1
|
Berton Giachetti PPM, Curigliano G. Pyrotinib in combination with first-line trastuzumab and docetaxel for patients with HER2-positive metastatic breast cancer: a new therapeutic option? Nat Rev Clin Oncol 2024; 21:171-172. [PMID: 38114789 DOI: 10.1038/s41571-023-00854-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Affiliation(s)
- Pier Paolo M Berton Giachetti
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
2
|
Li Y, Chen T, Du F, Wang H, Ma L. Concordance of RT-qPCR with immunohistochemistry and its beneficial role in breast cancer subtyping. Medicine (Baltimore) 2023; 102:e35272. [PMID: 37746948 PMCID: PMC10519502 DOI: 10.1097/md.0000000000035272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/12/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
This study was to compare the concordance of transcription-quantitative polymerase chain reaction (RT-qPCR) with immunohistochemistry (IHC) in determining estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and tumor proliferation index (Ki67) status in breast cancer, and to assess the prognosis based on different subtypes. Totally 323 breast cancer patients were selected, including 216 in the training set and 107 in the validation set. Logistic regression models were constructed using 5-fold cross-validation with the mRNA expression of each biomarker as the predictor and the corresponding IHC expression level as the binary response variable. Receiver operating characteristic curve was used to determine the cutoff value. When the thresholds of ER, PR, HER2, and Ki67 were 0.764, 0.709, 0.161, and 0.554, there existed high concordance rates between IHC and RT-qPCR in ER (94.4%), PR (88.0%) and HER2 (89.4%) and a medium concordance rate in Ki67 (67.8%), which were further confirmed in the validation set (ER: 81.3%, PR: 78.3%, HER2: 80.4%, and Ki67: 69.1%). Based on the subtyping stratified by RT-qPCR, the 5-year recurrence-free interval rates of patients with luminal, HER2-enriched, and triple-negative subtypes were 88% (95% CI: 0.84-0.93), 82% (95% CI: 0.73-0.92) and 58% (95% CI: 0.42-0.80), respectively, which were similar to those assessed by IHC (88%, 78% and 47%). RT-qPCR may be a complementary method to IHC, which can not only provide additional useful information in clinic, but also show more advantages over IHC in determining certain subtypes of breast cancer.
Collapse
Affiliation(s)
- Yilun Li
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Furong Du
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics CO., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Huimin Wang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics CO., Ltd., Nanjing, China
- Department of Medicine, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
| | - Li Ma
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
3
|
Correlation of ER, PR, and HER2 at protein and mRNA levels in the Asian patients with operable breast cancer. Biosci Rep 2022; 42:230628. [PMID: 35006257 PMCID: PMC8766827 DOI: 10.1042/bsr20211706] [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: 07/24/2021] [Revised: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer is the most common cancer and the leading cause of cancer-related deaths in women. The estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are the important biomarkers in the prognosis of breast cancer, and their expression is used to categorize breast cancer into subtypes. We aimed to analyze the concordance among ER, PR, and HER2 expression levels and breast cancer subtyping results obtained by immunohistochemistry (IHC, for protein) and reverse transcriptase-polymerase chain reaction (RT-PCR, for mRNA) and to assess the recurrence-free survival (RFS) of the different subtypes as determined by the two methods. We compared biomarker expression by IHC and RT-PCR in 397 operable breast cancer patients and categorized all patients into luminal, HER2, and triple-negative (TN) subtypes. The concordance of biomarker expression between the two methods was 81.6% (κ = 0.4075) for ER, 87.2% (κ = 0.5647) for PR, and 79.1% (κ = 0.2767) for HER2. The κ-statistic was 0.3624 for the resulting luminal, HER2, and TN subtypes. The probability of 5-year RFS was 0.78 for the luminal subtype versus 0.77 for HER2 and 0.51 for TN, when determined by IHC (P=0.007); and 0.80, 0.71, and 0.61, respectively, when determined by the RT-PCR method (P=0.008). Based on the current evidence, subtyping by RT-PCR performs similar to conventional IHC with regard to the 5-year prognosis. The PCR method may thus provide a complementary means of subtyping when IHC results are ambiguous.
Collapse
|
4
|
Abstract
Objective We aimed to describe the differences in clinicopathological characteristics and overall survival (OS) between male and female breast cancer patients, and to develop a prognostic nomogram to predict survival in patients with male breast cancer (MBC). Methods Using the Surveillance, Epidemiology, and End Results database, we compared age, race, histological type, histological grade, tumor size, lymph node status, metastases, estrogen/progesterone receptor (ER/PR) and HER-2 status between male and female patients, and analyzed their relationships with OS. We established a nomogram and produced a calibration curve to observe its predictive effect. Results Age, race, T stage, N stage, bone and lung metastases, and histological type and grade differed between male and female patients. OS in male patients was related to age, tumor size, metastatic site, ER/PR status, and histological grade, but not to race or lymph node status. A nomogram was established, which showed good predictive performance for survival in MBC patients (area under the curve = 0.7). Conclusion MBC has a worse prognosis than female breast cancer, mainly characterized by late onset age, late staging, high proportion of invasive non-specific histological types, high histological grade, and luminal breast cancer.
Collapse
Affiliation(s)
- Xinli Wang
- Xi'an International Medical Center Hospital, Xi'an, Shaanxi province, China
| | - Shusong Liu
- Xi'an International Medical Center Hospital, Xi'an, Shaanxi province, China
| | - Yan Xue
- Xi'an International Medical Center Hospital, Xi'an, Shaanxi province, China
| |
Collapse
|
5
|
Cardoso F, MacNeill F, Penault-Llorca F, Eniu A, Sardanelli F, Nordström EB, Poortmans P. Why is appropriate healthcare inaccessible for many European breast cancer patients? - The EBCC 12 manifesto. Breast 2021; 55:128-135. [PMID: 33461060 PMCID: PMC7817501 DOI: 10.1016/j.breast.2020.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
In Europe, inappropriate reimbursement and funding rules and regulations act as disincentives to best breast cancer care or, at worst, hinder best care. This problem was the focus of the 12th European Breast Cancer Conference (EBCC) manifesto, discussed during the virtual conference. As patient involvement is indispensable in driving changes to clinical practice, Europa Donna the European patient advocacy group was closely involved in the 12th manifesto. Reimbursement policies have rarely evolved with advances in breast cancer care such as outpatient (ambulatory) care rather than inpatient admission, use of oral or subcutaneous anti-cancer drugs rather than day-hospital intravenous administration, oncoplastic surgery techniques to minimize mastectomy rates, breast reconstructive surgery, risk-reducing surgery for BRCA mutation carriers, or use of hypo-fractionated breast radiation therapy. Although each European country, region and centre will have to understand how their reimbursement policies may hinder best care and find their own solutions, the problems are similar throughout Europe and some solutions can be broadly applied. This manifesto is not calling for more funding or demanding changes that will result in more expensive care. Reimbursement, if better aligned with guidelines and optimal clinical practice, will deliver more cost-effective healthcare. This will release resources, support more equitable use of finite funding and resources, so allowing more European breast cancer patients to benefit from evidence-based treatment recommended by national and international guidelines.
Collapse
Affiliation(s)
- Fatima Cardoso
- Breast Unit, Champalimaud Clinical Centre/ Champalimaud Foundation, Lisbon, Portugal.
| | - Fiona MacNeill
- Royal Marsden Hospital London, Division of Breast Surgery (UEMS), UK.
| | | | - Alexandru Eniu
- Cancer Institute Ion Chiricuta, Department of Breast Tumors, Day Hospital Unit, Cluj-Napoca, Romania; Hopital Riviera-Chablais, Switzerland.
| | - Francesco Sardanelli
- Università Degli Studi di Milano, Department of Biomedical Sciences for Health, Milan, Italy; IRCCS Policlinico San Donato, Department of Radiology, San Donato Milanese, Milan, Italy.
| | | | - Philip Poortmans
- Iridium Kankernetwerk and University of Antwerp, Wilrijk, Antwerp, Belgium.
| |
Collapse
|
6
|
Martuszewski A, Paluszkiewicz P, Nowak M, Szewczyk K, Staszek-Szewczyk U. Triple Discordances in Receptor Status During Breast Cancer Local Progression and Metastases: Case Report and Literature Review. Onco Targets Ther 2020; 13:10343-10349. [PMID: 33116614 PMCID: PMC7569034 DOI: 10.2147/ott.s260848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/11/2020] [Indexed: 11/23/2022] Open
Abstract
Breast cancer is the most common female malignant neoplasm in Poland and around the world. Precise determination of tumor molecular profile allows application of appropriate anticancer therapy, increasing the chances of recovery. A 28-year-old woman detected a thickening in her left breast. Mammography showed a change measuring 60 mm (radiologically BIRADS 5). The biopsy revealed invasive ductal carcinoma, luminal subtype B, HER2 positive (cT3N1M0). Neoadjuvant chemotherapy was administered and then breast conserving surgery was performed. In postoperative histopathology cancer, biological subtype was evaluated: HER2 positive, nonluminal (ypT2ypN0cM0). Then, postoperative radiotherapy was performed. After 14 months, breast ultrasonography (US) and mammography (MGF) revealed the presence of suspicious changes (BIRADS 4). Tru-cut biopsy confirmed cancer recurrence (luminal subtype B, HER2 negative, ER negative, PgR: 10%, Ki-67: 70%). Despite implemented and modified chemotherapy regimens, local progression occurred. Genetic testing excluded BRCA gene mutation. The patient qualified for radical mastectomy modo Halsted (ypT4bN0cM0). Postoperative microscopic examination revealed triple negative breast invasive carcinoma of no special type. After 22 months, metastatic lesions in lungs and left retrosternal nodes appeared. Due to the limited possibilities of systemic treatment, the patient qualified for stereotactic radiotherapy of tumors in the lungs' and left retrosternal nodes. Advancement, histological type and molecular profile should be controlled at each stage of the disease, as they may change several times and require modification of therapy.
Collapse
Affiliation(s)
| | | | | | - Krzysztof Szewczyk
- Wroclaw Medical University, Department of Oncology, Division of Surgical Oncology, Wroclaw, Poland.,Wroclaw Regional Comprehensive Cancer Center, 1st Department of Surgical Oncology, Wroclaw, Poland
| | - Urszula Staszek-Szewczyk
- Wroclaw Medical University, Department of Oncology, Division of Surgical Oncology, Wroclaw, Poland.,Wroclaw Regional Comprehensive Cancer Center, Department of Brachytherapy, Wroclaw, Poland
| |
Collapse
|
7
|
Estrogen, progesterone, and human epidermal growth factor receptor 2 discordance between primary and metastatic breast cancer. Breast Cancer Res Treat 2020; 183:137-144. [PMID: 32613540 PMCID: PMC7375990 DOI: 10.1007/s10549-020-05746-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 06/01/2020] [Indexed: 12/24/2022]
Abstract
Background The estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) statuses are frequently discordant between the primary tumor and metastatic lesions in metastatic breast cancer. This can have important therapeutic implications. Patients and methods In all, 541 patients with available receptor statuses from both primary tumor and metastatic lesion treated at Heidelberg and Tuebingen University Hospitals between 1982 and 2018 were included. Results Statistically significant discordance rates of 14% and 32% were found for ER and PR. HER2 status was statistically insignificantly discordant in 15% of patients. Gain in HER2 positivity was associated with an improved overall survival, whereas loss of HR positivity was associated with worse overall survival. Antiendocrine treatment differed in 20% of cases before and after biopsy and HER2-directed treatment in 14% of cases. Conclusions Receptor statuses are discordant between primary tumor and metastasis in a considerable fraction of patients with metastatic breast cancer. Next to a highly presumed predictive value with respect to efficacy of endocrine and HER2-targeted therapy, discordance seems to provide prognostically relevant information. Where feasible, metastatic lesions should be biopsied in accordance with current guidelines. Electronic supplementary material The online version of this article (10.1007/s10549-020-05746-8) contains supplementary material, which is available to authorized users.
Collapse
|
8
|
Eghlimi R, Shi X, Hrovat J, Xi B, Gu H. Triple Negative Breast Cancer Detection Using LC-MS/MS Lipidomic Profiling. J Proteome Res 2020; 19:2367-2378. [PMID: 32397718 DOI: 10.1021/acs.jproteome.0c00038] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Breast cancer (BC) is a heterogeneous malignancy that is responsible for a great portion of female cancer cases and cancer-related deaths in the United States. In comparison to other major BC subtypes, triple negative breast cancer (TNBC) presents with a relatively low survival rate and a high rate of metastasis. This has led to a strong, though largely unmet, need for more sensitive and specific methods of early-stage TNBC (ES-TNBC) detection to combat its high-grade pathology and relatively low survival rate. The current study employs a liquid chromatography-tandem mass spectrometry assay capable of targeted, highly specific, and sensitive detection of lipids to propose two diagnostic biomarker panels for TNBC/ES-TNBC. Using this approach, 110 lipids were reliably detected in 166 human plasma samples, 45 controls, and 121 BC (96 non-TNBC and 25 TNBC) subjects. Univariate and multivariate analyses allowed the construction and application of a 19-lipid biomarker panel capable of distinguishing TNBC (and ES-TNBC) from controls, as well as a 5-lipid biomarker panel capable of differentiating TNBC from non-TNBC and ES-TNBC from ES-non-TNBC. Receiver operating characteristic curves with notable classification performances were generated from the biomarker panels according to their orthogonal partial least-squares discrimination analysis models. TNBC was distinguished from controls with an area under the receiving operating characteristic curve (AUROC) = 0.93, sensitivity = 0.96, and specificity = 0.76 and ES-TNBC from controls with an AUROC = 0.96, sensitivity = 0.95, and specificity = 0.89. TNBC was differentiated from non-TNBC with an AUROC = 0.88, sensitivity = 0.88, and specificity = 0.79 and ES-TNBC from ES-non-TNBC with an AUROC = 0.95, sensitivity = 0.95, and specificity = 0.87. A pathway enrichment analysis between TNBC and controls also revealed significant disturbances in choline metabolism, sphingolipid signaling, and glycerophospholipid metabolism. To the best of our knowledge, this is the first study to propose a diagnostic lipid biomarker panel for TNBC detection. All raw mass spectrometry data have been deposited to MassIVE (dataset identifier MSV000085324).
Collapse
Affiliation(s)
- Ryan Eghlimi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Jonathan Hrovat
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Bowei Xi
- Department of Statistics, Purdue University, West Lafayette, Indiana 47907, United States
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| |
Collapse
|
9
|
Estrogen Receptor, Progesterone Receptor, and Human Epidermal Growth Factor Receptor-2 Testing in Breast Cancer: Assessing the Value of Repeated Centralized Testing in Excision Specimens. Appl Immunohistochem Mol Morphol 2020; 27:1-7. [PMID: 28549033 DOI: 10.1097/pai.0000000000000525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
At some tertiary breast care centers, where many patients are referred from other institutions, it is routine to repeat testing for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2/neu) in excision specimens if these tests were performed on the preceding biopsy at the referring facility. The goal of this study is to assess the value of this practice. We documented results from ER, PR, and HER2 testing in 541 consecutive invasive breast cancers excised over a 2.5-year period and analyzed the subset (n=153) for which testing was performed on the excision specimen solely due to the fact that testing on the preceding biopsy was performed at an outside institution. The rates and directions of biopsy-to-excision change were as follows: ER [1.3% (2/153), 100% from (+) to (-)]; PR [4% (6/153), 83% from (+) to (-)]; HER2/neu assessed by immunohistochemistry [21% (29/137)]; HER2/neu assessed by fluorescence in situ hybridization [3.3% (2/61); 50% from amplified to nonamplified and 50% vice versa]. There were no ER(-) and PR(-) biopsy cases that became ER and/or PR(+) in the excision. By coordinate analysis for the hormone receptors [ie, ER and/or PR(+) being indicative of "hormone receptor" (HR) positivity], there were no cases that changed from HR(+) in the biopsy to HR(-) in the excision (or vice versa), which suggests that repeat testing for ER and PR in this setting is of limited value. In an analysis that incorporated both immunohistochemistry and in situ fluorescence hybridization results, there were 2 cases with a clinically significant biopsy-to-excision change in HER2/neu status in which that change was detected primarily because the excision was retested. These findings provide baseline data for formulating policies on whether repeat testing should routinely be performed in the described scenario.
Collapse
|
10
|
Wahdan-Alaswad R, Liu B, Thor AD. Targeted lapatinib anti-HER2/ErbB2 therapy resistance in breast cancer: opportunities to overcome a difficult problem. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:179-198. [PMID: 35582612 PMCID: PMC9090587 DOI: 10.20517/cdr.2019.92] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
Abstract
Approximately 20% of invasive breast cancers have upregulation/gene amplification of the oncogene human epidermal growth factor receptor-2 (HER2/ErbB2). Of these, some also express steroid receptors (the so-called Luminal B subtype), whereas others do not (the HER2 subtype). HER2 abnormal breast cancers are associated with a worse prognosis, chemotherapy resistance, and sensitivity to selected anti-HER2 targeted therapeutics. Transcriptional data from over 3000 invasive breast cancers suggest that this approach is overly simplistic; rather, the upregulation of HER2 expression resulting from gene amplification is a driver event that causes major transcriptional changes involving numerous genes and pathways in breast cancer cells. Most notably, this includes a shift from estrogenic dependence to regulatory controls driven by other nuclear receptors, particularly the androgen receptor. We discuss members of the HER receptor tyrosine kinase family, heterodimer formation, and downstream signaling, with a focus on HER2 associated pathology in breast carcinogenesis. The development and application of anti-HER2 drugs, including selected clinical trials, are discussed. In light of the many excellent reviews in the clinical literature, our emphasis is on recently developed and successful strategies to overcome targeted therapy resistance. These include combining anti-HER2 agents with programmed cell death-1 ligand or cyclin-dependent kinase 4/6 inhibitors, targeting crosstalk between HER2 and other nuclear receptors, lipid/cholesterol synthesis to inhibit receptor tyrosine kinase activation, and metformin, a broadly inhibitory drug. We seek to facilitate a better understanding of new approaches to overcome anti-HER2 drug resistance and encourage exploration of two other therapeutic interventions that may be clinically useful for HER+ invasive breast cancer patients.
Collapse
Affiliation(s)
- Reema Wahdan-Alaswad
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO 80014, USA
| | - Bolin Liu
- Department of Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ann D Thor
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO 80014, USA
| |
Collapse
|
11
|
Ho-Pun-Cheung A, Bazin H, Boissière-Michot F, Mollevi C, Simony-Lafontaine J, Landas E, Bleuse JP, Chardès T, Prost JF, Pèlegrin A, Jacot W, Mathis G, Lopez-Crapez E. Quantification of HER1, HER2 and HER3 by time-resolved Förster resonance energy transfer in FFPE triple-negative breast cancer samples. Br J Cancer 2019; 122:397-404. [PMID: 31792349 PMCID: PMC7000684 DOI: 10.1038/s41416-019-0670-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) has a worse prognosis compared with other breast cancer subtypes, and biomarkers to identify patients at high risk of recurrence are needed. Here, we investigated the expression of human epidermal receptor (HER) family members in TNBC and evaluated their potential as biomarkers of recurrence. Methods We developed Time Resolved-Förster Resonance Energy Transfer (TR-FRET) assays to quantify HER1, HER2 and HER3 in formalin-fixed paraffin-embedded (FFPE) tumour tissues. After assessing the performance and precision of our assays, we quantified HER protein expression in 51 TNBC specimens, and investigated the association of their expression with relapse-free survival. Results The assays were quantitative, accurate, and robust. In TNBC specimens, HER1 levels ranged from ≈4000 to more than 2 million receptors per cell, whereas HER2 levels varied from ≈1000 to 60,000 receptors per cell. HER3 expression was very low (less than 5500 receptors per cell in all samples). Moderate HER2 expression was significantly associated with higher risk of recurrence (HR = 3.93; P = 0.003). Conclusions Our TR-FRET assays accurately quantify HER1, HER2 and HER3 in FFPE breast tumour specimens. Moderate HER2 expression may represent a novel prognostic marker in patients with TNBC.
Collapse
Affiliation(s)
- Alexandre Ho-Pun-Cheung
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | | | - Caroline Mollevi
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | | | | | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - William Jacot
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Evelyne Lopez-Crapez
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France. .,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France.
| |
Collapse
|
12
|
Pu T, Shui R, Shi J, Liang Z, Yang W, Bu H, Li Q, Zhang Z. External quality assessment (EQA) program for the immunohistochemical detection of ER, PR and Ki-67 in breast cancer: results of an interlaboratory reproducibility ring study in China. BMC Cancer 2019; 19:978. [PMID: 31640622 PMCID: PMC6805628 DOI: 10.1186/s12885-019-6210-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 09/26/2019] [Indexed: 02/05/2023] Open
Abstract
Background An External Quality Assessment (EQA) program was developed to investigate the status of estrogen receptor (ER), progesterone receptor (PR), and Ki-67 immunohistochemical (IHC) detection in breast cancer and to evaluate the reproducibility of staining and interpretation in 44 pathology laboratories in China. Methods This program was implemented through three specific steps. In study I, three revising centres defined the reference value for 11 sections. In study II, 41 participating centres (PC) stained and interpreted 11 sections by their own daily practice IHC protocols. In study III, all cases received second interpretation opinions. Results The stained slides of 44 laboratories were up to the interpretation standard. The overall interpretation concordance rate of this study was over 90%. A perfect agreement was reached among the PCs for the cases with ER+ and PR+ > 50% and Ki-67 > 30%, whereas a moderate agreement was observed for intermediate categories. After second interpretations, the misclassification rates for ER were reduced by 12.20%, for PR were reduced by 17.07%, and for Ki-67 were reduced by 4.88%. Up to 31 PCs observed a benefit from the second opinion strategy. Conclusions This project is the first EQA study performed on a national scale for assessment of ER, PR and Ki-67 status by IHC in China. In the whole IHC evaluation process, the intermediate categories were less reproducible than those with high expression rates. Second opinions can significantly improve the diagnostic agreement of pathologists’ interpretations.
Collapse
Affiliation(s)
- Tianjie Pu
- Department of Pathology, West China Hospital, Sichuan University, Guo Xue Xiang 37#, Chengdu, 610041, Sichuan, China.,Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruohong Shui
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Jie Shi
- Department of Pathology, Peking Union Medical College Hospital, China Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, China Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wentao Yang
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Guo Xue Xiang 37#, Chengdu, 610041, Sichuan, China.,Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin Li
- Department of Hospital Infection Control, Women's and Children's Hospital of Sichuan Province, Chengdu, China
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, Guo Xue Xiang 37#, Chengdu, 610041, Sichuan, China.
| | | |
Collapse
|
13
|
De Jesus J, Rosa M. Suboptimal concordance in testing and retesting results of triple-negative breast carcinoma cases among laboratories: one institution experience. Cancer Cell Int 2019; 19:263. [PMID: 31632197 PMCID: PMC6788018 DOI: 10.1186/s12935-019-0987-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Background Triple-negative breast carcinoma (TNBC) patients do not benefit from hormone- or human epidermal growth factor receptor 2- (HER2-) targeted therapies. Accurate testing is pivotal for these patients. Methods TNBC cases that were retested at our institution during a 3-year period were evaluated for concordance rates in estrogen (ER) and progesterone (PR) receptor and HER2 results. Results We found 19 (22%) discrepancies (13 major/6 minor) among 86 cases. Minor discrepancies were in HER2 changes by immunohistochemistry, and all cases were demonstrated to be negative by and dual in situ hybridization. All major discrepancies were in ER/PR expression changes. In only 2 cases the treatment changed based on repeated results and/or patient history. Conclusions Discrepancies in prognostic/predictive testing continue to be frequent despite rigorous regulations. However, since for the majority of patients in our setting, the treatment plan did not change, reflex retesting for TNBC has been deemed unnecessary in our institution.
Collapse
Affiliation(s)
- Jose De Jesus
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612 USA
| | - Marilin Rosa
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612 USA
| |
Collapse
|
14
|
Basal-like Breast Cancers: From Pathology to Biology and Back Again. Stem Cell Reports 2019; 10:1676-1686. [PMID: 29874626 PMCID: PMC6117459 DOI: 10.1016/j.stemcr.2018.04.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/17/2022] Open
Abstract
Human breast cancers referred to as "basal-like" are of interest because they lack effective therapies and their biology is poorly understood. The term basal-like derives from studies demonstrating tumor gene expression profiles that include some transcripts characteristic of the basal cells of the normal adult human mammary gland and others associated with a subset of normal luminal cells. Elucidating the mechanisms responsible for the profiles of basal-like tumors is an active area of investigation. More refined molecular analysis of patients' samples and genetic strategies to produce breast cancers de novo from defined populations of normal mouse mammary cells have served as complementary approaches to identify relevant pathway alterations. However, both also have limitations. Here, we review some of the underlying reasons, including the unifying concept that some normal luminal cells have both luminal and basal features, as well as some emerging new avenues of investigation.
Collapse
|
15
|
Wang J, Xu B. Targeted therapeutic options and future perspectives for HER2-positive breast cancer. Signal Transduct Target Ther 2019; 4:34. [PMID: 31637013 PMCID: PMC6799843 DOI: 10.1038/s41392-019-0069-2] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past 2 decades, there has been an extraordinary progress in the regimens developed for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Trastuzumab, pertuzumab, lapatinib, and ado-trastuzumab emtansine (T-DM1) are commonly recommended anti-HER2 target agents by the U.S. Food and Drug Administration. This review summarizes the most significant and updated research on clinical scenarios related to HER2-positive breast cancer management in order to revise the guidelines of everyday clinical practices. In this article, we present the data on anti-HER2 clinical research of neoadjuvant, adjuvant, and metastatic studies from the past 2 decades. We also highlight some of the promising strategies that should be critically considered. Lastly, this review lists some of the ongoing clinical trials, findings of which may soon be available.
Collapse
Affiliation(s)
- Jiani Wang
- 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, No. 17, Panjiayuannanli, Chaoyang District, 100021 Beijing, China
| | - Binghe Xu
- 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, No. 17, Panjiayuannanli, Chaoyang District, 100021 Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuannanli, Chaoyang District, 100021 Beijing, China
| |
Collapse
|
16
|
Laible M, Hartmann K, Gürtler C, Anzeneder T, Wirtz R, Weber S, Keller T, Sahin U, Rees M, Ramaswamy A. Impact of molecular subtypes on the prediction of distant recurrence in estrogen receptor (ER) positive, human epidermal growth factor receptor 2 (HER2) negative breast cancer upon five years of endocrine therapy. BMC Cancer 2019; 19:694. [PMID: 31307414 PMCID: PMC6631550 DOI: 10.1186/s12885-019-5890-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/27/2019] [Indexed: 12/24/2022] Open
Abstract
Background Current evidence suggests that patients with Luminal A early breast cancer can skip chemotherapy or extended endocrine therapy, but immunohistochemistry-based biomarker analysis for St Gallen subtyping may not be reproducible. We asked whether RT-qPCR can be used instead to address this clinical question. Methods RNA was extracted from tumor material derived from ER+/HER2- patients receiving adjuvant endocrine treatment for low-risk cancers and was semi-quantified by RT-qPCR with the MammaTyper®. St Gallen subtypes were based on the mRNA expression of ERBB2/HER2, ESR1/ER, PGR/PR and MKI67/Ki67 after dichotomizing at predefined cut-offs. Differences in distant disease-free survival (DDFS) were assessed by Kaplan Meier analysis and Cox regression. Results With a median follow up of 7.8 years, there were ten events in the group of 195 Luminal A-like tumors (5.1%) and 18 events in the remaining 127 tumors (14.1%), consisting mostly of Luminal B-like cases (N = 119). Luminal A-like had significantly better DDFS over the entire follow-up period (HR 0.35, 95% CIs 0.16–0.76, p = 0.0078) with a trend towards reduced probability of recurrences also in the late phase (> 5 years) (HR 0.20, p = 0.052). The survival advantage spanning the entire follow-up period persisted in the pN0 or pN0-N1 subgroups or after correcting for clinicopathological parameters. MKI67 alone significantly predicted for worse DDFS (HR 2.62, 95% CIs 1.24–5.56, p = 0.0088). Conclusions St Gallen Luminal A-like tumors identified by RT-qPCR display markedly low rates of distant recurrence at ten years follow-up. Patients with such tumors could be spared chemotherapy due to the obviously unfavourable benefit/toxicity ratio.
Collapse
Affiliation(s)
- Mark Laible
- BioNTech Diagnostics GmbH, An der Goldgrube 12, 55131, Mainz, Germany.
| | - Kerstin Hartmann
- BioNTech Diagnostics GmbH, An der Goldgrube 12, 55131, Mainz, Germany
| | - Claudia Gürtler
- BioNTech Diagnostics GmbH, An der Goldgrube 12, 55131, Mainz, Germany
| | | | - Ralph Wirtz
- Stratifyer Molecular Pathology GmbH, Werthmannstr. 1c, 50935, Köln, Germany
| | - Stephan Weber
- ACOMED Statistik, Fockestraße 57, 04275, Leipzig, Germany
| | - Thomas Keller
- ACOMED Statistik, Fockestraße 57, 04275, Leipzig, Germany
| | - Ugur Sahin
- BioNTech Diagnostics GmbH, An der Goldgrube 12, 55131, Mainz, Germany
| | - Martin Rees
- Gemeinschaftspraxis für Pathologie, Brustzentrum am St.-Johannes-Hospital, Amalienstraße 21, 44137, Dortmund, Germany
| | - Annette Ramaswamy
- Institut für Pathologie, Universitätsklinikum Giessen und Marburg, Baldingerstraße, 35043, Marburg, Germany
| |
Collapse
|
17
|
Hormone- and HER2-receptor assessment in 33,046 breast cancer patients: a nationwide comparison of positivity rates between pathology laboratories in the Netherlands. Breast Cancer Res Treat 2019; 175:487-497. [PMID: 30825048 PMCID: PMC6533417 DOI: 10.1007/s10549-019-05180-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/19/2019] [Indexed: 01/13/2023]
Abstract
Purpose Patient management of invasive breast cancer (IBC) is to a large extent based on hormone- and HER2-receptor assessment. High-quality, reliable receptor assessment is of key importance as false results may lead to under- or overtreatment of patients. Surveillance of case-mix adjusted positivity rates has been suggested as a tool to identify laboratories with insufficient testing assays, as this covers the whole process of receptor assessment and enables laboratories to benchmark their positivity rates against other laboratories. We studied laboratory-specific variation in hormone- and HER2 positivity rates of 33,046 breast cancer patients using real-life nationwide data. Methods All synoptic pathology reports of IBC resection-specimens, obtained between 2013 and 2016, were retrieved from the nationwide Dutch pathology registry (PALGA). Absolute and case-mix adjusted receptor positivity rates were compared to the mean national proportion and presented in funnel plots in separate analyses for estrogen (ER), progesterone (PR) and HER2. Case-mix adjustment was performed by multivariable logistic regression. Results 33,794 IBC lesions from 33,046 patients of 39 pathology laboratories were included. After case-mix adjustment, mean positivity rates were 87.2% for ER (range 80.4–94.3), 71.3% for PR (62.5–77.5%), and 9.9% for HER2 (5.5–12.7%). Overall, 14 (35.9%), 17 (43.6%) and 11 (28.2%) laboratories showed positivity rates outside the 95% confidence interval for ER, PR and HER2, respectively. Conclusion This nationwide study shows that absolute variation in hormone- and HER2-receptor positivity rates between Dutch pathology laboratories is limited. Yet, the considerable number of outlying laboratories shows that there is still need for improvement. Continuous monitoring and benchmarking of positivity rates may help to realize this.
Collapse
|
18
|
Bogen SA. A Root Cause Analysis Into the High Error Rate in Clinical Immunohistochemistry. Appl Immunohistochem Mol Morphol 2019; 27:329-338. [PMID: 30807309 PMCID: PMC6706333 DOI: 10.1097/pai.0000000000000750] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The field of Clinical Immunohistochemistry (IHC) is beset with a high error rate, an order of magnitude higher than in other types of clinical laboratory testing. Despite the many improvements in the field, these errors have persisted over the last 2 decades. The improvements over the years include an extensive literature describing the potential causes of errors and how to avoid them. More stringent regulatory guidelines have also been implemented. These measures reflect the standard view is that fixing the broad confluence of causes of error will address the problem. This review takes a different tack. To understand the high error rates, this review compares Clinical IHC laboratory practice to practices of other clinical laboratory disciplines. What aspects of laboratory testing that minimize errors in other clinical laboratory disciplines are not found in Clinical IHC? In this review, we seek to identify causal factors and underlying root causes that are unique to the field of Clinical IHC in comparison to other laboratory testing disciplines. The most important underlying root cause is the absence of traceable units of measure, international standards, calibrators that are traceable to standards, and quantitative monitoring of controls. These tools and practices (in other clinical laboratory disciplines) provide regular accurate feedback to laboratory personnel on analytic test performance.
Collapse
Affiliation(s)
- Steven A Bogen
- Department of Pathology & Laboratory Medicine, Tufts Medical Center and MDP LLC, Boston, MA
| |
Collapse
|
19
|
Concordance between Immunohistochemistry and Microarray Gene Expression Profiling for Estrogen Receptor, Progesterone Receptor, and HER2 Receptor Statuses in Breast Cancer Patients in Lebanon. Int J Breast Cancer 2018; 2018:8530318. [PMID: 30009057 PMCID: PMC6020516 DOI: 10.1155/2018/8530318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 11/17/2022] Open
Abstract
Introduction Accurate evaluation of estrogen and progesterone receptors and HER2 is critical when diagnosing invasive breast cancer for optimal treatment. The current evaluation method is via immunohistochemistry (IHC). In this paper, we compared results of ER, PR, and HER2 from microarray gene expression to IHC in 81 fresh breast cancer specimens. Methods Gene expression profiling was performed using the GeneChip Human Genome U133 Plus 2.0 arrays (Affymetrix Inc). Immunohistochemical staining for estrogen receptor, progesterone receptor, and HER2 status was performed using standard methods at a CAP-accredited pathology laboratory. Concordance rates, agreement measures, and kappa scores were calculated for both methods. Results For ER, Kappa score was 0.918 (95% CI, 0.77.3-1.000) and concordance rate was 97.5% (95% CI, 91.4%-99.7%). For PR, Kappa score was 0.652 (95% CI, 0.405-0.849) and concordance rate was 86.4% (95% CI, 77%-93%). For HER2, Kappa score was 0.709 (95% CI, 0.428-0.916) and concordance rate was 97.5% (95% CI, 91.4%-99.7%). Conclusion Our results are in line with the available evidence with the concordance rate being the lowest for the progesterone receptor. In general, microarray gene expression and IHC proved to have high concordance rates. Several factors can increase the discordance rate such as differences in sample processing.
Collapse
|
20
|
Pfitzner BM, Lederer B, Lindner J, Solbach C, Engels K, Rezai M, Dohnal K, Tesch H, Hansmann ML, Salat C, Beer M, Schneeweiss A, Sinn P, Bankfalvi A, Darb-Esfahani S, von Minckwitz G, Sinn BV, Kronenwett R, Weber K, Denkert C, Loibl S. Clinical relevance and concordance of HER2 status in local and central testing-an analysis of 1581 HER2-positive breast carcinomas over 12 years. Mod Pathol 2018; 31:607-615. [PMID: 29271415 DOI: 10.1038/modpathol.2017.171] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/11/2017] [Accepted: 10/14/2017] [Indexed: 01/07/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a central predictive biomarker in breast cancer. Inaccurate HER2 results in different laboratories could be as high as 20%. However, this statement is based on data generated more than 13 years ago and may not reflect the standards of modern diagnostic pathology. We compared central and local HER2 testing in a total of 1581 HER2-positive tumors from five clinical trials. We evaluated the clinical relevance for pathological complete response (pCR) and disease-free survival in a subgroup of 677 tumors, which received an anti-HER2 therapy. Over the period of 12 years, the discordance rate for HER2 decreased from 52.4 (GeparTrio) to 8.4% (GeparSepto). Discordance rates were significantly higher in hormone receptor (HR)-positive tumors (26.6%), compared to HR-negative tumors (16.3%, P<0.0001), which could be explained by a different distribution of HER2 mRNA levels in HR-positive and HR-negative tumors. pCR rates were significantly lower in discordant tumors (13.7%) compared to concordant tumors (32.2%, GeparQuattro and GeparQuinto, P<0.001). In survival analysis, tumors with discordant HER2 testing had a reduced overall survival (OS) in the HR-negative group (P=0.019) and a trend for improved OS in the HR-positive group (P=0.125). The performance of local HER2 testing was considerably improved over time and has reached a 92% concordance, which shows that quality initiatives in diagnostic pathology are working. Tumors with discordant HER2 testing had a reduced therapy response and different survival rates.
Collapse
Affiliation(s)
- Berit M Pfitzner
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Judith Lindner
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Knut Engels
- Center of Pathology, Cytology and Molecular Pathology Neuss, Neuss, Germany
| | - Mahdi Rezai
- European Breast Center, Luisen Hospital, Düsseldorf, Germany
| | - Karel Dohnal
- Center of Pathology and Cytology Düsseldorf, Düsseldorf, Germany
| | - Hans Tesch
- Oncology Bethanien, Frankfurt/Main, Germany
| | - Martin L Hansmann
- Institute of Pathology, University Hospital Frankfurt, Frankfurt/ Main, Germany
| | | | | | - Andreas Schneeweiss
- Division Gynecologic Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Agnes Bankfalvi
- Institute of Pathology Essen, University Hospital Essen, Essen, Germany
| | - Silvia Darb-Esfahani
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute of Pathology Spandau, Berlin, Germany
| | - Gunter von Minckwitz
- German Breast Group, Neu-Isenburg, Germany.,University Women's Hospital Frankfurt, Frankfurt/ Main, Germany
| | - Bruno V Sinn
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Carsten Denkert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany
| | - Sibylle Loibl
- German Breast Group, Neu-Isenburg, Germany.,Gynecological Hospital Sana Klinikum Offenbach, Offenbach, Germany
| |
Collapse
|
21
|
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
|
22
|
Abstract
There is a global mandate even in countries with low resources to improve the accuracy of testing biomarkers in breast cancer viz. oestrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2neu) given their critical impact in the management of patients. The steps taken include compulsory participation in an external quality assurance (EQA) programme, centralized testing, and regular performance audits for laboratories. This review addresses the status of ER/PR and HER2neu testing in India and possible reasons for the delay in development of guidelines and mandate for testing in the country. The chief cause of erroneous ER and PR testing in India continues to be easily correctable issues such as fixation and antigen retrieval, while for HER2neu testing, it is the use of low-cost non-validated antibodies and interpretative errors. These deficiencies can however, be rectified by (i) distributing the accountability and responsibility to surgeons and oncologist, (ii) certification of centres for testing in oncology, and (iii) initiation of a national EQA system (EQAS) programme that will help with economical solutions and identifying the centres of excellence and instill a system for reprimand of poorly performing laboratories.
Collapse
Affiliation(s)
- Tanuja Shet
- Department of Histopathology, Tata Memorial Centre, Mumbai, India
| |
Collapse
|
23
|
Van Bockstal M, Floris G, Galant C, Lambein K, Libbrecht L. A plea for appraisal and appreciation of immunohistochemistry in the assessment of prognostic and predictive markers in invasive breast cancer. Breast 2018; 37:52-55. [DOI: 10.1016/j.breast.2017.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022] Open
|
24
|
Chi Y, Yao L, Hu X, Huang S, Huang N, Li S, Shao Z, Wu J. The BMP inhibitor DAND5 in serum predicts poor survival in breast cancer. Oncotarget 2017; 7:14951-62. [PMID: 26908452 PMCID: PMC4924764 DOI: 10.18632/oncotarget.7498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/29/2016] [Indexed: 01/20/2023] Open
Abstract
Background & Aims Breast cancer (BC) is prevalent worldwide malignant cancer. Improvements in timely and effective diagnosis and prediction are needed. As reported, secreted DAND5 is contributed to BC metastasis. We aim to assess whether DAND5 in peripheral blood serum could determine BC-specific mortality. Methods We used immunohistochemistry staining to detect DAND5 expression in our BC tissue array including 250 samples. Angiogenesis assay and xenograft mice model were used to examine the secreted DAND5 function in BC progression. Serum concentration of DAND5 was examined by ELISA in 1730 BC patients. Kaplan-Meier and adjusted Cox proportional hazards models were utilized to analyze the prognosis and survival of BC patients. Results Tissue array results showed that positive DAND5 staining cases displayed a higher likelihood of occurrence of disease events (HR=5.494; 95% CI: 1.008-2.353; P=0.048) in univariate analysis and remained the same trend in multivariate analysis (HR=2.537; 95% CI: 1.056-6.096; P=0.037). DAND5 positive patients exerted generally poor DFS (P=0.041) in the Kaplan-Meier survival analysis. Furthermore, secreted DAND5 promoted tumor growth and angiogenesis in vitro and in vivo. In addition, positive DAND5 in BC patients serum was associated with increased risk of disease events occurrence (univariate: HR=1.58; 95% CI: 1.206-2.070; P=0.001; multivariate: HR=1.4; 95% CI: 1.003-1.954; P=0.048) in univariate and multivariate survival analysis. In the Kaplan-Meier analysis, serum DAND5 positively correlated with poor DFS (P=0.001) and DDFS (P=0.002). Conclusions DAND5 was correlated with poor survival and could serve as an easily detectable serum biomarker to predict the survival of breast cancer.
Collapse
Affiliation(s)
- Yayun Chi
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling Yao
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Hu
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Sheng Huang
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Naisi Huang
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shan Li
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhiming Shao
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiong Wu
- Department of Breast Surgery, Breast Cancer Institute, Shanghai Cancer Center, Collaborative Innovation Center of Cancer Medicine, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
25
|
Immunohistochemical Performance of Estrogen and Progesterone Receptor Antibodies on the Dako Omnis Staining Platform: Evaluation in Multicenter Studies. Appl Immunohistochem Mol Morphol 2017; 25:313-319. [PMID: 26657878 PMCID: PMC5447781 DOI: 10.1097/pai.0000000000000311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The analysis of estrogen receptor (ER) and progesterone receptor (PR) expression levels by immunohistochemistry is an important part of the initial evaluation of breast cancer and critically important in treatment planning. Anti-ERα (clone EP1) and anti-PR (clone PgR 1294) antibodies are in development for the Dako Omnis automated staining platform. These antibodies are not yet commercially available and are in performance evaluation, including the 4 international, multicenter studies reported here. For each antibody, a reproducibility study and a method comparison study was done in a randomized manner in order to test the antibodies under conditions closest to real-world user conditions. The reproducibility studies included 5 staining runs on the Dako Omnis with 20 formalin-fixed and paraffin-embedded human breast carcinoma specimens in 3 independent laboratories, and the method comparison studies included several hundred specimens stained on the Dako Omnis and on the Autostainer Link 48 platforms. Stained slides were evaluated for nuclear ER or PR expression according to American Society of Clinical Oncology/College of American Pathologists guidelines (≥1% cut-off for positive) by pathologists who were blinded from the staining method and specimen ID. For both anti-ERα (clone EP1) and anti-PR (clone PgR 1294) on the Dako Omnis, high reproducibility agreement rates were obtained on the interrun, interlaboratory, and interobserver endpoints. High concordance rates were observed between the specimens stained on the Dako Omnis platform and the Autostainer Link 48 platform. Staining quality was excellent for both anti-ERα (clone EP1) and anti-PR (clone PgR 1294) on the Dako Omnis. These results suggest that these antibodies are reliable and reproducible tools for immunohistochemistry analysis of ER and PR expression levels in formalin-fixed and paraffin-embedded breast carcinoma tissues on the Dako Omnis platform.
Collapse
|
26
|
Thomas JSJ, Provenzano E, Hiller L, Dunn J, Blenkinsop C, Grybowicz L, Vallier AL, Gounaris I, Abraham J, Hughes-Davies L, McAdam K, Chan S, Ahmad R, Hickish T, Houston S, Rea D, Caldas C, Bartlett JM, Cameron DA, Hayward RL, Earl HM. Central pathology review with two-stage quality assurance for pathological response after neoadjuvant chemotherapy in the ARTemis Trial. Mod Pathol 2017; 30:1069-1077. [PMID: 28548129 DOI: 10.1038/modpathol.2017.30] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/15/2017] [Accepted: 02/19/2017] [Indexed: 12/17/2022]
Abstract
The ARTemis Trial tested standard neoadjuvant chemotherapy±bevacizumab in the treatment of HER2-negative early breast cancer. We compare data from central pathology review with report review and also the reporting behavior of the two central pathologists. Eight hundred women with HER2-negative early invasive breast cancer were recruited. Response to chemotherapy was assessed from local pathology reports for pathological complete response in breast and axillary lymph nodes. Sections from the original core biopsy and surgical excision were centrally reviewed by one of two trial pathologists blinded to the local pathology reports. Pathologists recorded response to chemotherapy descriptively and also calculated residual cancer burden. 10% of cases were double-reported to compare the central pathologists' reporting behavior. Full sample retrieval was obtained for 681 of the 781 patients (87%) who underwent surgery within the trial and were evaluable for pathological complete response. Four hundred and eighty-three (71%) were assessed by JSJT, and 198 (29%) were assessed by EP. Residual cancer burden calculations were possible in 587/681 (86%) of the centrally reviewed patients, as 94/681 (14%) had positive sentinel nodes removed before neoadjuvant chemotherapy invalidating residual cancer burden scoring. Good concordance was found between the two pathologists for residual cancer burden classes within the 65-patient quality assurance exercise (kappa 0.63 (95% CI: 0.57-0.69)). Similar results were obtained for the between-treatment arm comparison both from the report review and the central pathology review. For pathological complete response, report review was as good as central pathology review but for minimal residual disease, report review overestimated the extent of residual disease. In the ARTemis Trial central pathology review added little in the determination of pathological complete response but had a role in evaluating low levels of residual disease. Calculation of residual cancer burden was a simple and reproducible method of quantifying response to neoadjuvant chemotherapy as demonstrated by performance comparison of the two pathologists.
Collapse
Affiliation(s)
| | - Elena Provenzano
- NIHR Cambridge Biomedical Research Centre, Addenbrookes Hospital, Cambridge, UK
- Cambridge Breast Cancer Research Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Histopathology, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Louise Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Janet Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Clare Blenkinsop
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Louise Grybowicz
- Cambridge Clinical Trials Unit-Cancer Theme, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anne-Laure Vallier
- Cambridge Clinical Trials Unit-Cancer Theme, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ioannis Gounaris
- The Queen Elizabeth Hospital King's Lynn NHS Foundation Trust, King's Lynn, UK
| | - Jean Abraham
- NIHR Cambridge Biomedical Research Centre, Addenbrookes Hospital, Cambridge, UK
- Cambridge Breast Cancer Research Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- University of Cambridge, Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | - Luke Hughes-Davies
- University of Cambridge, Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Karen McAdam
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Peterborough City Hospital, Edith Cavell Campus, Peterborough, UK
| | - Stephen Chan
- Nottingham University Hospital (City Campus), Nottingham, UK
| | | | - Tamas Hickish
- Poole Hospital NHS Foundation Trust, Poole, UK
- Bournemouth University, Poole, UK
| | - Stephen Houston
- The Royal Surrey County Hospital NHS Foundation Trust, Guildford, UK
| | - Daniel Rea
- Institute for Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Carlos Caldas
- NIHR Cambridge Biomedical Research Centre, Addenbrookes Hospital, Cambridge, UK
- Cambridge Breast Cancer Research Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- University of Cambridge, Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - John Ms Bartlett
- Ontario Institute for Cancer Research, MaRS Centre, Toronto, ON, Canada
| | - David Allan Cameron
- University of Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh, UK
| | | | - Helena Margaret Earl
- NIHR Cambridge Biomedical Research Centre, Addenbrookes Hospital, Cambridge, UK
- Cambridge Breast Cancer Research Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- University of Cambridge, Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
27
|
Hepatic Resection or Ablation for Isolated Breast Cancer Liver Metastasis: A Case-control Study With Comparison to Medically Treated Patients. Ann Surg 2017; 264:147-154. [PMID: 26445472 DOI: 10.1097/sla.0000000000001371] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To evaluate the efficacy of surgical treatment for patients with isolated breast cancer liver metastases (BCLM). BACKGROUND Single-arm retrospective studies have shown promising results associated with surgery for isolated BCLM; however, this treatment remains controversial and its role is not well-defined. METHODS A review of 2150 patients with BCLM who underwent treatment in a single institution was conducted, and 167 (8%) patients with isolated BCLM were identified. A case-control study was conducted to compare outcomes in patients with isolated BCLM who underwent surgery and/or ablation to patients who underwent conventional medical therapy. RESULTS A total of 167 patients were included (surgery/ablation: 69; medical: 98), with a median follow-up for survivors of 73 months. Patients in the surgical cohort more frequently had estrogen receptor-positive tumors and received adjuvant chemotherapy and radiotherapy for their primary breast tumor. The hepatic tumor burden was less and the interval from breast cancer diagnosis to BCLM was significantly longer (53 vs 30 months) in the surgical cohort. Patients undergoing surgical treatment had a median recurrence-free interval of 28.5 months (95% confidence interval (CI): 19-38) with 10 patients (15%) recurrence free after 5 years. There was no significant difference in overall survival (OS) between the surgical and medical cohorts (median OS: 50 vs 45 months; 5-year OS: 38% vs 39%). CONCLUSIONS Hepatic resection and/or ablation was not associated with a survival advantage. However, significant recurrence-free intervals can be accomplished with surgical treatment. Surgical intervention might be considered in highly selected patients with the goal of providing time off of systemic chemotherapy.
Collapse
|
28
|
Prat A, Navarro A, Paré L, Reguart N, Galván P, Pascual T, Martínez A, Nuciforo P, Comerma L, Alos L, Pardo N, Cedrés S, Fan C, Parker JS, Gaba L, Victoria I, Viñolas N, Vivancos A, Arance A, Felip E. Immune-Related Gene Expression Profiling After PD-1 Blockade in Non-Small Cell Lung Carcinoma, Head and Neck Squamous Cell Carcinoma, and Melanoma. Cancer Res 2017; 77:3540-3550. [PMID: 28487385 DOI: 10.1158/0008-5472.can-16-3556] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/02/2017] [Accepted: 05/02/2017] [Indexed: 11/16/2022]
Abstract
Antibody targeting of the immune checkpoint receptor PD1 produces therapeutic activity in a variety of solid tumors, but most patients exhibit partial or complete resistance to treatment for reasons that are unclear. In this study, we evaluated tumor specimens from 65 patients with melanoma, lung nonsquamous, squamous cell lung or head and neck cancers who were treated with the approved PD1-targeting antibodies pembrolizumab or nivolumab. Tumor RNA before anti-PD1 therapy was analyzed on the nCounter system using the PanCancer 730-Immune Panel, and we identified 23 immune-related genes or signatures linked to response and progression-free survival (PFS). In addition, we evaluated intra- and interbiopsy variability of PD1, PD-L1, CD8A, and CD4 mRNAs and their relationship with tumor-infiltrating lymphocytes (TIL) and PD-L1 IHC expression. Among the biomarkers examined, PD1 gene expression along with 12 signatures tracking CD8 and CD4 T-cell activation, natural killer cells, and IFN activation associated significantly with nonprogressive disease and PFS. These associations were independent of sample timing, drug used, or cancer type. TIL correlated moderately (∼0.50) with PD1 and CD8A mRNA levels and weakly (∼0.35) with CD4 and PD-L1. IHC expression of PD-L1 correlated strongly with PD-L1 (0.90), moderately with CD4 and CD8A, and weakly with PD1. Reproducibility of gene expression in intra- and interbiopsy specimens was very high (total SD <3%). Overall, our results support the hypothesis that identification of a preexisting and stable adaptive immune response as defined by mRNA expression pattern is reproducible and sufficient to predict clinical outcome, regardless of the type of cancer or the PD1 therapeutic antibody administered to patients. Cancer Res; 77(13); 3540-50. ©2017 AACR.
Collapse
Affiliation(s)
- Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain. .,Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain.,Translational Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Alejandro Navarro
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Laia Paré
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Noemí Reguart
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Patricia Galván
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain.,Translational Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Tomás Pascual
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Alex Martínez
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Laura Comerma
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Llucia Alos
- Pathology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Nuria Pardo
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Susana Cedrés
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Cheng Fan
- Department of Bioinformatics, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | - Joel S Parker
- Department of Bioinformatics, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | - Lydia Gaba
- Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Iván Victoria
- Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Nuria Viñolas
- Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Arance
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Medical Oncology Department, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron Hospital, Barcelona, Spain
| |
Collapse
|
29
|
Subgroup Analyses and Information from Clinical Trials on Breast Cancer. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
30
|
Stein RC, Dunn JA, Bartlett JMS, Campbell AF, Marshall A, Hall P, Rooshenas L, Morgan A, Poole C, Pinder SE, Cameron DA, Stallard N, Donovan JL, McCabe C, Hughes-Davies L, Makris A. OPTIMA prelim: a randomised feasibility study of personalised care in the treatment of women with early breast cancer. Health Technol Assess 2016; 20:xxiii-xxix, 1-201. [PMID: 26867046 DOI: 10.3310/hta20100] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is uncertainty about the chemotherapy sensitivity of some oestrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancers. Multiparameter assays that measure the expression of several tumour genes simultaneously have been developed to guide the use of adjuvant chemotherapy for this breast cancer subtype. The assays provide prognostic information and have been claimed to predict chemotherapy sensitivity. There is a dearth of prospective validation studies. The Optimal Personalised Treatment of early breast cancer usIng Multiparameter Analysis preliminary study (OPTIMA prelim) is the feasibility phase of a randomised controlled trial (RCT) designed to validate the use of multiparameter assay directed chemotherapy decisions in the NHS. OBJECTIVES OPTIMA prelim was designed to establish the acceptability to patients and clinicians of randomisation to test-driven treatment assignment compared with usual care and to select an assay for study in the main RCT. DESIGN Partially blinded RCT with adaptive design. SETTING Thirty-five UK hospitals. PARTICIPANTS Patients aged ≥ 40 years with surgically treated ER-positive HER2-negative primary breast cancer and with 1-9 involved axillary nodes, or, if node negative, a tumour at least 30 mm in diameter. INTERVENTIONS Randomisation between two treatment options. Option 1 was standard care consisting of chemotherapy followed by endocrine therapy. In option 2, an Oncotype DX(®) test (Genomic Health Inc., Redwood City, CA, USA) performed on the resected tumour was used to assign patients either to standard care [if 'recurrence score' (RS) was > 25] or to endocrine therapy alone (if RS was ≤ 25). Patients allocated chemotherapy were blind to their randomisation. MAIN OUTCOME MEASURES The pre-specified success criteria were recruitment of 300 patients in no longer than 2 years and, for the final 150 patients, (1) an acceptance rate of at least 40%; (2) recruitment taking no longer than 6 months; and (3) chemotherapy starting within 6 weeks of consent in at least 85% of patients. RESULTS Between September 2012 and 3 June 2014, 350 patients consented to join OPTIMA prelim and 313 were randomised; the final 150 patients were recruited in 6 months, of whom 92% assigned chemotherapy started treatment within 6 weeks. The acceptance rate for the 750 patients invited to participate was 47%. Twelve out of the 325 patients with data (3.7%, 95% confidence interval 1.7% to 5.8%) were deemed ineligible on central review of receptor status. Interviews with researchers and recordings of potential participant consultations made as part of the integral qualitative recruitment study provided insights into recruitment barriers and led to interventions designed to improve recruitment. Patient information was changed as the result of feedback from three patient focus groups. Additional multiparameter analysis was performed on 302 tumour samples. Although Oncotype DX, MammaPrint(®)/BluePrint(®) (Agendia Inc., Irvine, CA, USA), Prosigna(®) (NanoString Technologies Inc., Seattle, WA, USA), IHC4, IHC4 automated quantitative immunofluorescence (AQUA(®)) [NexCourse BreastTM (Genoptix Inc. Carlsbad, CA, USA)] and MammaTyper(®) (BioNTech Diagnostics GmbH, Mainz, Germany) categorised comparable numbers of tumours into low- or high-risk groups and/or equivalent molecular subtypes, there was only moderate agreement between tests at an individual tumour level (kappa ranges 0.33-0.60 and 0.39-0.55 for tests providing risks and subtypes, respectively). Health economics modelling showed the value of information to the NHS from further research into multiparameter testing is high irrespective of the test evaluated. Prosigna is currently the highest priority for further study. CONCLUSIONS OPTIMA prelim has achieved its aims of demonstrating that a large UK clinical trial of multiparameter assay-based selection of chemotherapy in hormone-sensitive early breast cancer is feasible. The economic analysis shows that a trial would be economically worthwhile for the NHS. Based on the outcome of the OPTIMA prelim, a large-scale RCT to evaluate the clinical effectiveness and cost-effectiveness of multiparameter assay-directed chemotherapy decisions in hormone-sensitive HER2-negative early breast would be appropriate to take place in the NHS. TRIAL REGISTRATION Current Controlled Trials ISRCTN42400492. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 20, No. 10. See the NIHR Journals Library website for further project information. The Government of Ontario funded research at the Ontario Institute for Cancer Research. Robert C Stein received additional support from the NIHR University College London Hospitals Biomedical Research Centre.
Collapse
Affiliation(s)
- Robert C Stein
- Department of Oncology, University College London Hospitals, London, UK
| | - Janet A Dunn
- Warwick Medical School, University of Warwick, Coventry, UK
| | | | - Amy F Campbell
- Warwick Medical School, University of Warwick, Coventry, UK
| | | | - Peter Hall
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Leila Rooshenas
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | | | | - Sarah E Pinder
- Research Oncology, Division of Cancer Studies, King's College London, London, UK
| | - David A Cameron
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, UK
| | - Nigel Stallard
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Jenny L Donovan
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Christopher McCabe
- Department of Emergency Medicine, University of Alberta, Edmonton, AB, Canada
| | - Luke Hughes-Davies
- Oncology Centre, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundations Trust, Cambridge, UK
| | - Andreas Makris
- Department of Clinical Oncology, Mount Vernon Cancer Centre, Mount Vernon Hospital, Northwood, UK
| | | |
Collapse
|
31
|
Orlando L, Viale G, Bria E, Lutrino ES, Sperduti I, Carbognin L, Schiavone P, Quaranta A, Fedele P, Caliolo C, Calvani N, Criscuolo M, Cinieri S. Discordance in pathology report after central pathology review: Implications for breast cancer adjuvant treatment. Breast 2016; 30:151-155. [PMID: 27750105 DOI: 10.1016/j.breast.2016.09.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 11/28/2022] Open
Abstract
AIM Pathological predictive factors are the most important markers when selecting early breast cancer adjuvant therapy. In randomized clinical trials the variability in pathology report after central pathology review is noteworthy. We evaluated the discordance rate (DR) and inter-rater agreement between local and central histopathological report and the clinical implication on treatment decision. METHODS A retrospective analysis was conducted in a series of consecutive early breast cancer tumors diagnosed by local pathologists and subsequently reviewed at the Pathology Division of European Institute of Oncology. The inter-rater agreement (k) between local and central pathology was calculated for Ki-67, grading, hormone receptors (ER/PgR) and HER2/neu. The Bland-Altman plots were derived to determine discrepancies in Ki-67, ER and PgR. DR was calculated for ER/PgR and HER2. RESULTS From 2007 to 2013, 187 pathology specimens from 10 Cancer Centers were reviewed. Substantial agreement was observed for ER (k0.612; 95% CI, 0538-0.686), PgR (k0.659; 95% CI, 0580-0.737), Ki-67 (k0.609; 95% CI, 0.534-0.684) and grading (k0.669; 95% CI, 0.569-0.769). Moderate agreement was found for HER2 (k0.546; 95% CI, 0444-0.649). DR was 9.5% (negativity to positivity) and 31.7% (positivity to negativity) for HER2 and 26.2% (negativity to positivity) and 12.5% (positivity to negativity) for ER/PgR. According to changes in Her2 and ER/PgR status, 23 (12.2%) and 33 (17.6%) systemic prescription were respectively modified. CONCLUSIONS In our retrospective analysis, central pathological review has a significant impact in the decision-making process in early breast cancer, as shown in clinical trials. Further studies are warranted to confirm these provocative results.
Collapse
Affiliation(s)
- Laura Orlando
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy.
| | - Giuseppe Viale
- Pathology Division, European Institute of Oncology, University of Milan, 20141 Milan, Italy
| | - Emilio Bria
- Medical Oncology & Breast Unit, University of Verona, Azienda Ospedaliera Universitaria Integrata, 37134 Verona, Italy
| | | | - Isabella Sperduti
- Biostatistics, Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Luisa Carbognin
- Medical Oncology & Breast Unit, University of Verona, Azienda Ospedaliera Universitaria Integrata, 37134 Verona, Italy
| | - Paola Schiavone
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy
| | | | - Palma Fedele
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy
| | - Chiara Caliolo
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy
| | - Nicola Calvani
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy
| | - Mario Criscuolo
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy
| | - Saverio Cinieri
- Medical Oncology Division & Breast Unit, 72100 Brindisi, Italy
| |
Collapse
|
32
|
Catenacci DVT, Liao WL, Zhao L, Whitcomb E, Henderson L, O'Day E, Xu P, Thyparambil S, Krizman D, Bengali K, Uzzell J, Darfler M, Cecchi F, Blackler A, Bang YJ, Hart J, Xiao SY, Lee SM, Burrows J, Hembrough T. Mass-spectrometry-based quantitation of Her2 in gastroesophageal tumor tissue: comparison to IHC and FISH. Gastric Cancer 2016; 19:1066-1079. [PMID: 26581548 PMCID: PMC4871781 DOI: 10.1007/s10120-015-0566-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/31/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Trastuzumab has shown a survival benefit in cases of Her2-positive gastroesophageal cancer (GEC). Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) currently determine eligibility for trastuzumab-based therapy. However, these low-throughput assays often produce discordant or equivocal results. METHODS We developed a targeted proteomic assay based on selected reaction monitoring mass spectrometry (SRM-MS) and quantified levels (amol/μg) of Her2-SRM protein in cell lines (n = 27) and GEC tissues (n = 139). We compared Her2-SRM protein expression with IHC/FISH, seeking to determine optimal SRM protein expression cutoffs in order to identify HER2 gene amplification. RESULTS After demonstrating assay development, precision, and stability, Her2-SRM protein measurement was observed to be highly concordant with the HER2/CEP17 ratio, particularly in a multivariate regression model adjusted for SRM expression of the covariates Met, Egfr, Her3, and HER2 heterogeneity, as well as their interactions (cell lines r (2) = 0.9842; FFPE r (2) = 0.7643). In GEC tissues, Her2-SRM protein was detected at any level in 71.2 % of cases. ROC curves demonstrated that Her2-SRM protein levels have a high specificity (100 %) at an upper-level cutoff of >750 amol/µg and sensitivity of 75 % at a lower-level cutoff of <450 amol/μg for identifying HER2 FISH-amplified tumors. An "equivocal zone" of 450-750 amol/µg of Her2-SRM protein was analogous to IHC2+ but represented fewer cases (9-16 % of cases versus 36-41 %). CONCLUSIONS Compared to IHC, targeted SRM-Her2 proteomics provided more objective and quantitative Her2 expression with excellent HER2/CEP17 FISH correlation and fewer equivocal cases. Along with its multiplex capability for other relevant oncoproteins, these results demonstrate a refined HER2 protein expression assay for clinical application.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA.
| | - Wei-Li Liao
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Lei Zhao
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Emma Whitcomb
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Les Henderson
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Emily O'Day
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Peng Xu
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Sheeno Thyparambil
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - David Krizman
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Kathleen Bengali
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | | | - Marlene Darfler
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Fabiola Cecchi
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Adele Blackler
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, Korea
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Sang Mee Lee
- Department of Public Health Studies, University of Chicago, Chicago, IL, USA
| | - Jon Burrows
- OncoPlex Diagnostics Inc., Rockville, MD, USA
| | - Todd Hembrough
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| |
Collapse
|
33
|
Kotoula V, Lakis S, Vlachos IS, Giannoulatou E, Zagouri F, Alexopoulou Z, Gogas H, Pectasides D, Aravantinos G, Efstratiou I, Pentheroudakis G, Papadopoulou K, Chatzopoulos K, Papakostas P, Sotiropoulou M, Nicolaou I, Razis E, Psyrri A, Kosmidis P, Papadimitriou C, Fountzilas G. Tumor Infiltrating Lymphocytes Affect the Outcome of Patients with Operable Triple-Negative Breast Cancer in Combination with Mutated Amino Acid Classes. PLoS One 2016; 11:e0163138. [PMID: 27685159 PMCID: PMC5042538 DOI: 10.1371/journal.pone.0163138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/02/2016] [Indexed: 12/26/2022] Open
Abstract
Background Stromal tumor infiltrating lymphocytes (TILs) density is an outcome predictor in triple-negative breast cancer (TNBC). Herein we asked whether TILs are related to coding mutation load and to the chemical class of the resulting mutated amino acids, i.e., charged, polar, and hydrophobic mutations. Methods We examined paraffin tumors from TNBC patients who had been treated with adjuvant chemotherapy mostly within clinical trials (training cohort, N = 133; validation, N = 190) for phenotype concordance; TILs density; mutation load and types. Results Concordance of TNBC phenotypes was 42.1% upon local / central, and 72% upon central / central pathology assessment. TILs were not associated with mutation load, type and class of mutated amino acids. Polar and charged mutation patterns differed between TP53 and PIK3CA (p<0.001). Hydrophobic mutations predicted for early relapse in patients with high nodal burden and <50% TILs tumors (training: HR 3.03, 95%CI 1.11–8.29, p = 0.031; validation: HR 2.90, 95%CI 0.97–8.70, p = 0.057), especially if compared to patients with >50% TILs tumors (training p = 0.003; validation p = 0.015). Conclusions TILs density is unrelated to mutation load in TNBC, which may be regarded as an unstable phenotype. If further validated, hydrophobic mutations along with TILs density may help identifying TNBC patients in higher risk for relapse.
Collapse
Affiliation(s)
- Vassiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
- * E-mail:
| | - Sotiris Lakis
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis S. Vlachos
- Molecular Diagnostics Laboratory, INRASTES, NCSR 'Demokritos', Athens, Greece
- DIANA-Lab, Department of Computer and Communication Engineering, University of Thessaly, Volos, Greece
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- The University of New South Wales, New South Wales, Australia
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Zoi Alexopoulou
- Department of Biostatistics, Health Data Specialists Ltd, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, Greece
| | - Gerasimos Aravantinos
- Second Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| | | | | | - Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriakos Chatzopoulos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Irene Nicolaou
- Department of Histopathology, Agii Anagriri Cancer Hospital, Athens, Greece
| | - Evangelia Razis
- Third Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Amanda Psyrri
- Division of Oncology, Second Department of Internal Medicine, Attikon University Hospital, Athens, Greece
| | - Paris Kosmidis
- Second Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Christos Papadimitriou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
- Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
34
|
Laible M, Schlombs K, Kaiser K, Veltrup E, Herlein S, Lakis S, Stöhr R, Eidt S, Hartmann A, Wirtz RM, Sahin U. Technical validation of an RT-qPCR in vitro diagnostic test system for the determination of breast cancer molecular subtypes by quantification of ERBB2, ESR1, PGR and MKI67 mRNA levels from formalin-fixed paraffin-embedded breast tumor specimens. BMC Cancer 2016; 16:398. [PMID: 27389414 PMCID: PMC4936300 DOI: 10.1186/s12885-016-2476-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/30/2016] [Indexed: 01/03/2023] Open
Abstract
Background MammaTyper is a novel CE-marked in vitro diagnostic RT-qPCR assay which assigns routinely processed breast cancer specimens into the molecular subtypes Luminal A-like, Luminal B-like (HER2 positive or negative), HER2 positive (non-luminal) and Triple negative (ductal) according to the mRNA expression of ERBB2, ESR1, PGR and MKI67 and the St Gallen consensus surrogate clinical definition. Until now and regarding formalin-fixed, paraffin-embedded material (FFPE), this has been a task mostly accomplished by immunohistochemistry (IHC). However the discrepancy rates of IHC for the four breast cancer biomarkers are frequently under debate, especially for Ki-67 which carries the highest degree of inter- and even intra-observer variability. Herein we describe a series of studies in FFPE specimens which aim to fully validate the analytical performance of the MammaTyper assay, including the site to site reproducibility of the individual marker measurements. Methods Tumor RNA was extracted with the novel RNXtract RNA extraction kit. Synthetic RNA was used to assess the sensitivity of the RNXtract kit. DNA and RNA specific qPCR assays were used so as to determine analyte specificity of RNXtract. For the assessment of limit of blank, limit of detection, analytical measurement range and PCR efficiency of the MammaTyper kit serial dilutions of samples were used. Analytical precision studies of MammaTyper were built around two different real time PCR platforms and involved breast tumor samples belonging to different subtypes analyzed across multiple sites and under various stipulated conditions. The MammaTyper assay robustness was tested against RNA input variations, alternative extraction methods and tumor cell content. Results Individual assays were linear up to at least 32.33 and 33.56 Cqs (quantification cycles) for the two qPCR platforms tested. PCR efficiency ranged from 99 to 109 %. In qPCR platform 1, estimates for assay specific inter-site standard deviations (SD) were between 0.14 and 0.20 Cqs accompanied by >94 % concordant single marker assignments for all four markers. In platform 2, the inter-site SD estimates were between 0.40 and 0.66 Cqs while the concordance for single marker assignments was >94 % for all four markers. The agreement reached between the two qPCR systems located in one site was 100 % for ERBB2, 96.9 % for ESR1, 97.2 % for PGR and 98.6 % for MKI67. RT-qPCR for individual markers was stable up to a 64-fold dilution for a typical clinical sample. There was no change in assay performance detected at the level of individual markers or subtypes after using different RNA isolation methods. The presence of up to 80 % of surrounding non-tumor tissue including in situ carcinoma did not affect the assay output. Sixteen out of 20 RNXtract eluates yielded more than 50 ng/μl of RNA (average RNA output: 233 ng/μl), whereas DNA contamination per sample was restricted to less than 15 ng/μl. Median recovery rate of RNA extraction was 91.0 %. Conclusions In this study the performance characteristics of MammaTyper were successfully validated. The various sources of analytical perturbations resulted in negligible variations in individual marker assessments. Therefore, MammaTyper may serve as a technical improvement to current standards for decentralized FFPE-based routine assessment of the commonly used breast cancer biomarkers and for molecular subtyping of breast cancer specimens. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2476-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Elke Veltrup
- STRATIFYER Molecular Pathology GmbH, Cologne, Germany
| | - Stefanie Herlein
- Institute of Pathology, University of Erlangen, Erlangen, Germany
| | - Sotiris Lakis
- STRATIFYER Molecular Pathology GmbH, Cologne, Germany
| | - Robert Stöhr
- Institute of Pathology, University of Erlangen, Erlangen, Germany
| | - Sebastian Eidt
- Institut für Pathologie am St. Elisabeth-Krankenhaus, Cologne, Germany
| | - Arndt Hartmann
- Institute of Pathology, University of Erlangen, Erlangen, Germany
| | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH, Cologne, Germany
| | - Ugur Sahin
- BioNTech Diagnostics GmbH, Mainz, Germany
| |
Collapse
|
35
|
Wesseling J, Tinterri C, Sapino A, Zanconati F, Lutke-Holzik M, Nguyen B, Deck KB, Querzoli P, Perin T, Giardina C, Seitz G, Guinebretière JM, Barone J, Dekker L, de Snoo F, Stork-Sloots L, Roepman P, Watanabe T, Cusumano P. An international study comparing conventional versus mRNA level testing (TargetPrint) for ER, PR, and HER2 status of breast cancer. Virchows Arch 2016; 469:297-304. [DOI: 10.1007/s00428-016-1979-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 05/30/2016] [Accepted: 06/27/2016] [Indexed: 01/05/2023]
|
36
|
Allott EH, Geradts J, Sun X, Cohen SM, Zirpoli GR, Khoury T, Bshara W, Chen M, Sherman ME, Palmer JR, Ambrosone CB, Olshan AF, Troester MA. Intratumoral heterogeneity as a source of discordance in breast cancer biomarker classification. Breast Cancer Res 2016; 18:68. [PMID: 27349894 PMCID: PMC4924300 DOI: 10.1186/s13058-016-0725-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/27/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Spatial heterogeneity in biomarker expression may impact breast cancer classification. The aims of this study were to estimate the frequency of spatial heterogeneity in biomarker expression within tumors, to identify technical and biological factors contributing to spatial heterogeneity, and to examine the impact of discordant biomarker status within tumors on clinical record agreement. METHODS Tissue microarrays (TMAs) were constructed using two to four cores (1.0 mm) for each of 1085 invasive breast cancers from the Carolina Breast Cancer Study, which is part of the AMBER Consortium. Immunohistochemical staining for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) was quantified using automated digital imaging analysis. The biomarker status for each core and for each case was assigned using clinical thresholds. Cases with core-to-core biomarker discordance were manually reviewed to distinguish intratumoral biomarker heterogeneity from misclassification of biomarker status by the automated algorithm. The impact of core-to-core biomarker discordance on case-level agreement between TMAs and the clinical record was evaluated. RESULTS On the basis of automated analysis, discordant biomarker status between TMA cores occurred in 9 %, 16 %, and 18 % of cases for ER, PR, and HER2, respectively. Misclassification of benign epithelium and/or ductal carcinoma in situ as invasive carcinoma by the automated algorithm was implicated in discordance among cores. However, manual review of discordant cases confirmed spatial heterogeneity as a source of discordant biomarker status between cores in 2 %, 7 %, and 8 % of cases for ER, PR, and HER2, respectively. Overall, agreement between TMA and clinical record was high for ER (94 %), PR (89 %), and HER2 (88 %), but it was reduced in cases with core-to-core discordance (agreement 70 % for ER, 61 % for PR, and 57 % for HER2). CONCLUSIONS Intratumoral biomarker heterogeneity may impact breast cancer classification accuracy, with implications for clinical management. Both manually confirmed biomarker heterogeneity and misclassification of biomarker status by automated image analysis contribute to discordant biomarker status between TMA cores. Given that manually confirmed heterogeneity is uncommon (<10 % of cases), large studies are needed to study the impact of heterogeneous biomarker expression on breast cancer classification and outcomes.
Collapse
Affiliation(s)
- Emma H Allott
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Epidemiology, University of North Carolina at Chapel Hill, 135 Dauer Drive, CB 7435, Chapel Hill, NC, 27599, USA
| | - Joseph Geradts
- Department of Pathology, Brigham & Women's Hospital, Boston, MA, USA
| | - Xuezheng Sun
- Department of Epidemiology, University of North Carolina at Chapel Hill, 135 Dauer Drive, CB 7435, Chapel Hill, NC, 27599, USA
| | - Stephanie M Cohen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gary R Zirpoli
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Thaer Khoury
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Mengjie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark E Sherman
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Julie R Palmer
- Slone Epidemiology Center, Boston University, Boston, MA, USA
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Andrew F Olshan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Epidemiology, University of North Carolina at Chapel Hill, 135 Dauer Drive, CB 7435, Chapel Hill, NC, 27599, USA
| | - Melissa A Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Department of Epidemiology, University of North Carolina at Chapel Hill, 135 Dauer Drive, CB 7435, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
37
|
Abramovitz M, Williams C, Loibl S, Leyland-Jones B. Dual Blockade of HER-2 Provides a Greater Magnitude of Benefit in Patients With Hormone-Negative Versus Hormone-Positive Breast Cancer. Clin Breast Cancer 2016; 16:444-455. [PMID: 27435628 DOI: 10.1016/j.clbc.2016.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/12/2016] [Accepted: 06/05/2016] [Indexed: 10/21/2022]
Abstract
The dual small molecule tyrosine kinase inhibitor lapatinib blocks both human epidermal growth factor receptor (HER-1) and human epidermal growth factor receptor 2 (HER-2) tyrosine kinase activity by binding reversibly to the ATP-binding site of the receptor's intracellular domain. Lapatinib, in combination with capecitabine, has been approved in 2007 for the treatment of patients with advanced HER-2+ breast cancer upon progressive disease following standard chemotherapy. Approval was also extended to the treatment of postmenopausal women with advanced hormone receptor (HR)-positive and HER-2-positive breast cancer in 2010. More recently, clinical trials that have investigated the efficacy of dual HER-2 blockade in both the metastatic and neoadjuvant breast cancer settings. For example, in 2013 the European Medicines Agency approved the combination of lapatinib and trastuzumab in HER-2+/HR- patients. We review the efficacy results from dual HER-2 blockade studies and present new post hoc analysis efficacy data according to HR status. We show that dual blockade of HER-2 appears to provide a greater magnitude of benefit in the HR- versus the HR+ subgroup of patients. Finally, we examine the potential of molecularly subtyping HER-2+ tumors using the PAM50 test as a predictor of response to treatment with the combination of trastuzumab and lapatinib.
Collapse
|
38
|
Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol 2016; 13:674-690. [PMID: 27184417 DOI: 10.1038/nrclinonc.2016.66] [Citation(s) in RCA: 1763] [Impact Index Per Article: 220.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chemotherapy is the primary established systemic treatment for patients with triple-negative breast cancer (TNBC) in both the early and advanced-stages of the disease. The lack of targeted therapies and the poor prognosis of patients with TNBC have fostered a major effort to discover actionable molecular targets to treat patients with these tumours. Massively parallel sequencing and other 'omics' technologies have revealed an unexpected level of heterogeneity of TNBCs and have led to the identification of potentially actionable molecular features in some TNBCs, such as germline BRCA1/2 mutations or 'BRCAness', the presence of the androgen receptor, and several rare genomic alterations. Whether these alterations are molecular 'drivers', however, has not been clearly established. A subgroup of TNBCs shows a high degree of tumour-infiltrating lymphocytes that also correlates with a lower risk of disease relapse and a higher likelihood of benefit from chemotherapy. Proof-of-principle studies with immune-checkpoint inhibitors in advanced-stage TNBC have yielded promising results, indicating the potential benefit of immunotherapy for patients with TNBC. In this Review, we discuss the most relevant molecular findings in TNBC from the past decade and the most promising therapeutic opportunities derived from these data.
Collapse
|
39
|
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
|
40
|
Gluz O, Nitz UA, Christgen M, Kates RE, Shak S, Clemens M, Kraemer S, Aktas B, Kuemmel S, Reimer T, Kusche M, Heyl V, Lorenz-Salehi F, Just M, Hofmann D, Degenhardt T, Liedtke C, Svedman C, Wuerstlein R, Kreipe HH, Harbeck N. West German Study Group Phase III PlanB Trial: First Prospective Outcome Data for the 21-Gene Recurrence Score Assay and Concordance of Prognostic Markers by Central and Local Pathology Assessment. J Clin Oncol 2016; 34:2341-9. [PMID: 26926676 DOI: 10.1200/jco.2015.63.5383] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The 21-gene Recurrence Score (RS) assay is a validated prognostic/predictive tool in early hormone receptor-positive breast cancer (BC); however, only a few prospective outcome results have been available so far. In the phase III PlanB trial, RS was prospectively used to define a subset of patients who received only endocrine therapy. We present 3-year outcome data and concordance analysis (among biomarkers/RS). PATIENTS AND METHODS Central tumor bank was established prospectively from PlanB (intermediate and high-risk, locally human epidermal growth factor receptor 2-negative BC). After an early amendment, HR-positive, pN0-1 patients with RS ≤ 11 were recommended to omit chemotherapy. RESULTS From 2009 to 2011, PlanB enrolled 3,198 patients with a median age of 56 years; 41.1% had node-positive and 32.5% grade 3 disease. In 348 patients (15.3%), chemotherapy was omitted based on RS ≤ 11. After 35 months median follow-up, 3-year disease-free survival in patients with RS ≤ 11 and endocrine therapy alone was 98% versus 92% and 98% in RS > 25 and RS 12 to 25 in chemotherapy-treated patients, respectively. Nodal status, central and local grade, the Ki-67 protein encoded by the MKI67 gene, estrogen receptor, progesterone receptor, tumor size, and RS were univariate prognostic factors for disease-free survival; only nodal status, both central and local grade, and RS were independent multivariate factors. Histologic grade was discordant between central and local laboratories in 44%. RS was positively but moderately correlated with the Ki-67 protein encoded by the MKI67 gene and grade and negatively correlated with progesterone receptor and estrogen receptor. CONCLUSION In this prospective trial, patients with enhanced clinical risk and omitted chemotherapy on the basis of RS ≤ 11 had excellent 3-year survival. The substantial discordance observed between traditional prognostic markers and RS emphasizes the need for standardized assessment and supports the potential integration of standardized, well-validated genomic assays such as RS with clinicopathologic prognostic factors for chemotherapy indication in early hormone receptor-positive BC.
Collapse
Affiliation(s)
- Oleg Gluz
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA.
| | - Ulrike A Nitz
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Matthias Christgen
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Ronald E Kates
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Steven Shak
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Michael Clemens
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Stefan Kraemer
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Bahriye Aktas
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Sherko Kuemmel
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Toralf Reimer
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Manfred Kusche
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Volker Heyl
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Fatemeh Lorenz-Salehi
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Marianne Just
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Daniel Hofmann
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Tom Degenhardt
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Cornelia Liedtke
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Christer Svedman
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Rachel Wuerstlein
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Hans H Kreipe
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| | - Nadia Harbeck
- Oleg Gluz, Ulrike A. Nitz, Ronald E. Kates, Daniel Hofmann, Cornelia Liedtke, Rachel Wuerstlein, and Nadia Harbeck, West German Study Group; Oleg Gluz and Ulrike A. Nitz, Evangelical Hospital Bethesda, Moenchengladbach; Matthias Christgen and Hans H. Kreipe, Medical School Hannover, Hannover; Michael Clemens, Mutterhaus der Borromäerinnen, Trier; Stefan Kraemer, University Clinics Cologne, Cologne; Bahriye Aktas, University Clinics Essen; Sherko Kuemmel, Clinics Essen-Mitte, Essen; Toralf Reimer, Clinics Suedstadt, Rostock; Manfred Kusche, Marienhospital Aachen, Aachen; Volker Heyl, Asklepios Paulinen Clinics; Fatemeh Lorenz-Salehi, Dr Horst-Schmidt Clinics, Wiesbaden; Marianne Just, Oncologic Practice, Bielefeld; Tom Degenhardt, Rachel Wuerstlein, and Nadia Harbeck, University of Munich, Munich; Cornelia Liedtke, University Hospital Schleswig-Holstein Campus Luebeck, Luebeck, Germany; and Steven Shak and Christer Svedman, Genomic Health, Redwood City, CA
| |
Collapse
|
41
|
Han HS, Magliocco AM. Molecular Testing and the Pathologist's Role in Clinical Trials of Breast Cancer. Clin Breast Cancer 2016; 16:166-79. [PMID: 27103546 DOI: 10.1016/j.clbc.2016.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 01/11/2016] [Accepted: 02/03/2016] [Indexed: 01/26/2023]
Abstract
Molecular characterization of breast cancer is pivotal for identifying new molecular targets and determining the appropriate treatment choices. Advances in molecular profiling technology have given greater insight into this heterogeneous disease, over and above hormone receptor and human epidermal growth factor receptor 2 status. Agents targeting recently characterized molecular biomarkers are under clinical development; the success of these targeted agents is likely to depend on identifying the patient population most likely to benefit. Therefore, clinical trials of breast cancer often require prescreening for, or stratification by, relevant molecular markers or exploratory analyses of biomarkers that can predict or monitor the response to treatment. Consequently, the role of the pathologist has become increasingly important. The key considerations for pathologists include tissue availability, ownership of archival tissue, type of diagnostic/biomarker test required, method of sample processing, concordance between different tests and testing centers, and tumor heterogeneity. In the present review, we explore how pathology is used in current clinical trials of breast cancer and describe the various technologies available for molecular testing. Furthermore, the factors required for the successful application of pathology in clinical trials of breast cancer and the issues that can arise and how these can be circumvented are discussed.
Collapse
Affiliation(s)
- Hyo Sook Han
- Department of Women's Oncology, Moffitt Cancer Center, Tampa, FL
| | | |
Collapse
|
42
|
Allott EH, Cohen SM, Geradts J, Sun X, Khoury T, Bshara W, Zirpoli GR, Miller CR, Hwang H, Thorne LB, O'Connor S, Tse CK, Bell MB, Hu Z, Li Y, Kirk EL, Bethea TN, Perou CM, Palmer JR, Ambrosone CB, Olshan AF, Troester MA. Performance of Three-Biomarker Immunohistochemistry for Intrinsic Breast Cancer Subtyping in the AMBER Consortium. Cancer Epidemiol Biomarkers Prev 2015; 25:470-8. [PMID: 26711328 DOI: 10.1158/1055-9965.epi-15-0874] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/09/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Classification of breast cancer into intrinsic subtypes has clinical and epidemiologic importance. To examine accuracy of IHC-based methods for identifying intrinsic subtypes, a three-biomarker IHC panel was compared with the clinical record and RNA-based intrinsic (PAM50) subtypes. METHODS Automated scoring of estrogen receptor (ER), progesterone receptor (PR), and HER2 was performed on IHC-stained tissue microarrays comprising 1,920 cases from the African American Breast Cancer Epidemiology and Risk (AMBER) consortium. Multiple cores (1-6/case) were collapsed to classify cases, and automated scoring was compared with the clinical record and to RNA-based subtyping. RESULTS Automated analysis of the three-biomarker IHC panel produced high agreement with the clinical record (93% for ER and HER2, and 88% for PR). Cases with low tumor cellularity and smaller core size had reduced agreement with the clinical record. IHC-based definitions had high agreement with the clinical record regardless of hormone receptor positivity threshold (1% vs. 10%), but a 10% threshold produced highest agreement with RNA-based intrinsic subtypes. Using a 10% threshold, IHC-based definitions identified the basal-like intrinsic subtype with high sensitivity (86%), although sensitivity was lower for luminal A, luminal B, and HER2-enriched subtypes (76%, 40%, and 37%, respectively). CONCLUSION Three-biomarker IHC-based subtyping has reasonable accuracy for distinguishing basal-like from nonbasal-like, although additional biomarkers are required for accurate classification of luminal A, luminal B, and HER2-enriched cancers. IMPACT Epidemiologic studies relying on three-biomarker IHC status for subtype classification should use caution when distinguishing luminal A from luminal B and when interpreting findings for HER2-enriched cancers.
Collapse
Affiliation(s)
- Emma H Allott
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie M Cohen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Translational Pathology Laboratory, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph Geradts
- Department of Pathology, Duke University, Durham, North Carolina
| | - Xuezheng Sun
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Thaer Khoury
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Wiam Bshara
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Gary R Zirpoli
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - C Ryan Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Translational Pathology Laboratory, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Helena Hwang
- Department of Pathology, University of Texas Southwestern, Dallas, Texas
| | - Leigh B Thorne
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Siobhan O'Connor
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chiu-Kit Tse
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mary B Bell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yan Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Erin L Kirk
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Traci N Bethea
- Slone Epidemiology Center, Boston University, Boston, Massachusetts
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Julie R Palmer
- Slone Epidemiology Center, Boston University, Boston, Massachusetts
| | | | - Andrew F Olshan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melissa A Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
43
|
Hicks DG, Fitzgibbons P, Hammond E. Core vs Breast Resection Specimen: Does It Make a Difference for HER2 Results? Am J Clin Pathol 2015; 144:533-5. [PMID: 26386073 DOI: 10.1309/ajcpp28eqmvaejih] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- David G. Hicks
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | | | - Elizabeth Hammond
- Department of Pathology, University of Utah School of Medicine and Intermountain Healthcare, Salt Lake City
| |
Collapse
|
44
|
Abstract
Gene-expression profiling has had a considerable impact on our understanding of breast cancer biology. During the last 15 years, 5 intrinsic molecular subtypes of breast cancer (Luminal A, Luminal B, HER2-enriched, Basal-like and Claudin-low) have been identified and intensively studied. In this review, we will focus on the current and future clinical implications of the intrinsic molecular subtypes beyond the current pathological-based classification endorsed by the 2013 St. Gallen Consensus Recommendations. Within hormone receptor-positive and HER2-negative early breast cancer, the Luminal A and B subtypes predict 10-year outcome regardless of systemic treatment administered as well as residual risk of distant recurrence after 5 years of endocrine therapy. Within clinically HER2-positive disease, the 4 main intrinsic subtypes can be identified and dominate the biological and clinical phenotype. From a clinical perspective, patients with HER2+/HER2-enriched disease seem to benefit the most from neoadjuvant trastuzumab, or dual HER2 blockade with trastuzumab/lapatinib, in combination with chemotherapy, and patients with HER2+/Luminal A disease seem to have a relative better outcome compared to the other subtypes. Finally, within triple-negative breast cancer (TNBC), the Basal-like disease predominates (70-80%) and, from a biological perspective, should be considered a cancer-type by itself. Importantly, the distinction between Basal-like versus non-Basal-like within TNBC might predict survival following (neo)adjvuvant multi-agent chemotherapy, bevacizumab benefit in the neoadjuvant setting (CALGB40603), and docetaxel vs. carboplatin benefit in first-line metastatic disease (TNT study). Overall, this data suggests that intrinsic molecular profiling provides clinically relevant information beyond current pathology-based classifications.
Collapse
|
45
|
Guarneri V, Dieci MV, Frassoldati A, Maiorana A, Ficarra G, Bettelli S, Tagliafico E, Bicciato S, Generali DG, Cagossi K, Bisagni G, Sarti S, Musolino A, Ellis C, Crescenzo R, Conte P. Prospective Biomarker Analysis of the Randomized CHER-LOB Study Evaluating the Dual Anti-HER2 Treatment With Trastuzumab and Lapatinib Plus Chemotherapy as Neoadjuvant Therapy for HER2-Positive Breast Cancer. Oncologist 2015; 20:1001-10. [PMID: 26245675 DOI: 10.1634/theoncologist.2015-0138] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/26/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The CHER-LOB randomized phase II study showed that the combination of lapatinib and trastuzumab plus chemotherapy increases the pathologic complete remission (pCR) rate compared with chemotherapy plus either trastuzumab or lapatinib. A biomarker program was prospectively planned to identify potential predictors of sensitivity to different treatments and to evaluate treatment effect on tumor biomarkers. MATERIALS AND METHODS Overall, 121 breast cancer patients positive for human epidermal growth factor 2 (HER2) were randomly assigned to neoadjuvant chemotherapy plus trastuzumab, lapatinib, or both trastuzumab and lapatinib. Pre- and post-treatment samples were centrally evaluated for HER2, p95-HER2, phosphorylated AKT (pAKT), phosphatase and tensin homolog, Ki67, apoptosis, and PIK3CA mutations. Fresh-frozen tissue samples were collected for genomic analyses. RESULTS A mutation in PIK3CA exon 20 or 9 was documented in 20% of cases. Overall, the pCR rates were similar in PIK3CA wild-type and PIK3CA-mutated patients (33.3% vs. 22.7%; p = .323). For patients receiving trastuzumab plus lapatinib, the probability of pCR was higher in PIK3CA wild-type tumors (48.4% vs. 12.5%; p = .06). Ki67, pAKT, and apoptosis measured on the residual disease were significantly reduced from baseline. The degree of Ki67 inhibition was significantly higher in patients receiving the dual anti-HER2 blockade. The integrated analysis of gene expression and copy number data demonstrated that a 50-gene signature specifically predicted the lapatinib-induced pCR. CONCLUSION PIK3CA mutations seem to identify patients who are less likely to benefit from dual anti-HER2 inhibition. p95-HER2 and markers of phosphoinositide 3-kinase pathway deregulation are not confirmed as markers of different sensitivity to trastuzumab or lapatinib. IMPLICATIONS FOR PRACTICE HER2 is currently the only validated marker to select breast cancer patients for anti-HER2 treatment; however, it is becoming evident that HER2-positive breast cancer is a heterogeneous disease. In addition, more and more new anti-HER2 treatments are becoming available. There is a need to identify markers of sensitivity to different treatments to move in the direction of treatment personalization. This study identified PIK3CA mutations as a potential predictive marker of resistance to dual anti-HER2 treatment that should be further studied in breast cancer.
Collapse
Affiliation(s)
- Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Antonio Frassoldati
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Antonino Maiorana
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Guido Ficarra
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Stefania Bettelli
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Enrico Tagliafico
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Silvio Bicciato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Daniele Giulio Generali
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Katia Cagossi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Giancarlo Bisagni
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Samanta Sarti
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Antonino Musolino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Catherine Ellis
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Rocco Crescenzo
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - PierFranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Division of Medical Oncology 2, Istituto Oncologico Veneto Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Italy; Division of Oncology, University Hospital, Ferrara, Italy; Division of Pathology, Modena University Hospital, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Unità Operativa Multidisciplinare di Patologia Mammaria, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy; Division of Medical Oncology, Ramazzini Hospital, Carpi, Italy; Department of Medical Oncology, Azienda Ospedaliera ASMN, IRCCS, Reggio Emilia, Italy; Division of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy; Division of Medical Oncology, University Hospital, Parma, Italy; GlaxoSmithKline, Collegeville, Pennsylvania, USA
| |
Collapse
|
46
|
Witzel I, Müller V. Targeted Therapies in Breast Cancer: New Approaches and Old Challenges. Breast Care (Basel) 2015; 10:157-8. [PMID: 26557819 DOI: 10.1159/000431067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Isabell Witzel
- Dept. of Gynecology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Müller
- Dept. of Gynecology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
47
|
Current HER2 Testing Recommendations and Clinical Relevance as a Predictor of Response to Targeted Therapy. Clin Breast Cancer 2015; 15:171-80. [DOI: 10.1016/j.clbc.2014.11.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 11/18/2022]
|
48
|
Nuciforo P, Radosevic-Robin N, Ng T, Scaltriti M. Quantification of HER family receptors in breast cancer. Breast Cancer Res 2015; 17:53. [PMID: 25887735 PMCID: PMC4389676 DOI: 10.1186/s13058-015-0561-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The clinical success of trastuzumab in breast cancer taught us that appropriate tumor evaluation is mandatory for the correct identification of patients eligible for targeted therapies. Although HER2 protein expression by immunohistochemistry (IHC) and gene amplification by fluorescence in situ hybridization (FISH) assays are routinely used to select patients to receive trastuzumab, both assays only partially predict response to the drug. In the case of epidermal growth factor receptor (EGFR), the link between the presence of the receptor or its amplification and response to anti-EGFR therapies could not be demonstrated. Even less is known for HER3 and HER4, mainly due to lack of robust and validated assays detecting these proteins. It is becoming evident that, besides FISH and IHC, we need better assays to quantify HER receptors and categorize the patients for individualized treatments. Here, we present the current available methodologies to measure HER family receptors and discuss the clinical implications of target quantification.
Collapse
Affiliation(s)
- Paolo Nuciforo
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain.
- Universitat Autònoma de Barcelona, Barcelona, 08035, Spain.
| | - Nina Radosevic-Robin
- ERTICa Research Group, University of Auvergne EA4677, 63000, Clermont-Ferrand, France.
- Biopathology, Jean Perrin Comprehensive Cancer Center, 58 rue Montalembert, 63011, Clermont-Ferrand, France.
| | - Tony Ng
- Richard Dimbleby Department of Cancer Research, Randall Division of Cell and Molecular Biophysics and Division of Cancer Studies, King's College London, London, SE1 1UL, UK.
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, WC1E 6DD, UK.
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital King's College London School of Medicine, London, SE1 9RT, UK.
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 20, New York, NY, 10065, USA.
| |
Collapse
|
49
|
Goel S, Krop IE. Deciphering the role of phosphatidylinositol 3-kinase mutations in human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2015; 33:1407-9. [PMID: 25800754 DOI: 10.1200/jco.2014.60.0742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Shom Goel
- Dana-Farber Cancer Institute, Boston, MA
| | - Ian E Krop
- Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
50
|
O'Reilly EA, Gubbins L, Sharma S, Tully R, Guang MHZ, Weiner-Gorzel K, McCaffrey J, Harrison M, Furlong F, Kell M, McCann A. The fate of chemoresistance in triple negative breast cancer (TNBC). BBA CLINICAL 2015; 3:257-75. [PMID: 26676166 PMCID: PMC4661576 DOI: 10.1016/j.bbacli.2015.03.003] [Citation(s) in RCA: 272] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Treatment options for women presenting with triple negative breast cancer (TNBC) are limited due to the lack of a therapeutic target and as a result, are managed with standard chemotherapy such as paclitaxel (Taxol®). Following chemotherapy, the ideal tumour response is apoptotic cell death. Post-chemotherapy, cells can maintain viability by undergoing viable cellular responses such as cellular senescence, generating secretomes which can directly enhance the malignant phenotype. SCOPE OF REVIEW How tumour cells retain viability in response to chemotherapeutic engagement is discussed. In addition we discuss the implications of this retained tumour cell viability in the context of the development of recurrent and metastatic TNBC disease. Current adjuvant and neo-adjuvant treatments available and the novel potential therapies that are being researched are also reviewed. MAJOR CONCLUSIONS Cellular senescence and cytoprotective autophagy are potential mechanisms of chemoresistance in TNBC. These two non-apoptotic outcomes in response to chemotherapy are inextricably linked and are neglected outcomes of investigation in the chemotherapeutic arena. Cellular fate assessments may therefore have the potential to predict TNBC patient outcome. GENERAL SIGNIFICANCE Focusing on the fact that cancer cells can bypass the desired cellular apoptotic response to chemotherapy through cellular senescence and cytoprotective autophagy will highlight the importance of targeting non-apoptotic survival pathways to enhance chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Elma A O'Reilly
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Luke Gubbins
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Shiva Sharma
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Riona Tully
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Matthew Ho Zhing Guang
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Karolina Weiner-Gorzel
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - John McCaffrey
- Department of Oncology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Michele Harrison
- Department of Pathology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Fiona Furlong
- School of Pharmacy, Queens University Belfast, Belfast BT7 1NN, UK
| | - Malcolm Kell
- Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Amanda McCann
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| |
Collapse
|