and ISH scores were available in 1,212 cases from TMAs, and the overall rate of false-negative cases was 1.6% (16 of 978 cases). CONCLUSION Our observation confirms that IHC is an adequate test to predict negative HER2 status in primary breast cancer in surgical excision specimens, even when different antibodies and IHC platforms are used. The study supports the American Society of Clinical Oncology/College of American Pathologists and Canadian testing algorithms of using IHC followed by ISH for equivocal cases.
Collapse
Affiliation(s)
- Wedad M Hanna
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL.
| | - Penny J Barnes
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Martin C Chang
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - C Blake Gilks
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Anthony M Magliocco
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Henrike Rees
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Louise Quenneville
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Susan J Robertson
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Sandip K SenGupta
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| | - Sharon Nofech-Mozes
- Wedad M. Hanna and Sharon Nofech-Mozes, Sunnybrook Health Sciences Centre, University of Toronto; Wedad M. Hanna, Martin C. Chang, and Sharon Nofech-Mozes, University of Toronto; Martin C. Chang, Mount Sinai Hospital, Toronto; Susan J. Robertson, Ottawa General Hospital and University of Ottawa, Ottawa; Sandip K. SenGupta, Kingston General Hospital and Queen's University, Kingston, Ontario; Penny J. Barnes, Capital Health District Authority and Dalhousie University, Halifax, Nova Scotia; C. Blake Gilks, Vancouver General Hospital and University of British Columbia, Vancouver, British Columbia; Henrike Rees and Louise Quenneville, Saskatoon City Hospital and University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and Anthony M. Magliocco, Esoteric Laboratory Services, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
28
|
Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JMS, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med 2014; 138:241-56. [PMID: 24099077 PMCID: PMC4086638 DOI: 10.5858/arpa.2013-0953-sa] [Citation(s) in RCA: 813] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To update the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guideline recommendations for human epidermal growth factor receptor 2 (HER2) testing in breast cancer to improve the accuracy of HER2 testing and its utility as a predictive marker in invasive breast cancer. METHODS ASCO/CAP convened an Update Committee that included coauthors of the 2007 guideline to conduct a systematic literature review and update recommendations for optimal HER2 testing. RESULTS The Update Committee identified criteria and areas requiring clarification to improve the accuracy of HER2 testing by immunohistochemistry (IHC) or in situ hybridization (ISH). The guideline was reviewed and approved by both organizations. RECOMMENDATIONS The Update Committee recommends that HER2 status (HER2 negative or positive) be determined in all patients with invasive (early stage or recurrence) breast cancer on the basis of one or more HER2 test results (negative, equivocal, or positive). Testing criteria define HER2-positive status when (on observing within an area of tumor that amounts to >10% of contiguous and homogeneous tumor cells) there is evidence of protein overexpression (IHC) or gene amplification (HER2 copy number or HER2/CEP17 ratio by ISH based on counting at least 20 cells within the area). If results are equivocal (revised criteria), reflex testing should be performed using an alternative assay (IHC or ISH). Repeat testing should be considered if results seem discordant with other histopathologic findings. Laboratories should demonstrate high concordance with a validated HER2 test on a sufficiently large and representative set of specimens. Testing must be performed in a laboratory accredited by CAP or another accrediting entity. The Update Committee urges providers and health systems to cooperate to ensure the highest quality testing.
Collapse
Affiliation(s)
- Antonio C Wolff
- Antonio C. Wolff, Johns Hopkins Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth H. Hammond, University of Utah School of Medicine and Intermountain Healthcare, Salt Lake City, UT; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Mitch Dowsett, Royal Marsden Hospital, London, United Kingdom; Kimberly H. Allison, Stanford University Medical Center, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Donald C. Allred, Washington University School of Medicine, St Louis, MO; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada; Michael Bilous, University of Western Sydney and Healthscope Pathology, Sydney, New South Wales, Australia; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Soonmyung Paik, National Surgical Adjuvant Breast and Bowel Project, Pitsburgh, PA; Edith A. Perez, Mayo Clinic, Jacksonville, FL; Patricia A. Spears, North Carolina State University, Raleigh, NC; Gail H. Vance, Indiana University Medical Center, Indianapolis, IN; Giuseppe Viale, University of Milan, European Institute of Oncology, Milan, Italy; and Daniel F. Hayes, University of Michigan Comprehensive Cancer Care Center, Ann Arbor, MI
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity. Mod Pathol 2014; 27:4-18. [PMID: 23807776 DOI: 10.1038/modpathol.2013.103] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/05/2013] [Accepted: 04/11/2013] [Indexed: 12/19/2022]
Abstract
Trastuzumab-containing therapy is a standard of care for patients with HER2+ breast cancer. HER2 status is routinely assigned using in situ hybridization to assess HER2 gene amplification, but interpretation of in situ hybridization results may be challenging in tumors with chromosome 17 polysomy or intratumoral genetic heterogeneity. Apparent chromosome 17 polysomy, defined by increased chromosome enumeration probe 17 (CEP17) signal number, is a common genetic aberration in breast cancer and represents an alternative mechanism for increasing HER2 copy number. Some studies have linked elevated CEP17 count ('polysomy') with adverse clinicopathologic features and HER2 overexpression, although there are numerous discrepancies in the literature. There is evidence that elevated CEP17 ('polysomy') count might account for trastuzumab response in tumors with normal HER2:CEP17 ratios. Nonetheless, recent studies establish that apparent 'polysomy' (CEP17 increase) is usually related to focal pericentromeric gains rather than true polysomy. Assigning HER2 status may also be complex where multiple cell subclones with distinct HER2 amplification characteristics coexist within the same tumor. Such genetic heterogeneity affects up to 40% of breast cancers when assessed according to a College of American Pathologists guideline, although other definitions have been proposed. Recent data have associated heterogeneity with unfavorable clinicopathologic variables and poor prognosis. Genetically heterogeneous tumors harboring HER2-amplified subclones have the potential to benefit from trastuzumab, but this has yet to be evaluated in clinical studies. In this review, we discuss the implications of apparent polysomy 17 and genetic heterogeneity for assigning HER2 status in clinical practice. Among our recommendations, we support the use of mean HER2 copy number rather than HER2:CEP17 ratio to define HER2 positivity in cases where coamplification of the centromere might mask HER2 amplification. We also highlight a need to harmonize in situ hybridization scoring methodology to support accurate HER2 status determination, particularly where there is evidence of heterogeneity.
Collapse
|
30
|
Fitzgibbons PL, Dillon DA, Alsabeh R, Berman MA, Hayes DF, Hicks DG, Hughes KS, Nofech-Mozes S. Template for reporting results of biomarker testing of specimens from patients with carcinoma of the breast. Arch Pathol Lab Med 2013; 138:595-601. [PMID: 24236805 DOI: 10.5858/arpa.2013-0566-cp] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Patrick L Fitzgibbons
- From the Department of Pathology, St Jude Medical Center, Fullerton, California (Dr Fitzgibbons); the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Dr Dillon); the Department of Pathology, Kaiser Permanente-Los Angeles Medical Center, Los Angeles, California (Dr Alsabeh); the Department of Pathology, Jefferson Hospital, Jefferson Hills, Pennsylvania (Dr Berman); the Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor (Dr Hayes); the Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York (Dr Hicks); the Division of Surgical Oncology, Massachusetts General Hospital, Boston (Dr Hughes); and the Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada (Dr Nofech-Mozes)
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JMS, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol 2013; 31:3997-4013. [PMID: 24101045 DOI: 10.1200/jco.2013.50.9984] [Citation(s) in RCA: 2911] [Impact Index Per Article: 264.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE To update the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guideline recommendations for human epidermal growth factor receptor 2 (HER2) testing in breast cancer to improve the accuracy of HER2 testing and its utility as a predictive marker in invasive breast cancer. METHODS ASCO/CAP convened an Update Committee that included coauthors of the 2007 guideline to conduct a systematic literature review and update recommendations for optimal HER2 testing. RESULTS The Update Committee identified criteria and areas requiring clarification to improve the accuracy of HER2 testing by immunohistochemistry (IHC) or in situ hybridization (ISH). The guideline was reviewed and approved by both organizations. RECOMMENDATIONS The Update Committee recommends that HER2 status (HER2 negative or positive) be determined in all patients with invasive (early stage or recurrence) breast cancer on the basis of one or more HER2 test results (negative, equivocal, or positive). Testing criteria define HER2-positive status when (on observing within an area of tumor that amounts to > 10% of contiguous and homogeneous tumor cells) there is evidence of protein overexpression (IHC) or gene amplification (HER2 copy number or HER2/CEP17 ratio by ISH based on counting at least 20 cells within the area). If results are equivocal (revised criteria), reflex testing should be performed using an alternative assay (IHC or ISH). Repeat testing should be considered if results seem discordant with other histopathologic findings. Laboratories should demonstrate high concordance with a validated HER2 test on a sufficiently large and representative set of specimens. Testing must be performed in a laboratory accredited by CAP or another accrediting entity. The Update Committee urges providers and health systems to cooperate to ensure the highest quality testing. This guideline was developed through a collaboration between the American Society of Clinical Oncology and the College of American Pathologists and has been published jointly by invitation and consent in both Journal of Clinical Oncology and the Archives of Pathology & Laboratory Medicine.
Collapse
Affiliation(s)
- Antonio C Wolff
- Antonio C. Wolff, Johns Hopkins Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth H. Hammond, University of Utah School of Medicine and Intermountain Healthcare, Salt Lake City, UT; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Mitch Dowsett, Royal Marsden Hospital, London, United Kingdom; Kimberly H. Allison, Stanford University Medical Center, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Donald C. Allred, Washington University School of Medicine, St Louis, MO; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada; Michael Bilous, University of Western Sydney and Healthscope Pathology, Sydney, New South Wales, Australia; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Soonmyung Paik, National Surgical Adjuvant Breast and Bowel Project, Pittsburgh, PA; Edith A. Perez, Mayo Clinic, Jacksonville, FL; Patricia A. Spears, North Carolina State University, Raleigh, NC; Gail H. Vance, Indiana University Medical Center, Indianapolis, IN; Giuseppe Viale, University of Milan, European Institute of Oncology, Milan, Italy; and Daniel F. Hayes, University of Michigan Comprehensive Cancer Care Center, Ann Arbor, MI
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lambein K, Van Bockstal M, Vandemaele L, Geenen S, Rottiers I, Nuyts A, Matthys B, Praet M, Denys H, Libbrecht L. Distinguishing score 0 from score 1+ in HER2 immunohistochemistry-negative breast cancer: clinical and pathobiological relevance. Am J Clin Pathol 2013; 140:561-6. [PMID: 24045554 DOI: 10.1309/ajcp4a7ktayhzsoe] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVES To investigate the clinical and pathobiological significance of distinguishing score 0 and score 1+ within the group of immunohistochemistry (IHC)-negative invasive breast cancers. METHODS We studied HER2 status using both IHC and fluorescence in situ hybridization (FISH) in 150 consecutive breast tumors submitted to our laboratory after a negative IHC result in local testing centers. RESULTS We were able to discern a group of score 0 tumors that had a lower HER2 copy number than the group consisting of score 1+ tumors. In contrast with the group of score 1+ tumors, HER2 FISH was consistently negative for both copy number-based and ratio-based tumors without equivocal results. CONCLUSIONS In a setting with stringent quality assurance, score 0 and score 1+ tumors emerge as distinct and clinically important subgroups within the HER2 IHC-negative population.
Collapse
Affiliation(s)
- Kathleen Lambein
- Department of Pathology, Ghent University, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Mieke Van Bockstal
- Department of Pathology, Ghent University, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Lies Vandemaele
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Sofie Geenen
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | | | - Ann Nuyts
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Bart Matthys
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Marleen Praet
- Department of Pathology, Ghent University, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Hannelore Denys
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Louis Libbrecht
- Department of Pathology, Ghent University, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
33
|
Sapino A, Goia M, Recupero D, Marchiò C. Current Challenges for HER2 Testing in Diagnostic Pathology: State of the Art and Controversial Issues. Front Oncol 2013; 3:129. [PMID: 23734345 PMCID: PMC3659312 DOI: 10.3389/fonc.2013.00129] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022] Open
Abstract
HER2 overexpression and anti-HER2 agents represent probably the best story of success of individualized therapy in breast cancer. Due to the important therapeutic implications, the issue under the spotlight has been, since ever, the correct identification of true HER2 positivity on tissue specimens. Eligibility to anti-HER2 agents is strictly dependent on the demonstration of HER2 overexpression (by immunohistochemistry) or of HER2 gene amplification by in situ techniques (fluorescence in situ hybridization, FISH), however there are controversial issues involving cases with "equivocal" HER2 status based on conventional techniques (about 20% of specimens). In terms of HER2 expression a major debate is the presence of full-length and truncated forms of the protein and controversial clinical data have been reported on the therapeutic implications of these HER2 fragments. In terms of HER2 gene assessment, the occurrence of amplification of the chromosome 17 centromeric region (CEP17) has been proven responsible for misleading HER2 FISH results, precluding anti-HER2 based therapy to some patients. Finally HER2 activating mutations have been recently described as a biological mechanisms alternative to HER2 gene amplification. In this review we will focus on the controversies that pathologists and oncologists routinely face in the attempt to design the most tailored treatment for breast cancer patients. We will focus on the HER2 gene and on the protein, both at technical and interpretational levels.
Collapse
Affiliation(s)
- Anna Sapino
- Department of Medical Sciences, University of TurinTurin, Italy
| | - Margherita Goia
- Department of Medical Sciences, University of TurinTurin, Italy
| | | | | |
Collapse
|
34
|
Valent A, Penault-Llorca F, Cayre A, Kroemer G. Change in HER2 (ERBB2) gene status after taxane-based chemotherapy for breast cancer: polyploidization can lead to diagnostic pitfalls with potential impact for clinical management. Cancer Genet 2013; 206:37-41. [DOI: 10.1016/j.cancergen.2012.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 10/29/2012] [Accepted: 12/03/2012] [Indexed: 12/31/2022]
|
35
|
HER2 testing in breast cancer: an overview of current techniques and recent developments. Pathology 2012; 44:587-95. [DOI: 10.1097/pat.0b013e328359cf9a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
36
|
Portier BP, Gruver AM, Huba MA, Minca EC, Cheah AL, Wang Z, Tubbs RR. From morphologic to molecular: established and emerging molecular diagnostics for breast carcinoma. N Biotechnol 2012; 29:665-81. [PMID: 22504737 DOI: 10.1016/j.nbt.2012.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/26/2012] [Accepted: 03/31/2012] [Indexed: 12/15/2022]
Abstract
Diagnostics in the field of breast carcinoma are constantly evolving. The recent wave of molecular methodologies, both microscope and non-microscope based, have opened new ways to gain insight into this disease process and have moved clinical diagnostics closer to a 'personalized medicine' approach. In this review we highlight some of the advancements that laboratory medicine technology is making toward guiding the diagnosis, prognosis, and therapy selection for patients affected by breast carcinoma. The content of the article is largely structured by methodology, with a distinct emphasis on both microscope based and non-microscope based diagnostic formats. Where possible, we have attempted to emphasize the potential benefits as well as limitations to each of these technologies. Successful molecular diagnostics, applied in concert within the morphologic context of a patient's tumor, are what will lay the foundation for personalized therapy and allow a more sophisticated approach to clinical trial stratification. The future of breast cancer diagnostics looks challenging, but it is also a field of great opportunity. Never before have there been such a plethora of new tools available for disease investigation or candidate therapy selection.
Collapse
Affiliation(s)
- Bryce P Portier
- Department of Molecular Pathology, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Prognostic value of TOP2A gene amplification and chromosome 17 polysomy in early breast cancer. Pathol Oncol Res 2012; 18:885-94. [PMID: 22426810 DOI: 10.1007/s12253-012-9518-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 03/07/2012] [Indexed: 12/13/2022]
Abstract
The aim of this study was to analyze the occurrence of TOP2A gene amplification and chromosome 17 polysomy in patients with early breast cancer and to correlate the status of these alterations with the prognostic significance expressed as patients' clinical features and survival. Such concurrent analyses of TOP2A gene status and chromosome 17 polysomy have not been performed before. Study group included 149 consecutive stage I-III patients administered standard multimodality treatment. TOP2A abnormalities were examined by standard fluorescence in situ hybridization (FISH) and developed by our group quantitative real-time PCR (qPCR). TOP2A amplification and deletion assessed by FISH were found in 23% and 7% of the tumours, respectively, and by qPCR in 31% and 11% of the tumours, respectively. Chromosome 17 polysomy was detected in 40% of the cases. TOP2A amplification (by qPCR) correlated with shorter disease-free survival (p = 0.03) and overall survival (p = 0.047), and the prognostic value of TOP2A was confirmed in the multivariate analysis (HR = 3.22, 95% CI 1.09-9.56, p = 0.03). TOP2A gene amplification, but not chromosome 17 polysomy, carries negative prognostic information in early breast cancer. Given the aforementioned results, qPCR might serve as a prognostic tool in determining the patient's prognosis.
Collapse
|
38
|
Goud KI, Dayakar S, Vijayalaxmi K, Babu SJ, Vijay ARP. Evaluation of HER-2/neu status in breast cancer specimens using immunohistochemistry (IHC) & fluorescence in-situ hybridization (FISH) assay. Indian J Med Res 2012; 135:312-7. [PMID: 22561616 PMCID: PMC3361866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND & OBJECTIVES Fluorescence in situ hybridization (FISH) is increasingly being recognized as the most accurate and predictive test for HER 2/neu gene amplification and response to therapy in breast cancer. In the present study we investigated HER-2/neu gene amplification by FISH in breast carcinoma tissue specimens and compared the results with that of immunohistochemical (IHC) analysis. METHODS A total of 90 breast carcinoma tissue samples were used for immunohistochemical (IHC) and FISH analysis. IHC was performed by using mouse monoclonal antibody to the intracellular domain of HER-2/neu protein. Each slide was scored in a blinded fashion by two pathologists according to the manufacturer's recommended criteria. FISH analysis was performed on paraffin embedded breast tumour tissue sections. The polysomy for centromere 17 (Spec green signal) was read as green signals less than 4 as moderate polysomy, and more than 4 as highly polysomy. RESULTS Thirty of the 90 patients had negative results by IHC and FISH. Of the 28 patients with the score of 2+ by IHC, 20 were FISH positive for HER-2/neu gene amplification, three were FISH negative and five patients showed equivocal (1.8-2.2) results by FISH. These five cases were retested for IHC and FISH on different paraffin embedded tissue blocks, and all five were found positive for HER-2/neu gene amplification. Twenty five patients with the score of 3+ by IHC were FISH positive for HER-2/neu gene amplification (>2.2). Seven cases with the score of 3+ by IHC were FISH negative for HER-2/neu gene amplification (>2.2), and showed polysomy of chromosome number 17 high polysomy > 4. INTERPRETATION & CONCLUSIONS Our results indicated that HER-2/neu status by FISH should be performed in all cases of breast tumour with a 2+ score by IHC. Cases demonstrating a 3+ score by IHC may be subjected to FISH to rule out polysomy of chromosome 17 which could be falsely interpreted as HER-2/neu overexpression by IHC analysis. There is also a need for establishing a clinically validated cut-off value for HER-2/neu FISH amplification against IHC which may be further compared and calibrated.
Collapse
Affiliation(s)
- Kalal Iravathy Goud
- Department of Molecular Biology & Cytogenetics, Apollo Health City, Hyderabad, India,Reprint requests: Dr Kalal Iravathy Goud, Molecular Biology & Cytogenetics Laboratory, Apollo Health City, Jubilee Hills, Hyderabad 500 033, India e-mail:
| | - Seetha Dayakar
- Department of Molecular Biology & Cytogenetics, Apollo Health City, Hyderabad, India
| | | | - Saidam Jangu Babu
- Department of Molecular Biology & Cytogenetics, Apollo Health City, Hyderabad, India
| | | |
Collapse
|
39
|
Allison KH, Dintzis SM, Schmidt RA. Frequency of HER2 heterogeneity by fluorescence in situ hybridization according to CAP expert panel recommendations: time for a new look at how to report heterogeneity. Am J Clin Pathol 2011; 136:864-71. [PMID: 22095371 DOI: 10.1309/ajcpxtzskbrip07w] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In 2009, a College of American Pathologists expert panel published supplemental HER2 testing recommendations suggesting that cases with between 5% and 50% individual cells amplified by fluorescence in situ hybridization be reported as "heterogeneous for HER2 gene amplification." We examined the implications of applying these recommendations to clinical practice in 1,329 consecutive breast cancer cases. By ratio criteria, 23.2% of cases met the proposed criteria for heterogeneity, of which 81.6% were not amplified and 15.5% were equivocal by standard criteria. In contrast, the proposed criteria based on HER2 signals per cell classified only 6.5% of cases as heterogeneous, of which only 8% (7/87) were not amplified and 79% (69/87) were equivocal by standard criteria. These results show that the 2 proposed criteria sets are not equivalent and that the ratio-based definition results in large numbers of nonamplified cases being classified as heterogeneous. Further definition of optimal criteria with clinical relevance is needed before HER2 heterogeneity reporting is adopted in routine practice.
Collapse
Affiliation(s)
| | - Suzanne M. Dintzis
- Department of Pathology, University of Washington Medical Center, Seattle
| | - Rodney A. Schmidt
- Department of Pathology, University of Washington Medical Center, Seattle
| |
Collapse
|
40
|
Kasaian K, Jones SJ. A new frontier in personalized cancer therapy: mapping molecular changes. Future Oncol 2011; 7:873-94. [PMID: 21732758 DOI: 10.2217/fon.11.63] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in the genome of a normal cell can affect the function of its many genes and pathways. These alterations could eventually transform the cell from a normal to a malignant state by allowing an uncontrolled proliferation of the cell and formation of a cancer tumor. Each tumor in an individual patient can have hundreds of mutated genes and perturbed pathways. Cancers clinically presenting as the same type or subtype could potentially be very different at the molecular level and thus behave differently in response to therapy. The challenge is to distinguish the key mutations driving the cancer from the background of mutational noise and find ways to effectively target them. The promise is that such a molecular approach to classifying cancer will lead to better diagnostic, prognostic and personalized treatment strategies. This article provides an overview of advances in the molecular characterization of cancers and their applications in therapy.
Collapse
Affiliation(s)
- Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | |
Collapse
|
41
|
Davis JD, Lin SY. DNA damage and breast cancer. World J Clin Oncol 2011; 2:329-38. [PMID: 21909479 PMCID: PMC3168783 DOI: 10.5306/wjco.v2.i9.329] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 08/08/2011] [Accepted: 08/15/2011] [Indexed: 02/06/2023] Open
Abstract
Cancer is intimately related to the accumulation of DNA damage, and repair failures (including mutation prone repair and hyperactive repair systems). This article relates current clinical categories for breast cancer and their common DNA damage repair defects. Information is included on the potential for accumulation of DNA damage in the breast tissue of a woman during her lifetime and the role of DNA damage in breast cancer development. We then cover endogenous and exogenous sources of DNA damage, types of DNA damage repair and basic signal transduction pathways for three gene products involved in the DNA damage response system; namely BRCA1, BRIT1 and PARP-1. These genes are often considered tumor suppressors because of their roles in DNA damage response and some are under clinical investigation as likely sources for effective new drugs to treat breast cancers. Finally we discuss some of the problems of DNA damage repair systems in cancer and the conundrum of hyper-active repair systems which can introduce mutations and confer a survival advantage to certain types of cancer cells.
Collapse
Affiliation(s)
- Jennifer D Davis
- Jennifer D Davis, Shiaw-Yih Lin, Department of Systems Biology, Unit 950, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | | |
Collapse
|
42
|
Moelans CB, Reis-Filho JS, van Diest PJ. Implications of rarity of chromosome 17 polysomy in breast cancer. Lancet Oncol 2011; 12:1087-9. [PMID: 21903472 DOI: 10.1016/s1470-2045(11)70234-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Utrecht 3508 GA, Netherlands
| | | | | |
Collapse
|
43
|
Egervari K, Kosa C, Szollosi Z. Impact of chromosome 17 centromere region assessment on HER2 status reported in breast cancer. Pathol Res Pract 2011; 207:468-71. [DOI: 10.1016/j.prp.2011.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 05/18/2011] [Indexed: 11/30/2022]
|
44
|
Gruver AM, Portier BP, Tubbs RR. Molecular pathology of breast cancer: the journey from traditional practice toward embracing the complexity of a molecular classification. Arch Pathol Lab Med 2011; 135:544-57. [PMID: 21526953 DOI: 10.5858/2010-0734-rair.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Adenocarcinoma of the breast is the most frequent cancer affecting women in both developed and developing regions of the world. From the moment of clinical presentation until the time of pathologic diagnosis, patients affected by this disease will face daunting questions related to prognosis and treatment options. While improvements in targeted therapies have led to increased patient survival, these same advances have created the imperative to accurately stratify patients to achieve maximum therapeutic efficacy while minimizing side effects. In this evolving era of personalized medicine, there is an ever-increasing need to overcome the limitations of traditional diagnostic practice. OBJECTIVE To summarize the molecular diagnostics traditionally used to guide prognostication and treatment of breast carcinomas, to highlight published data on the molecular classification of these tumors, and to showcase molecular assays that will supplement traditional methods of categorizing the disease. DATA SOURCES A review of the literature covering the molecular diagnostics of breast carcinomas with a focus on the gene expression and array studies used to characterize the molecular signatures of the disease. Special emphasis is placed on summarizing evolving technologies useful in the diagnosis and characterization of breast carcinoma. CONCLUSIONS Available and emerging molecular resources will allow pathologists to provide superior diagnostic, prognostic, and predictive information about individual breast carcinomas. These advances should translate into earlier identification and tailored therapy and should ultimately improve outcome for patients affected by this disease.
Collapse
Affiliation(s)
- Aaron M Gruver
- Department of Molecular Pathology, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH 44195, USA
| | | | | |
Collapse
|