1
|
Aswani BS, Hegde M, Vishwa R, Alqahtani MS, Abbas M, Almubarak HA, Sethi G, Kunnumakkara AB. Tackling exosome and nuclear receptor interaction: an emerging paradigm in the treatment of chronic diseases. Mil Med Res 2024; 11:67. [PMID: 39327610 PMCID: PMC11426102 DOI: 10.1186/s40779-024-00564-1] [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: 12/02/2023] [Accepted: 08/06/2024] [Indexed: 09/28/2024] Open
Abstract
Nuclear receptors (NRs) function as crucial transcription factors in orchestrating essential functions within the realms of development, host defense, and homeostasis of body. NRs have garnered increased attention due to their potential as therapeutic targets, with drugs directed at NRs demonstrating significant efficacy in impeding chronic disease progression. Consequently, these pharmacological agents hold promise for the treatment and management of various diseases. Accumulating evidence emphasizes the regulatory role of exosome-derived microRNAs (miRNAs) in chronic inflammation, disease progression, and therapy resistance, primarily by modulating transcription factors, particularly NRs. By exploiting inflammatory pathways such as protein kinase B (Akt)/mammalian target of rapamycin (mTOR), nuclear factor kappa-B (NF-κB), signal transducer and activator of transcription 3 (STAT3), and Wnt/β-catenin signaling, exosomes and NRs play a pivotal role in the panorama of development, physiology, and pathology. The internalization of exosomes modulates NRs and initiates diverse autocrine or paracrine signaling cascades, influencing various processes in recipient cells such as survival, proliferation, differentiation, metabolism, and cellular defense mechanisms. This comprehensive review meticulously examines the involvement of exosome-mediated NR regulation in the pathogenesis of chronic ailments, including atherosclerosis, cancer, diabetes, liver diseases, and respiratory conditions. Additionally, it elucidates the molecular intricacies of exosome-mediated communication between host and recipient cells via NRs, leading to immunomodulation. Furthermore, it outlines the implications of exosome-modulated NR pathways in the prophylaxis of chronic inflammation, delineates current limitations, and provides insights into future perspectives. This review also presents existing evidence on the role of exosomes and their components in the emergence of therapeutic resistance.
Collapse
Affiliation(s)
- Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Hassan Ali Almubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, 61421, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
2
|
Zhu XD, Yu JH, Ai FL, Wang Y, Lv W, Yu GL, Cao XK, Lin J. Construction and Validation of a Novel Nomogram for Predicting the Risk of Metastasis in a Luminal B Type Invasive Ductal Carcinoma Population. World J Oncol 2023; 14:476-487. [PMID: 38022397 PMCID: PMC10681780 DOI: 10.14740/wjon1553] [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: 02/04/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Postoperative distant metastasis is the main cause of death in breast cancer patients. We aimed to construct a nomogram to predict the risk of metastasis of luminal B type invasive ductal carcinoma. Methods We applied the data of 364 luminal B type breast cancer patients between 2008 and 2013. Patients were categorized into modeling group and validation group randomly (1:1). The breast cancer metastasis nomogram was developed from the logistic regression model using clinicopathological variables. The area under the receiver-operating characteristic curve (AUC) was calculated in modeling group and validation group to evaluate the predictive accuracy of the nomogram. Results The multivariate logistic regression analysis showed that tumor size, No. of the positive level 1 axillary lymph nodes, human epidermal growth factor receptor 2 (HER2) status and Ki67 index were the independent predictors of the breast cancer metastasis. The AUC values of the modeling group and the validation group were 0.855 and 0.818, respectively. The nomogram had a well-fitted calibration curve. The positive and negative predictive values were 49.3% and 92.7% in the modeling group, and 47.9% and 91.0% in the validation group. Patients who had a score of 60 or more were thought to have a high risk of breast cancer metastasis. Conclusions The nomogram has a great predictive accuracy of predicting the risk of breast cancer metastasis. If patients had a score of 60 or more, necessary measures, like more standard treatment methods and higher treatment adherence of patients, are needed to take to lower the risk of metastasis and improve the prognosis.
Collapse
Affiliation(s)
- Xu Dong Zhu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Jia Hui Yu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Fu Lu Ai
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
| | - Yue Wang
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
| | - Wu Lv
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
| | - Gui Lin Yu
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
| | - Xian Kui Cao
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
| | - Jie Lin
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
3
|
Liu D, Li X, Lan Y, Zhang L, Wu T, Cui H, Li Z, Sun P, Tian P, Tian J. Models for Predicting Sentinel and Non-sentinel Lymph Nodes Based on Pre-operative Ultrasonic Breast Imaging to Optimize Axillary Strategies. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3101-3110. [PMID: 34362583 DOI: 10.1016/j.ultrasmedbio.2021.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/04/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
Axillary strategy decisions have become more complex and controversial in considering minimally traumatic therapy instead of sentinel lymph node biopsy, axillary lymph node dissection or regional nodal irradiation for people with breast cancer. The purpose of this study was to noninvasively predict sentinel lymph node (SLN) and non-sentinel lymph node (NSLN) status based on pre-operative sonographic and clinicopathologic features to determine optimal decisions regarding axillary therapy. In total, 701 patients with breast cancer from two independent centers were retrospectively analyzed. The SLN model (SLNM) for predicting SLN status and the NSLN model (NSLNM) for predicting NSLN status were trained based on a training set using the random-forest algorithm, and their performance was validated using an independent external test set. A receiver operating characteristic curve was drawn to obtain the area under the curve, which was used to assess performance. The area under the curve for the SLNM in the training and test, respectively, was 94.2% and 83.0%, and for the NSLNM, 99.5% and 92.7%. The SLNM and NSLNM accurately predicted that 61.46% (319/519) and 17.53% (91/519), respectively, of our participants were non-metastatic. The overall benefit of the three models was 78.99% in our participants. The two models for predicting SLN and NSLN status showed excellent application potential in optimizing axillary strategies.
Collapse
Affiliation(s)
- Dongmei Liu
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lei Zhang
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tong Wu
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hao Cui
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ziyao Li
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Peng Tian
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
4
|
Sánchez-López EF, Corigliano MG, Oliferuk S, Ramos-Duarte VA, Rivera M, Mendoza-Morales LF, Angel SO, Sander VA, Clemente M. Oral Immunization With a Plant HSP90-SAG1 Fusion Protein Produced in Tobacco Elicits Strong Immune Responses and Reduces Cyst Number and Clinical Signs of Toxoplasmosis in Mice. FRONTIERS IN PLANT SCIENCE 2021; 12:726910. [PMID: 34675949 PMCID: PMC8525317 DOI: 10.3389/fpls.2021.726910] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/30/2021] [Indexed: 05/17/2023]
Abstract
Plant 90kDa heat shock protein (HSP90) is a potent adjuvant that increases both humoral and cellular immune responses to diverse proteins and peptides. In this study, we explored whether Arabidopsis thaliana HSP90 (AtHsp81.2) can improve the immune effects of a Toxoplasma gondii surface antigen 1 (SAG1). We designed two constructs containing the sequence of mature antigen (SAG1m), from aa77 to aa322, and B- and T-cell antigenic epitope-containing SAG1HC, from aa221 to aa319 fused to AtHsp81.2 sequence. When comparing the transient expression in Nicotiana tabacum X-27-8 leaves, which overexpress the suppressor helper component protease HC-Pro-tobacco etch virus (TEV), to that in N. benthamiana leaves, co-agroinfiltrated with the suppressor p19, optimal conditions included 6-week-old N. benthamiana plants, 7-day time to harvest, Agrobacterium tumefaciens cultures with an OD600nm of 0.6 for binary vectors and LED lights. While AtHsp81.2-SAG1m fusion protein was undetectable by Western blot in any of the evaluated conditions, AtHsp81.2-SAG1HC was expressed as intact fusion protein, yielding up to 90μg/g of fresh weight. Besides, the AtHsp81.2-SAG1HC mRNA was strongly expressed compared to the endogenous Nicotiana tabacum elongation factor-alpha (NtEFα) gene, whereas the AtHsp81.2-SAG1m mRNA was almost undetectable. Finally, mice were orally immunized with AtHsp81.2-SAG1HC-infiltrated fresh leaves (plAtHsp81.2-SAG1HC group), recombinant AtHsp81.2-SAG1HC purified from infiltrated leaves (rAtHsp81.2-SAG1HC group), non-infiltrated fresh leaves (control group), or phosphate-buffered saline (PBS group). Serum samples from plAtHsp81.2-SAG1HC-immunized mice had significantly higher levels of IgGt, IgG2a, and IgG2b anti-SAG1HC antibodies than serum from rAtHsp81.2-SAG1HC, control, and PBS groups. The number of cysts per brain in the plAtHsp81.2-SAG1HC-immunized mice was significantly reduced, and the parasite load in brain tissue was also lower in this group compared with the remaining groups. In an immunoblot assay, plant-expressed AtHsp81.2-SAG1HC was shown to react with antibodies present in sera from T. gondii-infected people. Therefore, the plant expression of a T. gondii antigen fused to the non-pathogenic adjuvant and carrier plant HSP90 as formulations against T. gondii can improve the vaccine efficacy, and plant extract can be directly used for vaccination without the need to purify the protein, making this platform a suitable and powerful biotechnological system for immunogenic antigen expression against toxoplasmosis.
Collapse
Affiliation(s)
- Edwin F. Sánchez-López
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Mariana G. Corigliano
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Sonia Oliferuk
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Victor A. Ramos-Duarte
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Maximiliano Rivera
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Luisa F. Mendoza-Morales
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Sergio O. Angel
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Valeria A. Sander
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| | - Marina Clemente
- Laboratorio de Molecular Farming y Vacunas, Instituto Tecnológico Chascomús (INTECH), Universidad Nacional de General San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Chascomús, Argentina
| |
Collapse
|
5
|
Itkin B, Garcia A, Straminsky S, Adelchanow ED, Pereyra M, Haab GA, Bardach A. Prevalence of HER2 overexpression and amplification in cervical cancer: A systematic review and meta-analysis. PLoS One 2021; 16:e0257976. [PMID: 34591928 PMCID: PMC8483403 DOI: 10.1371/journal.pone.0257976] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
The reported rates of HER2 positivity in cervical cancer (CC) range from 0% to 87%. The importance of HER2 as an actionable target in CC would depend on HER2 positivity prevalence. Our aim was to provide precise estimates of HER2 overexpression and amplification in CC, globally and by relevant subgroups. We conducted a PRISMA compliant meta-analytic systematic review. We searched Medline, EMBASE, Cochrane database, and grey literature for articles reporting the proportion of HER2 positivity in CC. Studies assessing HER2 status by immunohistochemistry or in situ hybridization in invasive disease were eligible. We performed descriptive analyses of all 65 included studies. Out of these, we selected 26 studies that used standardized American Society of Clinical Oncology / College of American Pathologists (ASCO/CAP) Guidelines compliant methodology. We conducted several meta-analyses of proportions to estimate the pooled prevalence of HER2 positivity and subgroup analyses using geographic region, histology, tumor stage, primary antibody brand, study size, and publication year as moderators. The estimated pooled prevalence of HER2 overexpression was 5.7% (CI 95%: 1.5% to 11.7%) I2 = 87% in ASCO/CAP compliant studies and 27.0%, (CI 95%: 19.9% to 34.8%) I2 = 96% in ASCO/CAP non-compliant ones, p < 0.001. The estimated pooled prevalence of HER2 amplification was 1.2% (CI 95%: 0.0% to 5.8%) I2 = 0% and 24.9% (CI 95%: 12.6% to 39.6%) I2 = 86%, respectively, p = 0.004. No other factor was significantly associated with HER2 positivity rates. Our results suggest that a small, but still meaningful proportion of CC is expected to be HER2-positive. High heterogeneity was the main limitation of the study. Variations in previously reported HER2 positivity rates are mainly related to methodological issues.
Collapse
Affiliation(s)
- Boris Itkin
- Department of Oncology, Juan A Fernández Hospital, Buenos Aires, Argentina
| | - Agustin Garcia
- Department of Oncology, María Curie Hospital, Buenos Aires, Argentina
| | - Samanta Straminsky
- Department of Oncology, Juan A Fernández Hospital, Buenos Aires, Argentina
| | | | - Matias Pereyra
- Department of Pathology, Juan A Fernández Hospital, Buenos Aires, Argentina
| | | | - Ariel Bardach
- Center for Research in Epidemiology and Public Health, Institute for Clinical Effectiveness and Health Policy (IECS)—National Scientific and Technical Research Council—Argentina, Buenos Aires, Argentina
| |
Collapse
|
6
|
Liu D, Lan Y, Zhang L, Wu T, Cui H, Li Z, Sun P, Tian P, Tian J, Li X. Nomograms for Predicting Axillary Lymph Node Status Reconciled With Preoperative Breast Ultrasound Images. Front Oncol 2021; 11:567648. [PMID: 33898303 PMCID: PMC8058421 DOI: 10.3389/fonc.2021.567648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 03/16/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction The axillary lymph node (ALN) status of breast cancer patients is an important prognostic indicator. The use of primary breast mass features for the prediction of ALN status is rare. Two nomograms based on preoperative ultrasound (US) images of breast tumors and ALNs were developed for the prediction of ALN status. Methods A total of 743 breast cancer cases collected from 2016 to 2019 at the Second Affiliated Hospital of Harbin Medical University were randomly divided into a training set (n = 523) and a test set (n = 220). A primary tumor feature model (PTFM) and ALN feature model (ALNFM) were separately generated based on tumor features alone, and a combination of features was used for the prediction of ALN status. Logistic regression analysis was used to construct the nomograms. A receiver operating characteristic curve was plotted to obtain the area under the curve (AUC) to evaluate accuracy, and bias-corrected AUC values and calibration curves were obtained by bootstrap resampling for internal and external verification. Decision curve analysis was applied to assess the clinical utility of the models. Results The AUCs of the PTFM were 0.69 and 0.67 for the training and test sets, respectively, and the bias-corrected AUCs of the PTFM were 0.67 and 0.67, respectively. Moreover, the AUCs of the ALNFM were 0.86 and 0.84, respectively, and the bias-corrected AUCs were 0.85 and 0.81, respectively. Compared with the PTFM, the ALNFM showed significantly improved prediction accuracy (p < 0.001). Both the calibration and decision curves of the ALNFM nomogram indicated greater accuracy and clinical practicality. When the US tumor size was ≤21.5 mm, the Spe was 0.96 and 0.92 in the training and test sets, respectively. When the US tumor size was greater than 21.5 mm, the Sen was 0.85 in the training set and 0.87 in the test set. Our further research showed that when the US tumor size was larger than 35 mm, the Sen was 0.90 in the training set and 0.93 in the test set. Conclusion The ALNFM could effectively predict ALN status based on US images especially for different US tumor size.
Collapse
Affiliation(s)
- Dongmei Liu
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lei Zhang
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Tong Wu
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Hao Cui
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Ziyao Li
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Peng Tian
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital, Harbin, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
7
|
Kim MC, Kang SH, Choi JE, Bae YK. Impact of the Updated Guidelines on Human Epidermal Growth Factor Receptor 2 (HER2) Testing in Breast Cancer. J Breast Cancer 2020; 23:484-497. [PMID: 33154824 PMCID: PMC7604374 DOI: 10.4048/jbc.2020.23.e53] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/13/2020] [Indexed: 01/13/2023] Open
Abstract
Purpose In 2007, the American Society of Clinical Oncology and the College of American Pathologists had established a human epidermal growth factor receptor 2 (HER2) testing guideline, which was updated in 2013 and subsequently in 2018. We assessed the clinical impact of the recent update by comparing the in situ hybridization (ISH) results based on the 2007, 2013, and 2018 guidelines. Methods We assessed 2 cohorts. The first cohort included 1,161 primary invasive breast cancer (IBC) samples including 18 bilateral IBC cases, with both immunohistochemistry (IHC) and silver-enhanced ISH (SISH) results available for the HER2 status. The second cohort included 160 IBC cases with equivocal HER2 IHC, assessed using SISH. We retrospectively evaluated and compared the HER2 SISH results. Results There were 22 (1.9%) and 20 (12.5%) cases with altered SISH results according to the 2013 guidelines in cohorts 1 and 2, respectively. As per the 2018 guidelines, final HER2 statuses of 16 (1.4%) and 14 (8.5%) cases changed in cohorts 1 and 2, respectively. The 2013 guidelines increased the positive rate compared to the 2007 guidelines, in both cohorts (0.6% and 6.2%, respectively). Most equivocal cases in cohorts 1 (92.3%) and 2 (100%) as per the 2013 guidelines were reclassified as HER2-negative according to the 2018 guidelines. The 2018 guidelines increased the negative rates (1.3% in cohort 1 and 8.7% in cohort 2) and slightly decreased the positive rates (−0.2% in cohort 1 and −3.1% in cohort 2), compared to the 2013 guidelines. With each update, minor changes in the positive and negative rates were observed in whole breast cancer samples (cohort 1). However, the 2018 guidelines affected previously defined HER2-positive IBC with equivocal IHC results. Conclusion Under the 2013 guidelines, the positive and equivocal cases increased. However, the 2018 guidelines eliminated ambiguous cases by reclassifying them as HER2-negative.
Collapse
Affiliation(s)
- Min Chong Kim
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| | - Su Hwan Kang
- Department of Breast Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Jung Eun Choi
- Department of Breast Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Young Kyung Bae
- Department of Pathology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
8
|
The impact of 2018 ASCO-CAP HER2 testing guidelines on breast cancer HER2 results. An audit of 2132 consecutive cases evaluated by immunohistochemistry and in situ hybridization. Mod Pathol 2020; 33:1783-1790. [PMID: 32366941 DOI: 10.1038/s41379-020-0555-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 11/08/2022]
Abstract
The 2018 iteration of the ASCO-CAP HER2 testing guidelines proposes significant changes with an emphasis on the integration of concurrent immunohistochemistry (IHC) and in situ hybridization (ISH). We wished to evaluate the impact of these changes on clinical practice. Between Jan 2012 to Feb 2017, 2132 consecutive invasive breast carcinomas were evaluated with IHC and ISH for HER2. The sample tested was the breast primary or axillary nodes in all but 57 (2.7%) distant metastases. For 1824 cases with both dual-probe ISH and IHC results, the ISH subgroup was 1: 299 (16.4%), 2: 19 (1.0%), 1.0%, 3: 6 (0.3%), 4: 48 (2.6%) and 5: 1452 (79.6%). Ultimately 21% of group 2 and 4 cases and 80% of group 4 cases were positive. The change in HER2 status between the 2018 vs 2013 was: amplified in 323 (15.2%) vs 15.5%; not amplified in 1804 (84.6%) vs 82.2%; equivocal in 0 vs 2.3% previously. In 22 of 2127 cases (1.03%) the 2013 and 2018 results were discordant, all in groups 2-4. The discrepant cases included 15 of 331 (4.5%) of 2013 amplified cancers, now negative (all in groups 2 or 3) and 7 of 1796 (0.4%) 2013 nonamplified cases, now positive (all in group 4). Because of routine testing with both IHC and ISH, we found 6 of 1147 (0.52%) IHC negative (0 or 1+) cases were amplified by ISH. Further, 19 of 289 (6.6%) of IHC 3+ cases were nonamplified by ISH, circumstances not covered by these guidelines. In summary at the population level, the 2018 ASCO-CAP guidelines have a 99% agreement with the 2013 results. A major advantage is the abolishment of the clinically problematic equivocal category. Routine performance of both IHC and ISH uncovers a small proportion of cancers whose HER2 status is not addressed by these guidelines.
Collapse
|
9
|
Zhang W, Xu J, Wang K, Tang XJ, Liang H, He JJ. Independent risk factors for axillary lymph node metastasis in breast cancer patients with one or two positive sentinel lymph nodes. BMC WOMENS HEALTH 2020; 20:143. [PMID: 32646416 PMCID: PMC7350751 DOI: 10.1186/s12905-020-01004-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 06/26/2020] [Indexed: 12/24/2022]
Abstract
Background The benefit of axillary lymph node dissection (ALND) in breast cancer patients with one or two positive sentinel lymph nodes (SLNs) remains inconclusive. The purpose of this study was to identify risk factors independently associated with axillary lymph node (ALN) metastasis. Methods We retrospectively analyzed data from 389 Chinese breast cancer patients with one or two positive SLNs who underwent ALND. Univariate and multivariate logistic regression analyses were performed to identify ALN metastasis-associated risk factors. Results Among the 389 patients, 174 (44.7%) had ALN metastasis, while 215 (55.3%) showed no evidence of ALN metastasis. Univariate analysis revealed significant differences in age (< 60 or ≥ 60 years), human epidermal growth factor receptor-2 (Her-2) status, and the ratio of positive to total SLNs between the ALN metastasis and non-metastasis groups (P < 0.05). The multivariate analysis indicated that age, the ratio of positive to total SLNs, and occupations were significantly different between the two groups. Lastly, younger age (< 60 years), a higher ratio of positive to total SLNs, and manual labor jobs were independently associated with ALN metastasis (P < 0.05). Conclusions The risk of ALN metastasis in breast cancer patients with one or two positive SLNs can be further increased by younger age, manual labor jobs, and a high ratio of positive to total SLNs. Our findings may also aid in identifying which patients with one or two positive SLNs may not require ALND.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd., Xi'an, 710061, Shaanxi, China
| | - Jing Xu
- Department of Geriatric Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ke Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd., Xi'an, 710061, Shaanxi, China
| | - Xiao-Jiang Tang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd., Xi'an, 710061, Shaanxi, China
| | - Hua Liang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian-Jun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd., Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Urothelial carcinoma (UC) is a common malignancy with an urgent need for more effective and less toxic treatment strategies. Antibody-drug conjugate (ADC) represents a novel therapeutic approach, which combines the high specificity of monoclonal antibodies covalently linked with highly active cytotoxic agents. UC is an appropriate candidate for these drugs, as it expresses unique cell surface antigens that allow for specific targeting of these cells. We hereby present a review of the current literature and future perspectives of ADC treatment in early-stage and metastatic UC. RECENT FINDINGS Several ADCs are in advanced stages of development and approval, such as intravesical oportuzumab monatox in BCG-refractory non-muscle invasive bladder cancer and enfortumab vedotin and sacituzumab govitecan in pretreated metastatic UC. Other agents are in earlier stages of development, including some promising anti-Her2 agents. The favorable toxicity profile of these agents led to several combination strategies, especially with checkpoint inhibitors. In light of the encouraging results presented in this review and the recent FDA approval of enfortumab vedotin, ADCs will likely be incorporated in the management of UC in the near future.
Collapse
Affiliation(s)
- Michal Sarfaty
- Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA. .,Oncology Department, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Jonathan E Rosenberg
- Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA. .,Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
11
|
Geiersbach KB, Meyer RG, Sill DR, Mounajjed T, Chen B, Sukov WR, Jenkins RB. Working Up Group 4 Equivocal HER2 Samples Tested by Fluorescence in Situ Hybridization in a Reference Laboratory Setting: Past, Present, and Future. J Clin Oncol 2020; 38:175-176. [DOI: 10.1200/jco.19.02482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Katherine B. Geiersbach
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Reid G. Meyer
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Daniel R. Sill
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Taofic Mounajjed
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Beiyun Chen
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - William R. Sukov
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| | - Robert B. Jenkins
- Katherine B. Geiersbach, MD; Reid G. Meyer; Daniel R. Sill; Taofic Mounajjed, MD; Beiyun Chen, MD, PhD; William R. Sukov, MD; and Robert B. Jenkins, MD, PhD, Mayo Clinic School of Medicine, Rochester, MN
| |
Collapse
|
12
|
Kalari KR, Sinnwell JP, Thompson KJ, Tang X, Carlson EE, Yu J, Vedell PT, Ingle JN, Weinshilboum RM, Boughey JC, Wang L, Goetz MP, Suman V. PANOPLY: Omics-Guided Drug Prioritization Method Tailored to an Individual Patient. JCO Clin Cancer Inform 2019; 2:1-11. [PMID: 30652605 DOI: 10.1200/cci.18.00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The majority of patients with cancer receive treatments that are minimally informed by omics data. We propose a precision medicine computational framework, PANOPLY (Precision Cancer Genomic Report: Single Sample Inventory), to identify and prioritize drug targets and cancer therapy regimens. MATERIALS AND METHODS The PANOPLY approach integrates clinical data with germline and somatic features obtained from multiomics platforms and applies machine learning and network analysis approaches in the context of the individual patient and matched controls. The PANOPLY workflow uses the following four steps: selection of matched controls to the patient of interest; identification of patient-specific genomic events; identification of suitable drugs using the driver-gene network and random forest analyses; and provision of an integrated multiomics case report of the patient with prioritization of anticancer drugs. RESULTS The PANOPLY workflow can be executed on a stand-alone virtual machine and is also available for download as an R package. We applied the method to an institutional breast cancer neoadjuvant chemotherapy study that collected clinical and genomic data as well as patient-derived xenografts to investigate the prioritization offered by PANOPLY. In a chemotherapy-resistant patient-derived xenograft model, we found that that the prioritized drug, olaparib, was more effective than placebo in treating the tumor ( P < .05). We also applied PANOPLY to in-house and publicly accessible multiomics tumor data sets with therapeutic response or survival data available. CONCLUSION PANOPLY shows promise as a means to prioritize drugs on the basis of clinical and multiomics data for an individual patient with cancer. Additional studies are needed to confirm this approach.
Collapse
Affiliation(s)
| | | | | | | | | | - Jia Yu
- All authors: Mayo Clinic, Rochester, MN
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Assessing the impact of the 2018 American Society of Clinical Oncology/College of American Pathologists recommendations on human epidermal growth factor receptor 2 testing by fluorescence in situ hybridization in breast carcinoma. Virchows Arch 2019; 476:367-372. [PMID: 31375912 DOI: 10.1007/s00428-019-02636-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/13/2019] [Accepted: 07/24/2019] [Indexed: 10/26/2022]
Abstract
The American Society of Clinical Oncology/College of American Pathologists recently updated their recommendations on human epidermal growth factor receptor 2 (HER2) testing by fluorescence in situ hybridization (FISH) in invasive breast cancer, with a focus on the clarification of less common test patterns of ISH. We assessed the impact of the updated ASCO/CAP guidelines on 1044 FISH tested tumors by comparing categorization according to the 2007, 2013, and 2018 ISH classification criteria. The 2013 guidelines increased the number of positive cases (17.4% vs 10.7%) identifying 70 (6.7%) additional patients who met the eligibility criteria for consideration for HER2-targeted therapy compared with the 2007 guidelines. There was a reduction in equivocal tumors (7.7%) with tumors classified as equivocal by the 2007 guidelines (n = 136) redistributed into positive (74, 54.4%) and negative (49, 36.0%) groups. The 2018 guidelines reclassified 10.8% of tumors in our series with a reduction in the number of positive tumors (7.1%). While the proportion of positive tumors (10.2%) was similar to that in 2007 (10.7%), the composition of this group was significantly altered. HER2 equivocal cases, a group which under the 2013 guidelines caused diagnostic and treatment difficulties, were largely eliminated. Our findings suggest that the 2018 update represents a potentially significant change in therapeutic options for a substantial proportion of patients with 2.9% of FISH-positive tumors according to the 2007 and 2013 guidelines now categorized as HER2 negative and, thus, ineligible for HER2-targeted therapy.
Collapse
|
14
|
Li J, Ma W, Jiang X, Cui C, Wang H, Chen J, Nie R, Wu Y, Li L. Development and Validation of Nomograms Predictive of Axillary Nodal Status to Guide Surgical Decision-Making in Early-Stage Breast Cancer. J Cancer 2019; 10:1263-1274. [PMID: 30854136 PMCID: PMC6400691 DOI: 10.7150/jca.32386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose: To develop and validate nomogram models using noninvasive imaging parameters with related clinical variables to predict the extent of axillary nodal involvement and stratify treatment options based on the essential cut-offs for axillary surgery according to the ACOSOG Z0011 criteria. Materials and Methods: From May 2007 to December 2017, 1799 patients who underwent preoperative breast and axillary magnetic resonance imaging (MRI) were retrospectively studied. Patients with data on axillary ultrasonography (AUS) were enrolled. The MRI images were interpreted according to Breast Imaging Reporting and Data system (BI-RADS). Using logistic regression analyses, nomograms were developed to visualize the associations between the predictors and each lymph node (LN) status endpoint. Predictive performance was assessed based on the area under the receiver operating characteristic curve (AUC). Bootstrap resampling was performed for internal validation. Goodness-of-fit of the models was evaluated using the Hosmer-Lemeshow test. Results: Of 397 early breast cancer patients, 200 (50.4%) had disease-free axilla, 119 (30.0%) had 1 or 2 positive LNs, and 78 (19.6%) had ≥3 positive LNs. Patient age, MRI features (mass margin, LN margin, presence/absence of LN hilum, and LN symmetry/asymmetry), and AUS descriptors (presence of cortical thickening or hilum) were identified as predictors of nodal disease. Nomograms with these predictors showed good calibration and discrimination; the AUC was 0.809 for negative axillary node (N0) vs. any LN metastasis, 0.749 for 1 or 2 involved nodes vs. N0, and 0.874 for ≥3 nodes vs. ≤2 metastatic nodes. The predictive ability of the 3 nomograms with additional pathological variables was significantly greater. Conclusion: The nomograms could predict the extent of ALN metastasis and facilitate decision-making preoperatively.
Collapse
Affiliation(s)
- Jiao Li
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Weimei Ma
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Xinhua Jiang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Chunyan Cui
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Hongli Wang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiewen Chen
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Runcong Nie
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Yaopan Wu
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| | - Li Li
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China
| |
Collapse
|
15
|
Murray C, D'Arcy C, Gullo G, Flanagan L, Quinn CM. Human Epidermal Growth Factor Receptor 2 Testing by Fluorescent In Situ Hybridization: Positive or Negative? American Society of Clinical Oncology/College of American Pathologists Guidelines 2007, 2013, and 2018. Arch Pathol Lab Med 2019; 143:412-413. [PMID: 30605368 DOI: 10.5858/arpa.2018-0905-le] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ciara Murray
- 1 St Vincent's University Hospital, Dublin, Ireland
| | - Clare D'Arcy
- 1 St Vincent's University Hospital, Dublin, Ireland
| | | | | | - Cecily M Quinn
- 1 St Vincent's University Hospital, Dublin, Ireland.,2 University College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Murray C, D'Arcy C, Gullo G, Flanagan L, Quinn CM, Quinn CM. Human Epidermal Growth Factor Receptor 2 Testing by Fluorescent In Situ Hybridization: Positive or Negative? ASCO/College of American Pathologists Guidelines 2007, 2013, and 2018. J Clin Oncol 2018; 36:JCO1800788. [PMID: 30346899 DOI: 10.1200/jco.18.00788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Ciara Murray
- Ciara Murray, Clare D'Arcy, Giuseppe Gullo, Louise Flanagan, and Cecily M. Quinn, St Vincent's University Hospital, Dublin, Ireland; and Cecily M. Quinn, University College Dublin, Dublin, Ireland
| | - Clare D'Arcy
- Ciara Murray, Clare D'Arcy, Giuseppe Gullo, Louise Flanagan, and Cecily M. Quinn, St Vincent's University Hospital, Dublin, Ireland; and Cecily M. Quinn, University College Dublin, Dublin, Ireland
| | - Giuseppe Gullo
- Ciara Murray, Clare D'Arcy, Giuseppe Gullo, Louise Flanagan, and Cecily M. Quinn, St Vincent's University Hospital, Dublin, Ireland; and Cecily M. Quinn, University College Dublin, Dublin, Ireland
| | - Louise Flanagan
- Ciara Murray, Clare D'Arcy, Giuseppe Gullo, Louise Flanagan, and Cecily M. Quinn, St Vincent's University Hospital, Dublin, Ireland; and Cecily M. Quinn, University College Dublin, Dublin, Ireland
| | - Cecily M Quinn
- Ciara Murray, Clare D'Arcy, Giuseppe Gullo, Louise Flanagan, and Cecily M. Quinn, St Vincent's University Hospital, Dublin, Ireland; and Cecily M. Quinn, University College Dublin, Dublin, Ireland
| | - Cecily M Quinn
- Ciara Murray, Clare D'Arcy, Giuseppe Gullo, Louise Flanagan, and Cecily M. Quinn, St Vincent's University Hospital, Dublin, Ireland; and Cecily M. Quinn, University College Dublin, Dublin, Ireland
| |
Collapse
|
17
|
Allison KH. Ancillary Prognostic and Predictive Testing in Breast Cancer: Focus on Discordant, Unusual, and Borderline Results. Surg Pathol Clin 2018; 11:147-176. [PMID: 29413654 DOI: 10.1016/j.path.2017.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ancillary testing in breast cancer has become standard of care to determine what therapies may be most effective for individual patients with breast cancer. Single-marker tests are required on all newly diagnosed and newly metastatic breast cancers. Markers of proliferation are also used, and include both single-marker tests like Ki67 as well as panel-based gene expression tests, which have made more recent contributions to prognostic and predictive testing in breast cancers. This review focuses on pathologist interpretation of these ancillary test results, with a focus on expected versus unexpected results and troubleshooting borderline, unusual, or discordant results.
Collapse
Affiliation(s)
- Kimberly H Allison
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Lane 235, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
Lai J, Wang H, Peng J, Chen P, Pan Z. Establishment and external validation of a prognostic model for predicting disease-free survival and risk stratification in breast cancer patients treated with neoadjuvant chemotherapy. Cancer Manag Res 2018; 10:2347-2356. [PMID: 30122984 PMCID: PMC6078091 DOI: 10.2147/cmar.s171129] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background The eighth edition of the American Joint Committee on Cancer (AJCC) tumor-node-metastasis (TNM) staging system for survival prediction and risk stratification in breast cancer (BC) patients after neoadjuvant chemotherapy (NCT) is of limited efficacy. This study aimed to establish a novel prognostic nomogram for predicting disease-free survival (DFS) in BC patients after NCT. Patients and methods A total of 567 BC patients treated with NCT, from two independent centers, were included in this study. Cox proportional-hazards regression (CPHR) analysis was conducted to identify the independent prognostic factors for DFS, in order to develop a model. Subsequently, the discrimination and calibration ability of the prognostic model were assessed in terms of its concordance index (C-index), risk group stratification, and calibration curve. The performance of the nomogram was compared with that of the eighth edition of the AJCC TNM staging system via C-index. Results Based on the CPHR model, eight prognostic predictors were screened and entered into the nomogram. The prognostic model showed better performance (p<0.01) in terms of DFS prediction (C-index: 0.738; 95% CI: 0.698-0.779) than the eighth edition of the AJCC TNM staging system (C-index: 0.644; 95% CI: 0.604-0.684). Stratification into three risk groups highlighted significant differences between the survival curves in the training cohort and those in the validation cohort. The calibration curves for likelihood of 3- and 5-year DFS indicated optimal agreement between nomogram predictions and actual observations. Conclusion We constructed and externally validated a novel nomogram scoring system for individualized DFS estimation in BC patients treated with NCT. This user-friendly predictive tool may help oncologists to make optimal clinical decisions.
Collapse
Affiliation(s)
- Jianguo Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China, .,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China,
| | - Hongli Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China, .,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China,
| | - Jingwen Peng
- Department of Rehabilitation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Peixian Chen
- Department of Breast surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Zihao Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China, .,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China,
| |
Collapse
|
19
|
Norton N, Fox N, McCarl CA, Tenner KS, Ballman K, Erskine CL, Necela BM, Northfelt D, Tan WW, Calfa C, Pegram M, Colon-Otero G, Perez EA, Clynes R, Knutson KL. Generation of HER2-specific antibody immunity during trastuzumab adjuvant therapy associates with reduced relapse in resected HER2 breast cancer. Breast Cancer Res 2018; 20:52. [PMID: 29898752 PMCID: PMC6000975 DOI: 10.1186/s13058-018-0989-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/15/2018] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Resected HER2 breast cancer patients treated with adjuvant trastuzumab and chemotherapy have superior survival compared to patients treated with chemotherapy alone. We previously showed that trastuzumab and chemotherapy induce HER2-specific antibodies which correlate with improved survival in HER2 metastatic breast cancer patients. It remains unclear whether the generation of immunity required trastuzumab and whether endogenous antibody immunity is associated with improved disease-free survival in the adjuvant setting. In this study, we addressed this question by analyzing serum anti-HER2 antibodies from a subset of patients enrolled in the NCCTG trial N9831, which includes an arm (Arm A) in which trastuzumab was not used. Arms B and C received trastuzumab sequentially or concurrently to chemotherapy, respectively. METHODS Pre-and post-treatment initiation sera were obtained from 50 women enrolled in N9831. Lambda IgG antibodies (to avoid detection of trastuzumab) to HER2 were measured and compared between arms and with disease-free survival. RESULTS Prior to therapy, across all three arms, N9831 patients had similar mean anti-HER2 IgG levels. Following treatment, the mean levels of antibodies increased in the trastuzumab arms but not the chemotherapy-only arm. The proportion of patients who demonstrated antibodies increased by 4% in Arm A and by 43% in the Arms B and C combined (p = 0.003). Cox modeling demonstrated that larger increases in antibodies were associated with improved disease-free survival in all patients (HR = 0.23; p = 0.04). CONCLUSIONS These results show that the increased endogenous antibody immunity observed in adjuvant patients treated with combination trastuzumab and chemotherapy is clinically significant, in view of its correlation with improved disease-free survival. The findings may have important implications for predicting treatment outcomes in patients treated with trastuzumab in the adjuvant setting. TRIAL REGISTRATION ClinicalTrials.gov, NCT00005970 . Registered on July 5, 2000.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/immunology
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Biomarkers, Tumor/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Chemotherapy, Adjuvant/adverse effects
- Combined Modality Therapy
- Disease-Free Survival
- Female
- Humans
- Kaplan-Meier Estimate
- Middle Aged
- Neoplasm Metastasis
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/pathology
- Receptor, ErbB-2/immunology
- Recurrence
- Trastuzumab/administration & dosage
- Trastuzumab/adverse effects
- Treatment Outcome
Collapse
Affiliation(s)
- Nadine Norton
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Nicholas Fox
- Department of Pathology, Medicine and Dermatology, Columbia University Medical Center, New York, NY 10032 USA
| | - Christie-Ann McCarl
- Department of Pathology, Medicine and Dermatology, Columbia University Medical Center, New York, NY 10032 USA
| | - Kathleen S. Tenner
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Karla Ballman
- Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY USA
| | | | - Brian M. Necela
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Donald Northfelt
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ 85259 USA
| | - Winston W. Tan
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Carmen Calfa
- Sylvester Cancer Center, University of Miami, Miami, FL 33136 USA
| | - Mark Pegram
- Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Gerardo Colon-Otero
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Edith A. Perez
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Raphael Clynes
- Department of Pathology, Medicine and Dermatology, Columbia University Medical Center, New York, NY 10032 USA
| | - Keith L. Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL 32224 USA
| |
Collapse
|
20
|
Dhiman VK, Bolt MJ, White KP. Nuclear receptors in cancer — uncovering new and evolving roles through genomic analysis. Nat Rev Genet 2017; 19:160-174. [DOI: 10.1038/nrg.2017.102] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
Serra M, Ferraro D, Pereiro I, Viovy JL, Descroix S. The power of solid supports in multiphase and droplet-based microfluidics: towards clinical applications. LAB ON A CHIP 2017; 17:3979-3999. [PMID: 28948991 DOI: 10.1039/c7lc00582b] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Multiphase and droplet microfluidic systems are growing in relevance in bioanalytical-related fields, especially due to the increased sensitivity, faster reaction times and lower sample/reagent consumption of many of its derived bioassays. Often applied to homogeneous (liquid/liquid) reactions, innovative strategies for the implementation of heterogeneous (typically solid/liquid) processes have recently been proposed. These involve, for example, the extraction and purification of target analytes from complex matrices or the implementation of multi-step protocols requiring efficient washing steps. To achieve this, solid supports such as functionalized particles (micro or nanometric) presenting different physical properties (e.g. magnetic, optical or others) are used for the binding of specific entities. The manipulation of such supports with different microfluidic principles has both led to the miniaturization of existing biomedical protocols and the development of completely new strategies for diagnostics and research. In this review, multiphase and droplet-based microfluidic systems using solid suspensions are presented and discussed with a particular focus on: i) working principles and technological developments of the manipulation strategies and ii) applications, critically discussing the level of maturity of these systems, which can range from initial proofs of concept to real clinical validations.
Collapse
Affiliation(s)
- M Serra
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.
| | | | | | | | | |
Collapse
|
22
|
Ercolani C, Marchiò C, Di Benedetto A, Fabi A, Perracchio L, Vici P, Sperati F, Buglioni S, Arena V, Pescarmona E, Sapino A, Terrenato I, Mottolese M. Breast carcinomas with low amplified/equivocal HER2 by Ish: potential supporting role of multiplex ligation-dependent probe amplification. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:143. [PMID: 29029640 PMCID: PMC5640946 DOI: 10.1186/s13046-017-0613-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/05/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND This is a retrospective cross sectional study aimed to verify whether Multiplex Ligation-dependent Probe Amplification (MLPA), a quantitative molecular assay, may represent a valuable reflex test in breast cancer with equivocal HER2 expression by immunohistochemistry and HER2 gene signals/nucleus (s/n) ranging between 4.0 and 5.9 by in situ hybridization. METHODS A series of 170 breast carcinomas scored as 2+ for HER2 expression by immunohistochemistry, were selected from our files and analyzed in parallel by silver in situ hybridization and by MLPA. According to ASCO-CAP 2013 guidelines, 54/170 tumors, displaying 4.0-5.9 HER2 gene s/n, were defined as low amplified (ratio ≥ 2) or equivocal (ratio < 2) on the basis of centromere enumeration probe 17 (CEP17) status. An independent set of 108 score 2+ breast cancers represented the external validation set. Concordance between the two techniques was assessed through the use of Cohen's K statistic. RESULTS A concordance rate of 78.2% (Cohen's K statistic: 0,548 95% CI:[0,419-0,677]) between in situ hybridization and MLPA was found in the whole series of 170 cases and of 55.5% (Cohen's K statistic: -0,043 95% CI:[-0,271-0,184]) in the 54 tumors presenting 4.0-5.9 HER2 gene s/n. By MLPA, we found HER2 amplification or gain in 14% of the 21 BC presenting a disomic status and in 18% of the 33 BC presenting a CEP17 > 2.0. These data were further confirmed in the external validation set. Interestingly, the 54 low amplified/equivocal breast carcinomas presented a frequency of hormonal receptor positivity significantly higher than that observed in the amplified tumors and similar to the non-amplified one (p = 0.016 for estrogen receptor and p = 0.001 for progesterone receptor). CONCLUSIONS To avoid to offer patients an ineffective therapy, HER2 status should be studied more thoroughly in low amplified and equivocal cases which can have lower response rates and shorter time to progression to trastuzumab. In this context, our data indicate that MLPA may be a reliable, objective supporting test in selecting HER2 positive breast cancer patients.
Collapse
Affiliation(s)
- Cristiana Ercolani
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Pathology Unit, Via Santena 7, 10126, Turin, Italy
| | - Anna Di Benedetto
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Alessandra Fabi
- Medical Oncology 1, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Letizia Perracchio
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Patrizia Vici
- Medical Oncology 2, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Francesca Sperati
- Biostatistic Unit, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Simonetta Buglioni
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Vincenzo Arena
- Department of Pathology, Catholic University of Sacred Heart, Foundation Policlinico A. Gemelli, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Turin, Pathology Unit, Via Santena 7, 10126, Turin, Italy.,Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO), IRCCS, Str. Prov. 142, km 3.95, Candiolo, 10060, To, Italy
| | - Irene Terrenato
- Biostatistic Unit, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Marcella Mottolese
- Department of Pathology, Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
23
|
Zhang X, Bleiweiss I, Jaffer S, Nayak A. The Impact of 2013 Updated ASCO/CAP HER2 Guidelines on the Diagnosis and Management of Invasive Breast Cancer: A Single-Center Study of 1739 Cases. Clin Breast Cancer 2017; 17:486-492. [DOI: 10.1016/j.clbc.2017.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/15/2017] [Accepted: 03/22/2017] [Indexed: 10/19/2022]
|
24
|
Comparison of estrogen receptor, progesterone receptor and HER2 results in concurrent ipsilateral samples with invasive breast carcinoma: a retrospective study of 246 biopsies from 119 patients. Hum Pathol 2017; 65:123-132. [DOI: 10.1016/j.humpath.2017.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/31/2017] [Accepted: 04/19/2017] [Indexed: 11/23/2022]
|
25
|
Zhang J, Li X, Huang R, Feng WL, Kong YN, Xu F, Zhao L, Song QK, Li J, Zhang BN, Fan JH, Qiao YL, Xie XM, Zheng S, He JJ, Wang K. A nomogram to predict the probability of axillary lymph node metastasis in female patients with breast cancer in China: A nationwide, multicenter, 10-year epidemiological study. Oncotarget 2017; 8:35311-35325. [PMID: 27852049 PMCID: PMC5471057 DOI: 10.18632/oncotarget.13330] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/26/2016] [Indexed: 01/17/2023] Open
Abstract
Axillary lymph node dissection (ALND) or sentinel lymph node biopsy (SLNB) alone may lead to postoperative complications. Among patients with positive ALN in the preoperative examination, approximately 40% patients do not have SLN metastasis. Herein, we aimed to develop a model to predict the probability of ALN metastasis as a preoperative tool to support clinical decision-making. We retrospectively analyzed the clinicopathological features of 4211 female patients with breast cancer who were diagnosed in seven breast cancer centers representing entire China, over 10 years (1999-2008). The patients were randomly categorized into a training cohort or validation cohort (3:1 ratio). Multivariate logistic regression analysis was performed for 1869 patients with complete information on the study variables. Age at diagnosis, tumor size, tumor quadrant, clinical nodal status, local invasion status, pathological type, and molecular subtypes were the independent predictors of ALN metastasis. The nomogram was then developed using the seven variables. Further, it was subsequently validated in 642 patients with complete data on variables in the validation cohort. Coefficient of determination (R²) and the area under the receiver-operating characteristic (ROC) curve (AUC) were calculated to be 0.979 and 0.7007, showing good calibration and discrimination of the model, respectively. The false-negative rates of the nomogram were 0 and 6.9% for the predicted risk cut-off values of 14.03% and 20%, respectively. Therefore, when the predicted risk is less than 20%, SLNB may be avoided. After further validation in various patient populations, this model may support increasingly limited axillary surgery in breast cancer.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P.R. China
| | - Xiao Li
- Department of Breast Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P.R. China
| | - Rong Huang
- Department of Cancer Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
- Department of Epidemiology, West China School of Public Health, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Wei-Liang Feng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | - Ya-Nan Kong
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Feng Xu
- Department of Breast-thyroid Surgery, Xiangya Second Hospital, Central South University, Changsha, P.R. China
| | - Lin Zhao
- Department of Breast Surgery, Liaoning Cancer Hospital, Shenyang, P.R. China
| | - Qing-Kun Song
- Department of Cancer Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Jing Li
- Department of Cancer Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Bao-Ning Zhang
- Center of Breast Disease, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Jin-Hu Fan
- Department of Cancer Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - You-Lin Qiao
- Department of Cancer Epidemiology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Xiao-Ming Xie
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, P.R. China
| | - Shan Zheng
- Department of Pathology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P.R. China
| | - Jian-Jun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P.R. China
| | - Ke Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P.R. China
| |
Collapse
|
26
|
Arihiro K, Oda M, Ogawa K, Kaneko Y, Shimizu T, Tanaka Y, Marubashi Y, Ishida K, Takai C, Taoka C, Kimura S, Shiroma N. Utility of cytopathological specimens and an automated image analysis for the evaluation of HER2 status and intratumor heterogeneity in breast carcinoma. Pathol Res Pract 2016; 212:1126-1132. [PMID: 27712974 DOI: 10.1016/j.prp.2016.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 09/09/2016] [Accepted: 09/19/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Although updated HER2 testing guidelines have been improved by a collaboration between the American Society of Clinical Oncology (ASCO) and the College of American Pathologists (CAP) in 2013, HER2 evaluation is still problematic because of issues involving CEP17 polysomy, heterogeneity, and HER2 score 2+ cases. The aim of this retrospective study was to evaluate the relationship between HER2 gene heterogeneity, or so called CEP17 polysomy, using breast carcinoma cells sampled by scraping and the IHC score graded by automated image analysis using whole slide image. MATERIAL AND METHODS We randomly selected 23 breast carcinoma cases with a HER2 score 0, 24 cases with a HER2 score 1+, 24 cases with HER2 score 2+, and 23 cases with HER2 score 3+ from the records of patients with breast cancer at Hiroshima University Hospital. We compared the results of fluorescent in situ hybridization (FISH) using formalin-fixed, paraffin-embedded (FFPE) tissues and cytological samples and compared the HER2 score calculated using an automated image analysis using wholly scanned slide images and visual counting. RESULTS We successfully performed the FISH assay in 78 of 94 cases (83%) using FFPE tissues and in all 94 (100%) cases using cytological samples. Frequency of both HER2 amplification and CEP17 polysomy was higher when cytological samples were used than when FFPE tissue was used. Frequency of HER2 heterogeneity using cytological samples was higher that than using FFPE tissue, except for the IHC score 3+ cases. CONCLUSIONS When assessment of HER2 status based on FISH using FFPE tissue cannot be accomplished, FISH using cytological samples should be considered. When intensity of HER2 is heterogeneous in the tumor tissue, particularly in cases regarded as score 2+, they should be evaluated by automated image analysis using the whole slide image.
Collapse
Affiliation(s)
- Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan.
| | - Miyo Oda
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Katsunari Ogawa
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Yoshie Kaneko
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Tomomi Shimizu
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Yuna Tanaka
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Yukari Marubashi
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Katsunari Ishida
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Chikako Takai
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Chie Taoka
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Shuji Kimura
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| | - Noriyuki Shiroma
- Department of Anatomical Pathology, Hiroshima University Hospital, Japan
| |
Collapse
|
27
|
Ferraro D, Champ J, Teste B, Serra M, Malaquin L, Viovy JL, de Cremoux P, Descroix S. Microfluidic platform combining droplets and magnetic tweezers: application to HER2 expression in cancer diagnosis. Sci Rep 2016; 6:25540. [PMID: 27157697 PMCID: PMC4860594 DOI: 10.1038/srep25540] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/18/2016] [Indexed: 01/25/2023] Open
Abstract
The development of precision medicine, together with the multiplication of targeted therapies and associated molecular biomarkers, call for major progress in genetic analysis methods, allowing increased multiplexing and the implementation of more complex decision trees, without cost increase or loss of robustness. We present a platform combining droplet microfluidics and magnetic tweezers, performing RNA purification, reverse transcription and amplification in a fully automated and programmable way, in droplets of 250nL directly sampled from a microtiter-plate. This platform decreases sample consumption about 100 fold as compared to current robotized platforms and it reduces human manipulations and contamination risk. The platform’s performance was first evaluated on cell lines, showing robust operation on RNA quantities corresponding to less than one cell, and then clinically validated with a cohort of 21 breast cancer samples, for the determination of their HER2 expression status, in a blind comparison with an established routine clinical analysis.
Collapse
Affiliation(s)
- Davide Ferraro
- Institut Curie, PSL Research University, Laboratoire Physicochimie, CNRS/UMR 168, Institut Pierre-Gilles de Gennes, MMBM group, Paris, France
| | - Jérôme Champ
- APHP Hôpital Saint-Louis, Molecular Oncology Unit, University Paris-Diderot, INSERM/CNRS, UMR944/7212, Paris, France
| | - Bruno Teste
- Institut Curie, PSL Research University, Laboratoire Physicochimie, CNRS/UMR 168, Institut Pierre-Gilles de Gennes, MMBM group, Paris, France
| | - Marco Serra
- Institut Curie, PSL Research University, Laboratoire Physicochimie, CNRS/UMR 168, Institut Pierre-Gilles de Gennes, MMBM group, Paris, France
| | | | - Jean-Louis Viovy
- Institut Curie, PSL Research University, Laboratoire Physicochimie, CNRS/UMR 168, Institut Pierre-Gilles de Gennes, MMBM group, Paris, France
| | - Patricia de Cremoux
- APHP Hôpital Saint-Louis, Molecular Oncology Unit, University Paris-Diderot, INSERM/CNRS, UMR944/7212, Paris, France
| | - Stephanie Descroix
- Institut Curie, PSL Research University, Laboratoire Physicochimie, CNRS/UMR 168, Institut Pierre-Gilles de Gennes, MMBM group, Paris, France
| |
Collapse
|
28
|
Peretyatko LP, Avrelkina EV, Malyshkina AI. [Immunohistochemical changes in the lungs from fetuses and newborn infants at 20- 40 weeks' gestation with false left-sided congenital diaphragmatic hernia]. Arkh Patol 2016; 77:21-28. [PMID: 26841646 DOI: 10.17116/patol201577621-28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE to study changes in the expression of growth factors in the lungs from fetuses and newborn infants at 20-40 weeks' gestation with false left-sided congenital diaphragmatic hernia (CDH). MATERIAL AND METHODS Lung fragments obtained at autopsies were examined using immunohistochemically stained paraffin sections. The expression indices of insulin-like growth factor-1 (IGF-1), platelet-derived growth factor (PDGF), epidermal growth factor (EGF), and transforming grow factor-β (TGF-β) in the fetal lung sections were compared with the results of survey and elective staining techniques, the morphometric data of digitized lung sections, and the indicators obtained in the comparison group. RESULTS Morphofunctional changes in the connective tissue, pulmonary vessels, and bronchial and alveolar epithelium at 20-24, 25-27, and 28-40 weeks' gestation were characterized, partly explaining the development of bilateral pulmonary hypoplasia in CDH. There was evidence for the involvement of PDGF, IGF-1, and EGF in primary pulmonary hypoplasia. PDGF deficiency plays a certain role in secondary pulmonary hypoplasia. CONCLUSION The findings can be used in researches and in the practical work of pathologists and forensic medical experts.
Collapse
Affiliation(s)
- L P Peretyatko
- Laboratory of Pathomorphology and Electron Microscopy, V.N. Gorodkov Ivanovo Research Institute of Maternity and Childhood, Ministry of Health of the Russian Federation, Ivanovo, Russia
| | - E V Avrelkina
- Laboratory of Pathomorphology and Electron Microscopy, V.N. Gorodkov Ivanovo Research Institute of Maternity and Childhood, Ministry of Health of the Russian Federation, Ivanovo, Russia
| | - A I Malyshkina
- Laboratory of Pathomorphology and Electron Microscopy, V.N. Gorodkov Ivanovo Research Institute of Maternity and Childhood, Ministry of Health of the Russian Federation, Ivanovo, Russia
| |
Collapse
|
29
|
Current challenges in HER2-positive breast cancer. Crit Rev Oncol Hematol 2016; 98:211-21. [DOI: 10.1016/j.critrevonc.2015.10.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 10/06/2015] [Accepted: 10/28/2015] [Indexed: 12/13/2022] Open
|
30
|
Martin-Castillo B, Lopez-Bonet E, Buxó M, Dorca J, Tuca-Rodríguez F, Ruano MA, Colomer R, Menendez JA. Cytokeratin 5/6 fingerprinting in HER2-positive tumors identifies a poor prognosis and trastuzumab-resistant basal-HER2 subtype of breast cancer. Oncotarget 2016; 6:7104-22. [PMID: 25742793 PMCID: PMC4466672 DOI: 10.18632/oncotarget.3106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/08/2015] [Indexed: 12/19/2022] Open
Abstract
There is an urgent need to refine the prognostic taxonomy of HER2+ breast carcinomas and develop easy-to-use, clinic-based prediction algorithms to distinguish between good- and poor-responders to trastuzumab-based therapy. Building on earlier studies suggesting that HER2+ tumors enriched with molecular and morpho-immunohistochemical features classically ascribed to basal-like tumors are highly aggressive and refractory to trastuzumab, we investigated the prognostic and predictive value of the basal-HER2+ phenotype in HER2-overexpressing tumors. Our retrospective cohort study of a consecutive series of 152 HER2+ primary invasive ductal breast carcinomas first confirmed the existence of a distinct subgroup co-expressing HER2 protein and basal cytokeratin markers CK5/6, the so-called basal-HER2+ phenotype. Basal-HER2+ phenotype (≥10% of cells showing positive CK5/6 staining), but not estrogen receptor status, was significantly associated with inferior overall survival by univariate analysis and predicted worsened disease free survival after accounting for strong prognostic variables such as tumor size at diagnosis in stepwise multivariate analysis. In the sub-cohort of HER2+ patients treated with trastuzumab-based adjuvant/neoadjuvant therapy, basal-HER2+ phenotype was found to be the sole independent prognostic marker for a significantly inferior time to treatment failure in multivariate analysis. A CK5/6-based immunohistochemical fingerprint may provide a simple, rapid, and accurate method for re-classifying women diagnosed with HER2+ breast cancer in a manner that can improve prognosis and therapeutic planning in patients with clinically aggressive basal-HER2+ tumors who are not likely to benefit from trastuzumab-based therapy.
Collapse
Affiliation(s)
- Begoña Martin-Castillo
- Unit of Clinical Research, Catalan Institute of Oncology, Girona, Catalonia, Spain.,Girona Biomedical Research Institute (IDIBGI), Molecular Oncology Group, Girona, Catalonia, Spain
| | - Eugeni Lopez-Bonet
- Girona Biomedical Research Institute (IDIBGI), Molecular Oncology Group, Girona, Catalonia, Spain.,Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona, Catalonia, Spain
| | - Maria Buxó
- Girona Biomedical Research Institute (IDIBGI), Molecular Oncology Group, Girona, Catalonia, Spain.,Epidemiology Unit and Cancer Registry of Girona (UERCG), Catalan Cancer Plan, Catalan Health Government, Girona, Catalonia, Spain.,Department of Nursing, Universitat de Girona (UdG), Girona, Catalonia, Spain
| | - Joan Dorca
- Medical Oncology Department, Catalan Institute of Oncology, Girona, Catalonia, Spain
| | | | - Miguel Alonso Ruano
- Department of Gynecology, Dr. Josep Trueta Hospital of Girona, Girona, Catalonia, Spain
| | - Ramon Colomer
- Breast Cancer Clinical Research Unit, CNIO-Spanish National Cancer Research Center, Madrid, Spain.,Medical Oncology Department, Hospital La Princesa, Madrid, Spain
| | - Javier A Menendez
- Girona Biomedical Research Institute (IDIBGI), Molecular Oncology Group, Girona, Catalonia, Spain.,Translational Research Laboratory, Catalan Institute of Oncology (ICO), Girona, Catalonia, Spain
| |
Collapse
|
31
|
PIK3CA(H1047R)- and Her2-initiated mammary tumors escape PI3K dependency by compensatory activation of MEK-ERK signaling. Oncogene 2015; 35:2961-70. [PMID: 26640141 PMCID: PMC4896860 DOI: 10.1038/onc.2015.377] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 08/27/2015] [Accepted: 08/31/2015] [Indexed: 12/20/2022]
Abstract
Human breast cancers that have HER2 amplification/overexpression frequently carry PIK3CA mutations, and are often associated with a worse prognosis. However, the role of PIK3CA mutations in the initiation and maintenance of these breast cancers remains elusive. In the present study, we generated a compound mouse model that genetically mimics HER2 positive breast cancer with coexisting PIK3CAH1047R. Induction of PIK3CAH1047R expression in mouse mammary glands with constitutive expression of activated Her2/Neu resulted in accelerated mammary tumorigenesis with enhanced metastatic potential. Interestingly, inducible expression of mutant PIK3CA resulted in a robust activation of PI3K/AKT signaling but attenuation of Her2/Her3 signaling, and this can be reversed by deinduction of PIK3CAH1047R expression. Strikingly, while these Her2+ PIK3CAH1047R initiated primary mammary tumors are refractory to HER2-targeted therapy, all tumors responded to inactivation of the oncogenic PIK3CAH1047R, a situation closely mimicking the use of a highly effective inhibitor specifically targeting the mutant PIK3CA/p110a. Notably, these tumors eventually resumed growth, and a fraction of them escaped PI3K dependence by compensatory ERK activation, which can be blocked by combined inhibition of Her2 and MEK. Together, these results suggest that PIK3CA-specific inhibition as a monotherapy followed by combination therapy targeting MAPK and HER2 in a timely manner may be an effective treatment approach against HER2 positive cancers with coexisting PIK3CA-activating mutations.
Collapse
|
32
|
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
|
33
|
Advani PP, Crozier JA, Perez EA. HER2 testing and its predictive utility in anti-HER2 breast cancer therapy. Biomark Med 2015; 9:35-49. [PMID: 25605454 DOI: 10.2217/bmm.14.95] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Breast cancer treatment is dependent on accurate pathologic diagnosis. HER2 testing is now universally recommended as part of evaluation of invasive breast cancer. HER2 testing is available via various slide and non-slide based assays, and interpretation of results continues to evolve. Herein we review these testing modalities and their incorporation into the 2013 ASCO/CAP guidelines. Once accurate HER2 status has been established the proper treatment based on recent clinical trials can be instituted.
Collapse
Affiliation(s)
- Pooja P Advani
- Division of Hematology & Oncology, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL 32224, USA
| | | | | |
Collapse
|
34
|
Valdivieso M, Corn BW, Dancey JE, Wickerham DL, Horvath LE, Perez EA, Urton A, Cronin WM, Field E, Lackey E, Blanke CD. The Globalization of Cooperative Groups. Semin Oncol 2015; 42:693-712. [PMID: 26433551 DOI: 10.1053/j.seminoncol.2015.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The National Cancer Institute (NCI)-supported adult cooperative oncology research groups (now officially Network groups) have a longstanding history of participating in international collaborations throughout the world. Most frequently, the US-based cooperative groups work reciprocally with the Canadian national adult cancer clinical trial group, NCIC CTG (previously the National Cancer Institute of Canada Clinical Trials Group). Thus, Canada is the largest contributor to cooperative groups based in the United States, and vice versa. Although international collaborations have many benefits, they are most frequently utilized to enhance patient accrual to large phase III trials originating in the United States or Canada. Within the cooperative group setting, adequate attention has not been given to the study of cancers that are unique to countries outside the United States and Canada, such as those frequently associated with infections in Latin America, Asia, and Africa. Global collaborations are limited by a number of barriers, some of which are unique to the countries involved, while others are related to financial support and to US policies that restrict drug distribution outside the United States. This article serves to detail the cooperative group experience in international research and describe how international collaboration in cancer clinical trials is a promising and important area that requires greater consideration in the future.
Collapse
Affiliation(s)
- Manuel Valdivieso
- Division of Hematology/Oncology, University of Michigan; and SWOG, Executive Officer, Quality Assurance and International Initiatives, Ann Arbor, MI.
| | - Benjamin W Corn
- Institute of Radiotherapy, Tel Aviv Medical Center, Tel Aviv, Israel; and Department of Radiation Oncology, Jefferson Medical College, Philadelphia, PA
| | - Janet E Dancey
- Director, NCIC Clinical Trials Group; Scientific Director Canadian Cancer Clinical Trials Network; Program Leader, High Impact Clinical Trials, Ontario Institute for Cancer Research; Professor of Oncology, Queen's University, Kingston, Ontario, Canada
| | - D Lawrence Wickerham
- Deputy Chairman, NRG Oncology, Pittsburgh, PA; Department of Human Oncology, Pittsburgh Campus, Drexel University School of Medicine; Allegheny Cancer Center at Allegheny General Hospital, Pittsburgh, PA
| | - L Elise Horvath
- Executive Officer, Alliance for Clinical Trials in Oncology, Chicago, IL
| | - Edith A Perez
- Deputy Director at Large, Mayo Clinic Cancer Center; Group Vice Chair, Alliance for Clinical Trials in Oncology; Hematology/Oncology and Cancer Biology Mayo Clinic, Jacksonville, FL
| | - Alison Urton
- Group Administrator, NCIC Clinical Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Walter M Cronin
- Associate Director, NRG Oncology Statistics and Data Management Center (SDMC); Associate Director, Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA
| | - Erica Field
- Project Specialist III, RTOG, Philadelphia, PA
| | - Evonne Lackey
- Coordinating Center Manager, SWOG Statistical Center, Seattle, WA
| | - Charles D Blanke
- Chair, SWOG; Department of Medicine, Division of Hematology and Medical Oncology, Oregon Health & Science University and Knight Cancer Institute, Portland, OR
| |
Collapse
|
35
|
Mates M, Fletcher GG, Freedman OC, Eisen A, Gandhi S, Trudeau ME, Dent SF. Systemic targeted therapy for her2-positive early female breast cancer: a systematic review of the evidence for the 2014 Cancer Care Ontario systemic therapy guideline. ACTA ACUST UNITED AC 2015; 22:S114-22. [PMID: 25848335 DOI: 10.3747/co.22.2322] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND This systematic review addresses the question "What is the optimal targeted therapy for female patients with early-stage human epidermal growth factor receptor 2 (her2)-positive breast cancer?" METHODS The medline and embase databases were searched for the period January 2008 to May 2014. The Standards and Guidelines Evidence directory of cancer guidelines and the Web sites of major guideline organizations were also searched. RESULTS Sixty publications relevant to the targeted therapy portion of the systematic review were identified. In four major trials (hera, National Surgical Adjuvant Breast and Bowel Project B-31, North Central Cancer Treatment Group N9831, and Breast Cancer International Research Group 006), adjuvant trastuzumab for 1 year was superior in disease-free survival (dfs) and overall survival (os) to no trastuzumab; trastuzumab showed no benefit in one trial (pacs 04). A shorter duration of trastuzumab (less than 1 year compared with 1 year) was evaluated, with mixed results for dfs: one trial showed superiority (finher), one trial could not demonstrate noninferiority (phare), another trial showed equivalent results (E 2198), and one trial is still ongoing (persephone). Longer trastuzumab duration (hera: 2 years vs. 1 year) showed no improvement in dfs or os and a higher rate of cardiac events. Newer her2-targeted agents (lapatinib, pertuzumab, T-DM1, neratinib) have been or are still being evaluated in both adjuvant and neoadjuvant trials, either by direct comparison with trastuzumab alone or combined with trastuzumab. In the neoadjuvant setting (neoaltto, GeparQuinto, Neosphere), trastuzumab alone or in combination with another anti-her2 agent (lapatinib, pertuzumab) was compared with either lapatinib or pertuzumab alone and showed superior or equivalent rates of pathologic complete response. In the adjuvant setting, lapatinib alone or in combination with trastuzumab, compared with trastuzumab alone (altto) or with placebo (teach), was not superior in dfs. The results of the completed aphinity trial, evaluating the role of dual her2 blockade with trastuzumab and pertuzumab, are highly anticipated. Ongoing trials are evaluating trastuzumab as a single agent without adjuvant chemotherapy (respect) and in patients with low her2 expression (National Surgical Adjuvant Breast and Bowel Project B-47). CONCLUSIONS Taking into consideration disease characteristics and patient preference, 1 year of trastuzumab should be offered to all patients with her2-positive breast cancer who are receiving adjuvant chemotherapy. Cardiac function should be regularly assessed in this patient population.
Collapse
Affiliation(s)
- M Mates
- Cancer Centre of Southeastern Ontario, Kinston General Hospital; and Queen's University, Kingston, ON
| | - G G Fletcher
- Program in Evidence-Based Care, Cancer Care Ontario; and Department of Oncology, McMaster University, Hamilton, ON
| | | | - A Eisen
- Sunnybrook Health Science Centre, Toronto, ON
| | - S Gandhi
- Sunnybrook Health Science Centre, Toronto, ON
| | - M E Trudeau
- Sunnybrook Health Science Centre, Toronto, ON
| | - S F Dent
- The Ottawa Hospital Cancer Centre and University of Ottawa, Ottawa, ON
| |
Collapse
|
36
|
Pivot X, Mansi L, Chaigneau L, Montcuquet P, Thiery-Vuillemin A, Bazan F, Dobi E, Sautiere JL, Rigenbach F, Algros MP, Butler S, Jamshidian F, Febbo P, Svedman C, Paget-Bailly S, Bonnetain F, Villanueva C. In the era of genomics, should tumor size be reconsidered as a criterion for neoadjuvant chemotherapy? Oncologist 2015; 20:344-50. [PMID: 25795632 PMCID: PMC4391758 DOI: 10.1634/theoncologist.2014-0198] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 12/18/2014] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The Oncotype DX recurrence score (RS) assay has been validated for prediction of 10-year risk of distant recurrence and likelihood of benefit from chemotherapy in patients with estrogen receptor (ER)-positive, HER2-negative early breast cancer. Patients with high RS tumors have substantial benefit, and patients with low RS tumors have minimal if any benefit from chemotherapy. Tumor size is used as a key parameter when selecting patients for neoadjuvant chemotherapy. The aim of this study was to assess the distribution of RS in patients selected for neoadjuvant chemotherapy primarily according to tumor size. PATIENTS AND METHODS Patients with ER-positive and HER2-negative tumors that were node-negative or had no more than 1 positive node from three trials were included in this study. Oncotype DX was performed at Genomic Health, Inc., blinded to the clinical data. Descriptive statistics were calculated for distribution of RS for all cases. RESULTS Of 277 patients, 96 met eligibility criteria, and 81 had sufficient material for analysis. Median tumor size was 40 mm (interquartile range [IQR], 30-50 mm). Grade I, II, and III were observed in 13, 49, and 17 cases, respectively. There was a wide distribution of RS with a median of 21.4 (IQR, 16.05-26.75). In total, 23 (28.3%) had high, 28 (34.6%) intermediate, and 30 (37%) low RS results. CONCLUSION The RS may provide relevant information for neoadjuvant treatment decisions in select patients both in clinical practice and in studies. Inclusion of low RS disease patients in neoadjuvant trials will likely only dilute the ability to look at treatment effects.
Collapse
Affiliation(s)
- Xavier Pivot
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Laura Mansi
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Loic Chaigneau
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Philippe Montcuquet
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Antoine Thiery-Vuillemin
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Fernando Bazan
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Erion Dobi
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Jean L Sautiere
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Frederic Rigenbach
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Marie P Algros
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Steve Butler
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Farid Jamshidian
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Phillip Febbo
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Christer Svedman
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Sophie Paget-Bailly
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Franck Bonnetain
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| | - Christian Villanueva
- Departments of Medical Oncology, Gynecology, Pathology, and Statistics, University Hospital Jean Minjoz, Besançon, France; University of Franche-Comté, UMR1098, SFR IBCT, Besançon, France; INSERM, UMR1098, Besançon, France; EFS Bourgogne Franche-Comté, UMR1098, Besançon, France; Genomic Health, Inc., Redwood City, California, USA
| |
Collapse
|
37
|
Eisen A, Fletcher G, Gandhi S, Mates M, Freedman O, Dent S, Trudeau M. Optimal systemic therapy for early breast cancer in women: a clinical practice guideline. Curr Oncol 2015; 22:S67-81. [PMID: 25848340 PMCID: PMC4381792 DOI: 10.3747/co.22.2320] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The Breast Cancer Disease Site Group of Cancer Care Ontario identified the need for new guidelines for the adjuvant systemic therapy of early-stage breast cancer. The specific question to be addressed was "What is the optimal adjuvant systemic therapy for female patients with early-stage operable breast cancer, when patient and disease factors are considered?" A systematic review was prepared based on literature searches conducted using the medline and embase databases for the period January 2008 to March 5, 2012, and updated to May 12, 2014. Guidelines were located from that search, from the Standards and Guidelines Evidence directory of cancer guidelines, and from the Web sites of major guideline organizations. The literature located was subdivided into the broad categories of chemotherapy, hormonal therapy, and therapy targeted to her2 (human epidermal growth factor receptor 2). Although several of the systemic therapies discussed in this guideline can be considered in the neoadjuvant setting, the review focused on trials with rates of disease-free and overall survival as endpoints and thus excluded several trials that used pathologic complete response as a primary endpoint. Based on the systematic review, the working group drafted recommendations on the use of chemotherapy, hormonal therapy, and targeted therapy; based on their professional experience, they also drafted recommendations on patient and disease characteristics and recurrence risk. The literature review and draft recommendations were circulated to a consensus panel of medical oncologists who had expertise in breast cancer and who represented the regions of Ontario. Items without initial consensus were discussed at an in-person consensus meeting held in Toronto, November 23, 2012. The final recommendations are those for which consensus was reached before or at the meeting. Some of the key evidence was revised after the updated literature search. Evidence reviews for systemic chemotherapy, endocrine therapy, and targeted therapy for her2-positive disease are reported in separate articles in this supplement. The full three-part 1-21 evidence-based series, including complete details of the development and consensus processes, can be found on the Cancer Care Ontario Web site at https://www.cancercare.on.ca/toolbox/qualityguidelines/diseasesite/breast-ebs.
Collapse
Affiliation(s)
- A. Eisen
- Sunnybrook Health Science Centre, Toronto, ON
| | - G.G. Fletcher
- Program in Evidence-Based Care, Cancer Care Ontario; and Department of Oncology, McMaster University, Hamilton, ON
| | - S. Gandhi
- Sunnybrook Health Science Centre, Toronto, ON
| | - M. Mates
- Cancer Centre of Southeastern Ontario, Kinston General Hospital; and Queen’s University, Kingston, ON
| | | | - S.F. Dent
- The Ottawa Hospital Cancer Centre and University of Ottawa, Ottawa, ON
| | | | - members of the Early Breast Cancer Systemic Therapy Consensus Panel
- P. Bedard, Princess Margaret Hospital, Toronto, ON; N. Califaretti, Grand River Regional Cancer Centre, Kitchener, ON; B. Dhesy, Juravinski Hospital and Cancer Centre, Hamilton, ON; D.A. Dueck, Northwestern Ontario Regional Cancer Centre, Thunder Bay, ON; K. Enright, Peel Regional Cancer Centre, Mississauga, ON; V. Glenns, North York, ON; C. Hamm, Windsor Regional Cancer Centre, Windsor, ON; Y. Madarnas, Department of Oncology, Queen’s University, Kingston, ON; Y. Rahim, Southlake Regional Cancer Centre, Newmarket, ON; S. Rask, Royal Victoria Hospital, Barrie, ON; A. Robinson, Kingston General Hospital, Kingston, ON [formerly Health Sciences North, Sudbury, ON]; S. Spadafora, Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON; S. Verma, The Ottawa Hospital Regional Cancer Centre, Ottawa, ON; J. Younus, London Regional Cancer Program, London, ON
| |
Collapse
|
38
|
Sanguedolce F, Bufo P. HER2 assessment by silver in situ hybridization: where are we now? Expert Rev Mol Diagn 2015; 15:385-98. [PMID: 25578771 DOI: 10.1586/14737159.2015.992416] [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] [Indexed: 01/05/2023]
Abstract
HER2 testing in breast and gastric cancer is critical not only as a prognostic tool but also as a predictive marker for response to the humanized monoclonal antibody trastuzumab. Currently, HER2 status is assessed on histological and cytological specimens by conventional validated methods such as immunohistochemistry and FISH, while bright-field in situ hybridization techniques, such as silver in situ hybridization and chromogenic in situ hybridization, may offer performance benefits over FISH. The major points are first, technical issues, advantages and disadvantages relevant to each methods, and their clinical implications and second, the well-known genetic heterogeneity of HER2, and the occurrence of polysomy of chromosome 17. This review aims to summarize the growing body of literature on the accuracy of bright-field in situ techniques, notably silver in situ hybridization, in assessing HER2 status, and to discuss the role of such methods in pathology practice.
Collapse
|
39
|
Demidova IA, Tsepenshchikova EO, Gikalo MB. [Use of the new 2013 ASCO/CAP guidelines to study HERS status by fluorescence in situ hybridization in patients with breast cancer]. Arkh Patol 2015; 77:43-51. [PMID: 26978020 DOI: 10.17116/patol201577543-51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Despite the long-term history of HER2 testing in breast cancer (BC), the results of its study are frequently interpreted ambiguously. The introduction of the new 2013 ASCO/CAP guidelines is aimed to further optimize HERS testing in view of new scientific evidence for the biological characteristics of BC. OBJECTIVE to study the results of assessing HERS2 status in patients with the questionable level of protein expression according to the 2007 versus 2013 ASCO/CAP criteria. SUBJECTS AND METHODS Biopsy specimens from 572 patients diagnosed with BC after immunohistochemical examination, including HER2 status assessment, were investigated by fluorescence in situ hybridization. RESULTS The use of the new 2013 ASCO/CAP criteria versus the previous criteria to determine intratumor heterogeneity, HER2 gene copy number, and the so-called chromosome 17 polysomy was ascertained to cause a statistically significant increase in HER2-positive cases from 25 to 42.5% (OR=2.2178; 95% CI, 1.8254-2.6950; р=0.0005). CONCLUSION The use of the new guidelines additionally permits effective therapy in patients with BC, which may be another step to improve survival in this large patient group.
Collapse
Affiliation(s)
- I A Demidova
- Moscow City Cancer Hospital Sixty-Two, Moscow Department of Healthcare, Moscow, Russia
| | - E O Tsepenshchikova
- Moscow City Cancer Hospital Sixty-Two, Moscow Department of Healthcare, Moscow, Russia
| | - M B Gikalo
- Moscow City Cancer Hospital Sixty-Two, Moscow Department of Healthcare, Moscow, Russia
| |
Collapse
|
40
|
Lee SB, Lee JW, Yu JH, Ko BS, Kim HJ, Son BH, Gong G, Lee HJ, Kim SB, Jung KH, Ahn JH, Lee W, Sung J, Ahn SH. Preoperative serum HER2 extracellular domain levels in primary invasive breast cancer. BMC Cancer 2014; 14:929. [PMID: 25491647 PMCID: PMC4295268 DOI: 10.1186/1471-2407-14-929] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 12/03/2014] [Indexed: 11/13/2022] Open
Abstract
Background Despite the preclinical outcomes and biologic significance of the presence of the human epidermal growth factor receptor-2 (HER2) extracellular domain (ECD), there is little evidence supporting the measurement of ECD levels in any clinical setting. The aim of this study was to determine the prevalence of elevated serum HER2 ECD levels, the association between these levels and tissue HER2 overexpression, and the potential clinical prognostic value of HER2 ECD in primary invasive breast cancer. Methods Serum HER2 ECD levels were examined preoperatively in 2,862 consecutive stage I–III primary breast cancer patients between 2007 and 2009. Serum HER2 ECD levels were measured by chemiluminescence immunoassay (ADVIA Centaur), and the tissue HER2 status was assessed by immunohistochemistry and fluorescence in situ hybridization. The cutoff value for the serum level of HER2 ECD was set at 15.2 ng/ml. Results Among the 2,862 patients, 126 (4.4%) had elevated serum HER2 ECD levels, and HER2 was overexpressed in the tumor tissue of 692 patients (24.2%), with a concordance of 78.7%. Multivariate analysis revealed that elevated serum HER2 ECD was a significant independent prognostic factor for worse distant-metastasis-free survival [DMFS; hazard ratio (HR) = 2.50, 95% confidence interval (CI) = 1.5–4.3, P = 0.001] and breast-cancer-specific survival (BCSS; HR = 2.0, 95% CI = 1.1–3.8, P = 0.036), which were much stronger in patients with tissue HER2-positive tumors (DMFS: HR = 3.8, 95% CI = 2.0–7.0, P < 0.001; BCSS: HR = 2.6, 95% CI = 1.2-5.3, P = 0.012). Conclusions Given the prevalence of HER2 expression, its measurement as an independent prognostic factor can be clinically useful, particularly in patients with tissue HER2-positive tumors.
Collapse
Affiliation(s)
| | - Jong Won Lee
- Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, 388-1 Pungnap 2 dong, Songpa-Gu, Seoul 138-736, Korea.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Solier C, Langen H. Antibody-based proteomics and biomarker research - current status and limitations. Proteomics 2014; 14:774-83. [PMID: 24520068 DOI: 10.1002/pmic.201300334] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/08/2013] [Accepted: 12/16/2013] [Indexed: 11/09/2022]
Abstract
Antibody-based proteomics play a very important role in biomarker discovery and validation, facilitating the high-throughput evaluation of candidate markers. Most proteomics-driven discovery is nowadays based on the use of MS. MS has many advantages, including its suitability for hypothesis-free biomarker discovery, since information on protein content of a sample is not required prior to analysis. However, MS presents one main caveat which is the limited sensitivity in complex samples, especially for body fluids, where protein expression covers a huge dynamic range. Antibody-based technologies remain the main solution to address this challenge since they reach higher sensitivity. In this article, we review the benefits and limitations of antibody-based proteomics in preclinical and clinical biomarker research for discovery and validation in body fluids and tissue. The combination of antibodies and MS, utilizing the best of both worlds, opens new avenues in biomarker research.
Collapse
Affiliation(s)
- Corinne Solier
- Translational Technologies and Bioinformatics, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Basel, Switzerland
| | | |
Collapse
|
42
|
Cheng H, Ballman K, Vassilakopoulou M, Dueck AC, Reinholz MM, Tenner K, Gralow J, Hudis C, Davidson NE, Fountzilas G, McCullough AE, Chen B, Psyrri A, Rimm DL, Perez EA. EGFR expression is associated with decreased benefit from trastuzumab in the NCCTG N9831 (Alliance) trial. Br J Cancer 2014; 111:1065-71. [PMID: 25117817 PMCID: PMC4453859 DOI: 10.1038/bjc.2014.442] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 06/23/2014] [Accepted: 07/13/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) has been hypothesised to modulate the effectiveness of anti-HER2 therapy. We used a standardised, quantitative immunofluorescence assay and a novel EGFR antibody to evaluate the correlation between EGFR expression and clinical outcome in the North Central Cancer Treatment Group (NCCTG) N9831 trial. METHODS Tissue microarrays were constructed that allowed analysis of 1365 patients randomly assigned to receive chemotherapy alone (Arm A), sequential trastuzumab after chemotherapy (Arm B) and chemotherapy with concurrent trastuzumab (Arm C). Measurement of EGFR was performed using the EGFR antibody, D38B1, on the fluorescence-based AQUA platform. The result was validated using an independent retrospective metastatic breast cancer cohort (n=130). RESULTS Epidermal growth factor receptor assessed as a continuous (logarithmic transformed) variable shows an association with disease-free survival in Arm C (P=0.009) but not in Arm A or B. High EGFR expression was associated with worse outcome (Hazard ratio (HR)=2.15; 95% CI 1.28-3.60, P=0.004). Validation in a Greek metastatic breast cancer cohort showed an HR associated with high EGFR expression of 1.92 (P=0.0073). CONCLUSIONS High expression of EGFR appears to be associated with decreased benefit from adjuvant concurrent trastuzumab. Since other treatment options exist for HER2-driven tumours, further validation of these data may select patients for alternative or additive therapy.
Collapse
Affiliation(s)
- H Cheng
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street BML116, New Haven, CT 06520, USA
| | - K Ballman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - M Vassilakopoulou
- Department of Medical Oncology, Pitie-Salpetriere Hospital, 83 Boulevard de l'Hôpital, 75013 Paris, France
| | - A C Dueck
- Section of Biostatistics, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| | - M M Reinholz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - K Tenner
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - J Gralow
- Department of Medical Oncology, University of Washington/Seattle Cancer Care Alliance, 825 Eastlake Avenue East, Seattle, WA 98109, USA
| | - C Hudis
- Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 8, New York, NY 10065, USA
| | - N E Davidson
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute and UPMC Cancer Center, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
| | - G Fountzilas
- Department of Medical Oncology, Papageorgiou General Hospital, Aristotle University of Thessaloniki School of Medicine, Efkarpia Peripheral Road Stavroupoli, 56429 Thessaloniki, Greece
- Hellenic Cooperative Oncology Group (HeCOG), Laskaridou 1, 11524 Athens, Greece
| | - A E McCullough
- Anatomic Pathology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ 85259, USA
| | - B Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - A Psyrri
- Hellenic Cooperative Oncology Group (HeCOG), Laskaridou 1, 11524 Athens, Greece
- Second Department of Internal Medicine Propaedeutic, Oncology Section, Attikon University Hospital, University of Athens Medical School, 1 Rimini Street, Haidari, 12462 Athens, Greece
| | - D L Rimm
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street BML116, New Haven, CT 06520, USA
| | - E A Perez
- Department of Hematology/Oncology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
43
|
Vassilakopoulou M, Togun T, Dafni U, Cheng H, Bordeaux J, Neumeister VM, Bobos M, Pentheroudakis G, Skarlos DV, Pectasides D, Kotoula V, Fountzilas G, Rimm DL, Psyrri A. In situ quantitative measurement of HER2mRNA predicts benefit from trastuzumab-containing chemotherapy in a cohort of metastatic breast cancer patients. PLoS One 2014; 9:e99131. [PMID: 24968015 PMCID: PMC4072595 DOI: 10.1371/journal.pone.0099131] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 05/11/2014] [Indexed: 12/04/2022] Open
Abstract
Background We sought to determine the predictive value of in situ mRNA measurement compared to traditional methods on a cohort of trastuzumab-treated metastatic breast cancer patients. Methods A tissue microarray composed of 149, classified as HER2-positive, metastatic breast cancers treated with various trastuzumab-containing chemotherapy regimens was constructed. HER2 intracellular domain(ICD), HER2 extracellular domain(ECD) and HER2 mRNA were assessed using AQUA. For HER2 protein evaluation, CB11 was used to measure ICD and SP3 to measure ECD of the HER2 receptor. In addition, HER2 mRNA status was assessed using RNAscope assay ERRB2 probe. Kaplan – Meier estimates were used for depicting time-to-event endpoints. Multivariate Cox regression models with backward elimination were used to assess the performance of markers as predictors of TTP and OS, after adjusting for important covariates. Results HER2 mRNA was correlated with ICD HER2, as measured by CB11 HER2, with ECD HER2 as measured by SP3 (Pearson’s Correlation Coefficient, r = 0.66 and 0.51 respectively) and with FISH HER2 (Spearman’s Correlation Coefficient, r = 0.75). All markers, HER2 mRNA, ICD HER2 and ECD HER2, along with FISH HER2, were found prognostic for OS (Log-rank p = 0.007, 0.005, 0.009 and 0.043 respectively), and except for FISH HER2, they were also prognostic for TTP Log-rank p = 0.036, 0.068 and 0.066 respectively) in this trastuzumab- treated cohort. Multivariate analysis showed that in the presence of pre-specified set of prognostic factors, among all biomarkers only ECD HER2, as measured by SP3, is strong prognostic factor for both TTP (HR = 0.54, 95% CI: 0.31–0.93, p = 0.027) and OS (HR = 0.39, 95%CI: 0.22–0.70, p = 0.002). Conclusions The expression of HER2 ICD and ECD as well as HER2 mRNA levels was significantly associated with TTP and OS in this trastuzumab-treated metastatic cohort. In situ assessment of HER2 mRNA has the potential to identify breast cancer patients who derive benefit from Trastuzumab treatment.
Collapse
Affiliation(s)
- Maria Vassilakopoulou
- Yale University, School of Medicine, Department of Pathology, New Haven, Connecticut, United States of America
| | - Taiwo Togun
- Yale University, School of Public Health, Department of Biostatistics, New Haven, Connecticut, United States of America
| | - Urania Dafni
- Laboratory of Biostatistics, University of Athens School of Nursing, Athens, Greece
| | - Huan Cheng
- Yale University, School of Medicine, Department of Pathology, New Haven, Connecticut, United States of America
| | - Jennifer Bordeaux
- Yale University, School of Medicine, Department of Pathology, New Haven, Connecticut, United States of America
| | - Veronique M. Neumeister
- Yale University, School of Medicine, Department of Pathology, New Haven, Connecticut, United States of America
| | - Mattheos Bobos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | | | | | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, “Hippokration” Hospital, Athens, Greece
| | - Vassiliki Kotoula
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
- Department of Medical Oncology, “Papageorgiou” Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - David L. Rimm
- Yale University, School of Medicine, Department of Pathology, New Haven, Connecticut, United States of America
| | - Amanda Psyrri
- Division of Oncology, Second Department of Internal Medicine, University of Athens School of Medicine, Attikon University Hospital, Athens, Greece
- * E-mail:
| |
Collapse
|
44
|
Gannon LM, Cotter MB, Quinn CM. The classification of invasive carcinoma of the breast. Expert Rev Anticancer Ther 2014; 13:941-54. [PMID: 23984896 DOI: 10.1586/14737140.2013.820577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The classification of invasive breast carcinoma assists diagnosis, allows for comparison of different patient groups in clinical trials and facilitates epidemiological analysis. For the individual patient, accurate tumor classification informs clinical decision-making with emphasis on assessment of prognosis and treatment formulation. Tumor grade is an independent prognostic indicator and is calculated by assessing specific tumor characteristics microscopically. The Tumor Node Metastasis staging system, produced by the American Joint Committee on Cancer Union for International Cancer Control, combines information about the primary tumor size, the status of the regional lymph nodes and the presence or absence of distant metastases at diagnosis to classify disease. In recent years, the use of gene expression profiling technology has led to the development of the molecular classification of breast cancer and has highlighted the importance of hormone receptor and HER2 oncogenic pathways, with particular reference to targeted chemotherapy. Tumor typing involves the identification of 'no special type' carcinoma with variable clinical, histological and molecular characteristics and 'special type' carcinomas that are usually associated with a particular set of prognostic and predictive indices. Some special type carcinomas have unique biological features that influence diagnostic investigation and clinical management.
Collapse
Affiliation(s)
- Louisa M Gannon
- Medical student, Health Sciences Building, University College, Dublin 4, Ireland
| | | | | |
Collapse
|
45
|
Perez EA, Cortés J, Gonzalez-Angulo AM, Bartlett JM. HER2 testing: Current status and future directions. Cancer Treat Rev 2014; 40:276-84. [DOI: 10.1016/j.ctrv.2013.09.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 08/19/2013] [Accepted: 09/02/2013] [Indexed: 11/30/2022]
|
46
|
Wolff AC, Hammond MEH, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JMS, Bilous M, Fitzgibbons P, Hanna W, Jenkins RB, Mangu PB, Paik S, Perez EA, Press MF, Spears PA, Vance GH, Viale G, Hayes DF. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. Arch Pathol Lab Med 2014; 138:241-56. [PMID: 24099077 PMCID: PMC4086638 DOI: 10.5858/arpa.2013-0953-sa] [Citation(s) in RCA: 813] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To update the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guideline recommendations for human epidermal growth factor receptor 2 (HER2) testing in breast cancer to improve the accuracy of HER2 testing and its utility as a predictive marker in invasive breast cancer. METHODS ASCO/CAP convened an Update Committee that included coauthors of the 2007 guideline to conduct a systematic literature review and update recommendations for optimal HER2 testing. RESULTS The Update Committee identified criteria and areas requiring clarification to improve the accuracy of HER2 testing by immunohistochemistry (IHC) or in situ hybridization (ISH). The guideline was reviewed and approved by both organizations. RECOMMENDATIONS The Update Committee recommends that HER2 status (HER2 negative or positive) be determined in all patients with invasive (early stage or recurrence) breast cancer on the basis of one or more HER2 test results (negative, equivocal, or positive). Testing criteria define HER2-positive status when (on observing within an area of tumor that amounts to >10% of contiguous and homogeneous tumor cells) there is evidence of protein overexpression (IHC) or gene amplification (HER2 copy number or HER2/CEP17 ratio by ISH based on counting at least 20 cells within the area). If results are equivocal (revised criteria), reflex testing should be performed using an alternative assay (IHC or ISH). Repeat testing should be considered if results seem discordant with other histopathologic findings. Laboratories should demonstrate high concordance with a validated HER2 test on a sufficiently large and representative set of specimens. Testing must be performed in a laboratory accredited by CAP or another accrediting entity. The Update Committee urges providers and health systems to cooperate to ensure the highest quality testing.
Collapse
Affiliation(s)
- Antonio C Wolff
- Antonio C. Wolff, Johns Hopkins Kimmel Comprehensive Cancer Center, Baltimore; Lisa M. McShane, National Cancer Institute, Bethesda, MD; M. Elizabeth H. Hammond, University of Utah School of Medicine and Intermountain Healthcare, Salt Lake City, UT; David G. Hicks, University of Rochester Medical Center, Rochester, NY; Mitch Dowsett, Royal Marsden Hospital, London, United Kingdom; Kimberly H. Allison, Stanford University Medical Center, Stanford; Patrick Fitzgibbons, St Jude Medical Center, Fullerton; Michael F. Press, University of Southern California, Los Angeles, CA; Donald C. Allred, Washington University School of Medicine, St Louis, MO; John M.S. Bartlett, Ontario Institute for Cancer Research; Wedad Hanna, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada; Michael Bilous, University of Western Sydney and Healthscope Pathology, Sydney, New South Wales, Australia; Robert B. Jenkins, Mayo Clinic, Rochester, MN; Pamela B. Mangu, American Society of Clinical Oncology, Alexandria, VA; Soonmyung Paik, National Surgical Adjuvant Breast and Bowel Project, Pitsburgh, PA; Edith A. Perez, Mayo Clinic, Jacksonville, FL; Patricia A. Spears, North Carolina State University, Raleigh, NC; Gail H. Vance, Indiana University Medical Center, Indianapolis, IN; Giuseppe Viale, University of Milan, European Institute of Oncology, Milan, Italy; and Daniel F. Hayes, University of Michigan Comprehensive Cancer Care Center, Ann Arbor, MI
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Central pathology laboratory review of HER2 and ER in early breast cancer: an ALTTO trial [BIG 2-06/NCCTG N063D (Alliance)] ring study. Breast Cancer Res Treat 2014; 143:485-92. [PMID: 24395109 DOI: 10.1007/s10549-013-2827-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Choice of therapy for breast cancer relies on human epidermal growth factor receptor-2 (HER2) and estrogen receptor α (ER) status. Before randomization in the phase III Adjuvant Lapatinib and/or Trastuzumab Treatment Optimisation (ALTTO) trial for HER2-positive disease, HER2 and ER were centrally reviewed by Mayo Clinic (Rochester, MN, and Scottsdale, AZ) for North America and by the European Institute of Oncology (IEO; Milan, Italy) for the rest of world (except China). Discordance rates (local vs. central review) differed between Mayo and IEO. Among locally HER2-positive cases, 5.8 % (Mayo) and 14.5 % (IEO) were centrally HER2 negative. Among locally ER-positive cases, 16.2 % (Mayo) and 4.2 % (IEO) were centrally ER-negative. Among locally ER-negative cases, 3.4 % (Mayo) and 21.4 % (IEO) were centrally ER-positive. We, therefore, performed a ring study to identify features contributing to these differing discordance rates. Mayo and IEO exchanged slides for 25 HER2 and 35 ER locally/centrally discordant cases. Both laboratories performed IHC and FISH for HER2 using the HercepTest(®) and PathVysion HER2 DNA probe kit/HER2/centromere 17 probe mixture. IHC for ER was tested centrally using the monoclonal ER 1D5 antibody (Mayo) or the DAKO cocktail of ER 1D5 and 2.123 antibodies (IEO). Mayo and IEO confirmed the central HER2-negative result in 100 % of 25 cases. Mayo and IEO confirmed the central ER result in 29 (85 %) of 34 evaluable cases. The five Mayo-negative/IEO-positive cases were ER-positive when retested at Mayo using the DAKO ER cocktail. In this ring study, ALTTO ineligibility did not change when HER2 testing was performed by either IEO or Mayo central laboratories. However, a dual antibody ER assay had fewer false-negative test results than an assay with a single antibody, and there was more discordance between the two ER reagents than has been previously reported. Using even slightly different assay methods yielded different results, even between experienced central laboratories.
Collapse
|
48
|
HER2 in situ hybridization in breast cancer: clinical implications of polysomy 17 and genetic heterogeneity. Mod Pathol 2014; 27:4-18. [PMID: 23807776 DOI: 10.1038/modpathol.2013.103] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/05/2013] [Accepted: 04/11/2013] [Indexed: 12/19/2022]
Abstract
Trastuzumab-containing therapy is a standard of care for patients with HER2+ breast cancer. HER2 status is routinely assigned using in situ hybridization to assess HER2 gene amplification, but interpretation of in situ hybridization results may be challenging in tumors with chromosome 17 polysomy or intratumoral genetic heterogeneity. Apparent chromosome 17 polysomy, defined by increased chromosome enumeration probe 17 (CEP17) signal number, is a common genetic aberration in breast cancer and represents an alternative mechanism for increasing HER2 copy number. Some studies have linked elevated CEP17 count ('polysomy') with adverse clinicopathologic features and HER2 overexpression, although there are numerous discrepancies in the literature. There is evidence that elevated CEP17 ('polysomy') count might account for trastuzumab response in tumors with normal HER2:CEP17 ratios. Nonetheless, recent studies establish that apparent 'polysomy' (CEP17 increase) is usually related to focal pericentromeric gains rather than true polysomy. Assigning HER2 status may also be complex where multiple cell subclones with distinct HER2 amplification characteristics coexist within the same tumor. Such genetic heterogeneity affects up to 40% of breast cancers when assessed according to a College of American Pathologists guideline, although other definitions have been proposed. Recent data have associated heterogeneity with unfavorable clinicopathologic variables and poor prognosis. Genetically heterogeneous tumors harboring HER2-amplified subclones have the potential to benefit from trastuzumab, but this has yet to be evaluated in clinical studies. In this review, we discuss the implications of apparent polysomy 17 and genetic heterogeneity for assigning HER2 status in clinical practice. Among our recommendations, we support the use of mean HER2 copy number rather than HER2:CEP17 ratio to define HER2 positivity in cases where coamplification of the centromere might mask HER2 amplification. We also highlight a need to harmonize in situ hybridization scoring methodology to support accurate HER2 status determination, particularly where there is evidence of heterogeneity.
Collapse
|
49
|
Press OA, Guzman R, Cervantes M, Santiago A, Press MF. Characterization of HER2 status by fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC). Methods Mol Biol 2014; 1180:181-207. [PMID: 25015148 DOI: 10.1007/978-1-4939-1050-2_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The use of human epidermal growth factor receptor type 2 (HER2) gene amplification and overexpression as a molecular predictive marker has become critically important for proper selection of breast cancer patients for treatment with targeted therapeutic agents such as trastuzumab, lapatinib, pertuzumab, and T-DM1. A high level of sensitivity and specificity of molecular tests for this alteration is desirable. The American Society of Clinical Oncology and College of American Pathology have jointly established consensus guidelines to standardize characterization of this alteration in breast cancers. This chapter provides a brief overview of pre-analytic and analytical processing of breast specimens as well as subsequent molecular evaluation for HER2 status.
Collapse
Affiliation(s)
- Oliver A Press
- Department of Pathology, USC Keck School of Medicine, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
50
|
Affiliation(s)
- Ian E Krop
- Affiliation of authors: Dana-Farber Cancer Institute, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|