1
|
Goulas K, Farmakis D, Constantinidou A, Kadoglou NPE. Cardioprotective Agents for the Primary Prevention of Trastuzumab-Associated Cardiotoxicity: A Systematic Review and Meta-Analysis. Pharmaceuticals (Basel) 2023; 16:983. [PMID: 37513895 PMCID: PMC10383255 DOI: 10.3390/ph16070983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
There are significant considerations about the prevention of cardiotoxicity caused by trastuzumab therapy in patients with breast cancer, leading to discontinuation. Recently, randomized controlled trials (RCTs) have evaluated the effects of early commitment of beta-blockers (BBs), angiotensin receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs) during trastuzumab chemotherapy in order to prevent the related cardiotoxicity. The present systematic review and meta-analysis of six RCTs included patients who have predominantly non-metastatic, HER2-positive, breast cancer and received trastuzumab as primary or adjuvant therapy. Those patients did not have any obvious cardiac dysfunction or any previous therapy with cardioprotective agent. We evaluated the efficacy of the aforementioned medications for primary prevention of cardiotoxicity, using random effects models. Any preventive treatment did not reduce cardiotoxicity occurrence compared to controls (Odds ratios (OR) = 0.92, 95% CI 0.54-1.56, p = 0.75). Results were similar for ACEIs/ARBs and beta-blockers. Treatment with ACEIs/ARBs led to a slight, but significant, increase in LVEF in patients compared to the placebo group. Only two studies reported less likelihood of discontinuation of trastuzumab treatment. More adequately powered RCTs are needed to determine the efficacy of routine prophylactic therapy.
Collapse
|
2
|
Strand SH, Rivero-Gutiérrez B, Houlahan KE, Seoane JA, King LM, Risom T, Simpson LA, Vennam S, Khan A, Cisneros L, Hardman T, Harmon B, Couch F, Gallagher K, Kilgore M, Wei S, DeMichele A, King T, McAuliffe PF, Nangia J, Lee J, Tseng J, Storniolo AM, Thompson AM, Gupta GP, Burns R, Veis DJ, DeSchryver K, Zhu C, Matusiak M, Wang J, Zhu SX, Tappenden J, Ding DY, Zhang D, Luo J, Jiang S, Varma S, Anderson L, Straub C, Srivastava S, Curtis C, Tibshirani R, Angelo RM, Hall A, Owzar K, Polyak K, Maley C, Marks JR, Colditz GA, Hwang ES, West RB. Molecular classification and biomarkers of clinical outcome in breast ductal carcinoma in situ: Analysis of TBCRC 038 and RAHBT cohorts. Cancer Cell 2022; 40:1521-1536.e7. [PMID: 36400020 PMCID: PMC9772081 DOI: 10.1016/j.ccell.2022.10.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/29/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022]
Abstract
Ductal carcinoma in situ (DCIS) is the most common precursor of invasive breast cancer (IBC), with variable propensity for progression. We perform multiscale, integrated molecular profiling of DCIS with clinical outcomes by analyzing 774 DCIS samples from 542 patients with 7.3 years median follow-up from the Translational Breast Cancer Research Consortium 038 study and the Resource of Archival Breast Tissue cohorts. We identify 812 genes associated with ipsilateral recurrence within 5 years from treatment and develop a classifier that predicts DCIS or IBC recurrence in both cohorts. Pathways associated with recurrence include proliferation, immune response, and metabolism. Distinct stromal expression patterns and immune cell compositions are identified. Our multiscale approach employed in situ methods to generate a spatially resolved atlas of breast precancers, where complementary modalities can be directly compared and correlated with conventional pathology findings, disease states, and clinical outcome.
Collapse
MESH Headings
- Humans
- Female
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Disease Progression
- Breast Neoplasms/pathology
- Biomarkers
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/analysis
Collapse
Affiliation(s)
- Siri H Strand
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Belén Rivero-Gutiérrez
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kathleen E Houlahan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jose A Seoane
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Lorraine M King
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA
| | - Tyler Risom
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lunden A Simpson
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA
| | - Sujay Vennam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Luis Cisneros
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Timothy Hardman
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA
| | - Bryan Harmon
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA; TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA
| | - Fergus Couch
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Kristalyn Gallagher
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark Kilgore
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Shi Wei
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Angela DeMichele
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tari King
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Breast Oncology Program, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Priscilla F McAuliffe
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Julie Nangia
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston TX 77030, USA
| | - Joanna Lee
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Surgery, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer Tseng
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Anna Maria Storniolo
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Alastair M Thompson
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston TX 77030, USA; Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gaorav P Gupta
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Robyn Burns
- TBCRC Loco-Regional Working Group, Baltimore, MD 21287, USA; TBCRC, The EMMES Corporation, Rockville, MD 20850, USA
| | - Deborah J Veis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63108, USA; Departments of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Katherine DeSchryver
- Departments of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Chunfang Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Magdalena Matusiak
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jason Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shirley X Zhu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jen Tappenden
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daisy Yi Ding
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Dadong Zhang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27708, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shu Jiang
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren Anderson
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA
| | - Cody Straub
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA
| | - Sucheta Srivastava
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christina Curtis
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Rob Tibshirani
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Robert Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Allison Hall
- Department of Pathology, Duke University School of Medicine, Durham, NC 27708, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27708, USA; Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC 27708, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Carlo Maley
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Jeffrey R Marks
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA
| | - Graham A Colditz
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University School of Medicine, Durham, NC 27708, USA.
| | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Zhang H, Katerji H, Turner BM, Hicks DG. HER2-Low Breast Cancers. Am J Clin Pathol 2022; 157:328-336. [PMID: 34519765 DOI: 10.1093/ajcp/aqab117] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Recent clinical trials have demonstrated significant clinical benefits from novel therapeutic compounds in breast cancer patient with human epidermal growth factor receptor 2 (HER2) immunohistochemical (IHC) score of 1+ or 2+ and negative in situ hybridization (ISH) result. A new concept of "HER2-low" breast cancer has been proposed and applied in the recent and ongoing clinical trials. In this article, we review the literature on the topic of HER2-low breast cancer. METHODS A literature search in PubMed was performed using key words related to HER2-low breast cancer. Major relevant studies that were presented in international breast cancer conferences were also included. RESULTS HER2-low breast cancer is currently defined as breast cancer with HER2 IHC score of 1+ or 2+ and negative ISH result. It likely represents a group of tumors with significant biological heterogeneity. Reports of clinical activity using the next generation of HER2-targeting antibody-drug conjugates in HER2-low breast cancers suggest that some strategies of targeting HER2 might be effective in this patient population while raising considerable concerns over limitations in our current testing methodologies and our ability to accurately identify such patients. CONCLUSIONS The promising efficacy of novel HER2-targeted therapy in advanced HER2-low breast cancers has raised the possibility for changing the clinical interpretation of HER2 status in breast cancer to include a HER2-low category; however, the definition of HER2-low breast cancer, the corresponding reliable and accurate quantitative HER2 testing methodology, and the biology of HER2-low breast cancer remain poorly defined.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Hani Katerji
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| | - David G Hicks
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
4
|
Suteau V, Bukasa-Kakamba J, Virjogh-Cenciu B, Adenis A, Sabbah N, Drak Alsibai K. Pathological Significance of GLUT-1 Expression in Breast Cancer Cells in Diabetic and Obese Patients: The French Guiana Study. Cancers (Basel) 2022; 14:cancers14020437. [PMID: 35053598 PMCID: PMC8774256 DOI: 10.3390/cancers14020437] [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: 10/28/2021] [Revised: 12/09/2021] [Accepted: 01/12/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary This study describes the clinical, histological, and molecular features of breast cancer in French Guiana, and characterizes the expression of the tumor metabolic marker GLUT-1 in breast cancers cells in diabetic and obese patients compared to a control group. This study reveals an overall overexpression of GLUT-1 in 60% of invasive breast carcinomas and in all medullary pattern and carcinoma in situ lesions. Our results highlight the potential role of GLUT-1 as a tumor metabolic prognostic marker and also as an interesting target therapy, independently of patient metabolic disorder. Abstract The prevalence of obesity and type 2 diabetes is higher in French Guiana compared to mainland France. These metabolic disorders are associated with an increased risk of cancer. One of the factors involved is hyperinsulinemia that promotes the action of glucose transporter 1 (GLUT-1). The objective of this study is to characterize the expression of GLUT-1 in breast cancers cells in diabetic and obese patients compared to those who are not and to describe the clinical and histological prognostic factors of breast cancer in this population. We conducted a monocentric study including patients with breast cancer diagnosed between 2014 and 2020. Patients were classified into three groups: diabetes, obesity, and control group. The GLUT-1 expression was assessed by immunohistochemistry. In total, 199 patients were included in this study. The median age was 53.5 years, and the median tumor size was 2.8 cm. Luminal A was the most frequent molecular type (58.1%), followed by the triple-negative type (19.9%). The breast cancer in our population was characterized by a younger age at diagnosis, more aggressive molecular types, and larger tumor size. Thus, we suggest the advancement of the age of breast cancer screening in this territory. A total of 144 patients (31 diabetes, 22 obese, and 91 control group) were included for the study of GLUT-1 expression. Overexpression of GLUT-1 was observed in 60.4% of cases and in all carcinoma in situ lesions. GLUT-1 overexpression was associated with more aggressive cancers. This overexpression is correlated with high histological grade, high proliferation index, and aggressive molecular types. Our study found no difference in GLUT-1 expression between the diabetic or obese patients and the control group. These results highlight the potential role of GLUT-1 as a tumor metabolic prognostic marker and also as an interesting target therapy, independently of patient metabolic disorder.
Collapse
Affiliation(s)
- Valentin Suteau
- Department of Pathology, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
| | - John Bukasa-Kakamba
- Department of Endocrinology and Metabolic Diseases, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana; (J.B.-K.); (N.S.)
| | - Beatrice Virjogh-Cenciu
- Department of Medicine, Hôpital de jour Adults, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
| | - Antoine Adenis
- Clinical Investigation Center Antilles French Guiana (CIC INSERM 1424), Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
| | - Nadia Sabbah
- Department of Endocrinology and Metabolic Diseases, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana; (J.B.-K.); (N.S.)
| | - Kinan Drak Alsibai
- Department of Pathology, Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana;
- Center of Biological Resources (CRB Amazonie), Cayenne Hospital Center André Rosemon, F-97306 Cayenne, French Guiana
- Correspondence: ; Tel.: +594-594395231
| |
Collapse
|
5
|
Hurvitz SA, McAndrew NP, Bardia A, Press MF, Pegram M, Crown JP, Fasching PA, Ejlertsen B, Yang EH, Glaspy JA, Slamon DJ. A careful reassessment of anthracycline use in curable breast cancer. NPJ Breast Cancer 2021; 7:134. [PMID: 34625570 PMCID: PMC8501074 DOI: 10.1038/s41523-021-00342-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
It has been over three decades since anthracyclines took their place as the standard chemotherapy backbone for breast cancer in the curative setting. Though the efficacy of anthracycline chemotherapy is not debatable, potentially life-threatening and long-term risks accompany this class of agents, leading some to question their widespread use, especially when newer agents with improved therapeutic indices have become available. Critically assessing when to incorporate an anthracycline is made more relevant in an era where molecular classification is enabling not only the development of biologically targeted therapeutics but also is improving the ability to better select those who would benefit from cytotoxic agents. This comprehensive analysis will present the problem of overtreatment in early-stage breast cancer, review evidence supporting the use of anthracyclines in the pre-taxane era, analyze comparative trials evaluating taxanes with or without anthracyclines in biologically unselected and selected patient populations, and explore published work aimed at defining anthracycline-sensitive tumor types.
Collapse
Affiliation(s)
- Sara Alsterlind Hurvitz
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - Nicholas P. McAndrew
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - Aditya Bardia
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Michael F. Press
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Mark Pegram
- Stanford Comprehensive Cancer Institute, Palo Alto, CA USA
| | - John P. Crown
- grid.412751.40000 0001 0315 8143Department of Medical Oncology, St. Vincent’s University Hospital, Dublin, Ireland
| | - Peter A. Fasching
- grid.411668.c0000 0000 9935 6525Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Bent Ejlertsen
- grid.4973.90000 0004 0646 7373Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eric H. Yang
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - John A. Glaspy
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - Dennis J. Slamon
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| |
Collapse
|
6
|
Zhang H, Moisini I, Turner BM, Wang X, Dhakal A, Yang Q, Kovar S, Schiffhauer LM, Hicks DG. Significance of HER2 in Microinvasive Breast Carcinoma. Am J Clin Pathol 2021; 156:155-165. [PMID: 33491064 DOI: 10.1093/ajcp/aqaa222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES We compared the clinicopathologic features, clinical management, and outcomes of human epidermal growth factor receptor 2 (HER2)-expressing and nonexpressing microinvasive breast carcinomas (MiBC) to explore the significance of HER2 in MiBC. METHODS Clinicopathologic and follow-up information of cases with final diagnosis of MiBC with known HER2 status between 2007 and 2019 were analyzed. RESULTS Nineteen (41.3%) HER2-positive (HER2+) and 27 (58.7%) HER2-negative (HER2-) MiBCs were identified. HER2 positivity was likely to be associated with high nuclear grade, presence of tumor-infiltrating lymphocytes, hormonal receptor negativity, and increased Ki-67 in both microinvasive and associated in situ carcinomas. Nodal metastases were found in 2 ER+/HER2- cases (5.3%). One HER2+ case was found to have isolated tumor cells in the axillary node. The majority of patients with HER2+ MiBCs (76.5%) did not receive HER2-targeted therapy. All patients with available follow-up were alive without recurrence or distant metastasis, with a median follow-up of 38 months. CONCLUSIONS Similar to the larger size of invasive breast carcinomas, HER2 positivity is associated with high-grade morphologic features in MiBCs. However, HER2 overexpression in MiBCs does not appear to be associated with nodal metastasis or worse outcome in our study cohort. The role of HER2-targeted therapy in this clinical setting merits additional study.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Ioana Moisini
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Xi Wang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Ajay Dhakal
- Department of Hematology and Oncology, University of Rochester Medical Center, Rochester, NY
| | - Qi Yang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Sierra Kovar
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Linda M Schiffhauer
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - David G Hicks
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
7
|
Verma A, Kar AG, Meena RN, Patne SCU, Mishra SP, Khanna S, Khanna R. Evaluation of Estrogen Receptor, Progesterone Receptor, and Human Epidermal Growth Factor Receptor 2 Status Before and After Neoadjuvant Chemotherapy in Breast Cancer Patients. Indian J Surg 2021. [DOI: 10.1007/s12262-020-02380-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
8
|
The Presence of an In Situ Component on Pre-Treatment Biopsy Is Not Associated with Response to Neoadjuvant Chemotherapy for Breast Cancer. Cancers (Basel) 2021; 13:cancers13020235. [PMID: 33435265 PMCID: PMC7827327 DOI: 10.3390/cancers13020235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/25/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Identifying markers predictive of response and resistance to neoadjuvant chemotherapy (NAC) has become a major research objective. Ductal carcinoma in situ (DCIS) is associated with invasive disease in more than half of invasive breast cancer cases. It is generally assumed that DCIS does not respond to NAC, but the effect of chemotherapy on in situ components has been little studied. We assessed the predictive value of the presence of an in situ component on pre-NAC biopsy on pathological complete response (pCR) in a real-life cohort of patients treated by NAC. We included 1148 patients; 44% of tumors were luminal (n = 508), 31% triple negative breast cancer (TNBC) (n = 359) and 24% HER2-positive (n = 281). DCIS was found in 225 samples (19.6%) before NAC. The presence of a DCIS component on pre-NAC biopsy was not associated with pCR and did not seem to be a critical factor for the prediction of response to NAC. Abstract A ductal in situ (DCIS) component is often associated with invasive breast carcinoma (BC), and its effect on response to treatment is unknown. We assessed the predictive value of the DCIS component for pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC). We analyzed a cohort of 1148 T1–3NxM0 breast cancer (BC) patients treated by NAC at Institut Curie between 2002 and 2012. The presence of a DCIS component was retrospectively recorded from both the pre-NAC biopsy pathological report and surgical specimens. We included 1148 BC patients treated by NAC for whom pre- and post-NAC data concerning the in situ component were available. DCIS was present before NAC in 19.6% of the population. Overall, 283 patients (19.4%) achieved pCR after NAC. There was no significant association between the presence of DCIS on pre-NAC biopsy and pCR. In a multivariate analysis including subtype, tumor size, grade, mitotic index, and Ki67 index, only BC subtype (luminal/TNBC/HER2-positive) and Ki67 were significantly associated with pCR. The presence of a DCIS component on pre-NAC biopsy is not associated with pCR and does not seem to be a critical factor for predicting response to NAC.
Collapse
|
9
|
Jacob S, Davis AA, Gerratana L, Velimirovic M, Shah AN, Wehbe F, Katam N, Zhang Q, Flaum L, Siziopikou KP, Platanias LC, Gradishar WJ, Behdad A, Bardia A, Cristofanilli M. The Use of Serial Circulating Tumor DNA to Detect Resistance Alterations in Progressive Metastatic Breast Cancer. Clin Cancer Res 2020; 27:1361-1370. [PMID: 33323406 DOI: 10.1158/1078-0432.ccr-20-1566] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/18/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Circulating tumor DNA (ctDNA) is a promising tool for noninvasive longitudinal monitoring of genomic alterations. We analyzed serial ctDNA to characterize genomic evolution in progressive metastatic breast cancer. EXPERIMENTAL DESIGN This was a retrospective cohort between 2015 and 2019 obtained under an Institutional Review Board-approved protocol at Northwestern University (Chicago, IL). ctDNA samples were analyzed with Guardant360 next-generation sequencing (NGS) assay. A total of 86 patients had at least two serial ctDNA collections with the second drawn at first post-NGS progression (PN1) by imaging and clinical assessment. A total of 27 participants had ctDNA drawn at second post-NGS clinical progression (PN2). We analyzed alterations, mutant allele frequency (MAF), number of alterations (NOA), and sites of disease on imaging in close proximity to ctDNA evaluation. Matched pairs' variations in MAF, NOA, and alterations at progression were tested through Wilcoxon test. We identified an independent control cohort at Massachusetts General Hospital (Boston, MA) of 63 patients with serial ctDNA sampling and no evidence of progression. RESULTS We identified 44 hormone receptor-positive, 20 HER2+, and 22 triple-negative breast cancer cases. The significant alterations observed between baseline and PN1 were TP53 (P < 0.0075), PIK3CA (P < 0.0126), AR (P < 0.0126), FGFR1 (P < 0.0455), and ESR1 (P < 0.0143). Paired analyses revealed increased MAF and NOA from baseline to PN1 (P = 0.0026, and P < 0.0001, respectively). When compared with controls without progression, patients with ctDNA collection at times of progression were associated with increased MAF and NOA (P = 0.0042 and P < 0.0001, respectively). CONCLUSIONS Serial ctDNA testing identified resistance alterations and increased NOA and MAF were associated with disease progression. Prospective longitudinal ctDNA evaluation could potentially monitor tumor genomic evolution.
Collapse
Affiliation(s)
- Saya Jacob
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Andrew A Davis
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Division of Hematology and Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Lorenzo Gerratana
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Medicine, University of Udine, Udine, Italy
| | | | - Ami N Shah
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Firas Wehbe
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Neelima Katam
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Qiang Zhang
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Lisa Flaum
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Kalliopi P Siziopikou
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Pathology, Northwestern University, Chicago, Illinois
| | - Leonidas C Platanias
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - William J Gradishar
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Amir Behdad
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Pathology, Northwestern University, Chicago, Illinois
| | - Aditya Bardia
- Massachusetts General Hospital, Boston, Massachusetts
| | - Massimo Cristofanilli
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| |
Collapse
|
10
|
Kutova OM, Sencha LM, Pospelov AD, Dobrynina OE, Brilkina AA, Cherkasova EI, Balalaeva IV. Comparative Analysis of Cell-Cell Contact Abundance in Ovarian Carcinoma Cells Cultured in Two- and Three-Dimensional In Vitro Models. BIOLOGY 2020; 9:biology9120446. [PMID: 33291824 PMCID: PMC7761996 DOI: 10.3390/biology9120446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/28/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Simple Summary Tumor resistance to therapy is a crucial problem of today’s oncology. The emerging data indicate that tumor microenvironment is the key participant in the resistance development. One of the most basic aspect of tumor microenvironment is intercellular adhesion. Our data obtained using monolayer culture, matrix-free and matrix-based three-dimensional in vitro models indicate that the abundance of cell-cell contact proteins is varying depending on the microenvironment. These differences coincided with the degree of the resistance to therapeutics. The importance of adhesion proteins in tumor resistance may provide the fundamental basis for improving cancer treatment approaches and must be taken into account when screening candidate drugs. Abstract Tumor resistance to therapy is associated with the 3D organization and peculiarities of the tumor microenvironment, of which intercellular adhesion is a key participant. In this work, the abundance of contact proteins was compared in SKOV-3 and SKOV-3.ip human ovarian adenocarcinoma cell lines, cultivated in monolayers, tumor spheroids and collagen hydrogels. Three-dimensional models were characterized by extremely low expression of basic molecules of adherens junctions E-cadherin and demonstrated a simultaneous decrease in desmosomal protein desmoglein-2, gap junction protein connexin-43 and tight junction proteins occludin and ZO-1. The reduction in the level of contact proteins was most pronounced in collagen hydrogel, accompanied by significantly increased resistance to treatment with doxorubicin and targeted anticancer toxin DARPin-LoPE. Thus, we suggest that 3D models of ovarian cancer, especially matrix-based models, tend to recapitulate tumor microenvironment and treatment responsiveness to a greater extent than monolayer culture, so they can be used as a highly relevant platform for drug efficiency evaluation.
Collapse
|
11
|
Huang B, Yip WK, Wei N, Luo KQ. Acetyltanshinone IIA is more potent than lapatinib in inhibiting cell growth and degrading HER2 protein in drug-resistant HER2-positive breast cancer cells. Cancer Lett 2020; 490:1-11. [PMID: 32585412 DOI: 10.1016/j.canlet.2020.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 10/24/2022]
Abstract
High expression of human epidermal factor receptor 2 (HER2) is directly related to tumor progression, malignancy and drug resistance in HER2-positive breast cancer (HER2-PBC). The major limitation of current anti-HER2 therapies is that they cannot reduce the levels of HER2 protein. Here, we investigated the effect of acetyltanshinone IIA (ATA) in lapatinib-resistant HER2-PBC cells. Our data showed that ATA exhibited more potent effects than lapatinib against drug-resistant HER2-PBC cells in terms of (1) inhibiting cell growth, (2) reducing phosphorylated and total HER2 levels, (3) inhibiting tumor xenograft growth in nude mice, and (4) reducing HER2 protein levels in tumor xenografts. A mechanistic study revealed that ATA promoted HER2 degradation via increasing c-Cbl and CHIP-mediated HER2 ubiquitination and subsequent HER2 degradation by the proteasome or lysosome. ATA also reduced the levels of other tyrosine kinase receptors (TKRs), such as HER3, IGF-1R and MET, in lapatinib-resistant cells. Our findings suggest that direct degradation of HER2 and other TKRs can be an effective strategy for combatting drug resistance. They also indicate the potential utilization of ATA in treating breast cancer that is resistant or nonresponsive to current HER2-targeted therapies.
Collapse
Affiliation(s)
- Bin Huang
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Wai Kien Yip
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Na Wei
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China.
| |
Collapse
|
12
|
Peeney D, Jensen SM, Castro NP, Kumar S, Noonan S, Handler C, Kuznetsov A, Shih J, Tran AD, Salomon DS, Stetler-Stevenson WG. TIMP-2 suppresses tumor growth and metastasis in murine model of triple-negative breast cancer. Carcinogenesis 2020; 41:313-325. [PMID: 31621840 PMCID: PMC7221506 DOI: 10.1093/carcin/bgz172] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/12/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022] Open
Abstract
Metastasis is the primary cause of treatment failures and mortality in most cancers. Triple-negative breast cancer (TNBC) is refractory to treatment and rapidly progresses to disseminated disease. We utilized an orthotopic mouse model that molecularly and phenotypically resembles human TNBC to study the effects of exogenous, daily tissue inhibitor of metalloproteinase-2 (TIMP-2) treatment on tumor growth and metastasis. Our results demonstrated that TIMP-2 treatment maximally suppressed primary tumor growth by ~36-50% and pulmonary metastasis by >92%. Immunostaining assays confirmed disruption of the epithelial to mesenchymal transition (EMT) and promotion of vascular integrity in primary tumor tissues. Immunostaining and RNA sequencing analysis of lung tissue lysates from tumor-bearing mice identified significant changes associated with metastatic colony formation. Specifically, TIMP-2 treatment disrupts periostin localization and critical cell-signaling pathways, including canonical Wnt signaling involved in EMT, as well as PI3K signaling, which modulates proliferative and metastatic behavior through p27 phosphorylation/localization. In conclusion, our study provides evidence in support of a role for TIMP-2 in suppression of triple-negative breast cancer growth and metastasis through modulation of the epithelial to mesenchymal transition, vascular normalization, and signaling pathways associated with metastatic outgrowth. Our findings suggest that TIMP-2, a constituent of the extracellular matrix in normal tissues, may have both direct and systemic antitumor and metastasis suppressor effects, suggesting potential utility in the clinical management of breast cancer progression.
Collapse
Affiliation(s)
- David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sandra M Jensen
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Sarvesh Kumar
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Silvia Noonan
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chenchen Handler
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alex Kuznetsov
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Joanna Shih
- Biostatistics Branch, National Cancer Institute, Rockville, MD, USA
| | - Andy D Tran
- Confocal Core Facility, National Cancer Institute, Bethesda, MD, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
13
|
Zhang H, Moisini I, Ajabnoor RM, Turner BM, Hicks DG. Applying the New Guidelines of HER2 Testing in Breast Cancer. Curr Oncol Rep 2020; 22:51. [PMID: 32346807 DOI: 10.1007/s11912-020-0901-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The human epidermal growth factor receptor 2 (HER2) is an important prognostic and predictive biomarker in the breast cancer. The American Society of Clinical Oncology/College of American Pathology (ASCO/CAP) has published HER2 testing guidelines in breast cancer. We herein reviewed the HER2 testing guidelines in breast cancer with a focus on the application of the current guidelines. RECENT FINDINGS The continual investigation of HER2 testing in breast cancer has resulted in updates in the HER2 testing guidelines. The current guidelines focus on the uncommon clinical scenarios and emphasize the coordination between immunohistochemistry and in situ hybridization results, in an effort to improve clarity and accuracy. The ASCO/CAP guidelines provide valuable recommendations to ensure the accurate evaluation of HER2 status in breast cancer patients through standardization. Additional studies, particularly those with long-term outcome data are still needed to validate the guideline recommendations, especially the uncommon cases.
Collapse
Affiliation(s)
- Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ioana Moisini
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Rana M Ajabnoor
- Department of Pathology, Faculty of medicine, King Abdulaziz University, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Bradley M Turner
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - David G Hicks
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, NY, 14642, USA.
| |
Collapse
|
14
|
Discordant Marker Expression Between Invasive Breast Carcinoma and Corresponding Synchronous and Preceding DCIS. Am J Surg Pathol 2020; 43:1574-1582. [PMID: 31206365 DOI: 10.1097/pas.0000000000001306] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ductal carcinoma in situ (DCIS) is considered a potential precursor of invasive breast carcinoma (IBC). Studies aiming to find markers involved in DCIS progression generally have compared characteristics of IBC lesions with those of adjacent synchronous DCIS lesions. The question remains whether synchronous DCIS and IBC comparisons are a good surrogate for primary DCIS and subsequent IBC. In this study, we compared both primary DCIS and synchronous DCIS with the associated IBC lesion, on the basis of immunohistochemical marker expression. Immunohistochemical analysis of ER, PR, HER2, p53, and cyclo-oxygenase 2 (COX-2) was performed for 143 primary DCIS and subsequent IBC lesions, including 81 IBC lesions with synchronous DCIS. Agreement between DCIS and IBC was assessed using kappa, and symmetry tests were performed to assess the pattern in marker conversion. The primary DCIS and subsequent IBC more often showed discordant marker expression than synchronous DCIS and IBC. Strikingly, 18 of 49 (36%) women with HER2-positive primary DCIS developed an HER2-negative IBC. Such a difference in HER2 expression was not observed when comparing synchronous DCIS and IBC. The frequency of discordant marker expression did not increase with longer time between primary DCIS and IBC. In conclusion, comparison of primary DCIS and subsequent IBC yields different results than a comparison of synchronous DCIS and IBC, in particular with regard to HER2 status. To gain more insight into the progression of DCIS to IBC, it is essential to focus on the relationship between primary DCIS and subsequent IBC, rather than comparing IBC with synchronous DCIS.
Collapse
|
15
|
Miligy IM, Toss MS, Gorringe KL, Lee AHS, Ellis IO, Green AR, Rakha EA. The clinical and biological significance of HER2 over-expression in breast ductal carcinoma in situ: a large study from a single institution. Br J Cancer 2019; 120:1075-1082. [PMID: 31065110 PMCID: PMC6738110 DOI: 10.1038/s41416-019-0436-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Previous studies have reported up to 50% of ductal carcinoma in situ (DCIS), is HER2 positive, but the frequency of HER2-positive invasive breast cancer (IBC) is lower. The aim of this study is to characterise HER2 status in DCIS and assess its prognostic value. METHODS HER2 status was evaluated in a large series of DCIS (n = 868), including pure DCIS and DCIS associated with IBC, prepared as tissue microarrays (TMAs). HER2 status was assessed using immunohistochemistry (IHC) and chromogenic in situ hybridisation (CISH). RESULTS In pure DCIS, HER2 protein was over-expressed in 9% of DCIS (3+), whereas 15% were HER2 equivocal (2+). Using CISH, the final HER2 status was positive in 20%. In mixed DCIS, HER2 amplification of the DCIS component was detected in 15% with amplification in the invasive component of only 12%. HER2-positive DCIS was associated with features of aggressiveness (p < 0.0001) and more frequent local recurrence (p = 0.03). On multivariate analysis, combined HER2+/Ki67+ profile was an independent predictor of local recurrence (p = 0.006). CONCLUSIONS The frequency of HER2 positivity in DCIS is comparable to IBC- and HER2-positive DCIS is associated with features of poor prognosis. The majority of HER2 over-expression in DCIS is driven by gene amplification.
Collapse
Affiliation(s)
- Islam M Miligy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK.,Histopathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK.,Histopathology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, University of Melbourne, Parkville, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Andrew H S Lee
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK
| | - Ian O Ellis
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK. .,Histopathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| |
Collapse
|
16
|
Jasra S, Anampa J. Anthracycline Use for Early Stage Breast Cancer in the Modern Era: a Review. Curr Treat Options Oncol 2018; 19:30. [PMID: 29752560 DOI: 10.1007/s11864-018-0547-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OPINION STATEMENT Anthracycline-based regimens have been an important treatment component for patients with breast cancer. As demonstrated in the last Early Breast Cancer Trialists' Collaborative Group (EBCTCG) meta-analysis, anthracycline-based regimens decrease breast cancer mortality by 20-30%. Anthracycline toxicities include the rare-but potential morbid-cardiotoxicity or leukemogenic effect, and the almost universal-but very distressing-alopecia. Due to potential toxicities, and large number of patients being exposed, several worldwide trials have re-examined the role of anthracycline-based regimens in the management of breast cancer. Current literature supports that anthracyclines are not required for all patients with breast cancer and should be avoided in those with high cardiac risk. Recent results from the ABC trials suggest that anthracyclines should not be spared for patients with triple negative breast cancer (regardless of axillary node involvement) or HER2-/ER+ with significant node involvement. Based on current literature, for HER2-negative patients with low-risk breast cancer, anthracyclines could be spared with regimens such as cyclophosphamide, methotrexate, and fluorouracil (CMF) or docetaxel and cyclophosphamide (TC). Patients with intermediate or high-risk breast cancer should be considered for anthracycline-based regimens based on other factors such as age, comorbidities, tumor grade, lymphovascular invasion, and genomic profiling. Patients with HER2-positive breast cancer with low risk could be treated with paclitaxel and trastuzumab. For the remaining patients with HER2 overexpression, while docetaxel, carboplatin, and trastuzumab (TCH) has demonstrated to improve disease-free survival (DFS), anthracycline-containing regimens should be discussed, especially for those with very high-risk breast cancer. Although several biomarkers, such as topoisomerase II (TOP2A) and chromosome 17 centromeric duplication (Ch17CEP) have been proposed to predict benefit from anthracycline regimens, further research is required to delineate their proper utility in the clinical setting.
Collapse
Affiliation(s)
- Sakshi Jasra
- Department of Oncology, Section of Breast Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jesus Anampa
- Department of Oncology, Section of Breast Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Rd., Bronx, NY, 10461, USA.
| |
Collapse
|
17
|
Ribas A, Bellmunt J, Albanell J, De Torres I, Bermejo B, De Torres JA, Morote J, Gallardo E, Vera R, Carulla J, Sole-Calvo LA. Early Results of the Value of p53 in Predicting Survival in a Homogeneous Cohort of Patients with Invasive Bladder Cancer Treated with a Neoadjuvant Carboplatin-Based Regimen (M-CAVI). TUMORI JOURNAL 2018; 82:554-9. [PMID: 9061063 DOI: 10.1177/030089169608200608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aims and Background Several reports on prognostic factors for infiltrating bladder cancer have given controversial results. We assessed the prognostic value of p53 nuclear overexpression together with known prognostic factors for survival in patients with invasive T2-4 NO MO bladder cancer treated with neoadjuvant chemotherapy. Study Design Thirty-five paraffi-nized tumor samples from initial transurethral resection of patients with bladder cancer were analyzed immunohistochemi-cally to detect overexpression of p53 protein. Patients were treated with 3 to 4 cycles of neoadjuvant methotrexate, carboplatin, and vinblastine (M-CAVI) and then underwent radical cystectomy. Prechemotherapy, treatment, and postchemotherapy factors were analyzed for correlation with survival by univariate and multivariate analysis. Fifty-seven percent of tumors were positive for p53 protein, 71.5% had grade III-IV tumors, and 72% had organ-confined disease. The median follow-up was 20 months (range 5-71+). Results By univariate analysis, the significant pretreatment factors were initial tumor (T) stage ( P <0.0001) and the male sex ( P = 0.03). Five postchemotherapy variables were found significant: surgery performed according to protocol ( P = 0.003), overall clinical ( P = 0.004), and overall pathologic ( P = 0.02) response to therapy, postchemotherapy pathologic stage ( P = 0.0002), and tumor status after surgery ( P = 0.0006). By multivariate analysis, the initial prechemotherapy T stage was the only factor that demonstrated independent significance. Conclusions Although the median follow-up of the study is still too short, in this group of patients treated with a neoadjuvant carboplatin-based regimen, a classical variable (prechemotherapy T stage) rather than p53 nuclear overexpression was an independent prognostic factor for survival. Further follow-up will be required to assess the value of p53 overexpression as a prognostic factor in invasive bladder cancer patients treated with neoadjuvant carboplatin-based chemotherapy.
Collapse
Affiliation(s)
- A Ribas
- Medical Oncology Section, Hospital General Universitari Vall d'Hebron, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Quaranta M, Daniele A, Coviello M, Savonarola A, Abbate I, Venneri MT, Paradiso A, Stea B, Zito A, Labriola A, Schittulli F. C-Erbb-2 Protein Level in Tissue and Sera of Breast Cancer Patients: A Possibly Useful Clinical Correlation. TUMORI JOURNAL 2018; 92:311-7. [PMID: 17036522 DOI: 10.1177/030089160609200409] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background The aims of this study were to assess the clinical utility of circulating preoperative HER-2 extracellular domain p105 detected by enzyme immunoassay (ELISA), to compare the tissue expression of HER-2/neu determined by immunohistochemistry (IHC), to correlate prognostic factors including tumor size, nodal involvement, and hormone receptor status, and to analyze the prognostic significance of the marker in relation to clinical outcome as measured by disease-free and overall survival. Methods In this study, we enrolled 108 consecutive patients with breast carcinoma, and obtained serum samples and frozen tumor tissues. We compared them with 57 women with fibroadenoma and 63 healthy women as controls. Results Univariate ANOVA analysis showed no relationship between HER-2/neu in tissue and serum. Preoperative serum levels of p105 were significantly higher in breast cancer patients than in women with benign disease or healthy women. Concerning the correlation between p105, HER-2/neu tissue expression, and the other prognostic factors, a statistically significant correlation between high serum p105 levels and ER-negative status in breast cancer patients was found. Kaplan-Meier analysis confirmed that patients with positive HER-2/neu tissue expression had a significantly shorter survival than those with negative expression. Analysis with the Cox model demonstrated that tumor size was the only significant independent prognostic factor. Conclusions This research failed to demonstrate a relationship between preoperative tissue overexpression and circulating HER-2/neu, suggesting that p105 does not represent a valid alternative to predict a worsened prognosis in breast cancer, but it could be a diagnostic marker to discriminate healthy subjects from breast cancer patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Analysis of Variance
- Antigens, Nuclear/blood
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/blood
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Chromosomal Proteins, Non-Histone/blood
- Disease-Free Survival
- Enzyme-Linked Immunosorbent Assay
- Female
- Fibroadenoma/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Lymphatic Metastasis
- Middle Aged
- Predictive Value of Tests
- Prognosis
- Proportional Hazards Models
- Receptor, ErbB-2/blood
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Retrospective Studies
- Risk Factors
- Survival Analysis
- Up-Regulation
Collapse
Affiliation(s)
- Michele Quaranta
- Chemical-Clinical Microbiology and Immunology Laboratory Unit, National Oncology Institute of Bari, Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
c-erbB-2 is an oncoprotein which is overexpressed in up to 40% of primary breast cancers. c-erbB-2 overexpression is a bad prognostic factor in patients with lymph node-positive disease. Unfortunately, there has been no agreement to date on whether c-erbB-2 overexpression is of prognostic significance in patients with lymph node-negative disease. c-erbB-2 overexpression is correlated with the absence of estrogen receptor expression in a number of publications. Correlation between c-erbB-2 overexpression and hormone sensitivity in the clinical setting is less well established and is the focus of ongoing studies. Both preclinical and clinical studies support an association between c-erbB-2 receptor overexpression and resistance to alkylating agents. In contrast, the data for c-erbB-2 and anthracyclines should be viewed in a slightly different manner. Anthracyclines appear to have a greater therapeutic effect in c-erbB-2-positive disease which may be dose sensitive. In c-erbB-2-negative disease not only is the therapeutic effect reduced but there does not appear to be any improved response to higher doses of anthracyclines. The data for c-erbB-2 and the taxanes is still not clear enough to provide any definite conclusions. If there is a correlation it would at present appear to be between paclitaxel and response rates, but this needs to be confirmed in larger studies. Few studies have looked at changes in c-erbB-2 on therapy. Those that have seem to show no significant change on either tamoxifen or chemotherapy.
Collapse
Affiliation(s)
- E Tagliabue
- Division of Experimental Oncology E, Istituto Nazionale Tumori, Milan, Italy.
| | | | | | | |
Collapse
|
20
|
Koo JS, Jung W, Jeong J. Metastatic Breast Cancer Shows Different Immunohistochemical Phenotype According to Metastatic Site. TUMORI JOURNAL 2018; 96:424-32. [DOI: 10.1177/030089161009600308] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aims and background The study was performed to assess the status of immunohistochemical markers in primary and metastatic breast cancer and to determine the organ-specific characteristics of metastatic breast cancer. Methods Samples from 13 cases of paired primary and metastatic breast cancer and 34 cases of metastatic breast cancer were included. Results In the analysis of 13 cases of paired primary and metastatic breast cancer, estrogen receptor and progesterone receptor loss were noted in 1 (7.7%) case each. Androgen receptor loss and gain was noted in 2 (15.4%) cases, respectively. HER-2 showed 100% concordance with primary and metastatic tumors. C-kit was demonstrated in only 2 (15.4%) cases of metastatic breast cancer. In the analysis of 34 cases of metastatic breast cancer, when classified into triple-negative type (ER-, PR-, and HER-2-), HER-2+ type, and ER+ or PR+/HER-2- type according to immunohistochemical stain results, HER-2 type (66.7%) in brain metastasis and ER+ or PR+/HER-2- type (75.0%) in liver metastasis were predominant. Bone metastasis was composed of triple negative type (44.4%) and ER+ or PR+/HER-2- type (55.6%), and lung metastasis showed all of three subtypes in similar proportions. Conclusions Metastatic breast cancer shows different immunohistochemical phenotypes according to metastatic site (P = 0.048).
Collapse
Affiliation(s)
- Ja Seung Koo
- Department of Pathology, Yonsei University Health System, Seoul, Korea
| | - Woohee Jung
- Department of Pathology, Yonsei University Health System, Seoul, Korea
| | - Joon Jeong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Biganzoli E, Boracchi P, Marubini E. Biostatistics and Tumor Marker Studies in Breast Cancer: Design, Analysis and Interpretation Issues. Int J Biol Markers 2018; 18:40-8. [PMID: 12699062 DOI: 10.1177/172460080301800107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- E Biganzoli
- Unità di Statistica Medica e Biometria, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.
| | | | | |
Collapse
|
22
|
Xu L, Jia S, Li H, Yu Y, Liu G, Wu Y, Liu X, Liu C, Zhou Y, Zhang Z, Sheng Y. Characterization of circulating tumor cells in newly diagnosed breast cancer. Oncol Lett 2017; 15:2522-2528. [PMID: 29434968 DOI: 10.3892/ol.2017.7540] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 06/02/2017] [Indexed: 12/21/2022] Open
Abstract
Identification of circulating tumor cells (CTCs) by surface marker expression and ploidy analysis [immunostaining-fluorescence in situ hybridization (iFISH)] has been shown to be a highly sensitive method in the identification of certain solid cancers. In the present study, iFISH analysis was performed to identify CTCs in 184 patients with newly diagnosed non-metastatic breast cancer, and the distribution of CTC subtypes was characterized based on cytokeratin (CK) expression and ploidy status. It was revealed that CTCs of non-metastatic, aneuploid breast cancers, independent of CK expression profile, can be detected with high sensitivity (90.76%) by the iFISH system. Higher CTC counts and sensitivity were observed in patients with increased tumor size burden and unfavorable hormone receptor status. Investigation of CTC subtypes based on ploidy analysis indicated that triploid CTCs constituted the majority of CTCs evaluated. While CK-positive CTCs were detected in a small cohort of patients, an overall low rate of CK expression was observed in the 18 patient samples evaluated. Of note, CK expression was rare in CTCs detected in patients with Herceptin 2 (Her2)-positive or triple-negative [estrogen receptor (ER)-, progesterone receptor (PR)- and Her2-negative], indicating that lack of ER and PR may result in promotion of epithelial-mesenchymal transition and enhancement of tumor aggression.
Collapse
Affiliation(s)
- Lu Xu
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Songlin Jia
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Hengyu Li
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yue Yu
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Guoping Liu
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yanmei Wu
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Xishui Liu
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Chaoqian Liu
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yue Zhou
- Biotecan Medical Diagnostics Co., Ltd., Zhangjiang Center for Translational Medicine, Shanghai 201204, P.R. China
| | - Zhenzhen Zhang
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China.,Biotecan Medical Diagnostics Co., Ltd., Zhangjiang Center for Translational Medicine, Shanghai 201204, P.R. China
| | - Yuan Sheng
- Department of Breast Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
23
|
Haghnavaz N, Asghari F, Elieh Ali Komi D, Shanehbandi D, Baradaran B, Kazemi T. HER2 positivity may confer resistance to therapy with paclitaxel in breast cancer cell lines. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:518-523. [PMID: 28509576 DOI: 10.1080/21691401.2017.1326927] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION MicroRNAs (miRNAs) are short non-coding single-stranded RNAs. Involving in post-transcriptional gene silencing, miRNAs are thought to play important roles in many cancers such as breast cancer. Paclitaxel is used widely in the treatment of breast cancer. In this study, we investigated the effect of paclitaxel treatment on the expression levels of two oncomirs (oncomiRs), miR-21 and miR-203, in breast cancer cell lines. MATERIALS AND METHODS MTT assay was performed to determine IC50 of paclitaxel for human breast cancer cell lines including MCF-7, MDA-MB-231, SKBR3 and BT-474. After RNA extraction and cDNA synthesis, the expression levels of miRNAs were then quantitatively evaluated using real-time PCR. RESULTS Our results showed that after treatment, the expression levels of both miR-21 and miR-203 were significantly increased in HER2-positive cell lines, BT-474 and SKBR3. HER2-negative cell lines, MCF-7 and MDA-MB-231, in contrast had significantly decreased expression of both assessed oncomiRs. CONCLUSION Our results showed that the expression levels of oncomiRs were increased in HER-2 positive breast cancer cells and this finding is in line with previous studies. Our findings present a probable mechanism of resistance against paclitaxel chemotherapy in HER2-positive breast cancers.
Collapse
Affiliation(s)
- Navideh Haghnavaz
- a Immunology Research Center , Tabriz University of Medical Science , Tabriz , Iran.,b Department of Immunology, Faculty of Medicine , Tabriz University of Medical Science , Tabriz , Iran.,c Student research committee , Tabriz University of Medical Science , Tabriz , Iran
| | - Faezeh Asghari
- a Immunology Research Center , Tabriz University of Medical Science , Tabriz , Iran.,b Department of Immunology, Faculty of Medicine , Tabriz University of Medical Science , Tabriz , Iran
| | - Daniel Elieh Ali Komi
- a Immunology Research Center , Tabriz University of Medical Science , Tabriz , Iran.,b Department of Immunology, Faculty of Medicine , Tabriz University of Medical Science , Tabriz , Iran
| | - Dariush Shanehbandi
- b Department of Immunology, Faculty of Medicine , Tabriz University of Medical Science , Tabriz , Iran
| | - Behzad Baradaran
- a Immunology Research Center , Tabriz University of Medical Science , Tabriz , Iran.,b Department of Immunology, Faculty of Medicine , Tabriz University of Medical Science , Tabriz , Iran
| | - Tohid Kazemi
- a Immunology Research Center , Tabriz University of Medical Science , Tabriz , Iran.,b Department of Immunology, Faculty of Medicine , Tabriz University of Medical Science , Tabriz , Iran
| |
Collapse
|
24
|
Zoppoli G, Garuti A, Cirmena G, di Cantogno LV, Botta C, Gallo M, Ferraioli D, Carminati E, Baccini P, Curto M, Fregatti P, Isnaldi E, Lia M, Murialdo R, Friedman D, Sapino A, Ballestrero A. Her2 assessment using quantitative reverse transcriptase polymerase chain reaction reliably identifies Her2 overexpression without amplification in breast cancer cases. J Transl Med 2017; 15:91. [PMID: 28460632 PMCID: PMC5412048 DOI: 10.1186/s12967-017-1195-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 04/03/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Immunohistochemistry (IHC) and fluorescent-in situ hybridization (FISH) are standard methods to assess human epidermal growth factor receptor 2 (HER2) status in breast cancer (BC) patients. Real-time quantitative polymerase-chain-reaction (qRT-PCR) is able to detect HER2 overexpression. Here we compared FISH, IHC, quantitative PCR (qPCR), and qRT-PCR to determine the concordance rates and evaluate their relative roles in HER2 determination. PATIENTS AND METHODS We determined HER2 status in 153 BC patients, using IHC, FISH, Q-PCR and qRT-PCR. In discordant cases, we directly measured HER2 protein levels using Western blotting. RESULTS The overall agreement (OA) between FISH and Q-PCR was 94.1, with a k value of 0.87. Assuming FISH as the standard reference, Q-PCR showed an 86.1% sensitivity and a 99.0% specificity with a global accuracy of 91.6%. OA between FISH and qRT-PCR was 90.8% with a k value of 0.81. Of interest, the disagreement between FISH and qRT-PCR was mostly restricted to equivocal cases. HER2 protein analysis suggested that qRT-PCR correlates better than FISH with HER2 protein levels, particularly where FISH fails to provide conclusive results. SIGNIFICANCE qRT-PCR may outperform FISH in identifying patients overexpressing HER2 protein. Q-PCR cannot be used for HER2 status assessment, due to its suboptimal level of agreement with FISH. Both FISH and Q-PCR may be less accurate than qRT-PCR as surrogates of HER2 protein determination.
Collapse
Affiliation(s)
- Gabriele Zoppoli
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy.
| | - Anna Garuti
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Gabriella Cirmena
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | | | - Cristina Botta
- Department of Biomedical Sciences and Human Oncology, University of Turin, Turin, Italy
| | - Maurizio Gallo
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Domenico Ferraioli
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy.,Comprehensive Cancer Center Léon Bérard, Lyon, France
| | - Enrico Carminati
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Paola Baccini
- Department of Pathology, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Monica Curto
- Department of Pathology, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Piero Fregatti
- Department of Surgery, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Edoardo Isnaldi
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Michela Lia
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Roberto Murialdo
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Daniele Friedman
- Department of Surgery, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Anna Sapino
- Department of Biomedical Sciences and Human Oncology, University of Turin, Turin, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine (Di.M.I.), University of Genoa and IRCCS AOU San Martino-IST, Viale Benedetto XV, 6, 16132, Genoa, Italy
| |
Collapse
|
25
|
Lazzeroni M, Dunn BK, Pruneri G, Jereczek-Fossa BA, Orecchia R, Bonanni B, DeCensi A. Adjuvant therapy in patients with ductal carcinoma in situ of the breast: The Pandora's box. Cancer Treat Rev 2017; 55:1-9. [PMID: 28262606 DOI: 10.1016/j.ctrv.2017.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 12/23/2022]
Abstract
Most patients with ductal carcinoma in situ of the breast (DCIS) are eligible for breast conservation treatment. The key management decision is whether to add radiotherapy and/or endocrine therapy to minimize the risk of a subsequent recurrence. Recent analyses indicating a lack of benefit in terms of breast cancer-associated mortality have suggested that more conservative approaches, omitting adjuvant therapy or even surgery, may be advisable in selected patients. These mortality observations are directly influenced by widespread use of mammographic screening which has opened a Pandora's box of subclinical DCIS and early invasive lesions. Confusion as to how aggressively such possibly indolent lesions should be treated has led to misunderstandings among patients and medical professionals. While awaiting further prospective evidence from clinical trials, we endorse an active treatment of DCIS as the standard of care. Our rationale is twofold: invasive recurrences are associated with an increase in breast cancer mortality, which is not the only relevant endpoint for DCIS. The benefit of complete surgical excision, adjuvant radiotherapy and endocrine treatment in preventing recurrence and invasive progression has been demonstrated in DCIS. The challenge now is how to identify DCIS patients who will not progress to invasive carcinoma even without complete excision and, at the other extreme, those patients at the highest risk who require mastectomy for local control. The current controversies over whether and which adjuvant therapy should be implemented can at least in part be addressed by developing effective doctor-patient communications that enable mutual understanding about the management of this biologically heterogeneous disease.
Collapse
Affiliation(s)
- Matteo Lazzeroni
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Barbara K Dunn
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD 20892, USA
| | - Giancarlo Pruneri
- Pathology, European Institute of Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Radiotherapy, European Institute of Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Roberto Orecchia
- Radiotherapy, European Institute of Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Bernardo Bonanni
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Andrea DeCensi
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy; Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy; Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, United Kingdom.
| |
Collapse
|
26
|
Rathnam C, Lee S, Jiang X. An algorithm for direct causal learning of influences on patient outcomes. Artif Intell Med 2016; 75:1-15. [PMID: 28363452 DOI: 10.1016/j.artmed.2016.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/25/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVE This study aims at developing and introducing a new algorithm, called direct causal learner (DCL), for learning the direct causal influences of a single target. We applied it to both simulated and real clinical and genome wide association study (GWAS) datasets and compared its performance to classic causal learning algorithms. METHOD The DCL algorithm learns the causes of a single target from passive data using Bayesian-scoring, instead of using independence checks, and a novel deletion algorithm. We generate 14,400 simulated datasets and measure the number of datasets for which DCL correctly and partially predicts the direct causes. We then compare its performance with the constraint-based path consistency (PC) and conservative PC (CPC) algorithms, the Bayesian-score based fast greedy search (FGS) algorithm, and the partial ancestral graphs algorithm fast causal inference (FCI). In addition, we extend our comparison of all five algorithms to both a real GWAS dataset and real breast cancer datasets over various time-points in order to observe how effective they are at predicting the causal influences of Alzheimer's disease and breast cancer survival. RESULTS DCL consistently outperforms FGS, PC, CPC, and FCI in discovering the parents of the target for the datasets simulated using a simple network. Overall, DCL predicts significantly more datasets correctly (McNemar's test significance: p<<0.0001) than any of the other algorithms for these network types. For example, when assessing overall performance (simple and complex network results combined), DCL correctly predicts approximately 1400 more datasets than the top FGS method, 1600 more datasets than the top CPC method, 4500 more datasets than the top PC method, and 5600 more datasets than the top FCI method. Although FGS did correctly predict more datasets than DCL for the complex networks, and DCL correctly predicted only a few more datasets than CPC for these networks, there is no significant difference in performance between these three algorithms for this network type. However, when we use a more continuous measure of accuracy, we find that all the DCL methods are able to better partially predict more direct causes than FGS and CPC for the complex networks. In addition, DCL consistently had faster runtimes than the other algorithms. In the application to the real datasets, DCL identified rs6784615, located on the NISCH gene, and rs10824310, located on the PRKG1 gene, as direct causes of late onset Alzheimer's disease (LOAD) development. In addition, DCL identified ER category as a direct predictor of breast cancer mortality within 5 years, and HER2 status as a direct predictor of 10-year breast cancer mortality. These predictors have been identified in previous studies to have a direct causal relationship with their respective phenotypes, supporting the predictive power of DCL. When the other algorithms discovered predictors from the real datasets, these predictors were either also found by DCL or could not be supported by previous studies. CONCLUSION Our results show that DCL outperforms FGS, PC, CPC, and FCI in almost every case, demonstrating its potential to advance causal learning. Furthermore, our DCL algorithm effectively identifies direct causes in the LOAD and Metabric GWAS datasets, which indicates its potential for clinical applications.
Collapse
Affiliation(s)
- Chandramouli Rathnam
- Department of Biomedical Informatics, University of Pittsburgh, 5607 Baum Blvd, Pittsburgh, PA 15206, USA
| | - Sanghoon Lee
- Department of Biomedical Informatics, University of Pittsburgh, 5607 Baum Blvd, Pittsburgh, PA 15206, USA
| | - Xia Jiang
- Department of Biomedical Informatics, University of Pittsburgh, 5607 Baum Blvd, Pittsburgh, PA 15206, USA.
| |
Collapse
|
27
|
Büyükköroğlu G, Şenel B, Gezgin S, Dinh T. The simultaneous delivery of paclitaxel and Herceptin® using solid lipid nanoparticles: In vitro evaluation. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Gramlich TL, Fritsch C, Cohen C, Samuel M, Hunt D, Dean PJ, Gansler T. Oncogene Expression and Amplification in Barrett Adenocarcinoma. Int J Surg Pathol 2016. [DOI: 10.1177/106689699700400402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oncogene activation by amplification and/or overexpression has been implicated in the development and progression of a number of malignancies. To better define the role of oncogene activation in adenocarcinomas arising in the setting of Barrett esophagus, we examined 44 resected Barrett adenocarcinomas for c-erb B-2, epidermal growth factor receptor (EGFR), c-myc, and cyclin DI gene amplification as well as c-erb B-2 and EGFR expression. DNA was extracted from formalin-fixed paraffin-embedded tissue and analyzed by differential polymerase chain reaction, a technique that permits semiquantitative analysis of gene dosage on archival material. Formalinfixed paraffin-embedded sections were stained with monoclonal antibodies directed against either the internal domain of c-erb B-2 or the extracellular domain of EGFR with a labeled streptavidin biotin technique. C-erb B-2 amplification was identified in three (7%) cases. A plasma membrane pattern of c-erb B-2 immunoreactivity in >50% of tumor cells correlated highly with c-erb B-2 amplification ( P=.00008). EGFR amplification was noted in 18% of cases and correlated highly with the intensity of EGFR immunoreactivity ( P=.0004). C-myc amplification was present in 18% of cases. No tumors showed cyclin DI amplification. Follow-up was available regarding 29 patients and showed decreased mean survival ( P=.029) in patients with strong EGFR immunoreactivity (5.8 months) versus those with weak or absent EGFR labeling (11.8 months), and a trend toward decreased one-year survival (P=.069) for patients with (17%) versus those without (52%) c-myc amplification. Our results indicate one or more selected oncogenes are amplified/overexpressed in some Barrett adenocarcinomas and that EGFR and c-erb B-2 overexpression correlates with amplification. Additionally, strong EGFR expression in tumor cells indicates a poorer prognosis.
Collapse
Affiliation(s)
- Terry L. Gramlich
- Department of Pathology, Emory University Hospital, Atlanta, Georgia; Department of Anatomic Pathology (L25), Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | | | | | | | - Dirk Hunt
- Department of Pathology, Emory University Hospital, Atlanta, Georgia
| | - Patrick J. Dean
- Baptist Memorial Hospital, Memphis, Tennessee; Department of Pathology, Emory University Hospital, Atlanta, Georgia
| | - Ted Gansler
- Department of Pathology, Emory University Hospital, Atlanta, Georgia
| |
Collapse
|
29
|
Ko SS, Na YS, Yoon CS, Park JY, Kim HS, Hur MH, Lee HK, Chun YK, Kang SS, Park BW, Lee JH. The Significance of c-erbB-2 Overexpression and p53 Expression in Patients With Axillary Lymph Node—Negative Breast Cancer: A Tissue Microarray Study. Int J Surg Pathol 2016; 15:98-109. [PMID: 17478762 DOI: 10.1177/1066896906299124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We conducted this study to examine whether the expression of c-erbB-2 and p53 is the prognostic indicator for patients with early-stage breast cancer in which axillary lymph node metastasis is absent. We examined 326 patients with early-stage breast cancer in which axillary lymph node metastasis is absent. Tissue microarrays were constructed. Following this, immunohistochemical staining was done for estrogen receptor (ER), progesterone receptor (PR), c-erbB2, and p53. The results were as follows: (1) expression of c-erbB-2 was correlated with other clinicopathologic factors (eg, patient's age, presence of menopause, tumor size, histologic and nuclear grade, and presence of hormone receptors such as ER and PR); and (2) expression of p53 was correlated with survival rate, patient's age, presence of menopause, and tumor size. However, these results were not statistically significant. In conclusion, our results indicate that expression of c-erbB-2 and p53 did not have any prognostic value in patients with early-stage breast cancer in which axillary lymph node metastasis is absent.
Collapse
Affiliation(s)
- Seung-Sang Ko
- Department of Surgery, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
da Fonseca Reis Silva D, Ribeiro JM. Infracentimetric HER-2 positive breast tumours-review of the literature. Ecancermedicalscience 2015; 9:593. [PMID: 26635897 PMCID: PMC4659705 DOI: 10.3332/ecancer.2015.593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Indexed: 01/03/2023] Open
Abstract
Breast cancer is the most common malignant neoplasm in the world among women. As a result of the dissemination of population screening programmes, about half of non-metastatic breast cancers are now diagnosed at stage I. 10-15% of T1abN0 tumours over-express human epidermal growth factor (HER-2). These tumours have a globally excellent prognosis, however, treatment with chemotherapy and/or targeted therapy may further improve outcomes in selected cases. In this article, we will review studies with information on prognosis and benefit of adjuvant therapy for T1abN0 HER-2+ breast cancer.
Collapse
Affiliation(s)
- Danilo da Fonseca Reis Silva
- Instituto do Câncer do Estado de São Paulo - ICESP - Faculdade de Medicina da Universidade de São Paulo, Av. Doutor Arnaldo, 251 - Cerqueira César, São Paulo - SP 01246-000, Brazil
| | - Joana M Ribeiro
- Breast Unit, Champalimaud Clinical Centre, Av. de Brasília, s/n, Lisbon 1400-038, Portugal
| |
Collapse
|
31
|
Expression of Lipid Metabolism-Related Proteins in Metastatic Breast Cancer. PLoS One 2015; 10:e0137204. [PMID: 26334757 PMCID: PMC4559312 DOI: 10.1371/journal.pone.0137204] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/13/2015] [Indexed: 02/02/2023] Open
Abstract
PURPOSE The tumor biology of metastatic breast cancers differ according to the metastatic sites, and the features of cancer metabolism may also be different. The aim of this study is to investigate the expression of lipid metabolism-related proteins in metastatic breast cancer according to metastatic site and discuss the clinical significance thereof. METHODS Immunohistochemical staining for lipid metabolism-related proteins [fatty acid synthase (FASN), hormone-sensitive lipase (HSL), carnitine palmitoyltransferase IA (CPT-1A), acyl-CoA oxidase 1 (ACOX1), fatty acid binding protein 4 (FABP4,) and perilipin 1 (PLIN1)] was performed using a tissue microarray of 149 cases of metastatic breast cancer (bone metastasis = 39, brain metastasis = 37, liver metastasis = 21, and lung metastasis = 52). RESULTS The expression levels of ACOX1 (p = 0.009) and FASN (p = 0.007) varied significantly according to metastatic site, with the highest expression in brain metastasis and the lowest expression in liver metastasis. ACOX1 positivity (p = 0.005) and FASN positivity (p = 0.003) correlated with HER-2 positivity. The expression of FASN was significantly higher in HER-2 type breast cancer, and lower in luminal A and TNBC type breast cancer (p<0.001). Among lipid metabolism-related proteins, PLIN1 positivity was found to be an independent poor prognostic factor on multivariate analysis (Hazard ratio: 4.979, 95% CI: 1.054-22.59, p = 0.043). CONCLUSION Different expression levels of lipid metabolism-related proteins were observed according to metastatic site. The expression of ACOX1 and FASN was highest in brain metastasis. These results suggest that the metastatic site should be considered when using lipid metabolism inhibitors for targeted therapy.
Collapse
|
32
|
Small HER2-Positive Breast Cancer: Should Size Affect Adjuvant Treatment? Clin Breast Cancer 2015; 15:277-84. [DOI: 10.1016/j.clbc.2014.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/30/2014] [Accepted: 12/31/2014] [Indexed: 01/10/2023]
|
33
|
Liu N, Yu C, Shi Y, Jiang J, Liu Y. SMAD4 expression in breast ductal carcinoma correlates with prognosis. Oncol Lett 2015; 10:1709-1715. [PMID: 26622737 DOI: 10.3892/ol.2015.3442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 06/11/2015] [Indexed: 11/06/2022] Open
Abstract
The present study examined SMAD4 expression in fine-needle aspiration cell blocks from patients with breast ductal carcinoma, in order to assess its viability as a prognostic marker. Using immunohistochemistry, the SMAD4 protein status of 86 breast ductal carcinoma fine-needle biopsies, from patients who underwent tumor resection at Beihua University Affiliated Hospital (Jilin, China) between 2002 and 2008, was characterized. The association between SMAD4 expression and clinicopathological parameters, as well as prognosis was assessed using the Mantel-Haenszel method and Cox proportional hazards regression. SMAD4 staining was observed in the cytoplasm and nucleus, and its expression was found to be decreased in ductal breast carcinoma as compared with adjacent normal breast epithelia. Patients with reduced SMAD4 expression levels tended to exhibit more poorly differentiated tumors, a higher risk of recurrence and shorter overall survival. These results demonstrated that the evaluation of SMAD4 protein status in fine-needle biopsy specimens of breast ductal carcinoma may provide additional prognostic information.
Collapse
Affiliation(s)
- Nannan Liu
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin City, Jilin 132013, P.R. China
| | - Chunyan Yu
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin City, Jilin 132013, P.R. China
| | - Yanfen Shi
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Jing Jiang
- Department of Pathology, Beihua University Affiliated Hospital, Jilin City, Jilin 132011, P.R. China
| | - Yuhe Liu
- Department of Pathology, College of Basic Medicine, Beihua University, Jilin City, Jilin 132013, P.R. China
| |
Collapse
|
34
|
Borgquist S, Zhou W, Jirström K, Amini RM, Sollie T, Sørlie T, Blomqvist C, Butt S, Wärnberg F. The prognostic role of HER2 expression in ductal breast carcinoma in situ (DCIS); a population-based cohort study. BMC Cancer 2015; 15:468. [PMID: 26062614 PMCID: PMC4464713 DOI: 10.1186/s12885-015-1479-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/02/2015] [Indexed: 12/04/2022] Open
Abstract
Background HER2 is a well-established prognostic and predictive factor in invasive breast cancer. The role of HER2 in ductal breast carcinoma in situ (DCIS) is debated and recent data have suggested that HER2 is mainly related to in situ recurrences. Our aim was to study HER2 as a prognostic factor in a large population based cohort of DCIS with long-term follow-up. Methods All 458 patients diagnosed with a primary DCIS 1986–2004 in two Swedish counties were included. Silver-enhanced in situ hybridisation (SISH) was used for detection of HER2 gene amplification and protein expression was assessed by immunohistochemistry (IHC) in tissue microarrays. HER2 positivity was defined as amplified HER2 gene and/or HER2 3+ by IHC. HER2 status in relation to new ipsilateral events (IBE) and Invasive Breast Cancer Recurrences, local or distant (IBCR) was assessed by Kaplan-Meier survival analyses and Cox proportional hazards regression models. Results Primary DCIS was screening-detected in 75.5 % of cases. Breast conserving surgery (BCS) was performed in 78.6 % of whom 44.0 % received postoperative radiotherapy. No patients received adjuvant endocrine- or chemotherapy. The majority of DCIS could be HER2 classified (N = 420 (91.7 %)); 132 HER2 positive (31 %) and 288 HER2 negative (69 %)). HER2 positivity was related to large tumor size (P = 0.002), high grade (P < 0.001) and ER- and PR negativity (P < 0.001 for both). During follow-up (mean 184 months), 106 IBCRs and 105 IBEs were identified among all 458 cases corresponding to 54 in situ and 51 invasive recurrences. Eighteen women died from breast cancer and another 114 had died from other causes. The risk of IBCR was statistically significantly lower subsequent to a HER2 positive DCIS compared to a HER2 negative DCIS, (Log-Rank P = 0.03, (HR) 0.60 (95 % CI 0.38–0.94)). Remarkably, the curves did not separate until after 10 years. In ER-stratified analyses, HER2 positive DCIS was associated with lower risk of IBCR among women with ER negative DCIS (Log-Rank P = 0.003), but not for women with ER positive DCIS. Conclusions Improved prognostic tools for DCIS patients are warranted to tailor adjuvant therapy. Here, we demonstrate that HER2 positive disease in the primary DCIS is associated with lower risk of recurrent invasive breast cancer.
Collapse
Affiliation(s)
- Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village Building 404:B3, Scheelevägen 2, SE-223 81, Lund, Sweden.
| | - Wenjing Zhou
- Department of Surgical Science, Uppsala University, Uppsala, SE-75105, Sweden.
| | - Karin Jirström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village Building 404:B3, Scheelevägen 2, SE-223 81, Lund, Sweden.
| | - Rose-Marie Amini
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Thomas Sollie
- Department of Pathology, Örebro University, Örebro, Sweden.
| | - Therese Sørlie
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Montebello, 0310, Oslo, Norway.
| | - Carl Blomqvist
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
| | - Salma Butt
- Department of Surgery, Clinical Sciences, Lund University, Malmö, Sweden.
| | - Fredrik Wärnberg
- Department of Surgical Science, Uppsala University, Uppsala, SE-75105, Sweden.
| |
Collapse
|
35
|
X-ray CT guided fault-free photothermal ablation of metastatic lymph nodes with ultrafine HER-2 targeting W18O49 nanoparticles. Biomaterials 2014; 35:9155-66. [DOI: 10.1016/j.biomaterials.2014.07.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/21/2014] [Indexed: 12/29/2022]
|
36
|
Liang Y, Besch-Williford C, Aebi JD, Mafuvadze B, Cook MT, Zou X, Hyder SM. Cholesterol biosynthesis inhibitors as potent novel anti-cancer agents: suppression of hormone-dependent breast cancer by the oxidosqualene cyclase inhibitor RO 48-8071. Breast Cancer Res Treat 2014; 146:51-62. [PMID: 24878988 PMCID: PMC11121502 DOI: 10.1007/s10549-014-2996-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023]
Abstract
In most human breast cancers, tumor cell proliferation is estrogen dependent. Although hormone-responsive tumors initially respond to anti-estrogen therapies, most of them eventually develop resistance. Our goal was to identify alternative targets that might be regulated to control breast cancer progression. Sulforhodamine B assay was used to measure the viability of cultured human breast cancer cell lines exposed to various inhibitors. Protein expression in whole-cell extracts was determined by Western blotting. BT-474 tumor xenografts in nude mice were used for in vivo studies of tumor progression. RO 48-8071 ([4'-[6-(Allylmethylamino)hexyloxy]-4-bromo-2'-fluorobenzophenone fumarate]; RO), a small-molecule inhibitor of oxidosqualene cyclase (OSC, a key enzyme in cholesterol biosynthesis), potently reduced breast cancer cell viability. In vitro exposure of estrogen receptor (ER)-positive human breast cancer cells to pharmacological levels of RO or a dose close to the IC50 for OSC (nM) reduced cell viability. Administration of RO to mice with BT-474 tumor xenografts prevented tumor growth, with no apparent toxicity. RO degraded ERα while concomitantly inducing the anti-proliferative protein ERβ. Two other cholesterol-lowering drugs, Fluvastatin and Simvastatin, were less effective in reducing breast cancer cell viability and were found not to induce ERβ. ERβ inhibition or knockdown prevented RO-dependent loss of cell viability. Importantly, RO had no effect on the viability of normal human mammary cells. RO is a potent inhibitor of hormone-dependent human breast cancer cell proliferation. The anti-tumor properties of RO appear to be in part due to an off-target effect that increases the ratio of ERβ/ERα in breast cancer cells.
Collapse
Affiliation(s)
- Yayun Liang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65211, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Wolfe E, Corsetti R, Bolton JS, Stolier AJ, Fuhrman GM. The evolution in management of patients with subcentimeter, node-negative, triple-negative breast cancer. Am J Surg 2013; 206:888-92; discussion 892-3. [DOI: 10.1016/j.amjsurg.2013.08.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/29/2013] [Accepted: 08/21/2013] [Indexed: 12/25/2022]
|
38
|
Seemann I, te Poele JAM, Song JY, Hoving S, Russell NS, Stewart FA. Radiation- and anthracycline-induced cardiac toxicity and the influence of ErbB2 blocking agents. Breast Cancer Res Treat 2013; 141:385-95. [PMID: 24091769 PMCID: PMC3824562 DOI: 10.1007/s10549-013-2707-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/18/2013] [Indexed: 01/10/2023]
Abstract
In Her2-positive breast cancer patients, inhibition of epidermal growth factor receptor 2 (ErbB2)-signaling is often combined with chemotherapy and radiotherapy. The risk of cardiac toxicity after anthracyclines and radiotherapy is recognized, but little is known about increased risk when these treatments are combined with ErbB2 inhibition. This study investigated whether ErbB2 inhibition increased radiation or anthracycline-induced toxicity. In an in vitro study, human cardiomyocytes were treated with irradiation or doxorubicin, alone or in combination with trastuzumab, and evaluated for cell survival and growth. Groups of mice received 0 or 14 Gy to the heart, alone or in combination with lapatinib, or 3 × 4 mg/kg doxorubicin alone or in combination with lapatinib. Mice were evaluated 40 weeks after treatment for cardiac damage. Changes in cardiac function ((99m)Tc-Myoview gated SPECT) were related to histomorphology and microvascular damage. Radiation or doxorubicin-induced cardiomyocyte toxicity (in vitro) were not exacerbated by trastuzumab. Cardiac irradiation of mice decreased microvascular density (MVD) and increased endothelial damage in surviving capillaries (decrease alkaline phosphatase expression and increased von Willebrand factor), but these changes were not exacerbated by lapatinib. Inflammatory responses in the irradiated epicardium (CD45+ and F4/80+ cells) were significantly reduced in combination with lapatinib. Irradiation, doxorubicin, and lapatinib each induced cardiac fibrosis but this was not further enhanced when treatments were combined. At the ultra-structural level, both lapatinib and doxorubicin induced mitochondrial damage, which was enhanced in combined treatments. Lapatinib alone also induced mild changes in cardiac function but this was not enhanced in the combined treatments. Trastuzumab did not enhance direct radiation or anthracycline toxicity of cardiomyocytes in vitro. Lapatinib did not enhance the risk of radiation or anthracycline-induced cardiac toxicity in mice up to 40 weeks after treatment, but mitochondrial damage was more severe after doxorubicin combined with lapatinib.
Collapse
Affiliation(s)
- Ingar Seemann
- Division of Biological Stress Response (H3), The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
39
|
Kaur H, Mao S, Shah S, Gorski DH, Krawetz SA, Sloane BF, Mattingly RR. Next-generation sequencing: a powerful tool for the discovery of molecular markers in breast ductal carcinoma in situ. Expert Rev Mol Diagn 2013; 13:151-65. [PMID: 23477556 DOI: 10.1586/erm.13.4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mammographic screening leads to frequent biopsies and concomitant overdiagnosis of breast cancer, particularly ductal carcinoma in situ (DCIS). Some DCIS lesions rapidly progress to invasive carcinoma, whereas others remain indolent. Because we cannot yet predict which lesions will not progress, all DCIS is regarded as malignant, and many women are overtreated. Thus, there is a pressing need for a panel of molecular markers in addition to the current clinical and pathological factors to provide prognostic information. Genomic technologies such as microarrays have made major contributions to defining subtypes of breast cancer. Next-generation sequencing (NGS) modalities offer unprecedented depth of expression analysis through revealing transcriptional boundaries, mutations, rare transcripts and alternative splice variants. NGS approaches are just beginning to be applied to DCIS. Here, the authors review the applications and challenges of NGS in discovering novel potential therapeutic targets and candidate biomarkers in the premalignant progression of breast cancer.
Collapse
Affiliation(s)
- Hitchintan Kaur
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Lee JC, Chou LC, Lien JC, Wu JC, Huang CH, Chung CH, Lee FY, Huang LJ, Kuo SC, Way TD. CLC604 preferentially inhibits the growth of HER2-overexpressing cancer cells and sensitizes these cells to the inhibitory effect of Taxol in vitro and in vivo. Oncol Rep 2013; 30:1762-72. [PMID: 23900492 DOI: 10.3892/or.2013.2634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/25/2013] [Indexed: 11/06/2022] Open
Abstract
HER2 has become a solicitous therapeutic target in metastatic and clinical drug-resistant cancer. Here, we evaluated whether or not 1-benzyl-3-(5-hydroxymethyl-2-furyl)indazole (YC-1) and its furopyrazole and thienopyrazole analogues repress the expression of the HER2 protein. Among the test compounds, (1-benzyl-3-(p-hydroxymethylphenyl)-5-methylfuro[3,2-c]pyrazol) (CLC604), an isosteric analogue of YC-1, significantly suppressed the expression of HER2, and preferentially inhibited cell proliferation and induced apoptosis in HER2-overexpressing cancer cells. Our results revealed that CLC604 reduced HER2 expression through a post-transcriptional mechanism and involvement of proteasomal activity. CLC604 disrupted the association of 90-kDa heat shock protein (Hsp90) with HER2 resulting from the inhibition of Hsp90 ATPase activity. Moreover, we found that CLC604 significantly enhanced the antitumor efficacy of clinical drugs against HER2-overexpressing tumors and efficiently reduced HER2-induced drug resistance in vitro and in vivo. These findings suggest that CLC604 should be developed further as a novel antitumor drug candidate for the treatment of drug-resistant cancer.
Collapse
Affiliation(s)
- Jang-Chang Lee
- Graduate Institute of Pharmaceutical Chemistry, College of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Elias JM. It Doesn't Take a Crystal Ball to See the Handwriting on the Wall. J Histotechnol 2013. [DOI: 10.1179/his.1993.16.4.311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
42
|
Allred DC, Clark GM, Tandon AK, McGuire WL. Immunohistochemistry on Histological Sections from Small (50 mg) Samples of Pulverized Breast Cancer. J Histotechnol 2013. [DOI: 10.1179/his.1993.16.2.117] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
43
|
Mohsin SK, Allred DC. Immunohistochemical Biomarkers in Breast Cancer. J Histotechnol 2013. [DOI: 10.1179/his.1999.22.3.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
44
|
Wang RA, Li ZS, Zhang HZ, Zheng PJ, Li QL, Shi JG, Yan QG, Ye J, Wang JB, Guo Y, Huang XF, Yu YH. Invasive cancers are not necessarily from preformed in situ tumours - an alternative way of carcinogenesis from misplaced stem cells. J Cell Mol Med 2013; 17:921-6. [PMID: 23741988 PMCID: PMC3822897 DOI: 10.1111/jcmm.12078] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 03/29/2013] [Indexed: 11/28/2022] Open
Abstract
Cancers are thought to be the result of accumulated gene mutations in cells. Carcinomas, which are cancers arising from epithelial tissues usually go through several stages of development: atypical hyperplasia, carcinoma in situ and then invasive carcinoma, which might further metastasize. However, we think that the present pathological data are enough to prove that there might be an alternative way of carcinogenesis. We propose that majority of invasive cancers arise in the connective tissue stroma de novo, from the misplaced epithelial stem cells which come to the wrong land of connective tissue stroma by accident. The in situ carcinomas, which are mostly curable, should not be considered genuine cancer, but rather as quasi-cancer. We design this new theory of carcinogenesis as the stem cell misplacement theory (SCMT). Our SCMT theory chains together other carcinogenesis theories such as the inflammation-cancer chain, the stem cell theory and the tissue organization field theory. However, we deny the pathway of somatic mutation theory as the major pathway of carcinogenesis.
Collapse
Affiliation(s)
- Rui-An Wang
- State Key Lab of Cancer Biology, The Fourth Military Medical University, Xi'an, Shaanxi Pr., China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chang J, Lee A, Lee J, Lim W, Sung SH, Moon BI. Correlation between the molecular subtype of breast cancer and the in vitro adenosine triphosphate-based chemosensitivity assay. JOURNAL OF THE KOREAN SURGICAL SOCIETY 2013; 84:313-20. [PMID: 23741688 PMCID: PMC3670999 DOI: 10.4174/jkss.2013.84.6.313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/01/2013] [Accepted: 04/10/2013] [Indexed: 11/30/2022]
Abstract
Purpose The empirical use of a chemotherapy regimen shows different results in individual breast cancer patient treatment. Recent studies showed the effectiveness of the adenosine triphosphate-based chemotherapy response assay (ATP-CRA). However, little is known about the correlation between chemosensitivity and breast cancer molecular subtypes. Therefore, we investigated whether the result of ATP-CRA is associated with a molecular subtype of breast cancer. Methods Two hundred eighty-seven patients diagnosed with breast cancer and receiving ATP-CRA at Mokdong Hospital, Ewha Womans University between September 2007 and December 2010 were enrolled in this study. Hormone receptor status, HER2/neu expression, and results of chemosensitivity tests of the patients was analyzed. Results In all of four subtypes, the combination of two agents showed significant higher mean cell death rate than a single agent. Within the breast cancer cell lines in this study, the range of chemosensitivity response was very wide and varied for each patient. For this reason, the molecular subtype of breast cancer is inconclusive in choosing an effective chemotherapeutic agent and in vitro chemosensitivity test, prior to therapy, could be a useful method for planning chemotherapy for each patient. Conclusion Chemosensitivity response to anticancer agents was found to vary depending on the individual breast cancer patients. The molecular subtype of breast cancer is inconclusive to choose the effective chemotherapeutic agent and the in vitro chemosensitivity test, prior to therapy, could be more useful for planning chemotherapy for each patient.
Collapse
Affiliation(s)
- Jina Chang
- Department of Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
46
|
Duggal S, Julian TB. A multigene expression assay to predict local recurrence risk for ductal carcinoma in situ. J Natl Cancer Inst 2013; 105:681-3. [PMID: 23641041 DOI: 10.1093/jnci/djt098] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
47
|
Fountzilas G, Dafni U, Bobos M, Kotoula V, Batistatou A, Xanthakis I, Papadimitriou C, Kostopoulos I, Koletsa T, Tsolaki E, Televantou D, Timotheadou E, Koutras A, Klouvas G, Samantas E, Pisanidis N, Karanikiotis C, Sfakianaki I, Pavlidis N, Gogas H, Linardou H, Kalogeras KT, Pectasides D, Dimopoulos MA. Evaluation of the prognostic role of centromere 17 gain and HER2/topoisomerase II alpha gene status and protein expression in patients with breast cancer treated with anthracycline-containing adjuvant chemotherapy: pooled analysis of two Hellenic Cooperative Oncology Group (HeCOG) phase III trials. BMC Cancer 2013; 13:163. [PMID: 23537287 PMCID: PMC3621498 DOI: 10.1186/1471-2407-13-163] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/20/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The HER2 gene has been established as a valid biological marker for the treatment of breast cancer patients with trastuzumab and probably other agents, such as paclitaxel and anthracyclines. The TOP2A gene has been associated with response to anthracyclines. Limited information exists on the relationship of HER2/TOP2A gene status in the presence of centromere 17 (CEP17) gain with outcome of patients treated with anthracycline-containing adjuvant chemotherapy. METHODS Formalin-fixed paraffin-embedded tumor tissue samples from 1031 patients with high-risk operable breast cancer, enrolled in two consecutive phase III trials, were assessed in a central laboratory by fluorescence in situ hybridization for HER2/TOP2A gene amplification and CEP17 gain (CEP17 probe). Amplification of HER2 and TOP2A were defined as a gene/CEP17 ratio of >2.2 and ≥2.0, respectively, or gene copy number higher than 6. Additionally, HER2, TopoIIa, ER/PgR and Ki67 protein expression was assessed by immunohistochemistry (IHC) and patients were classified according to their IHC phenotype. Treatment consisted of epirubicin-based adjuvant chemotherapy followed by hormonal therapy and radiation, as indicated. RESULTS HER2 amplification was found in 23.7% of the patients and TOP2A amplification in 10.1%. In total, 41.8% of HER2-amplified tumors demonstrated TOP2A co-amplification. The median (range) of HER2, TOP2A and CEP17 gain was 2.55 (0.70-45.15), 2.20 (0.70-26.15) and 2.00 (0.70-26.55), respectively. Forty percent of the tumors had CEP17 gain (51% of those with HER2 amplification). Adjusting for treatment groups in the Cox model, HER2 amplification, TOP2A amplification, CEP17 gain and HER2/TOP2A co-amplification were not associated with time to relapse or time to death. CONCLUSION HER2 amplification, TOP2A amplification, CEP17 gain and HER2/TOP2A co-amplification were not associated with outcome in high-risk breast cancer patients treated with anthracycline-based adjuvant chemotherapy. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry (ANZCTR) ACTRN12611000506998 and ACTRN12609001036202.
Collapse
Affiliation(s)
- George Fountzilas
- Department of Medical Oncology, Papageorgiou Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim JE, Ahn HJ, Ahn JH, Yoon DH, Kim SB, Jung KH, Gong GY, Kim MJ, Son BH, Ahn SH. Impact of triple-negative breast cancer phenotype on prognosis in patients with stage I breast cancer. J Breast Cancer 2012; 15:197-202. [PMID: 22807937 PMCID: PMC3395743 DOI: 10.4048/jbc.2012.15.2.197] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 04/06/2012] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Although most patients with stage I breast cancer have a good prognosis, their clinical outcomes may vary significantly. We assessed clinical outcomes and prognostic factors in stage I breast cancer patients with and without triple-negative breast cancer (TNBC) phenotype. METHODS Of 2,489 patients undergoing breast cancer surgery between January 1998 and December 2002, 554 (22.3%) had stage I breast cancer (tumor size ≤2 cm, and lymph node-negative). TNBC was defined as a primary tumor negative for estrogen and progesterone receptors (Allred scores <3/8) and for HER2/neu (0-1+ by immunohistochemistry). RESULTS Of the 554 patients with stage I breast cancer, 78 (14.1%) had TNBC. A significant proportion of TNBC patients had histologic grade 3 tumors (47.4% vs. 34.5%, p=0.031) and tumors >1 cm (87.2% vs. 75.8%, p=0.028) and received adjuvant chemotherapy (79.5% vs. 44.7%, p<0.001). During a median follow-up time of 8.7 years, 72 patients experienced tumor recurrences; 18 (23.1%) in the TNBC group and 54 (11.3%) in the non-TNBC group (p=0.010), with cumulative 3-year rate of recurrence of 12.8% and 5.3%, respectively (p=0.010). Ten-year relapse-free survival (RFS; 75.6% vs. 87.5%, p=0.004) and overall survival (OS; 83.0% vs. 91.4%, p=0.002) rates were significantly lower in the TNBC group. Multivariate analysis showed that triple negativity and histologic grade were independent predictors of shorter RFS and OS. CONCLUSION TNBC had more aggressive clinicopathologic characteristics and was associated with poorer survival in patients with stage I breast cancer. More intensive adjuvant chemotherapy or a different therapeutic strategy targeting this population is warranted.
Collapse
Affiliation(s)
- Jeong Eun Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mukohara T. Role of HER2-Targeted Agents in Adjuvant Treatment for Breast Cancer. CHEMOTHERAPY RESEARCH AND PRACTICE 2011; 2011:730360. [PMID: 22295205 PMCID: PMC3263614 DOI: 10.1155/2011/730360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 03/04/2011] [Accepted: 06/16/2011] [Indexed: 01/05/2023]
Abstract
Approximately 20% of breast cancers overexpress human epidermal growth factor receptor 2 (HER2) protein, mainly as a result of gene amplification. The receptor tyrosine kinase is believed to play a critical role in the pathogenesis and further proliferation of these tumors. The application of trastuzumab, a humanized monoclonal antibody against the extracellular domain of HER2 protein, to HER2-positive metastatic breast cancer has significantly improved treatment outcomes. Following this success, several phase III trials have evaluated the role of trastuzumab in the adjuvant setting, with the result that trastuzumab use is now the standard of care for most HER2-positive early breast cancer patients. In this paper, we review these pivotal phase III trials. We also discuss unresolved issues in adjuvant treatment with trastuzumab, including target patient population, sequential or concurrent use with chemotherapy or radiation, treatment duration, cardiotoxicity, and the possibility of eliminating chemotherapy. Following confirmation of its ability to partially overcome trastuzumab resistance, we also discuss the role of lapatinib in adjuvant use.
Collapse
Affiliation(s)
- Toru Mukohara
- Department of Medical Oncology/Hematology, Cancer Center, Kobe University Hospital, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
50
|
Shah S, Chen B. Testing for HER2 in Breast Cancer: A Continuing Evolution. PATHOLOGY RESEARCH INTERNATIONAL 2010; 2011:903202. [PMID: 21188214 PMCID: PMC3005907 DOI: 10.4061/2011/903202] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 10/22/2010] [Indexed: 01/29/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is an important prognostic and predictive factor in breast cancer. HER2 is overexpressed in approximately 15%–20% of invasive breast carcinomas and is associated with earlier recurrence, shortened disease free survival, and poor prognosis. Trastuzumab (Herceptin) a “humanized” monoclonal antibody targets the extracellular domain of HER2 and is widely used in the management of HER2 positive breast cancers. Accurate assessment of HER2 is thus critical in the management of breast cancer. The aim of this paper is to present a comprehensive review of HER2 with reference to its discovery and biology, clinical significance, prognostic value, targeted therapy, current and new testing modalities, and the interpretation guidelines and pitfalls.
Collapse
Affiliation(s)
- Sejal Shah
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|