1
|
Kollara A, Burt BD, Ringuette MJ, Brown TJ. The adaptor protein VEPH1 interacts with the kinase domain of ERBB2 and impacts EGF signaling in ovarian cancer cells. Cell Signal 2023; 106:110634. [PMID: 36828346 DOI: 10.1016/j.cellsig.2023.110634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023]
Abstract
Upregulation of ERBB2 and activating mutations in downstream KRAS/BRAF and PIK3CA are found in several ovarian cancer histotypes. ERBB2 enhances signaling by the ERBB family of EGF receptors, and contains docking positions for proteins that transduce signaling through multiple pathways. We identified the adaptor protein ventricular zone-expressed pleckstrin homology domain-containing protein 1 (VEPH1) as a potential interacting partner of ERBB2 in a screen of proteins co-immunoprecipitated with VEPH1. In this study, we confirm a VEPH1 - ERBB2 interaction by co-immunoprecipitation and biotin proximity labelling and show that VEPH1 interacts with the juxtamembrane-kinase domain of ERBB2. In SKOV3 ovarian cancer cells, which bear a PIK3CA mutation and ERBB2 overexpression, ectopic VEPH1 expression enhanced EGF activation of ERK1/2, and mTORC2 activation of AKT. In contrast, in ES2 ovarian cancer cells, which bear a BRAFV600E mutation with VEPH1 amplification but low ERBB2 expression, loss of VEPH1 expression enabled further activation of ERK1/2 by EGF and enhanced EGF activation of AKT. VEPH1 expression in SKOV3 cells enhanced EGF-induced cell migration consistent with increased Snail2 and decreased E-cadherin levels. In comparison, loss of VEPH1 expression in ES2 cells led to decreased cell motility independent of EGF treatment despite higher levels of N-cadherin and Snail2. Importantly, we found that loss of VEPH1 expression rendered ES2 cells less sensitive to BRAF and MEK inhibition. This study extends the range of adaptor function of VEPH1 to ERBB2, and indicates VEPH1 has differential effects on EGF signaling in ovarian cancer cells that may be influenced by driver gene mutations.
Collapse
Affiliation(s)
- Alexandra Kollara
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Brian D Burt
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Maurice J Ringuette
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Theodore J Brown
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Kumar R, George B, Campbell MR, Verma N, Paul AM, Melo-Alvim C, Ribeiro L, Pillai MR, da Costa LM, Moasser MM. HER family in cancer progression: From discovery to 2020 and beyond. Adv Cancer Res 2020; 147:109-160. [PMID: 32593399 DOI: 10.1016/bs.acr.2020.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The human epidermal growth factor receptor (HER) family of receptor tyrosine kinases (RTKs) are among the first layer of molecules that receive, interpret, and transduce signals leading to distinct cancer cell phenotypes. Since the discovery of the tooth-lid factor-later characterized as the epidermal growth factor (EGF)-and its high-affinity binding EGF receptor, HER kinases have emerged as one of the commonly upregulated or hyperactivated or mutated kinases in epithelial tumors, thus allowing HER1-3 family members to regulate several hallmarks of cancer development and progression. Each member of the HER family exhibits shared and unique structural features to engage multiple receptor activation modes, leading to a range of overlapping and distinct phenotypes. EGFR, the founding HER family member, provided the roadmap for the development of the cell surface RTK-directed targeted cancer therapy by serving as a prototype/precursor for the currently used HER-directed cancer drugs. We herein provide a brief account of the discoveries, defining moments, and historical context of the HER family and guidepost advances in basic, translational, and clinical research that solidified a prominent position of the HER family in cancer research and treatment. We also discuss the significance of HER3 pseudokinase in cancer biology; its unique structural features that drive transregulation among HER1-3, leading to a superior proximal signaling response; and potential role of HER3 as a shared effector of acquired therapeutic resistance against diverse oncology drugs. Finally, we also narrate some of the current drawbacks of HER-directed therapies and provide insights into postulated advances in HER biology with extensive implications of these therapies in cancer research and treatment.
Collapse
Affiliation(s)
- Rakesh Kumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India; Department of Medicine, Division of Hematology & Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Bijesh George
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Marcia R Campbell
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Nandini Verma
- Advanced Centre for Treatment, Research and Education in Cancer, Mumbai, India
| | - Aswathy Mary Paul
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Cecília Melo-Alvim
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Leonor Ribeiro
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - M Radhakrishna Pillai
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Luis Marques da Costa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mark M Moasser
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
3
|
Jacob W, James I, Hasmann M, Weisser M. Clinical development of HER3-targeting monoclonal antibodies: Perils and progress. Cancer Treat Rev 2018; 68:111-123. [PMID: 29944978 DOI: 10.1016/j.ctrv.2018.06.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 12/16/2022]
Abstract
The human epidermal growth factor receptor (HER) family consists of four transmembrane receptor tyrosine kinases: epidermal growth factor receptor (EGFR), HER2, HER3, and HER4. They are part of a complex signalling network and stimulate intracellular pathways regulating cell growth and differentiation. So far, monoclonal antibodies (mAbs) and small molecule tyrosine kinase inhibitors targeting EGFR and HER2 have been developed and approved. Recently, focus has turned to HER3 as it may play an important role in resistance to EGFR- and HER2-targeting therapies. HER3-targeting agents have been undergoing clinical evaluation for the last 10 years and currently thirteen mAbs are in phase 1 or 2 clinical studies. Single agent activity has proven to be limited, however, the tolerability was favourable. Thus, combinations of HER3-binding mAbs with other HER-targeting therapies or chemotherapies have been pursued in various solid tumor entities. Data indicate that the HER3-binding ligand heregulin may serve as a response prediction marker for HER3-targeting therapy. Within this review the current status of clinical development of HER3-targeting compounds is described.
Collapse
Affiliation(s)
- Wolfgang Jacob
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany.
| | - Ian James
- A4P Consulting Ltd, Discovery Park, Sandwich, UK
| | - Max Hasmann
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Martin Weisser
- Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
4
|
Khalil HS, Langdon SP, Goltsov A, Soininen T, Harrison DJ, Bown J, Deeni YY. A novel mechanism of action of HER2 targeted immunotherapy is explained by inhibition of NRF2 function in ovarian cancer cells. Oncotarget 2018; 7:75874-75901. [PMID: 27713148 PMCID: PMC5342785 DOI: 10.18632/oncotarget.12425] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/21/2016] [Indexed: 12/16/2022] Open
Abstract
Nuclear erythroid related factor-2 (NRF2) is known to promote cancer therapeutic detoxification and crosstalk with growth promoting pathways. HER2 receptor tyrosine kinase is frequently overexpressed in cancers leading to uncontrolled receptor activation and signaling. A combination of HER2 targeting monoclonal antibodies shows greater anticancer efficacy than the single targeting antibodies, however, its mechanism of action is largely unclear. Here we report novel actions of anti-HER2 drugs, Trastuzumab and Pertuzumab, involving NRF2. HER2 targeting by antibodies inhibited growth in association with persistent generation of reactive oxygen species (ROS), glutathione (GSH) depletion, reduction in NRF2 levels and inhibition of NRF2 function in ovarian cancer cell lines. The combination of antibodies produced more potent effects than single antibody alone; downregulated NRF2 substrates by repressing the Antioxidant Response (AR) pathway with concomitant transcriptional inhibition of NRF2. We showed the antibody combination produced increased methylation at the NRF2 promoter consistent with repression of NRF2 antioxidant function, as HDAC and methylation inhibitors reversed such produced transcriptional effects. These findings demonstrate a novel mechanism and role for NRF2 in mediating the response of cancer cells to the combination of Trastuzumab and Pertuzumab and reinforce the importance of NRF2 in drug resistance and as a key anticancer target.
Collapse
Affiliation(s)
- Hilal S Khalil
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - Simon P Langdon
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, United Kingdom
| | - Alexey Goltsov
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - Tero Soininen
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - David J Harrison
- School of Medicine, University of St Andrews, St Andrews, KY16 9TF, United Kingdom
| | - James Bown
- Division of Computing and Mathematics, School of Arts, Media, and Computer Games, Abertay University, Dundee, DD1 1HG, United Kingdom
| | - Yusuf Y Deeni
- Division of Science, School of Science, Engineering and Technology, Abertay University, Dundee, DD1 1HG, United Kingdom
| |
Collapse
|
5
|
N-glycans of growth factor receptors: their role in receptor function and disease implications. Clin Sci (Lond) 2017; 130:1781-92. [PMID: 27612953 DOI: 10.1042/cs20160273] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/08/2016] [Indexed: 11/17/2022]
Abstract
Numerous signal-transduction-related molecules are secreted proteins or membrane proteins, and the mechanism by which these molecules are regulated by glycan chains is a very important issue for developing an understanding of the cellular events that transpire. This review covers the functional regulation of epidermal growth factor receptor (EGFR), ErbB3 and the transforming growth factor β (TGF-β) receptor by N-glycans. This review shows that the N-glycans play important roles in regulating protein conformation and interactions with carbohydrate recognition molecules. These results point to the possibility of a novel strategy for controlling cell signalling and developing novel glycan-based therapeutics.
Collapse
|
6
|
Bax HJ, Josephs DH, Pellizzari G, Spicer JF, Montes A, Karagiannis SN. Therapeutic targets and new directions for antibodies developed for ovarian cancer. MAbs 2016; 8:1437-1455. [PMID: 27494775 PMCID: PMC5098446 DOI: 10.1080/19420862.2016.1219005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antibody therapeutics against different target antigens are widely used in the treatment of different malignancies including ovarian carcinomas, but this disease still requires more effective agents. Improved understanding of the biological features, signaling pathways, and immunological escape mechanisms involved in ovarian cancer has emerged in the past few years. These advances, including an appreciation of the cross-talk between cancer cells and the patient's immune system, have led to the identification of new targets. In turn, potential antibody treatments with various mechanisms of action, including immune activation or toxin-delivery, that are directed at these targets have been developed. Here, we identify established as well as novel targets for antibodies in ovarian cancer, and discuss how they may provide fresh opportunities to identify interventions with enhanced therapeutic potential.
Collapse
Affiliation(s)
- Heather J Bax
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK.,b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - Debra H Josephs
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK.,b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - Giulia Pellizzari
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK.,b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - James F Spicer
- b Division of Cancer Studies, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital , London , UK
| | - Ana Montes
- c Department of Medical Oncology , Guy's and St Thomas' NHS Foundation Trust , London , UK
| | - Sophia N Karagiannis
- a St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London & NIHR Biomedical Research Center at Guy's and St. Thomas' Hospital and King's College London, Guy's Hospital, King's College London , London , UK
| |
Collapse
|
7
|
Kurzeder C, Bover I, Marmé F, Rau J, Pautier P, Colombo N, Lorusso D, Ottevanger P, Bjurberg M, Marth C, Barretina-Ginesta P, Vergote I, Floquet A, del Campo JM, Mahner S, Bastière-Truchot L, Martin N, Oestergaard MZ, Kiermaier A, Schade-Brittinger C, Polleis S, du Bois A, Gonzalez-Martin A. Double-Blind, Placebo-Controlled, Randomized Phase III Trial Evaluating Pertuzumab Combined With Chemotherapy for Low Tumor Human Epidermal Growth Factor Receptor 3 mRNA–Expressing Platinum-Resistant Ovarian Cancer (PENELOPE). J Clin Oncol 2016; 34:2516-25. [DOI: 10.1200/jco.2015.66.0787] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The AGO-OVAR 2.29/ENGOT-ov14/PENELOPE prospectively randomized phase III trial evaluated the addition of pertuzumab to chemotherapy in patients with platinum-resistant ovarian carcinoma with low tumor human epidermal growth factor receptor 3 (HER3) mRNA expression. We report the results of the primary efficacy analysis. Patients and Methods Eligible patients had ovarian carcinoma that progressed during or within 6 months of completing four or more platinum cycles, centrally tested low tumor HER3 mRNA expression (concentration ratio ≤ 2.81 by quantitative reverse transcriptase polymerase chain reaction on cobas z480 [Roche Molecular Diagnostics, Pleasanton, CA]), and no more than two prior lines of chemotherapy. After investigators’ selection of the chemotherapy backbone (single-agent topotecan, weekly paclitaxel, or gemcitabine), patients were randomly assigned to also receive either placebo or pertuzumab (840-mg loading dose followed by 420 mg every 3 weeks). Stratification factors were selected chemotherapy, prior antiangiogenic therapy, and platinum-free interval. The primary end point was independent review committee–assessed progression-free survival (PFS). Additional end points included overall survival, investigator-assessed PFS, objective response rate, safety, patient-reported outcomes, and translational research. Results Overall, 156 patients were randomly assigned. Adding pertuzumab to chemotherapy did not significantly improve independent review committee–assessed PFS for the primary analysis (stratified hazard ratio, 0.74; 95% CI, 0.50 to 1.11; P = .14; median PFS, 4.3 months for pertuzumab plus chemotherapy v 2.6 months for placebo plus chemotherapy). Sensitivity analyses and secondary efficacy end point results were consistent with the primary analysis. The effect on PFS favoring pertuzumab was more pronounced in the gemcitabine and paclitaxel cohorts. No new safety signals were seen. Conclusion Although the primary objective was not met, subgroup analyses showed trends in PFS favoring pertuzumab in the gemcitabine and paclitaxel cohorts, meriting further exploration of pertuzumab in ovarian cancer.
Collapse
Affiliation(s)
- Christian Kurzeder
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Isabel Bover
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Frederik Marmé
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Joern Rau
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Patricia Pautier
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Nicoletta Colombo
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Domenica Lorusso
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Petronella Ottevanger
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Maria Bjurberg
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Christian Marth
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Pilar Barretina-Ginesta
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Ignace Vergote
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Anne Floquet
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Josep M. del Campo
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Sven Mahner
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Lydie Bastière-Truchot
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Nicolas Martin
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Mikkel Z. Oestergaard
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Astrid Kiermaier
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Carmen Schade-Brittinger
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Sandra Polleis
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Andreas du Bois
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Antonio Gonzalez-Martin
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| |
Collapse
|
8
|
Pertuzumab Plus Chemotherapy for Platinum-Resistant Ovarian Cancer: Safety Run-in Results of the PENELOPE Trial. Int J Gynecol Cancer 2016; 26:898-905. [DOI: 10.1097/igc.0000000000000695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
ObjectiveIn platinum-resistant ovarian cancer, adding pertuzumab to gemcitabine improved progression-free survival in the subgroup with low tumor HER3 messenger RNA expression. The 2-part PENELOPE trial (NCT01684878) is prospectively investigating pertuzumab plus chemotherapy in this population.Patients and MethodsPart 1 evaluated pertuzumab plus either topotecan or paclitaxel. Patients with platinum-refractory or platinum-resistant recurrent ovarian, primary peritoneal, or fallopian tube cancer and low HER3 messenger RNA expression (concentration ratio ≤2.81 by central quantitative reverse transcriptase-polymerase chain reaction testing on Cobas z480) received intravenous pertuzumab (840 mg loading dose then 420 mg every 3 weeks) with the investigator’s choice of topotecan (1.25 mg/m2days 1–5 every 3 weeks) or weekly paclitaxel (80 mg/m2) until disease progression or unacceptable toxicity. The primary objective was to assess safety and tolerability.ResultsFifty patients were treated in part 1 (22 topotecan; 28 paclitaxel). In both cohorts, disease progression was the most common primary reason for discontinuing pertuzumab, and the most common all-grade adverse events (AEs) were fatigue/asthenia, anemia, and diarrhea. The most common grade ≥3 AEs were anemia (36%), neutropenia (27%), and fatigue/asthenia (18%) for topotecan, and peripheral sensory neuropathy (14%) and anemia (11%) for paclitaxel. Two patients receiving paclitaxel-pertuzumab died from AEs (abdominal infection; unexplained death). Median progression-free survival was 4.1 months (95% confidence interval, 1.9–6.1) with topotecan-pertuzumab and 4.2 months (95% confidence interval, 3.5–6.0) with paclitaxel-pertuzumab.ConclusionsBased on part 1 tolerability, the Independent Data Monitoring Committee had no objection to PENELOPE proceeding to part 2, a double-blind randomized comparison of chemotherapy (topotecan, paclitaxel, or gemcitabine) plus pertuzumab or placebo.
Collapse
|
9
|
Takahashi M, Kizuka Y, Ohtsubo K, Gu J, Taniguchi N. Disease-associated glycans on cell surface proteins. Mol Aspects Med 2016; 51:56-70. [PMID: 27131428 DOI: 10.1016/j.mam.2016.04.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 04/23/2016] [Indexed: 01/02/2023]
Abstract
Most of membrane molecules including cell surface receptors and secreted proteins including ligands are glycoproteins and glycolipids. Therefore, identifying the functional significance of glycans is crucial for developing an understanding of cell signaling and subsequent physiological and pathological cellular events. In particular, the function of N-glycans associated with cell surface receptors has been extensively studied since they are directly involved in controlling cellular functions. In this review, we focus on the roles of glycosyltransferases that are involved in the modification of N-glycans and their target proteins such as epidermal growth factor receptor (EGFR), ErbB3, transforming growth factor β (TGF-β) receptor, T-cell receptors (TCR), β-site APP cleaving enzyme (BACE1), glucose transporter 2 (GLUT2), E-cadherin, and α5β1 integrin in relation to diseases and epithelial-mesenchymal transition (EMT) process. Above of those proteins are subjected to being modified by several glycosyltransferases such as N-acetylglucosaminyltransferase III (GnT-III), N-acetylglucosaminyltransferase IV (GnT-IV), N-acetylglucosaminyltransferase V (GnT-V), α2,6 sialyltransferase 1 (ST6GAL1), and α1,6 fucosyltransferase (Fut8), which are typical N-glycan branching enzymes and play pivotal roles in regulating the function of cell surface receptors in pathological cell signaling.
Collapse
Affiliation(s)
- Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, South-1 West-17, Chuo-ku, Sapporo 060-8556, Japan
| | - Yasuhiko Kizuka
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, Global Research Cluster, RIKEN, 2-1 Hirosawa, Wako 351-0198, Japan
| | - Kazuaki Ohtsubo
- Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-ku, Kumamoto 862-0976, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsusima, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Naoyuki Taniguchi
- Systems Glycobiology Research Group, RIKEN-Max Planck Joint Research Center, Global Research Cluster, RIKEN, 2-1 Hirosawa, Wako 351-0198, Japan.
| |
Collapse
|
10
|
Zhang N, Chang Y, Rios A, An Z. HER3/ErbB3, an emerging cancer therapeutic target. Acta Biochim Biophys Sin (Shanghai) 2016; 48:39-48. [PMID: 26496898 DOI: 10.1093/abbs/gmv103] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/10/2015] [Indexed: 01/24/2023] Open
Abstract
HER3 is a member of the HER (EGFR/ErbB) receptor family consisting of four closely related type 1 transmembrane receptors (EGFR, HER2, HER3, and HER4). HER receptors are part of a complex signaling network intertwined with the Ras/Raf/MAPK, PI3K/AKT, JAK/STAT, and PKC signaling pathways. Aberrant activation of the HER receptors and downstream signaling molecules tips the balance on cellular events, leading to various types of cancers. Monoclonal antibodies (mAbs) and small molecule inhibitors targeting EGFR and HER2 tyrosine kinase activities exhibit clinical benefits in the treatment of several types of cancers, but their clinical efficacy is limited by the occurrence of drug resistance. HER3 is the preferred dimerization partner of HER2 and it is well established that HER3 plays an important role in drug resistance to EGFR- and HER2-targeting therapies. Since HER3 has limited kinase activity, mAbs are being explored to target HER3 for cancer therapy. Currently, approximately a dozen of anti-HER3 mAbs are at different stages of clinical development. However, the lack of established biomarkers has made it more challenging to stratify cancer patients to whom HER3-targeting therapies can be more effective. In this review, we focus on the validation of HER3 as a cancer drug target, the recent development in biomarker discovery for anti-HER3 therapies, and the progress made in the clinical development of HER3-targeting mAbs.
Collapse
Affiliation(s)
- Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | - Adan Rios
- Division of Oncology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
11
|
NRF2 Regulates HER2 and HER3 Signaling Pathway to Modulate Sensitivity to Targeted Immunotherapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:4148791. [PMID: 26770651 PMCID: PMC4685121 DOI: 10.1155/2016/4148791] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/27/2022]
Abstract
NF-E2 related factor-2 (NRF2) is an essential transcription factor for multiple genes encoding antioxidants and detoxification enzymes. NRF2 is implicated in promoting cancer therapeutic resistance by its detoxification function and crosstalk with proproliferative pathways. However, the exact mechanism of this intricate connectivity between NRF2 and growth factor induced proliferative pathway remains elusive. Here, we have demonstrated that pharmacological activation of NRF2 by tert-butylhydroquinone (tBHQ) upregulates the HER family receptors, HER2 and HER3 expression, elevates pAKT levels, and enhances the proliferation of ovarian cancer cells. Preactivation of NRF2 also attenuates the combined growth inhibitory effects of HER2 targeting monoclonal antibodies, Pertuzumab and Trastuzumab. Further, tBHQ caused transcriptional induction of HER2 and HER3, while SiRNA-mediated knockdown of NRF2 prevented this and further caused transcriptional repression and enhanced cytotoxicity of the HER2 inhibitors. Hence, NRF2 regulates both HER2 and HER3 receptors to influence cellular responses to HER2 targeting monoclonal antibodies. This deciphered crosstalk mechanism reinforces the role of NRF2 in drug resistance and as a relevant anticancer target.
Collapse
|
12
|
Hodeib M, Serna-Gallegos T, Tewari KS. A review of HER2-targeted therapy in breast and ovarian cancer: lessons from antiquity - CLEOPATRA and PENELOPE. Future Oncol 2015; 11:3113-31. [PMID: 26597460 DOI: 10.2217/fon.15.266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although breast and ovarian cancer have notable distinctions, there may exist parallel pathways that can be exploited for therapeutic gain. For example, the therapeutic arena in breast cancer has benefited greatly from available endocrine therapies as well as novel drugs designed to target the HER2 receptor, including trastuzumab, lapatinib, T-DM1 and pertuzumab. CLEOPATRA, a Phase III randomized clinical trial studying pertuzumab in women with HER2-amplified metastatic breast cancer, was practice-changing in 2014. Its counterpart, the Phase III randomized PENELOPE trial, was activated following promising Phase II data and studied pertuzumab in an enriched ovarian cancer patient population with low HER3 mRNA. This review will trace the development of anti-HER2 therapies in breast and ovarian cancer.
Collapse
Affiliation(s)
- Melissa Hodeib
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, University of California, Irvine-Medical Center, Orange, CA, USA
| | - Tasha Serna-Gallegos
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, University of California, Irvine-Medical Center, Orange, CA, USA
| | - Krishnansu S Tewari
- Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, University of California, Irvine-Medical Center, Orange, CA, USA
| |
Collapse
|
13
|
McEvoy LM, O'Toole SA, Spillane CD, Martin CM, Gallagher MF, Stordal B, Blackshields G, Sheils O, O'Leary JJ. Identifying novel hypoxia-associated markers of chemoresistance in ovarian cancer. BMC Cancer 2015. [PMID: 26205780 PMCID: PMC4513971 DOI: 10.1186/s12885-015-1539-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Ovarian cancer is associated with poor long-term survival due to late diagnosis and development of chemoresistance. Tumour hypoxia is associated with many features of tumour aggressiveness including increased cellular proliferation, inhibition of apoptosis, increased invasion and metastasis, and chemoresistance, mostly mediated through hypoxia-inducible factor (HIF)-1α. While HIF-1α has been associated with platinum resistance in a variety of cancers, including ovarian, relatively little is known about the importance of the duration of hypoxia. Similarly, the gene pathways activated in ovarian cancer which cause chemoresistance as a result of hypoxia are poorly understood. This study aimed to firstly investigate the effect of hypoxia duration on resistance to cisplatin in an ovarian cancer chemoresistance cell line model and to identify genes whose expression was associated with hypoxia-induced chemoresistance. Methods Cisplatin-sensitive (A2780) and cisplatin-resistant (A2780cis) ovarian cancer cell lines were exposed to various combinations of hypoxia and/or chemotherapeutic drugs as part of a ‘hypoxia matrix’ designed to cover clinically relevant scenarios in terms of tumour hypoxia. Response to cisplatin was measured by the MTT assay. RNA was extracted from cells treated as part of the hypoxia matrix and interrogated on Affymetrix Human Gene ST 1.0 arrays. Differential gene expression analysis was performed for cells exposed to hypoxia and/or cisplatin. From this, four potential markers of chemoresistance were selected for evaluation in a cohort of ovarian tumour samples by RT-PCR. Results Hypoxia increased resistance to cisplatin in A2780 and A2780cis cells. A plethora of genes were differentially expressed in cells exposed to hypoxia and cisplatin which could be associated with chemoresistance. In ovarian tumour samples, we found trends for upregulation of ANGPTL4 in partial responders and down-regulation in non-responders compared with responders to chemotherapy; down-regulation of HER3 in partial and non-responders compared to responders; and down-regulation of HIF-1α in non-responders compared with responders. Conclusion This study has further characterized the relationship between hypoxia and chemoresistance in an ovarian cancer model. We have also identified many potential biomarkers of hypoxia and platinum resistance and provided an initial validation of a subset of these markers in ovarian cancer tissues. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1539-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lynda M McEvoy
- Department of Histopathology TCD, Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland. .,Department of Obstetrics and Gynaecology, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland.
| | - Sharon A O'Toole
- Department of Obstetrics and Gynaecology, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland.
| | - Cathy D Spillane
- Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin 8, Ireland.
| | - Cara M Martin
- Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin 8, Ireland.
| | - Michael F Gallagher
- Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin 8, Ireland.
| | - Britta Stordal
- Department of Histopathology TCD, Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.
| | - Gordon Blackshields
- Department of Histopathology TCD, Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.
| | - Orla Sheils
- Department of Histopathology TCD, Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland.
| | - John J O'Leary
- Department of Histopathology TCD, Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin 8, Ireland. .,Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin 8, Ireland.
| |
Collapse
|
14
|
Abstract
Over the past decade, trastuzumab was the only available monoclonal anti-HER2 antibody for the treatment of HER2 positive breast and gastric cancer. Recently, pertuzumab added to docetaxel and trastuzumab showed dramatic overall survival improvement in first line treatment of HER2 positive metastatic breast cancer. Pertuzumab is the first approved monoclonal antibody in a new class of drugs called dimerization inhibitors. This agent was also approved in association with trastuzumab for neoadjuvant HER2-positive breast cancer treatment. However, pertuzumab development was not confined to breast cancer and in the present review, we will focus on biological rational, preclinical data and clinical trial results of pertuzumab in solid tumors excluding breast cancer.
Collapse
|
15
|
Matsuoka T, Yashiro M. Recent advances in the HER2 targeted therapy of gastric cancer. World J Clin Cases 2015; 3:42-51. [PMID: 25610849 PMCID: PMC4295218 DOI: 10.12998/wjcc.v3.i1.42] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/09/2014] [Accepted: 11/19/2014] [Indexed: 02/05/2023] Open
Abstract
Recent advances in molecular targeted therapies, including targeting human epidermal growth factor receptor 2 (HER2), had a major forward step in the therapy for gastric cancer patients. Application of HER2-targeted therapies, in particular trastuzumab in combination with chemotherapy in metastatic HER2-positive gastric cancers, resulted in improvements in response rates, time to progression and overall survival. Nevertheless, as with breast cancer, many patients with gastric cancer develop resistance to trastuzumab. Several promising therapies are currently being developed in combination with chemotherapy to increase the efficacy and overcome the cancer-resistance. Here we review the current overview of clinical application of agents targeting HER2 in gastric cancer. We also discuss the ongoing trials supporting the use of HER2-targeted agents combined with cytotoxic agents or other monoclonal antibodies.
Collapse
|
16
|
Huang W, Wu QD, Zhang M, Kong YL, Cao PR, Zheng W, Xu JH, Ye M. Novel Hsp90 inhibitor FW-04-806 displays potent antitumor effects in HER2-positive breast cancer cells as a single agent or in combination with lapatinib. Cancer Lett 2015; 356:862-71. [DOI: 10.1016/j.canlet.2014.10.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/30/2014] [Accepted: 10/30/2014] [Indexed: 10/24/2022]
|
17
|
Thomas G, ChardèS T, Gaborit N, Mollevi C, Leconet W, Robert B, Radosevic-Robin N, Penault-Llorca F, Gongora C, Colombo PE, Lazrek Y, Bras-Goncalves R, Savina A, Azria D, Bazin H, PèLegrin A, Larbouret C. HER3 as biomarker and therapeutic target in pancreatic cancer: new insights in pertuzumab therapy in preclinical models. Oncotarget 2014; 5:7138-48. [PMID: 25216528 PMCID: PMC4196190 DOI: 10.18632/oncotarget.2231] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 07/16/2014] [Indexed: 02/05/2023] Open
Abstract
The anti-HER2 antibody pertuzumab inhibits HER2 dimerization and affects HER2/HER3 dimer formation and signaling. As HER3 and its ligand neuregulin are implicated in pancreatic tumorigenesis, we investigated whether HER3 expression could be a predictive biomarker of pertuzumab efficacy in HER2low-expressing pancreatic cancer. We correlated in vitro and in vivo HER3 expression and neuregulin dependency with the inhibitory effect of pertuzumab on cell viability and tumor progression. HER3 knockdown in BxPC-3 cells led to resistance to pertuzumab therapy. Pertuzumab treatment of HER3-expressing pancreatic cancer cells increased HER3 at the cell membrane, whereas the anti-HER3 monoclonal antibody 9F7-F11 down-regulated it. Both antibodies blocked HER3 and AKT phosphorylation and inhibited HER2/HER3 heterodimerization but affected differently HER2 and HER3 homodimers. The pertuzumab/9F7-F11 combination enhanced tumor inhibition and the median survival time in mice xenografted with HER3-expressing pancreatic cancer cells. Finally, HER2 and HER3 were co-expressed in 11% and HER3 alone in 27% of the 45 pancreatic ductal adenocarcinomas analyzed by immunohistochemistry. HER3 is essential for pertuzumab efficacy in HER2low-expressing pancreatic cancer and HER3 expression might be a predictive biomarker of pertuzumab efficacy in such cancers. Further studies in clinical samples are required to confirm these findings and the interest of combining anti-HER2 and anti-HER3 therapeutic antibodies.
Collapse
Affiliation(s)
- GaëLle Thomas
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
- Institut Roche de Recherche et Médecine Translationnelle, Boulogne Bilancourt, France
| | - Thierry ChardèS
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - NadèGe Gaborit
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Caroline Mollevi
- Unité de Biostatistiques, ICM Val d'Aurelle, Montpellier, France
| | - Wilhem Leconet
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Bruno Robert
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Nina Radosevic-Robin
- Department of Biopathology, The Jean Perrin Comprehensive Cancer Center and ERTICa Research Group, University of Auvergne EA4677, Clermont-Ferrand, France
| | - FréDéRique Penault-Llorca
- Department of Biopathology, The Jean Perrin Comprehensive Cancer Center and ERTICa Research Group, University of Auvergne EA4677, Clermont-Ferrand, France
| | - CéLine Gongora
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Pierre-Emmanuel Colombo
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Yassamine Lazrek
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
- Millegen SA, F-31681, Labège, France
| | - Rui Bras-Goncalves
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Ariel Savina
- Roche SAS Scientific Partnerships, Boulogne Billancourt, France
| | - David Azria
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | | | - André PèLegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| | - Christel Larbouret
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France; INSERM, Unit 896, Montpellier, F-34298, France; Université Montpellier1, Montpellier, F-34298, France; ICM, Montpellier, France
| |
Collapse
|
18
|
Goltsov A, Deeni Y, Khalil HS, Soininen T, Kyriakidis S, Hu H, Langdon SP, Harrison DJ, Bown J. Systems analysis of drug-induced receptor tyrosine kinase reprogramming following targeted mono- and combination anti-cancer therapy. Cells 2014; 3:563-91. [PMID: 24918976 PMCID: PMC4092865 DOI: 10.3390/cells3020563] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/14/2014] [Accepted: 05/19/2014] [Indexed: 12/12/2022] Open
Abstract
The receptor tyrosine kinases (RTKs) are key drivers of cancer progression and targets for drug therapy. A major challenge in anti-RTK treatment is the dependence of drug effectiveness on co-expression of multiple RTKs which defines resistance to single drug therapy. Reprogramming of the RTK network leading to alteration in RTK co-expression in response to drug intervention is a dynamic mechanism of acquired resistance to single drug therapy in many cancers. One route to overcome this resistance is combination therapy. We describe the results of a joint in silico, in vitro, and in vivo investigations on the efficacy of trastuzumab, pertuzumab and their combination to target the HER2 receptors. Computational modelling revealed that these two drugs alone and in combination differentially suppressed RTK network activation depending on RTK co-expression. Analyses of mRNA expression in SKOV3 ovarian tumour xenograft showed up-regulation of HER3 following treatment. Considering this in a computational model revealed that HER3 up-regulation reprograms RTK kinetics from HER2 homodimerisation to HER3/HER2 heterodimerisation. The results showed synergy of the trastuzumab and pertuzumab combination treatment of the HER2 overexpressing tumour can be due to an independence of the combination effect on HER3/HER2 composition when it changes due to drug-induced RTK reprogramming.
Collapse
Affiliation(s)
- Alexey Goltsov
- Scottish Informatics, Mathematics, Biology and Statistics Centre (SIMBIOS), Abertay University, Dundee, DD1 1HG, United Kingdom.
| | - Yusuf Deeni
- Scottish Informatics, Mathematics, Biology and Statistics Centre (SIMBIOS), Abertay University, Dundee, DD1 1HG, United Kingdom.
| | - Hilal S Khalil
- Scottish Informatics, Mathematics, Biology and Statistics Centre (SIMBIOS), Abertay University, Dundee, DD1 1HG, United Kingdom.
| | - Tero Soininen
- Scottish Informatics, Mathematics, Biology and Statistics Centre (SIMBIOS), Abertay University, Dundee, DD1 1HG, United Kingdom.
| | | | - Huizhong Hu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Simon P Langdon
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, United Kingdom.
| | - David J Harrison
- School of Medicine, University of St Andrews, St Andrews, KY16 9TF, United Kingdom.
| | - James Bown
- Scottish Informatics, Mathematics, Biology and Statistics Centre (SIMBIOS), Abertay University, Dundee, DD1 1HG, United Kingdom.
| |
Collapse
|
19
|
Lipton A, Goodman L, Leitzel K, Cook J, Sperinde J, Haddad M, Köstler WJ, Huang W, Weidler JM, Ali S, Newton A, Fuchs EM, Paquet A, Singer CF, Horvat R, Jin X, Banerjee J, Mukherjee A, Tan Y, Shi Y, Chenna A, Larson J, Lie Y, Sherwood T, Petropoulos CJ, Williams S, Winslow J, Parry G, Bates M. HER3, p95HER2, and HER2 protein expression levels define multiple subtypes of HER2-positive metastatic breast cancer. Breast Cancer Res Treat 2014; 141:43-53. [PMID: 23959396 PMCID: PMC3758835 DOI: 10.1007/s10549-013-2665-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/05/2013] [Indexed: 01/06/2023]
Abstract
Trastuzumab is effective in the treatment of HER2/neu over-expressing breast cancer, but not all patients benefit from it. In vitro data suggest a role for HER3 in the initiation of signaling activity involving the AKT–mTOR pathway leading to trastuzumab insensitivity. We sought to investigate the potential of HER3 alone and in the context of p95HER2 (p95), a trastuzumab resistance marker, as biomarkers of trastuzumab escape. Using the VeraTag® assay platform, we developed a dual antibody proximity-based assay for the precise quantitation of HER3 total protein (H3T) from formalin-fixed paraffin-embedded (FFPE) breast tumors. We then measured H3T in 89 patients with metastatic breast cancer treated with trastuzumab-based therapy, and correlated the results with progression-free survival and overall survival using Kaplan–Meier and decision tree analyses that also included HER2 total (H2T) and p95 expression levels. Within the sub-population of patients that over-expressed HER2, high levels of HER3 and/or p95 protein expression were significantly associated with poor clinical outcomes on trastuzumab-based therapy. Based on quantitative H3T, p95, and H2T measurements, multiple subtypes of HER2-positive breast cancer were identified that differ in their outcome following trastuzumab therapy. These data suggest that HER3 and p95 are informative biomarkers of clinical outcomes on trastuzumab therapy, and that multiple subtypes of HER2-positive breast cancer may be defined by quantitative measurements of H3T, p95, and H2T.
Collapse
Affiliation(s)
- Allan Lipton
- Breast Oncology Research, Division of Hematology/Oncology, Department of Medicine, Penn State Hershey Medical Center, 500 University Drive, Hershey, PA 17033, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Goltsov A, Langdon SP, Goltsov G, Harrison DJ, Bown J. Customizing the therapeutic response of signaling networks to promote antitumor responses by drug combinations. Front Oncol 2014; 4:13. [PMID: 24551596 PMCID: PMC3914444 DOI: 10.3389/fonc.2014.00013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 01/20/2014] [Indexed: 01/26/2023] Open
Abstract
Drug resistance, de novo and acquired, pervades cellular signaling networks (SNs) from one signaling motif to another as a result of cancer progression and/or drug intervention. This resistance is one of the key determinants of efficacy in targeted anti-cancer drug therapy. Although poorly understood, drug resistance is already being addressed in combination therapy by selecting drug targets where SN sensitivity increases due to combination components or as a result of de novo or acquired mutations. Additionally, successive drug combinations have shown low resistance potential. To promote a rational, systematic development of combination therapies, it is necessary to establish the underlying mechanisms that drive the advantages of combination therapies, and design methods to determine drug targets for combination regimens. Based on a joint systems analysis of cellular SN response and its sensitivity to drug action and oncogenic mutations, we describe an in silico method to analyze the targets of drug combinations. Our method explores mechanisms of sensitizing the SN through a combination of two drugs targeting vertical signaling pathways. We propose a paradigm of SN response customization by one drug to both maximize the effect of another drug in combination and promote a robust therapeutic response against oncogenic mutations. The method was applied to customize the response of the ErbB/PI3K/PTEN/AKT pathway by combination of drugs targeting HER2 receptors and proteins in the down-stream pathway. The results of a computational experiment showed that the modification of the SN response from hyperbolic to smooth sigmoid response by manipulation of two drugs in combination leads to greater robustness in therapeutic response against oncogenic mutations determining cancer heterogeneity. The application of this method in drug combination co-development suggests a combined evaluation of inhibition effects together with the capability of drug combinations to suppress resistance mechanisms before they become clinically manifest.
Collapse
Affiliation(s)
- Alexey Goltsov
- Centre for Research in Informatics and Systems Pathology (CRISP), University of Abertay Dundee , Dundee , UK
| | - Simon P Langdon
- Division of Pathology, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh , Edinburgh , UK
| | | | | | - James Bown
- Centre for Research in Informatics and Systems Pathology (CRISP), University of Abertay Dundee , Dundee , UK
| |
Collapse
|
21
|
Phillips GDL, Fields CT, Li G, Dowbenko D, Schaefer G, Miller K, Andre F, Burris HA, Albain KS, Harbeck N, Dieras V, Crivellari D, Fang L, Guardino E, Olsen SR, Crocker LM, Sliwkowski MX. Dual Targeting of HER2-Positive Cancer with Trastuzumab Emtansine and Pertuzumab: Critical Role for Neuregulin Blockade in Antitumor Response to Combination Therapy. Clin Cancer Res 2013; 20:456-68. [DOI: 10.1158/1078-0432.ccr-13-0358] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
O'Sullivan CC, Swain SM. Pertuzumab : evolving therapeutic strategies in the management of HER2-overexpressing breast cancer. Expert Opin Biol Ther 2013; 13:779-90. [PMID: 23530718 DOI: 10.1517/14712598.2013.783007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION HER2 overexpression or amplification is present in approximately one-fifth of breast cancers and historically was associated with aggressive disease and poorer prognosis. The introduction of the humanized monoclonal antibody trastuzumab dramatically improved disease-free survival (DFS) and overall survival (OS) in this subgroup. As the majority of patients with metastatic disease ultimately develop resistance to trastuzumab, a need exists for more effective targeted therapies. Pertuzumab is an anti-HER2/neu-targeted therapy in the late stages of clinical development. The combination of pertuzumab, trastuzumab and docetaxel has been found to have an OS benefit in patients with HER2 positive metastatic breast cancer (MBC) when used in the first-line setting. This reflects a new standard of care, and pertuzumab was recently approved for this indication by the Food and Drug Administration (FDA). The efficacy of pertuzumab and trastuzumab in conjunction with chemotherapy is currently being evaluated in the adjuvant setting. AREAS COVERED This article provides an overview of preclinical investigations in addition to reviewing pertinent Phase I, Phase II and Phase III clinical trials. EXPERT OPINION Pertuzumab, in combination with the humanized monoclonal antibody trastuzumab, and docetaxel is a standard of care for patients with previously untreated metastatic breast cancer based on the CLEOPATRA study showing a survival benefit. There is no increase in cardiac toxicity with the combined HER2-targeted therapy. Future issues will address appropriate sequencing and combination with other anti-HER2-targeted therapies and/or chemotherapy.
Collapse
Affiliation(s)
- Ciara C O'Sullivan
- Medical Oncology Branch, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
23
|
Leone Roberti Maggiore U, Bellati F, Ruscito I, Gasparri ML, Alessandri F, Venturini PL, Ferrero S. Monoclonal antibodies therapies for ovarian cancer. Expert Opin Biol Ther 2013; 13:739-64. [PMID: 23373587 DOI: 10.1517/14712598.2013.767328] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Despite aggressive debulking surgery, intraperitoneal therapies and the use of new drugs for chemotherapy, patients with ovarian cancer (OC) still have poor prognosis and, therefore, new strategies for its management are needed. Molecular-targeted agents can be considered a new option in drug research. Several antigens related to OC have been isolated and they could be potential target of monoclonal antibodies (mAbs); therefore, different mAbs have been developed and are emerging as new potential OC treatments. AREAS COVERED This article aims to review the literature on the use of mAbs in the treatment of OC. The purposes of this manuscript are to offer a brief explanation of the mechanisms of action of mAbs and to help readers in understanding the current role of mAbs in the treatment of OC. EXPERT OPINION A deeper knowledge of the molecular biology of OC has brought new developments in targeted therapies. Among these therapies, bevacizumab demonstrated the higher clinical efficacy. Further larger trials are needed to better define the role of the other mAbs in OC treatment. There is a strong need to identify and validate robust biomarkers for a more focused patient selection and for tailoring therapies, optimizing dose and assessing response.
Collapse
Affiliation(s)
- Umberto Leone Roberti Maggiore
- University of Genoa, San Martino Hospital and National Institute for Cancer Research, Department of Obstetrics and Gynecology, Largo R. Benzi 1, 16132 Genoa, Italy
| | | | | | | | | | | | | |
Collapse
|
24
|
Loewe RP. Combinational usage of next generation sequencing and qPCR for the analysis of tumor samples. Methods 2012. [PMID: 23178393 DOI: 10.1016/j.ymeth.2012.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The combination of multiple techniques especially those adding complementary information have proven to be beneficial in terms of data consistency. The employment of quantitative PCR (qPCR) prior to next generation sequencing (NGS) methods such as RNA-Seq and mutational analysis presented here does not only enhance data in terms of CNV integration and sample choice, but also allows a faster and more efficient workflow. Correct analysis of libraries prior to sequencing has proven to be a vital step for specific assumption and to some extent for a more parallel testing. By illustrating the combination of qPCR and NGS in oncological examples, the potential of this approach is presented.
Collapse
Affiliation(s)
- Robert P Loewe
- GeneWake GmbH, Floriansbogen 2-4, 82061 Neuried bei Muenchen, Germany.
| |
Collapse
|
25
|
Choi BK, Fan X, Deng H, Zhang N, An Z. ERBB3 (HER3) is a key sensor in the regulation of ERBB-mediated signaling in both low and high ERBB2 (HER2) expressing cancer cells. Cancer Med 2012; 1:28-38. [PMID: 23342251 PMCID: PMC3544427 DOI: 10.1002/cam4.10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/18/2012] [Accepted: 05/21/2012] [Indexed: 12/19/2022] Open
Abstract
Aberrant expression and activation of EGFR and ERBB2 (HER2) have been successfully targeted for cancer therapeutics. Recent evidence from both basic and clinical studies suggests that ERBB3 (HER3) serves as a key activator of downstream signaling through dimerization with other ERBB proteins and plays a critical role in the widespread clinical resistance to EGFR and HER2 targeting cancer therapies. As a result, HER3 is actively pursued as an antibody therapeutic target for cancer. Ligand binding is thought to be a prerequisite for dimerization of HER3 with other ERBB proteins, which results in phosphorylation of its c-terminal tyrosine residues and activation of downstream AKT and MAPK signaling pathways. In this study, we report that an anti-HER2 monoclonal antibody (HER2Mab), which blocks HER2 dimerization with HER3, induces HER3 dimerization with EGFR in both low and high HER2 expressing cancer cells. Treatment of the low HER2 expressing MCF7 cancer cells with HER2Mab promoted cell proliferation and migration in the absence of HER3 ligand stimulation. Follow-up studies revealed that HER2Mab-induced HER3 signaling via EGFR/HER3 dimerization and activation of downstream AKT signaling pathways. These results suggest that equilibrium of dimerization among the ERBB proteins can be perturbed by HER2Mab and HER3 plays a key role in sensing the perturbation.
Collapse
Affiliation(s)
- Byung-Kwon Choi
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston Houston, Texas, USA
| | | | | | | | | |
Collapse
|
26
|
Abstract
Ovarian cancer is the second most common gynaecological malignancy and the leading cause of death from gynaecological cancer. Although in some cases treatment is initially effective, there is a considerable risk of disease recurrence and resistance to therapy. Therapies targeting molecular alterations in tumours offer the promise of significantly improved treatment. So far, the most promising targeted agents are angiogenesis inhibitors and PARP inhibitors. Here, we review the various targeted therapeutic approaches under clinical investigation in phase I and II trials of ovarian cancer and the challenges facing their future success in the clinic.
Collapse
Affiliation(s)
- Susana Banerjee
- The Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom
| | | |
Collapse
|
27
|
Goltsov A, Faratian D, Langdon SP, Mullen P, Harrison DJ, Bown J. Features of the reversible sensitivity-resistance transition in PI3K/PTEN/AKT signalling network after HER2 inhibition. Cell Signal 2011; 24:493-504. [PMID: 21996585 DOI: 10.1016/j.cellsig.2011.09.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 09/15/2011] [Accepted: 09/27/2011] [Indexed: 12/19/2022]
Abstract
Systems biology approaches that combine experimental data and theoretical modelling to understand cellular signalling network dynamics offer a useful platform to investigate the mechanisms of resistance to drug interventions and to identify combination drug treatments. Extending our work on modelling the PI3K/PTEN/AKT signalling network (SN), we analyse the sensitivity of the SN output signal, phospho-AKT, to inhibition of HER2 receptor. We model typical aberrations in this SN identified in cancer development and drug resistance: loss of PTEN activity, PI3K and AKT mutations, HER2 overexpression, and overproduction of GSK3β and CK2 kinases controlling PTEN phosphorylation. We show that HER2 inhibition by the monoclonal antibody pertuzumab increases SN sensitivity, both to external signals and to changes in kinetic parameters of the proteins and their expression levels induced by mutations in the SN. This increase in sensitivity arises from the transition of SN functioning from saturation to non-saturation mode in response to HER2 inhibition. PTEN loss or PIK3CA mutation causes resistance to anti-HER2 inhibitor and leads to the restoration of saturation mode in SN functioning with a consequent decrease in SN sensitivity. We suggest that a drug-induced increase in SN sensitivity to internal perturbations, and specifically mutations, causes SN fragility. In particular, the SN is vulnerable to mutations that compensate for drug action and this may result in a sensitivity-to-resistance transition. The combination of HER2 and PI3K inhibition does not sensitise the SN to internal perturbations (mutations) in the PI3K/PTEN/AKT pathway: this combination treatment provides both synergetic inhibition and may prevent the SN from acquired mutations causing drug resistance. Through combination inhibition treatments, we studied the impact of upstream and downstream interventions to suppress resistance to the HER2 inhibitor in the SN with PTEN loss. Comparison of experimental results of PI3K inhibition in the PTEN upstream pathway with PDK1 inhibition in the PTEN downstream pathway shows that upstream inhibition abrogates resistance to pertuzumab more effectively than downstream inhibition. This difference in inhibition effect arises from the compensatory mechanism of an activation loop induced in the downstream pathway by PTEN loss. We highlight that drug target identification for combination anti-cancer therapy needs to account for the mutation effects on the upstream and downstream pathways.
Collapse
Affiliation(s)
- Alexey Goltsov
- Centre for Research in Informatics and Systems Pathology (CRISP), University of Abertay Dundee, Dundee, DD1 1HG, United Kingdom.
| | - Dana Faratian
- Edinburgh Breakthrough Research Unit and Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Simon P Langdon
- Edinburgh Breakthrough Research Unit and Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - Peter Mullen
- Edinburgh Breakthrough Research Unit and Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - David J Harrison
- Edinburgh Breakthrough Research Unit and Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - James Bown
- Centre for Research in Informatics and Systems Pathology (CRISP), University of Abertay Dundee, Dundee, DD1 1HG, United Kingdom
| |
Collapse
|
28
|
Faratian D, Zweemer AJM, Nagumo Y, Sims AH, Muir M, Dodds M, Mullen P, Um I, Kay C, Hasmann M, Harrison DJ, Langdon SP. Trastuzumab and pertuzumab produce changes in morphology and estrogen receptor signaling in ovarian cancer xenografts revealing new treatment strategies. Clin Cancer Res 2011; 17:4451-61. [PMID: 21571868 DOI: 10.1158/1078-0432.ccr-10-2461] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of this study was to investigate the antitumor effects of HER2-directed combination therapy in ovarian cancer xenograft models to evaluate their potential. The combinations of trastuzumab and pertuzumab, and trastuzumab and aromatase inhibitor therapy were investigated. EXPERIMENTAL DESIGN The effects of trastuzumab, pertuzumab, and letrozole on growth response, apoptosis, morphology, and gene and protein expression were evaluated in the SKOV3 ovarian cancer cell line xenograft and a panel of five human ovarian xenografts derived directly from clinical specimens. RESULTS The combination of HER2-directed antibodies showed enhanced antitumor activity compared with single antibody therapy in the SKOV3 xenograft model. Apoptosis, morphology, and estrogen-regulated gene expression were modulated by these antibodies in both spatial and temporal manners. A panel of ovarian cancer xenografts showed differential growth responses to the combination of trastuzumab and pertuzumab. High HER2 expression and increasing HER3 protein expression on treatment were associated with growth response. In trastuzumab-treated SKOV3 tumors, there was a change in tumor morphology, with a reduction in frequency of estrogen receptor alpha (ERα)-negative clear cell areas. Trastuzumab, but not pertuzumab, increased expression of ERα in SKOV3 xenografts when analyzed by quantitative immunofluorescence. ERα and downstream signaling targets were modulated by trastuzumab alone and in combination. Trastuzumab enhanced the responsiveness of SKOV3 xenografts to letrozole when given in combination. CONCLUSIONS These data suggest that trastuzumab in combination with pertuzumab could be an effective approach in high HER2-expressing ovarian cancers and could also enhance sensitivity to endocrine therapy in ERα-positive ovarian cancer.
Collapse
Affiliation(s)
- Dana Faratian
- Division of Pathology and Edinburgh Breakthrough Research Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
IMPORTANCE OF THE FIELD Heterodimerization of human EGF receptor (HER) 2 and HER3, a co-receptor of HER2, plays an important and dominant role in the functionality and transformation of HER-mediated pathways. Understanding the role of HER3 in oncogenesis as well as its place as a target for anticancer therapy is an ongoing area of research. Determination of biomarkers for clinical benefit from agents targeting HER3 is an essential component of translating basic science into real-world effective anticancer therapies, with the aim of ensuring the patients most likely to benefit from such treatments can be identified. AREAS COVERED IN THIS REVIEW This review focuses on the targeting of HER2 and HER3 by monoclonal antibodies and the potential for HER3 mRNA levels to predict treatment outcome in ovarian cancer. WHAT THE READER WILL GAIN An understanding of the value of biomarkers for clinical benefit to anticancer therapy and the current status of HER3 mRNA as a biomarker for clinical benefit of the HER2-HER3 dimerization inhibitor pertuzumab. TAKE HOME MESSAGE HER3 mRNA levels may be a biomarker for active ligand-induced HER2-HER3 signaling, with low HER3 mRNA levels correlated with clinical benefit from the HER2-HER3 dimerization inhibitor pertuzumab.
Collapse
Affiliation(s)
- Lukas C Amler
- Genentech, Inc., Oncology Biomarker Development, 1 DNA Way, South San Francisco, 94080-4990, USA.
| |
Collapse
|
30
|
Langdon SP, Faratian D, Nagumo Y, Mullen P, Harrison DJ. Pertuzumab for the treatment of ovarian cancer. Expert Opin Biol Ther 2010; 10:1113-20. [DOI: 10.1517/14712598.2010.487062] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
O'Brien C, Wallin JJ, Sampath D, GuhaThakurta D, Savage H, Punnoose EA, Guan J, Berry L, Prior WW, Amler LC, Belvin M, Friedman LS, Lackner MR. Predictive biomarkers of sensitivity to the phosphatidylinositol 3' kinase inhibitor GDC-0941 in breast cancer preclinical models. Clin Cancer Res 2010; 16:3670-83. [PMID: 20453058 DOI: 10.1158/1078-0432.ccr-09-2828] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The class I phosphatidylinositol 3' kinase (PI3K) plays a major role in proliferation and survival in a wide variety of human cancers. A key factor in successful development of drugs targeting this pathway is likely to be the identification of responsive patient populations with predictive diagnostic biomarkers. This study sought to identify candidate biomarkers of response to the selective PI3K inhibitor GDC-0941. EXPERIMENTAL DESIGN We used a large panel of breast cancer cell lines and in vivo xenograft models to identify candidate predictive biomarkers for a selective inhibitor of class I PI3K that is currently in clinical development. The approach involved pharmacogenomic profiling as well as analysis of gene expression data sets from cells profiled at baseline or after GDC-0941 treatment. RESULTS We found that models harboring mutations in PIK3CA, amplification of human epidermal growth factor receptor 2, or dual alterations in two pathway components were exquisitely sensitive to the antitumor effects of GDC-0941. We found that several models that do not harbor these alterations also showed sensitivity, suggesting a need for additional diagnostic markers. Gene expression studies identified a collection of genes whose expression was associated with in vitro sensitivity to GDC-0941, and expression of a subset of these genes was found to be intimately linked to signaling through the pathway. CONCLUSION Pathway focused biomarkers and the gene expression signature described in this study may have utility in the identification of patients likely to benefit from therapy with a selective PI3K inhibitor.
Collapse
Affiliation(s)
- Carol O'Brien
- Department of Development Oncology Diagnostics, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kristjansdottir K, Dizon D. HER-dimerization inhibitors: evaluating pertuzumab in women's cancers. Expert Opin Biol Ther 2010; 10:243-50. [PMID: 20001562 DOI: 10.1517/14712590903514090] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
IMPORTANCE OF THE FIELD Pertuzumab is a human EGF receptor (HER)-dimerization inhibitor that represent a novel class of agents aimed at blocking HER2 from pairing with other receptors of the HER family. In this review, we discuss the background and scientific rationale, related to pertuzumab as it has undergone development for women's cancers. AREAS COVERED IN THIS REVIEW Pre-clinical and clinical trials, published or presented at national meetings from 1995 to the present, are included in this review. WHAT THE READER WILL GAIN A Phase II trial in HER2-positive metastatic breast cancer showed promising activity of pertuzumab when it was combined with trastuzumab and a randomized Phase III trial is now underway. Pertuzumab, when evaluated in recurrent ovarian cancer showed limited activity when combined with chemotherapy in platinum-sensitive and platinum-resistant disease. However, a recent subset analysis suggests that HER3 mRNA levels may predict response to pertuzumab in ovarian cancer. TAKE HOME MESSAGE HER-dimerization inhibitors represent a novel mechanism of inhibition of HER pathways. Pertuzumab may play a role in the management of HER2-positive breast cancer. The potential benefit of pertuzumab in ovarian cancer is unclear, but low HER3 levels may suggest a sub-group of patients that can benefit from pertuzumab.
Collapse
Affiliation(s)
- Katrin Kristjansdottir
- Warren Alpert Medical School of Brown University, Women and Infants Hospital of Rhode Island - Program in Women's Oncology, 101 Dudley Street, Providence, Rhode Island, USA
| | | |
Collapse
|