1
|
Zhu M, Yu M, Meng Y, Yang J, Wang X, Li L, Liang Y, Kong F. HER3 receptor and its role in the therapeutic management of metastatic breast cancer. J Transl Med 2024; 22:665. [PMID: 39020378 PMCID: PMC11253420 DOI: 10.1186/s12967-024-05445-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Metastatic breast cancer (mBC) poses a significant threat to women's health and is a major cause of malignant neoplasms in women. Human epidermal growth factor receptor (HER)3, an integral member of the ErbB/HER receptor tyrosine kinase family, is a crucial activator of the phosphoinositide-3 kinase/protein kinase B signaling pathway. HER3 overexpression significantly contributes to the development of resistance to drugs targeting other HER receptors, such as HER2 and epidermal growth factor receptors, and plays a crucial role in the onset and progression of mBC. Recently, numerous HER3-targeted therapeutic agents, such as monoclonal antibodies (mAbs), bispecific antibodies (bAbs), and antibody-drug conjugates (ADCs), have emerged. However, the efficacy of HER3-targeted mAbs and bAbs is limited when used individually, and their combination may result in toxic adverse effects. On the other hand, ADCs are cytotoxic to cancer cells and can bind to target cells through antibodies, which highlights their use in targeted HER3 therapy for mBC. This review provides an overview of recent advancements in HER3 research, historical initiatives, and innovative approaches in targeted HER3 therapy for metastatic breast cancer. Evaluating the advantages and disadvantages of current methods may yield valuable insights and lessons.
Collapse
Affiliation(s)
- Meiying Zhu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin Cancer Institute of Traditional Chinese Medicine, Tianjin, China
| | - Minghui Yu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuan Meng
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jie Yang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xuerui Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Longhui Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yangyueying Liang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fanming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Anshanxi Road, Nankai District, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
2
|
Gandullo-Sánchez L, Ocaña A, Pandiella A. HER3 in cancer: from the bench to the bedside. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:310. [PMID: 36271429 PMCID: PMC9585794 DOI: 10.1186/s13046-022-02515-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
Abstract
The HER3 protein, that belongs to the ErbB/HER receptor tyrosine kinase (RTK) family, is expressed in several types of tumors. That fact, together with the role of HER3 in promoting cell proliferation, implicate that targeting HER3 may have therapeutic relevance. Furthermore, expression and activation of HER3 has been linked to resistance to drugs that target other HER receptors such as agents that act on EGFR or HER2. In addition, HER3 has been associated to resistance to some chemotherapeutic drugs. Because of those circumstances, efforts to develop and test agents targeting HER3 have been carried out. Two types of agents targeting HER3 have been developed. The most abundant are antibodies or engineered antibody derivatives that specifically recognize the extracellular region of HER3. In addition, the use of aptamers specifically interacting with HER3, vaccines or HER3-targeting siRNAs have also been developed. Here we discuss the state of the art of the preclinical and clinical development of drugs aimed at targeting HER3 with therapeutic purposes.
Collapse
Affiliation(s)
- Lucía Gandullo-Sánchez
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Alberto Ocaña
- grid.411068.a0000 0001 0671 5785Hospital Clínico San Carlos and CIBERONC, 28040 Madrid, Spain
| | - Atanasio Pandiella
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
3
|
Drago JZ, Ferraro E, Abuhadra N, Modi S. Beyond HER2: Targeting the ErbB receptor family in breast cancer. Cancer Treat Rev 2022; 109:102436. [PMID: 35870237 PMCID: PMC10478787 DOI: 10.1016/j.ctrv.2022.102436] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/28/2022]
Abstract
Targeting the HER2 oncogene represents one of the greatest advances in the treatment of breast cancer. HER2 is one member of the ERBB-receptor family, which includes EGFR (HER1), HER3 and HER4. In the presence or absence of underling genomic aberrations such as mutations or amplification events, intricate interactions between these proteins on the cell membrane lead to downstream signaling that encourages cancer growth and proliferation. In this Review, we contextualize efforts to pharmacologically target the ErbB receptor family beyond HER2, with a focus on EGFR and HER3. Preclinical and clinical efforts are synthesized. We discuss successes and failures of this approach to date, summarize lessons learned, and propose a way forward that invokes new therapeutic modalities such as antibody drug conjugates (ADCs), combination strategies, and patient selection through rational biomarkers.
Collapse
Affiliation(s)
- Joshua Z Drago
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA.
| | - Emanuela Ferraro
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nour Abuhadra
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| |
Collapse
|
4
|
Hayes DA, Kunde DA, Taylor RL, Pyecroft SB, Sohal SS, Snow ET. ERBB3: A potential serum biomarker for early detection and therapeutic target for devil facial tumour 1 (DFT1). PLoS One 2017; 12:e0177919. [PMID: 28591206 PMCID: PMC5462353 DOI: 10.1371/journal.pone.0177919] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Devil Facial Tumour 1 (DFT1) is one of two transmissible neoplasms of Tasmanian devils (Sarcophilus harrisii) predominantly affecting their facial regions. DFT1's cellular origin is that of Schwann cell lineage where lesions are evident macroscopically late in the disease. Conversely, the pre-clinical timeframe from cellular transmission to appearance of DFT1 remains uncertain demonstrating the importance of an effective pre-clinical biomarker. We show that ERBB3, a marker expressed normally by the developing neural crest and Schwann cells, is immunohistohemically expressed by DFT1, therefore the potential of ERBB3 as a biomarker was explored. Under the hypothesis that serum ERBB3 levels may increase as DFT1 invades local and distant tissues our pilot study determined serum ERBB3 levels in normal Tasmanian devils and Tasmanian devils with DFT1. Compared to the baseline serum ERBB3 levels in unaffected Tasmanian devils, Tasmanian devils with DFT1 showed significant elevation of serum ERBB3 levels. Interestingly Tasmanian devils with cutaneous lymphoma (CL) also showed elevation of serum ERBB3 levels when compared to the baseline serum levels of Tasmanian devils without DFT1. Thus, elevated serum ERBB3 levels in otherwise healthy looking devils could predict possible DFT1 or CL in captive or wild devil populations and would have implications on the management, welfare and survival of Tasmanian devils. ERBB3 is also a therapeutic target and therefore the potential exists to consider modes of administration that may eradicate DFT1 from the wild.
Collapse
Affiliation(s)
- Dane A. Hayes
- Department of Primary Industries, Parks Water and Environment, Animal Health Laboratory, Launceston, Tasmania, Australia
- Save the Tasmanian Devil Program, University of Tasmania, Hobart, Tasmania, Australia
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Dale A. Kunde
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Robyn L. Taylor
- Save the Tasmanian Devil Program, University of Tasmania, Hobart, Tasmania, Australia
- Department of Primary Industries, Parks Water and Environment, Resource Management and Conservation, Hobart, Tasmania, Australia
| | - Stephen B. Pyecroft
- School of Animal & Veterinary Sciences, Faculty of Science, University of Adelaide, Roseworthy Campus, Roseworthy, South Australia
| | - Sukhwinder Singh Sohal
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| | - Elizabeth T. Snow
- School of Health Sciences, Faculty of Health, University of Tasmania, Launceston, Tasmania, Australia
| |
Collapse
|
5
|
Lindsey S, Langhans SA. Epidermal growth factor signaling in transformed cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 314:1-41. [PMID: 25619714 DOI: 10.1016/bs.ircmb.2014.10.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Members of the epidermal growth factor receptor (EGFR/ErbB) family play a critical role in normal cell growth and development. However, many ErbB family members, especially EGFR, are aberrantly expressed or deregulated in tumors and are thought to play crucial roles in cancer development and metastatic progression. In this chapter, we provide an overview of key mechanisms contributing to aberrant EGFR/ErbB signaling in transformed cells, which results in many phenotypic changes associated with the earliest stages of tumor formation, including several hallmarks of epithelial-mesenchymal transition (EMT). These changes often occur through interaction with other major signaling pathways important to tumor progression, causing a multitude of transcriptional changes that ultimately impact cell morphology, proliferation, and adhesion, all of which are crucial for tumor progression. The resulting mesh of signaling networks will need to be taken into account as new regimens are designed for targeting EGFR for therapeutic intervention. As new insights are gained into the molecular mechanisms of cross talk between EGFR signaling and other signaling pathways, including their roles in therapeutic resistance to anti-EGFR therapies, a continual reassessment of clinical therapeutic regimes and strategies will be required. Understanding the consequences and complexity of EGF signaling and how it relates to tumor progression is critical for the development of clinical compounds and establishing clinical protocols for the treatment of cancer.
Collapse
Affiliation(s)
- Stephan Lindsey
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Sigrid A Langhans
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| |
Collapse
|
6
|
Kol A, Terwisscha van Scheltinga AG, Timmer-Bosscha H, Lamberts LE, Bensch F, de Vries EG, Schröder CP. HER3, serious partner in crime. Pharmacol Ther 2014; 143:1-11. [DOI: 10.1016/j.pharmthera.2014.01.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 09/27/2013] [Indexed: 02/07/2023]
|
7
|
Canbaz MÇ, Şimşek ÇS, Sezgintürk MK. Electrochemical biosensor based on self-assembled monolayers modified with gold nanoparticles for detection of HER-3. Anal Chim Acta 2014; 814:31-8. [DOI: 10.1016/j.aca.2014.01.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/18/2013] [Accepted: 01/16/2014] [Indexed: 11/25/2022]
|
8
|
Blackburn E, Zona S, Murphy ML, Brown IR, Chan SKW, Gullick WJ. A monoclonal antibody to the human HER3 receptor inhibits Neuregulin 1-beta binding and co-operates with Herceptin in inhibiting the growth of breast cancer derived cell lines. Breast Cancer Res Treat 2011; 134:53-9. [PMID: 22169894 DOI: 10.1007/s10549-011-1908-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/30/2011] [Indexed: 10/14/2022]
Abstract
The HER3 protein contributes to malignant transformation in breast and other cancer types as a consequence of elevated levels of expression, particularly in the presence of the HER2 protein. We show here that an antibody, called SGP1, to the extracellular domain of the HER3 receptor can inhibit completely Neuregulin stimulated growth of cultured breast cancer cells. Herceptin is a humanised monoclonal antibody to the HER2 protein which has an established role in the treatment of some patients with breast cancer. We demonstrate that Herceptin and SGP1 can bind simultaneously to breast cancer cells expressing both the HER2 and HER3 proteins. In the presence of moderate levels of Herceptin, addition of the SGP1 monoclonal antibody gave a dose-dependent inhibition of the growth of cells expressing both the high levels and moderate levels of HER2. The combination of Herceptin with SGP1 is effective in inhibiting breast cancer cell growth in cases where both HER2 and HER3 are expressed.
Collapse
Affiliation(s)
- Edith Blackburn
- Cancer Biology Laboratory, Microscopy and Imaging Facility, School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | | | | | | | | | | |
Collapse
|
9
|
Rajkumar T, Stamp GW, Hughes CM, Gullick WJ. c-erbB3 protein expression in ovarian cancer. Mol Pathol 2010; 49:M199-202. [PMID: 16696074 PMCID: PMC408058 DOI: 10.1136/mp.49.4.m199] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Aims-To study the prevalence of overexpression of c-erbB3 growth factor receptor in ovarian carcinomas and to analyse its relation to histological subtype, stage and grade of the tumours.Methods-Ninety eight ovarian carcinomas were evaluated immunohistochemically using the RTJ1 monoclonal antibody raised against a synthetic peptide, the sequence of which was derived from the cytoplasmic domain of the c-erbB3 protein.Results-Of the tumours, 16% (16/98) overexpressed c-erbB3 protein relative to normal ovarian epithelium, whereas 22% (22/98) were completely negative. There was a statistically significant association between overexpression and well differentiated grade.Conclusions-These findings suggest that c-erbB3 protein overexpression occurs in a significant proportion of ovarian cancers and is correlated with differentiation. Overexpression may merit further investigation as a potential prognostic indicator and as a target for new treatment.
Collapse
Affiliation(s)
- T Rajkumar
- ICRF Oncology Unit, Hammersmith Hospital, London W12 ONN
| | | | | | | |
Collapse
|
10
|
Bartlett JM, Munro A, Cameron DA, Thomas J, Prescott R, Twelves CJ. Type 1 Receptor Tyrosine Kinase Profiles Identify Patients With Enhanced Benefit From Anthracyclines in the BR9601 Adjuvant Breast Cancer Chemotherapy Trial. J Clin Oncol 2008; 26:5027-35. [PMID: 18768436 DOI: 10.1200/jco.2007.14.6597] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Patients with early breast cancer who receive anthracycline-containing chemotherapy experience improved relapse-free (RFS) and overall survival (OS) compared with those who receive non–anthracycline-containing chemotherapy. Such benefit, however, may be restricted to women whose tumors have specific molecular characteristics. We tested the hypothesis that HER2, epidermal growth factor receptor (EGFr)/HER1, HER3, Ki67, and topoisomerase IIα expression are predictive of outcome after anthracycline-based chemotherapy. Methods Tissue microarrays from 322 of 374 women in the BR9601 trial, which compared cyclophosphamide, methotrexate, and fluorouracil (CMF) with epirubicin followed by CMF (epi-CMF), were analyzed for HER1, 2, 3, 4; Ki67; and topoisomerase IIα protein expression and for HER2/topoisomerase IIα gene amplification. Their relationships to RFS and OS were investigated, and multiple regression analysis was used to identify interactions. Results A significant interaction was seen between tumors with normal HER1, HER2 fluorescent in situ hybridization (FISH), or HER3 levels and the enhanced benefit from epi-CMF versus CMF for RFS (hazard ratio [HR], 0.36; HR for overexpressed HER1 or HER2 FISH or HER3, 0.92; P = .035) and for OS (HR, 0.30; HR for overexpressed HER1 or HER2 FISH or HER3), 0.98; P = .023). Neither Ki67 nor TIIα expressions or gene alterations showed clear predictive value for benefit from the addition of the anthracycline. Conclusion Patients with HER2 amplified and those with HER1, HER2 FISH, or HER3-positive tumors did not benefit from the addition of epirubicin to CMF. Conversely, patients with HER2 nonamplified and HER1 through HER3–negative tumors showed significantly increased RFS and OS rates when treated with epi-CMF compared with CMF.
Collapse
Affiliation(s)
- John M.S. Bartlett
- From the Endocrine Cancer Research Group, Cancer Research United Kingdom Building, Western General Hospital; Western General Hospital; Medical Statistics Unit, Public Health Services, Medical School, Edinburgh; and the Cancer Research United Kingdom Clinical Centre, St James’ University Hospital, Leeds, United Kingdom
| | - Alison Munro
- From the Endocrine Cancer Research Group, Cancer Research United Kingdom Building, Western General Hospital; Western General Hospital; Medical Statistics Unit, Public Health Services, Medical School, Edinburgh; and the Cancer Research United Kingdom Clinical Centre, St James’ University Hospital, Leeds, United Kingdom
| | - David A. Cameron
- From the Endocrine Cancer Research Group, Cancer Research United Kingdom Building, Western General Hospital; Western General Hospital; Medical Statistics Unit, Public Health Services, Medical School, Edinburgh; and the Cancer Research United Kingdom Clinical Centre, St James’ University Hospital, Leeds, United Kingdom
| | - Jeremy Thomas
- From the Endocrine Cancer Research Group, Cancer Research United Kingdom Building, Western General Hospital; Western General Hospital; Medical Statistics Unit, Public Health Services, Medical School, Edinburgh; and the Cancer Research United Kingdom Clinical Centre, St James’ University Hospital, Leeds, United Kingdom
| | - Robin Prescott
- From the Endocrine Cancer Research Group, Cancer Research United Kingdom Building, Western General Hospital; Western General Hospital; Medical Statistics Unit, Public Health Services, Medical School, Edinburgh; and the Cancer Research United Kingdom Clinical Centre, St James’ University Hospital, Leeds, United Kingdom
| | - Chris J. Twelves
- From the Endocrine Cancer Research Group, Cancer Research United Kingdom Building, Western General Hospital; Western General Hospital; Medical Statistics Unit, Public Health Services, Medical School, Edinburgh; and the Cancer Research United Kingdom Clinical Centre, St James’ University Hospital, Leeds, United Kingdom
| |
Collapse
|
11
|
Murphy ML, Chan SKW, Gullick WJ. Why do most c-erbB-2/HER-2-positive breast cancer patients fail to respond to Herceptin? Breast Cancer Res 2008. [PMCID: PMC3300781 DOI: 10.1186/bcr1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
12
|
Koumakpayi IH, Diallo JS, Le Page C, Lessard L, Filali-Mouhim A, Bégin LR, Mes-Masson AM, Saad F. Low nuclear ErbB3 predicts biochemical recurrence in patients with prostate cancer. BJU Int 2007; 100:303-9. [PMID: 17532856 DOI: 10.1111/j.1464-410x.2007.06992.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To further evaluate the association between the cytoplasmic or nuclear localization of ErbB3 with biochemical recurrence (BCR) in patients with prostate cancer and positive surgical margins, as there is a greater risk of BCR for such patients after radical prostatectomy (RP). PATIENTS AND METHODS We recently noted that ErbB3, which is normally associated with the plasma membrane, can translocate to the nucleus, an event which appears to be associated with disease progression. We evaluated ErbB3 expression and localization using immunohistochemistry on tissue samples from 55 patients with positive surgical margins after RP; 30 of these 55 (55%) had BCR after 3 years of follow-up. The relationship between ErbB3 nuclear localization and BCR (prostate-specific antigen, PSA, >0.3 ng/mL) after RP was analysed by Kaplan-Meier survival analysis and Cox regression models. RESULTS The BCR-free survival probability at 3 years was 0.65 and 0.35 for positive and negative nuclear ErbB3, respectively (Kaplan-Meier, P = 0.029). Patients negative for nuclear ErbB3 had a 2.47-fold increase in BCR frequency in a univariate Cox model (P = 0.008) and it remained an independent prognostic marker when combined with clinical prognostic variables in a multivariate model (P = 0.023). CONCLUSION Low nuclear localization of ErbB3 is a predictor of BCR in patients with prostate cancer and positive surgical margins after RP.
Collapse
Affiliation(s)
- Ismaël H Koumakpayi
- Centre de recherche du Centre Hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Bouyain S, Leahy DJ. Structure-based mutagenesis of the substrate-recognition domain of Nrdp1/FLRF identifies the binding site for the receptor tyrosine kinase ErbB3. Protein Sci 2007; 16:654-61. [PMID: 17384230 PMCID: PMC2203331 DOI: 10.1110/ps.062700307] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The E3 ubiquitin ligase neuregulin receptor degrading protein 1 (Nrdp1) mediates the ligand-independent degradation of the epidermal growth factor receptor family member ErbB3/HER3. By regulating cellular levels of ErbB3, Nrdp1 influences ErbB3-mediated signaling, which is essential for normal vertebrate development. Nrdp1 belongs to the tripartite or RBCC (RING, B-box, coiled-coil) family of ubiquitin ligases in which the RING domain is responsible for ubiquitin ligation and a variable C-terminal region mediates substrate recognition. We report here the 1.95 A crystal structure of the C-terminal domain of Nrdp1 and show that this domain is sufficient to mediate ErbB3 binding. Furthermore, we have used site-directed mutagenesis to map regions of the Nrdp1 surface that are important for interacting with ErbB3 and mediating its degradation in transfected cells. The ErbB3-binding site localizes to a region of Nrdp1 that is conserved from invertebrates to vertebrates, in contrast to ErbB3, which is only found in vertebrates. This observation suggests that Nrdp1 uses a common binding site to recognize its targets in different species.
Collapse
Affiliation(s)
- Samuel Bouyain
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
14
|
van der Horst EH, Weber I, Ullrich A. Tyrosine phosphorylation of PYK2 mediates heregulin-induced glioma invasion: novel heregulin/HER3-stimulated signaling pathway in glioma. Int J Cancer 2005; 113:689-98. [PMID: 15499613 DOI: 10.1002/ijc.20643] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Receptor tyrosine kinases of the EGFR family transmit extracellular signals that control diverse cellular functions such as proliferation, differentiation and survival. Signaling function of a member of this family, HER3, is believed to be impaired due to deviations in its kinase consensus motifs. Here we address the functional role and signaling mechanisms of HER3. HER3 preferentially forms heterodimers with HER2 inducing the most potent mitogenic signal among EGFR family members. Our data show that in a glioma-derived cell line the cytoplasmic tyrosine kinase PYK2 is constitutively associated with HER3 and that stimulation with Heregulin results in PYK2 tyrosine phosphorylation. HER3, but not HER2, mediates the phosphorylation of the C-terminal region of PYK2 to promote a mitogenic response through activation of the MAPK pathway. A central role of PYK2 in signaling downstream of HER3 is substantiated by the demonstration that expression of a dominant-negative PYK2-KM construct abrogates the Heregulin-induced MAPK activity and inhibits the invasive potential of glioma cells. These results suggest a novel Heregulin/HER3-stimulated signaling pathway in glioblastoma-derived cell lines that involves phosphorylation of PYK2 and mediates invasiveness of glioma cells.
Collapse
|
15
|
Offterdinger M, Schöfer C, Weipoltshammer K, Grunt TW. c-erbB-3: a nuclear protein in mammary epithelial cells. J Cell Biol 2002; 157:929-39. [PMID: 12045181 PMCID: PMC2174048 DOI: 10.1083/jcb.200109033] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
c-erbB receptors are usually located in cell membranes and are activated by extracellular binding of EGF-like growth factors. Unexpectedly, using immunofluorescence we found high levels of c-erbB-3 within the nuclei of MTSV1-7 immortalized nonmalignant human mammary epithelial cells. Nuclear localization was mediated by the COOH terminus of c-erbB-3, and a nuclear localization signal was identified by site-directed mutagenesis and by transfer of the signal to chicken pyruvate kinase. A nuclear export inhibitor caused accumulation of c-erbB-3 in the nuclei of other mammary epithelial cell lines as demonstrated by immunofluorescence and biochemical cell fractionation, suggesting that c-erbB-3 shuttles between nuclear and nonnuclear compartments in these cells. Growth of MTSV1-7 on permeable filters induced epithelial polarity and concentration of c-erbB-3 within the nucleoli. However, the c-erbB-3 ligand heregulin beta1 shifted c-erbB-3 from the nucleolus into the nucleoplasm and then into the cytoplasm. The subcellular localization of c-erbB-3 obviously depends on exogenous stimuli and on the stage of epithelial polarity and challenges the specific function of c-erbB-3 as a transmembrane receptor protein arguing for additional, as yet unidentified, roles of c-erbB-3 within the nucle(ol)us of mammary epithelial cells.
Collapse
Affiliation(s)
- Martin Offterdinger
- Signaling Networks Program, Division of Oncology, Department of Internal Medicine I, University of Vienna, A-1097 Vienna, Austria
| | | | | | | |
Collapse
|
16
|
Hensel F, Brändlein S, Eck M, Schmidt K, Krenn V, Kloetzer A, Bachi A, Mann M, Müller-Hermelink HK, Vollmers HP. A novel proliferation-associated variant of CFR-1 defined by a human monoclonal antibody. J Transl Med 2001; 81:1097-108. [PMID: 11502861 DOI: 10.1038/labinvest.3780322] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The germline coded human monoclonal IgM antibody 103/51 was isolated from a gastric carcinoma patient. This antibody binds to a 130-kd membrane molecule and has a mitotic effect on tumor cells in vitro. To characterize the target, we sequenced the protein and showed that the antibody binds to the cysteine-rich fibroblast growth factor receptor (CFR)-1, which is highly homologous to MG-160 and the E-selectin-ligand (ESL)-1. The epitope was determined by glycosidase-digestion experiments to be an N-linked carbohydrate side chain. Immunohistochemistry was used to investigate the tissue distribution of CFR-1. Different healthy tissues were tested and only the collecting tubes of the kidney, the Golgi apparatus, and the glomerular and fascicular zones of the adrenal gland stained positive. However, on malignant tissue the receptor is overexpressed in nearly all tested stomach cancers (12 of 15) and other tested carcinomas (13 of 15). Most interestingly, the receptor is also present in Helicobacter pylori gastritis and gastric dysplasia, but absent on uninflamed stomach mucosa. This restricted tissue pattern indicates that antibody 103/51 reacts with a membrane-bound variant of CFR-1, which is mainly expressed on transformed cells and precursor lesions and is essential for proliferation processes. The possible activity of antibody 103/51 as an activating ligand in these proliferative changes of gastric epithelial mucosa is discussed.
Collapse
Affiliation(s)
- F Hensel
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Shimoyama M, Matsuoka H, Tamekane A, Ito M, Iwata N, Inoue R, Chihara K, Furuya A, Hanai N, Matsui T. T-cell-specific expression of kinase-defective Eph-family receptor protein, EphB6 in normal as well as transformed hematopoietic cells. Growth Factors 2000; 18:63-78. [PMID: 10831073 DOI: 10.3109/08977190009003234] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Although most kinase-defective growth factor receptor proteins are associated with pathogenic conditions, a kinase-defective Eph-family receptor protein, EphB6, is expressed in normal human tissues. We generated monoclonal antibodies specific for human EphB6 to characterize its expression on human hematopoietic cells. A very small population of normal human peripheral white blood cells (0.57 +/- 0.07%, n = 12) expressed EphB6. The EphB6-positive cells were CD2+, CD7+, CD3+ and CD4+ or CD8+ lymphocytes, but they did not express CD19 or CD11b. In human bone marrow, only 1.5 +/- 0.19% of lymphocytes expressed EphB6. Compared with the expression in peripheral lymphocytes, prominent expression of EphB6 protein was demonstrated in CD4+CD8+ double-positive mouse thymocytes. The T-cell lineage-specific expression was strictly conserved in human leukemia/lymphoma cells. Among T-cell-derived leukemia cells, the expression level of EphB6 seemed to decrease with maturation of the cells. These results suggest that EphB6 expression is regulated in T-cell development.
Collapse
Affiliation(s)
- M Shimoyama
- Department of Medicine, Kobe University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Lange CA, Richer JK, Shen T, Horwitz KB. Convergence of progesterone and epidermal growth factor signaling in breast cancer. Potentiation of mitogen-activated protein kinase pathways. J Biol Chem 1998; 273:31308-16. [PMID: 9813039 DOI: 10.1074/jbc.273.47.31308] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During late stages of breast cancer progression, tumors frequently acquire steroid hormone resistance with concurrent amplification of growth factor receptors; this alteration predicts a poor prognosis. We show here that following treatment with the progestin, R5020, breast cancer cells undergo a "biochemical shift" in the regulation of epidermal growth factor (EGF)-stimulated signaling pathways: R5020 potentiates the effects of EGF by up-regulating EGFR, c-ErbB2 and c-ErbB3 receptors, and by enhancing EGF-stimulated tyrosine phosphorylation of signaling molecules known to associate with activated type I receptors. Independently of EGF, R5020 increases Stat5 protein levels, association of Stat5 with phosphotyrosine-containing proteins, and tyrosine phosphorylation of JAK2 and Shc. Furthermore, progestins "prime" breast cancer cells for growth signals by potentiating EGF-stimulated p42/p44 mitogen-activated protein kinase (MAPK), p38 MAP kinase, and JNK activities. Although the levels of cyclin D1, cyclin E, and p21(WAF1), are up-regulated by R5020 alone, they are synergistically up-regulated by EGF in the presence of R5020. Up-regulation of cell cycle proteins by EGF is blocked by inhibition of p42/p44 MAPK only in the presence of R5020, supporting a shift in the regulation of these cell cycle mediators from MAPK-independent to MAPK-dependent pathways. In summary, progesterone selectively increases the sensitivity of key kinase cascades to growth factors, thereby priming cells for stimulation by latent growth signals. These data support a model in which breast cancer cell growth switches from steroid hormone to growth factor dependence.
Collapse
Affiliation(s)
- C A Lange
- Department of Medicine, Campus Box B151, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | | | |
Collapse
|
19
|
Offterdinger M, Schneider SM, Huber H, Grunt TW. Retinoids control the expression of c-erbB receptors in breast cancer cells. Biochem Biophys Res Commun 1998; 251:907-13. [PMID: 9791009 DOI: 10.1006/bbrc.1998.9570] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nuclear retinoid and membrane c-erbB receptors participate in signal transduction systems that control mammary epithelial cell proliferation and differentiation. Recently, we demonstrated that c-erbB receptor activation stimulates retinoic acid receptor-alpha expression. We now report that retinoids reduce SK-BR-3 breast cancer cell growth by inhibiting the cell cycle and by inducing apoptosis. This is accompanied with reduced c-erbB expression as determined by FACS, Western, Northern, RT-PCR, and reporter assays. All-trans (ATRA) and 9-cis retinoic acid (9cRA) reduce c-erbB-1 protein to 50-100%, c-erbB-2 to 20-30%, and c-erbB-3 to 10-50% of control, depending on the concentration, respectively, without influencing the tyrosine phosphorylation status. Down-regulation of c-erbB-2 and -3 was seen at all levels analyzed, whereas c-erbB-1 mRNA remained unchanged. Retinoic acid-mediated down-regulation of growth and c-erbB-2 and -3 expression was also seen in MCF-7 cells. We conclude that retinoic acids are efficient repressors of c-erbB-2 and -3 gene expression, whereas c-erbB-1 is not markedly affected.
Collapse
Affiliation(s)
- M Offterdinger
- Division of Oncology, Department of Internal Medicine I, University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | | | | | | |
Collapse
|
20
|
Rajkumar T, Stamp GW, Pandha HS, Waxman J, Gullick WJ. Expression of the type 1 tyrosine kinase growth factor receptors EGF receptor, c-erbB2 and c-erbB3 in bladder cancer. J Pathol 1996; 179:381-5. [PMID: 8869284 DOI: 10.1002/(sici)1096-9896(199608)179:4<381::aid-path603>3.0.co;2-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Expression of the c-erbB3 protein was determined in transitional cell carcinoma of the bladder by immunohistochemistry. Strong membrane staining was observed in 10 per cent of cases (7/70) and cytoplasmic and membrane overexpression in 20 per cent (14/70). Overexpression of the epidermal growth factor (EGF) receptor (36 per cent, 25/70) and c-erbB2 proteins (9 per cent 6/70) was determined in the same series of cases. c-erbB3 overexpression was positively correlated with EGF receptor expression (P < 0.025) but appeared to be inversely associated with c-erbB2 overexpression.
Collapse
Affiliation(s)
- T Rajkumar
- ICRF Oncology Unit, Royal Postgraduate Medical School, London, UK
| | | | | | | | | |
Collapse
|
21
|
Zhang K, Sun J, Liu N, Wen D, Chang D, Thomason A, Yoshinaga SK. Transformation of NIH 3T3 cells by HER3 or HER4 receptors requires the presence of HER1 or HER2. J Biol Chem 1996. [PMID: 8632008 DOI: 10.1074/jbc.271.7.3884] [Citation(s) in RCA: 143] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the epidermal growth factor receptor (EGFR) subfamily of receptor protein tyrosine kinases have been implicated in the pathogenesis of various malignancies. The ability of one EGFR subfamily member to influence, or function synergistically with, another is likely to be a general feature of these receptors. To assess the role of receptor heterodimerization, we analyzed the ability of Neu differentiation factor (NDF) to induce cell growth and transformation of NIH 3T3 cells transfected with different combinations of the EGFR subfamily of receptors. NDF induced mitogenesis, but not transformation, of cells expressing either HER3 or HER4 alone. However, NDF-induced cell transformation was observed when either HER1 or HER2 was coexpressed with HER3 or HER4. In analogous receptor phosphorylation experiments, NDF-induced transphosphorylation appears to be correlated with synergistic transformation of NIH 3T3 cells. Interestingly, transphosphorylation between HER1 and HER4 can be stimulated by either EGF or NDF.
Collapse
Affiliation(s)
- K Zhang
- Amgen Inc., Thousand Oaks, California 91320, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Chen X, Levkowitz G, Tzahar E, Karunagaran D, Lavi S, Ben-Baruch N, Leitner O, Ratzkin BJ, Bacus SS, Yarden Y. An Immunological Approach Reveals Biological Differences between the Two NDF/Heregulin Receptors, ErbB-3 and ErbB-4. J Biol Chem 1996. [DOI: 10.1074/jbc.271.13.7620] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
23
|
Binder C, Marx D, Overhoff R, Binder L, Schauer A, Hiddemann W. Bcl-2 protein expression in breast cancer in relation to established prognostic factors and other clinicopathological variables. Ann Oncol 1995; 6:1005-10. [PMID: 8750153 DOI: 10.1093/oxfordjournals.annonc.a059064] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Bel-2 inhibits most kinds of programmed cell death and provides a selective survival advantage to various cell types. Bcl-2 is physiologically expressed in ductal epithelia of the normal breast. The biological significance of Bcl-2 (over)expression for the development and progression of breast cancer has still to be evaluated. PATIENTS AND METHODS A series of 133 primary breast cancers was investigated for expression of the Bcl-2 protein by immunohistochemistry of paraffin-embedded tissue sections. Results were correlated with other variables of established or presumed predictive value. RESULTS A significant positive correlation was observed between Bcl-2 expression and positivity for estrogen and progesterone receptors (p < 0.001). High proliferative activity as assessed by Ki-67 staining correlated inversely with Bcl-2 expression (p < 0.001). Bcl-2 immunostaining was not related to positivity for c-erbB-1. It was negatively associated with overexpression of c-erbB-2 (p = 0.04), whereas a strong positive correlation was found with expression of c-erbB-3 (p = 0.01). There was a significant inverse correlation between histological grading and immunoreactivity for Bcl-2 (p < 0.001). N0 tumors tended to be Bcl-2 positive, but differences were not statistically significant. CONCLUSION Bcl-2 was detected predominantly in differentiated tumors. Expression was associated with other favorable histopathological features and predictors of positive clinical outcome. Loss of Bcl-2 expression seems to be linked to loss of hormonal regulatability, increased dedifferentiation and deregulated proliferation.
Collapse
Affiliation(s)
- C Binder
- Department of Haematology/Oncology, Georg-August-University, Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Hills D, Rowlinson-Busza G, Gullick WJ. Specific targeting of a mutant, activated FGF receptor found in glioblastoma using a monoclonal antibody. Int J Cancer 1995; 63:537-43. [PMID: 7591264 DOI: 10.1002/ijc.2910630414] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A truncated epidermal growth factor receptor (EGFR) expressed from a rearranged and amplified EGFR gene is present at high frequency in gliomas. In this work we show that when this receptor is expressed in NIH3T3 fibroblasts it is partially activated and confers tumorigenicity to this cell line in vivo but no growth advantage in in vitro anchorage-independent growth assays. Because the mutation occurs in the extracellular domain of the receptor, it can be considered to represent a glioma-specific tumour marker. Here we demonstrate that 2 monoclonal antibodies, DH1.1 and DH8.3, raised to a synthetic peptide spanning the unique junctional sequence, can recognise the mutant receptor but not the normal receptor in both denatured and native states. Furthermore, radiolabelled antibody DH8.3 successfully targets tumours expressing this antigen in nude mice.
Collapse
Affiliation(s)
- D Hills
- Imperial Cancer Research Fund Oncology Unit, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
25
|
Rajkumar T, Hollywood D, Hurst H, Gullick W. c-erbB3 expression in breast tumour derived cell lines. Breast 1995. [DOI: 10.1016/0960-9776(95)90001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
26
|
|