1
|
Xiong Q, Zhu J, Zhang Y, Deng H. CAR-NK cell therapy for glioblastoma: what to do next? Front Oncol 2023; 13:1192128. [PMID: 37404752 PMCID: PMC10315652 DOI: 10.3389/fonc.2023.1192128] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/29/2023] [Indexed: 07/06/2023] Open
Abstract
Glioblastoma is a malignant tumor with the highest morbidity and mortality in the central nervous system. Conventional surgical resection combined with radiotherapy or chemotherapy has a high recurrence rate and poor prognosis. The 5-year survival rate of patients is less than 10%. In tumor immunotherapy, CAR-T cell therapy represented by chimeric antigen receptor-modified T cells has achieved great success in hematological tumors. However, the application of CAR-T cells in solid tumors such as glioblastoma still faces many challenges. CAR-NK cells are another potential adoptive cell therapy strategy after CAR-T cells. Compared with CAR-T cell therapy, CAR-NK cells have similar anti-tumor effects. CAR-NK cells can also avoid some deficiencies in CAR-T cell therapy, a research hotspot in tumor immunity. This article summarizes the preclinical research status of CAR-NK cells in glioblastoma and the problems and challenges faced by CAR-NK in glioblastoma.
Collapse
|
2
|
Feldman L, Brown C, Badie B. Chimeric Antigen Receptor (CAR) T Cell Therapy for Glioblastoma. Neuromolecular Med 2022; 24:35-40. [PMID: 34665390 PMCID: PMC11220928 DOI: 10.1007/s12017-021-08689-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/13/2021] [Indexed: 11/24/2022]
Abstract
Glioblastoma (GBM) are the most common and aggressive primary brain tumors in adults. Current mainstay treatments include surgery, chemotherapy, and radiation; however, these are ineffective. As a result, immunotherapy treatment strategies are being developed to harness the body's natural defense mechanisms against gliomas. Adoptive cell therapy with chimeric antigen receptor (CAR) T cells uses patients' own T cells that are genetically modified to target tumor-associated antigens. These cells are harvested from patients, engineered to target specific proteins expressed by the tumor and re-injected into the patient with the goal of destroying tumor cells. In this mini review, we outline the history of CAR T cell therapy, describe current antigen targets, and review challenges this treatment faces specifically in targeting GBM.
Collapse
Affiliation(s)
- Lisa Feldman
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, CA, 91010, USA.
- Division of Neurosurgery, City of Hope National Medical Center, MOB 2001, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| | - Christine Brown
- Departments of Cancer Immunotherapy & Tumor Immunology and Hematology & Hematopoietic Call Transplantation, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Behnam Badie
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, CA, 91010, USA
| |
Collapse
|
3
|
Gatto L, Franceschi E, Di Nunno V, Maggio I, Lodi R, Brandes AA. Engineered CAR-T and novel CAR-based therapies to fight the immune evasion of glioblastoma: gutta cavat lapidem. Expert Rev Anticancer Ther 2021; 21:1333-1353. [PMID: 34734551 DOI: 10.1080/14737140.2021.1997599] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The field of cancer immunotherapy has achieved great advancements through the application of genetically engineered T cells with chimeric antigen receptors (CAR), that have shown exciting success in eradicating hematologic malignancies and have proved to be safe with promising early signs of antitumoral activity in the treatment of glioblastoma (GBM). AREAS COVERED We discuss the use of CAR T cells in GBM, focusing on limitations and obstacles to advancement, mostly related to toxicities, hostile tumor microenvironment, limited CAR T cells infiltration and persistence, target antigen loss/heterogeneity and inadequate trafficking. Furthermore, we introduce the refined strategies aimed at strengthening CAR T activity and offer insights in to novel immunotherapeutic approaches, such as the potential use of CAR NK or CAR M to optimize anti-tumor effects for GBM management. EXPERT OPINION With the progressive wide use of CAR T cell therapy, significant challenges in treating solid tumors, including central nervous system (CNS) tumors, are emerging, highlighting early disease relapse and cancer cell resistance issues, owing to hostile immunosuppressive microenvironment and tumor antigen heterogeneity. In addition to CAR T cells, there is great interest in utilizing other types of CAR-based therapies, such as CAR natural killer (CAR NK) or CAR macrophages (CAR M) cells for CNS tumors.
Collapse
Affiliation(s)
- Lidia Gatto
- Medical Oncology Department, Azienda USL, Bologna, Italy
| | - Enrico Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, Bologna, Italy
| | | | - Ilaria Maggio
- Medical Oncology Department, Azienda USL, Bologna, Italy
| | - Raffaele Lodi
- IrcssIstituto di Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alba Ariela Brandes
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, Bologna, Italy
| |
Collapse
|
4
|
Feldman L, Brown C, Badie B. Chimeric Antigen Receptor T-Cell Therapy: Updates in Glioblastoma Treatment. Neurosurgery 2021; 88:1056-1064. [PMID: 33575786 DOI: 10.1093/neuros/nyaa584] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) are the most common and among the deadliest brain tumors in adults. Current mainstay treatments are insufficient to treat this tumor, and therefore, more effective therapies are desperately needed. Immunotherapy, which takes advantage of the body's natural defense mechanism, is an exciting emerging field in neuro-oncology. Adoptive cell therapy with chimeric antigen receptor (CAR) T cells provides a treatment strategy based on using patients' own selected and genetically engineered cells that target tumor-associated antigens. These cells are harvested from patients, modified to target specific proteins expressed by the tumor, and re-introduced into the patient with the goal of destroying tumor cells. Here, we review the history of CAR T-cell therapy, and describe the characteristics of various generations of CAR T therapies, and the challenges inherent to treatment of GBM. Finally, we describe recent and current CAR T clinical trials designed to combat GBM.
Collapse
Affiliation(s)
- Lisa Feldman
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, California
| | - Christine Brown
- Department of Cancer Immunotherapy & Tumor Immunology, City of Hope National Medical Center, Duarte, California.,Department of Hematology & Hematopoietic Call Transplantation, City of Hope National Medical Center, Duarte, California
| | - Behnam Badie
- Division of Neurosurgery, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
5
|
Wang K, Liu J, Duan Y, Wu J, Dongye S, Wang Y, Liu Z, Han G. C-erbB-2 expression is related with pathological progression of gastric cancer: results of a non-radioactive in situ hybridization. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9649-9653. [PMID: 31966845 PMCID: PMC6965992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/23/2017] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To study the relationship of c-erbB-2 oncogene expression with major pathological characteristics of gastric cancer (GC) progression. METHODS Eighty-one GC specimens were studied for c-erbB-2 oncogene amplification using non-radioactive in situ hybridization method. The c-erbB-2 overexpression status was correlated with tumor differentiation, tumor invasion and lymph node metastasis. RESULTS Among the 81 pathology confirmed GC patients, 41 (50.6%) were found to have c-erbB-2 overexpression in cancer tissues. The rate of c-erbB-2 overexpression was significantly higher in those with poor tumor differentiation (63.0%, 29/46) than in those with well differentiated tumor (34.3%, 12/35) (χ 2=6.576, P<0.001); significantly higher in those that invaded into deep muscle and beyond (55.7%, 39/70) than in those with tumors limited to the superficial muscle (18.2%, 2/11) (χ 2=5.357, P<0.025); and significantly higher in those with lymph node metastases (59.6%, 34/57) than in those without lymph node involvement (29.2%, 7/24) (χ 2=6.278, P<0.025). CONCLUSIONS c-erbB-2 oncogene overexpression may indicate a more aggressive biological behavior of the tumor and could be used as a predictive marker for GC pathological progression.
Collapse
Affiliation(s)
- Keqiang Wang
- Department of Clinical Laboratory, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| | - Jiangang Liu
- Department of General Surgery, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| | - Yanchao Duan
- Department of Hematology, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| | - Jiafeng Wu
- Department of Clinical Laboratory, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| | - Shengyi Dongye
- Department of Pathology, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| | - Yiren Wang
- Medical Laboratory Class, Grade 2016, Medical College, Qingdao UniversityQingdao, Shandong Province, China
| | - Zhenzhong Liu
- Department of General Surgery, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| | - Guoxin Han
- Department of General Surgery, Affiliated Hospital of Taishan Medical UniversityTai’an, Shandong Province, China
| |
Collapse
|
6
|
Menendez JA, Rubio MA, Campisi J, Lupu R. Heregulin, a new regulator of telomere length in human cells. Oncotarget 2016; 6:39422-36. [PMID: 26318724 PMCID: PMC4741836 DOI: 10.18632/oncotarget.4964] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 07/12/2015] [Indexed: 01/12/2023] Open
Abstract
The growth factor heregulin (HRG) promotes breast cancer (BC) tumorigenesis and metastasis and differentially modulates BC cell responses to DNA-damaging agents via its dual extracellular and nuclear localization. Given the central role of telomere dysfunction to drive carcinogenesis and to alter the chemotherapeutic profile of transformed cells, we hypothesized that an unanticipated nuclear function of HRG might be to regulate telomere length. Engineered overexpression of the HRGβ2 isoform in non-aggressive, HRG-negative MCF-7 BC cells resulted in a significant shortening of telomeres (up to 1.3 kb) as measured by Southern blotting of telomere terminal restriction fragments. Conversely, antisense-mediated suppression of HRGβ2 in highly aggressive, HRG-overexpressing MDA-MB-231 and Hs578T cells increased telomere length up to 3.0 kb. HRGβ2 overexpression promoted a marked upregulation of telomere-binding protein 2 (TRF2) protein expression, whereas its knockdown profoundly decreased TRF2 expression. Double staining of endogenous HRGβ2 with telomere-specific peptide nucleic acid probe/fluorescence in situ hybridization (PNA/FISH) revealed the partial localization of HRG at the chromosome ends. Moreover, a predominantly nucleoplasmic staining pattern of endogenous HRGβ2 appeared to co-localize with TRF2 and, concomitantly with RAP1, a telomere regulator that specifically interacts with TRF2. Small interfering RNA-mediated knockdown of HRG decreased the expression of TRF2 and RAP1, decreased their presence at chromosome ends, and coincidentally resulted in the formation of longer telomeres. This study uncovers a new function for HRGβ2 in controlling telomere length, in part due to its ability to regulate and interact with the telomere-associated proteins TRF2 and RAP1.
Collapse
Affiliation(s)
- Javier A Menendez
- ProCURE (Program Against Cancer Therapeutic Resistance), Metabolism & Cancer Group, Catalan Institute of Oncology (ICO), Girona, Spain.,Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Miguel A Rubio
- Laboratory of Hematology Service, Institut d'Investigació Biomèdica Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Judith Campisi
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, CA, USA.,Buck Institute for Research on Aging, Novato, CA, USA
| | - Ruth Lupu
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Rochester, MN, USA.,Mayo Clinic Cancer Center, Rochester, MN, USA
| |
Collapse
|
7
|
Zhang L, Ren J, Zhang H, Cheng G, Xu Y, Yang S, Dong C, Fang D, Zhang J, Yang A. HER2-targeted recombinant protein immuno-caspase-6 effectively induces apoptosis in HER2-overexpressing GBM cells in vitro and in vivo. Oncol Rep 2016; 36:2689-2696. [DOI: 10.3892/or.2016.5088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 07/18/2016] [Indexed: 11/05/2022] Open
|
8
|
Zhou BG, Liu MY, Qiu XC, Xu YM, Fan QY, Yang AG, Zhang Y, Xia H. A novel recombinant immunocasp-6 fusion gene specifically and efficiently suppresses HER2-overexpressing osteosarcoma. Oncol Rep 2013; 29:276-82. [PMID: 23135254 DOI: 10.3892/or.2012.2122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 09/25/2012] [Indexed: 11/06/2022] Open
Abstract
Osteosarcoma is the most common primary malignant tumor of bone for adolescent or children. The poor prognosis of patients, due to its remote metastasis, has led to the exploration of more effective and less toxic treatments. Immunotherapy is a promising strategy for the treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing tumors. Herein, we describe experiments conducted with a fusion gene, immunocasp-6, which was generated by fusing a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A and an active caspase-6 which can directly cleave lamin A leading to nucleus damage inducing programmed cell death. We demonstrated that immunocasp-6 can specifically and efficiently recognize and induce apoptosis in HER2-overexpressing osteosarcoma cells in vitro. The immunocasp-6 was transferred into BALB/c athymic mice bearing human osteosarcoma by i.m. injection of liposome-encapsulated pCMV-immunocap-6. Expression of immunocasp-6 not only strongly inhibited tumor growth and significantly prolonged animal survival, but also greatly prevented tumor metastasis. Our data showed that the immuno-casp-6 can specifically recognize HER2-overexpressing osteosarcoma cells, can also promptly attack their nucleus and induce apoptotic death, suggesting the potential of this strategy for the treatment of human HER2-overexpressing tumors.
Collapse
Affiliation(s)
- Ben-Gen Zhou
- Graduate School of Southern Medical University, Department of Orthopaedics, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, Guangdong, PR China
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Affiliation(s)
- A S Leong
- Division of Tissue Pathology, Institute of Medical and Veterinary Science and Department of Pathology, University of Adelaide, Adelaide, South Australia
| | | |
Collapse
|
10
|
Khan IH, Zhao J, Ghosh P, Ziman M, Sweeney C, Kung HJ, Luciw PA. Microbead arrays for the analysis of ErbB receptor tyrosine kinase activation and dimerization in breast cancer cells. Assay Drug Dev Technol 2010; 8:27-36. [PMID: 20035613 DOI: 10.1089/adt.2009.0208] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) in the ErbB family (EGFR, ErbB2, ErbB3, and ErbB4) are implicated in a variety of human malignancies. Accordingly, determination of both expression and activation (dimerization/heterodimerization and phosphorylation) of ErbB proteins is critical in defining their functional role in cancer. Efficient and comprehensive methods to study molecular functions of ErbB family of RTKs are needed not only for improvements in diagnostics but also for early screening of targeted drugs (eg, small molecule inhibitors and therapeutic antibodies). We report development of 3 multiplex microbead immunoassays for simultaneous detection of expression, protein-protein interactions, and phosphorylation of these RTKs. These novel multiplex immunoassays were used to study ErbB RTKs under different cell activation conditions in 2 breast cancer cell lines (MDA-MB-453 and MDA-MB-468) and an epidermoid cancer cell line (A431). The results were confirmed by immunoprecipitation/western blot. Importantly, the multiplex immunoassay facilitated time-course studies in these cell lines after cell activation with EGF and neuregulin, revealing the kinetics of phosphorylation of the ErbB family RTKs. This study demonstrates the utility of the Luminex(R) multiplex system as an efficient and comprehensive approach to study different aspects of molecular roles of these RTKs. Importantly, the study provides proof-of-concept for the utility of the multiplex microbead immunoassay approach for potential use in efficient, robust, and rapid screening of drugs, particularly those targeting functional aspects of these potent signaling molecules. In addition, the assays described here may be useful for cancer diagnostics and monitoring efficacy of therapy targeting the ErbB family of RTKs.
Collapse
Affiliation(s)
- Imran H Khan
- Center for Comparative Medicine, University of California at Davis, Hutchison Drive and County Road 98, Davis, CA 95616, USA.
| | | | | | | | | | | | | |
Collapse
|
11
|
Feigin ME, Muthuswamy SK. ErbB receptors and cell polarity: new pathways and paradigms for understanding cell migration and invasion. Exp Cell Res 2008; 315:707-16. [PMID: 19022245 DOI: 10.1016/j.yexcr.2008.10.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 10/19/2008] [Accepted: 10/20/2008] [Indexed: 12/25/2022]
Abstract
The ErbB family of receptor tyrosine kinases is involved in initiation and progression of a number of human cancers, and receptor activation or overexpression correlates with poor patient survival. Research over the past two decades has elucidated the molecular mechanisms underlying ErbB-induced tumorigenesis, which has resulted in the development of effective targeted therapies. ErbB-induced signal transduction cascades regulate a wide variety of cell processes, including cell proliferation, apoptosis, cell polarity, migration and invasion. Within tumors, disruption of these core processes, through cooperative oncogenic lesions, results in aggressive, metastatic disease. This review will focus on the ErbB signaling networks that regulate migration and invasion and identify a potential role for cell polarity pathways during cancer progression.
Collapse
Affiliation(s)
- Michael E Feigin
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | | |
Collapse
|
12
|
Qiu XC, Xu YM, Wang F, Fan QY, Wang LF, Ma BA, Jia LT, Zhao J, Meng YL, Yao LB, Chen SY, Yang AG. Single-chain antibody/activated BID chimeric protein effectively suppresses HER2-positive tumor growth. Mol Cancer Ther 2008; 7:1890-1899. [PMID: 18644999 DOI: 10.1158/1535-7163.mct-07-2235] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BH3-interacting domain death agonist (BID) is a crucial element in death signaling pathways and is recognized as an intracellular link connecting the intrinsic mitochondrial apoptotic and extrinsic death receptor-mediated apoptotic pathways. Herein, we describe experiments conducted with a fusion protein, which was generated by fusing a human epidermal growth factor receptor-2 (HER2)-specific single-chain antibody with domain II of Pseudomonas exotoxin A and the truncated active BID (tBID). These experiments extend our previous work on several other immuno-proapoptotic proteins. Specifically, by excluding cells with undetectable HER2, we showed that the secreted immuno-tBID molecule selectively recognized and killed HER2-overexpressing tumor cells in vitro by attacking their mitochondria and inducing their apoptotic death. This apoptosis could only be inhibited partially by caspase pan-inhibitor zVAD and mitochondrial protector TAT-BH4. Subsequently, we transferred the immuno-tbid gene into BALB/c athymic mice bearing HER2-positive tumors together with other immuno-proapoptotic proteins using i.m. injections of liposome-encapsulated vectors. The expression of the immuno-tbid gene suppressed tumor growth and prolonged animal survival significantly. We also shortened the translocation domain of Pseudomonas exotoxin A II to only 10-amino acid sequence, which were crucial for furin cleavage. The new recombinant molecule retained the translocation efficiency and the ability of specific killing HER2-positive tumor cells. Our data showed that, compared with the toxins employed before, the chimeric immuno-tBID molecule can not only specifically recognize HER2-positive tumor cells but also certainly induce apoptosis even in the presence of zVAD and TAT-BH4, thereby suggesting an alternative approach to treating HER2/neu-positive tumors.
Collapse
Affiliation(s)
- Xiu-Chun Qiu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Fry DW. Protein tyrosine kinases as therapeutic targets in cancer chemotherapy and recent advances in the development of new inhibitors. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.3.6.577] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
14
|
Maegawa M, Takeuchi K, Funakoshi E, Kawasaki K, Nishio K, Shimizu N, Ito F. Growth stimulation of non-small cell lung cancer cell lines by antibody against epidermal growth factor receptor promoting formation of ErbB2/ErbB3 heterodimers. Mol Cancer Res 2007; 5:393-401. [PMID: 17426253 DOI: 10.1158/1541-7786.mcr-06-0303] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibodies are the most rapidly expanding class of human therapeutics, including their use in cancer therapy. Monoclonal antibodies (mAb) against epidermal growth factor (EGF) receptor (EGFR) generated for cancer therapy block the binding of ligand to various EGFR-expressing human cancer cell lines and abolish ligand-dependent cell proliferation. In this study, we show that our mAb against EGFRs, designated as B4G7, exhibited a growth-stimulatory effect on various human cancer cell lines including PC-14, a non-small cell lung cancer cell line; although EGF exerted no growth-stimulatory activity toward these cell lines. Tyrosine phosphorylation of EGFRs occurred after treatment of PC-14 cells with B4G7 mAb, and it was completely inhibited by AG1478, a specific inhibitor of EGFR tyrosine kinase. However, this inhibitor did not affect the B4G7-stimulated cell growth, indicating that the growth stimulation by B4G7 mAb seems to be independent of the activation of EGFR tyrosine kinase. Immunoprecipitation with anti-ErbB3 antibody revealed that B4G7, but not EGF, stimulated heterodimerization between ErbB2 and ErbB3. ErbB3 was tyrosine phosphorylated in the presence of B4G7 but not in the presence of EGF. Further, the phosphorylation and B4G7-induced increase in cell growth were inhibited by AG825, a specific inhibitor of ErbB2. These results show that the ErbB2/ErbB3 dimer functions to promote cell growth in B4G7-treated cells. Changes in receptor-receptor interactions between ErbB family members after inhibition of one of its members are of potential importance in optimizing current EGFR family-directed therapies for cancer.
Collapse
Affiliation(s)
- Mari Maegawa
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Human epidermal growth factor receptor 2 (HER-2) is over-expressed in 15% to 30% of breast cancers and is a poor prognostic marker in node-positive patients. HER-2 expression is an indicator of greater sensitivity to anthracycline-based chemotherapy and is the major criterion for selection for treatment with the anti-HER-2 antibody trastuzumab (Herceptin). Fluorescence in situ hybridization and immunohistochemistry (IHC) are the 2 most commonly used methods for detection of the gene and protein, respectively. Criticisms have been levied at the IHC method of identifying HER-2 overexpression but convenience and costs of this technique cannot be overlooked. Modifications to the IHC technique and scoring accommodate for many of the problems that derive from variables in preanalytical and analytic factors that influence results but standardization is currently impossible to attain. Deficiencies in fluorescence in situ hybridization assay also exist and alternative molecular methods of assay are explored in this review.
Collapse
Affiliation(s)
- Trishe Y-M Leong
- Department of Anatomical Pathology, Monash Medical Centre, Melbourne, Australia
| | | |
Collapse
|
16
|
Jiménez-Farfán D, Guevara J, Zenteno E, Malagón H, Hernández-Guerrero JC. EGF-R and erbB-2 in murine tooth development after ethanol exposure. ACTA ACUST UNITED AC 2005; 73:65-71. [PMID: 15690356 DOI: 10.1002/bdra.20113] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Alcohol consumption during pregnancy can frequently lead to a congenital disorder known as fetal alcohol syndrome (FAS); however, not all children born to alcoholic women develop FAS. Alcohol consumption may affect diverse organs and systems during embryonic development, including craniofacial structures. Small teeth, enamel alterations, and delayed eruption have been observed after ethanol exposure. Epidermal growth factor receptors (EGF-Rs) participate in dental proliferation and differentiation, and changes in these receptors were considered here to be a likely mechanism associated to the dental anomalies observed in this syndrome. Epidermal growth factor receptor type 1 (EGF-R) and epidermal growth factor receptor type 2 (erbB-2) immunoexpression during the lower first molar morphogenesis was investigated in mouse fetuses exposed to ethanol during gestation. METHODS Pregnant female mice were divided into groups, consuming either 5, 10, 15, 20, or 25% ethanol solutions, or water (control group). Heads were obtained from 16.5- and 18.5-day fetuses. Immunohistochemistry was applied to EGF-R and erbB-2. RESULTS At days 16.5 and 18.5, fetuses from 15%, 20%, and 25% ethanol groups showed delayed differentiation, degenerative changes in dental epithelial tissues and reduced dental size; additionally, they displayed an enhanced immunoreactivity to EGF-R and erbB-2. CONCLUSIONS Our results suggest that ethanol consumption during pregnancy affects the expression of EGF receptors and induces a delay in murine fetal dental morphogenesis. Dental development is a process that involves a number of growth factors; hence we consider that further research is required to show whether the changes in glycosylation and growth-factor signaling pathways observed in other cells are also involved in the alterations observed in this study.
Collapse
Affiliation(s)
- Dolores Jiménez-Farfán
- Laboratorio de Inmunologa, DEPeI, Facultad de Odontologa, Universidad Nacional Autónoma de México, México City, Mexico
| | | | | | | | | |
Collapse
|
17
|
Panutsopulos D, Arvanitis DL, Tsatsanis C, Papalambros E, Sigala F, Spandidos DA. Expression of heregulin in human coronary atherosclerotic lesions. J Vasc Res 2005; 42:463-74. [PMID: 16155362 DOI: 10.1159/000088100] [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] [Received: 03/07/2005] [Accepted: 07/03/2005] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Endothelial cells, monocytes/macrophages, and vascular smooth muscle cells contribute to the establishment and progression of atherosclerotic lesions by expressing growth and inflammatory factors. The aim of the present study was to determine whether heregulin (HRG) is associated with human coronary artery disease. METHODS Twenty-six fresh human coronary artery segments were collected at autopsy. Expression of cysteine-rich 61 (CYR61) and VEGF in response to HRG was studied in the human endothelial cell line EA.hy926, and expression of CYR61 and HRG was evaluated in activated macrophages isolated from peripheral blood of healthy donors. RESULTS We found that HRG was overexpressed at the protein and mRNA level in all lesions analyzed and gradually increased as the stages of the lesions progressed. Expression of HRG was observed in the intima primarily in macrophages. The same specimens were analyzed for the expression of CYR61, an angiogenetic factor regulated by HRG in breast cancer epithelial cells. CYR61 was expressed in both normal and atheromatic specimens, but its expression was significantly enhanced in macrophages of the intima. Activation of primary human macrophages results in increased expression of both HRG and CYR61. In addition, studies in endothelial cells where no endogenous HRG is present showed that HRG induces expression of CYR61 and secretion of VEGF. CONCLUSIONS HRG may, therefore, play an important role in the development of coronary artery disease and the expansion of the atherosclerotic plaque and may locally regulate the expression of the angiogenetic factor CYR61.
Collapse
|
18
|
Xu YM, Wang LF, Jia LT, Qiu XC, Zhao J, Yu CJ, Zhang R, Zhu F, Wang CJ, Jin BQ, Chen SY, Yang AG. A caspase-6 and anti-human epidermal growth factor receptor-2 (HER2) antibody chimeric molecule suppresses the growth of HER2-overexpressing tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2004; 173:61-67. [PMID: 15210759 DOI: 10.4049/jimmunol.173.1.61] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Clinical studies have suggested that human epidermal growth factor receptor-2 (HER2) provide a useful target for antitumor therapy. We previously described the generation of a chimeric HER2-targeted immunocasp-3 protein. In this study, we extend the repertoire of chimeric proapoptotic proteins with immunocasp-6, a construct that comprises a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A, and an active caspase-6, which can directly cleave lamin A leading to nucleus damage and inducing programmed cell death. We demonstrate that the secreted immunocasp-6 molecule selectively recognizes and induces apoptosis in HER2-overexpressing tumor cells in vitro, but not in cells with undetectable HER2. The immunocasp-6 gene was next transferred into BALB/c athymic mice bearing human breast SK-BR-3 tumors by i.m. injection of liposome-encapsulated vectors, by intratumor injection of adenoviral vectors, or by i.v. injection of PBMC modified by retroviral infection. Regardless of the method used, expression of immunocasp-6 suppressed tumor growth and prolonged animal survival significantly. Our data show that the chimeric immunocasp-6 molecule can recognize HER2-positive tumor cells, promptly attack their nucleus, and induce their apoptotic death, suggesting the potential of this strategy for the treatment of human cancers that overexpress HER2.
Collapse
Affiliation(s)
- Yan-Ming Xu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, 17 Changle West Road, 710-032 Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Summary. I have enjoyed reading previous historical sketches that have appeared in Journal of Thrombosis and Haemostasis, and especially those by Ted Tuddenham on factor VIII and Bjorn Dahlback on activated protein C resistance. Like those authors, I have tried to capture some of the excitement-as well as the disappointments-that occurred along the way to a long-term goal.
Collapse
Affiliation(s)
- R F Doolittle
- Center for Molecular Genetics, University of California, San Diego, La Jolla, CA 92093-0634, USA.
| |
Collapse
|
20
|
Abstract
More than a dozen crystal structures of fibrinogen and fibrin fragments have been reported, including a structure of a native fibrinogen. The majority of the other structures are fragments D and d-dimer crystallized in the presence or absence of synthetic peptide ligands patterned on the A and B 'knobs'. Overall, fibrinogens or their fragments from four different species--human, bovine, chicken and lamprey--have been studied so far, with only minor differences in the structures being observed. Although these studies have thrown much light on the details of the fibrinogen to fibrin conversion, much remains to be found out.
Collapse
Affiliation(s)
- R F Doolittle
- Center for Molecular Genetics, University of California, San Diego, La Jolla, California 92093-0634, USA.
| |
Collapse
|
21
|
Tsai MS, Shamon-Taylor LA, Mehmi I, Tang CK, Lupu R. Blockage of heregulin expression inhibits tumorigenicity and metastasis of breast cancer. Oncogene 2003; 22:761-8. [PMID: 12569369 DOI: 10.1038/sj.onc.1206130] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The growth factor heregulin (HRG), expressed in about 30% of breast cancer tumors, activates the erbB-2 receptor via induction of heterodimeric complexes of erbB-2 with erbB-3 or erbB-4. HRG induces tumorigenicity and metastasis of breast cancer cells. Our investigation into whether HRG is a factor likely to promote tumor formation independently of erbB-2 overexpression concludes that blockage of HRG expression suppresses the aggressive phenotype of MDA-MB-231 breast cancer cells by inhibiting cell proliferation, preventing anchorage-independent growth, and suppressing the invasive potential of the cells in vitro. More importantly, we observed a marked reduction in tumor formation, tumor size, and a lack of metastasis in vivo. These studies were achieved by blocking HRG expression in MDA-MB-231 cells using an HRG antisense cDNA. In the search for the mechanism by which blockage of HRG reverts this aggressive phenotype, we discovered that the cells in which HRG is blocked exhibit a marked decrease in erbB activation and a significant reduction in MMP-9 activity, demonstrating a direct causal role in HRG induction of tumorigenicity. Our study is the first report and serves as a proof of the concept that HRG is a key promoter of breast cancer tumorigenicity and metastasis independently of erbB-2 overexpression and should be deemed a potential target in developing therapies for breast cancer.
Collapse
Affiliation(s)
- Miaw-Sheue Tsai
- Ernest Orlando Lawrence Berkeley National Laboratory, University of California, 94720, USA
| | | | | | | | | |
Collapse
|
22
|
Tari AM, Lim SJ, Hung MC, Esteva FJ, Lopez-Berestein G. Her2/neu induces all-trans retinoic acid (ATRA) resistance in breast cancer cells. Oncogene 2002; 21:5224-32. [PMID: 12149644 DOI: 10.1038/sj.onc.1205660] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2001] [Revised: 04/19/2002] [Accepted: 05/10/2002] [Indexed: 11/09/2022]
Abstract
We observed that all-trans retinoic acid (ATRA) inhibited the growth of MCF-7 breast cancer cells, but not those transfected with HER2/NEU or its transactivating ligand HEREGULIN. This suggests that Her2/neu causes breast cancer cells to be resistant to the growth inhibitory effects of ATRA. To confirm this observation, MDA-MB-453 and BT-474 cells, which have high levels of Her2/neu and are resistant to ATRA, were incubated with the trastuzumab (Herceptin) antibody so that we could determine whether inhibition of the expression and function of Her2/neu would resensitize these cells to ATRA. Indeed, we found that MDA-MB-453 and BT-474 cells treated with trastuzumab were growth inhibitory by ATRA. We then determined whether Her2/neu uses Grb2 and Akt proteins to induce ATRA resistance. Liposome-incorporated Grb2 antisense oligonucleotides (L-Grb2) and a dominant negative (DN) AKT mutant were used to down-regulate Grb2 expression and inhibit Akt activity, respectively. When incubated with L-Grb2 or transfected with the DN AKT mutant, ATRA-resistant, Her2/neu-overexpressing cells became sensitive to ATRA. Our results indicate that Her2/neu utilizes Grb2 and Akt proteins to induce ATRA resistance in breast cancer cells. ATRA sensitivity was also correlated with RARalpha protein levels since higher RARalpha protein levels were observed in cells in which the Her2/neu pathway was inhibited.
Collapse
Affiliation(s)
- Ana M Tari
- Department of Bioimmunotherapy, Section of Immunobiology and Drug Carriers, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, TX 77030, USA
| | | | | | | | | |
Collapse
|
23
|
Li L, Cleary S, Mandarano MA, Long W, Birchmeier C, Jones FE. The breast proto-oncogene, HRGalpha regulates epithelial proliferation and lobuloalveolar development in the mouse mammary gland. Oncogene 2002; 21:4900-7. [PMID: 12118369 DOI: 10.1038/sj.onc.1205634] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2002] [Revised: 04/24/2002] [Accepted: 04/29/2002] [Indexed: 11/09/2022]
Abstract
Members of the EGF family of growth factors play critical roles during normal and neoplastic breast development. EGF family member HRGalpha is the only HRG1 isoform expressed in the mouse mammary gland and our previous experiments suggest that HRG1 has a unique role in mammary development. To determine the function of HRGalpha activity during mouse mammary gland development, we generated a HRGalpha-deficient mouse strain. Unlike mice with HRG1 or isoform specific HRGbeta gene deletions, HRGalpha-null mice survive to adulthood. HRGalpha-deficient mice display pronounced defects in mammary gland lobuloalveolar development at 17 days of pregnancy and 3 days post-partum. Terminal and lateral ductal alveoli were condensed and alveolar outgrowth during pregnancy was severely impaired. A dramatic reduction in beta-casein expression accompanied defective alveolar development in the HRGalpha-null mice, as determined by in situ hybridization and Northern blot analysis of HRGalpha-deficient mammary glands at 3 days post-partum. Expression of the milk-protein genes WAP and alpha-lactalbumin was not adversely affected. In situ incorporation of BrdU demonstrated that epithelial proliferation was significantly curtailed in mammary glands of HRGalpha-deficient mice at 17 days post-coitus and 3 days post-partum. These results demonstrate that HRGalpha is an important mammary gland mitogen regulating alveolar development and lactogenesis.
Collapse
Affiliation(s)
- Li Li
- Department of Medical Genetics, Max-Delbruck-Center (MDC) for Molecular Medicine, 13122 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Houimel M, Schneider P, Terskikh A, Mach JP. Selection of peptides and synthesis of pentameric peptabody molecules reacting specifically with ErbB-2 receptor. Int J Cancer 2001; 92:748-55. [PMID: 11340582 DOI: 10.1002/1097-0215(20010601)92:5<748::aid-ijc1258>3.0.co;2-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The HER-2/ErbB-2 oncoprotein is overexpressed in human breast and ovarian adenocarcinomas and is clearly associated with the malignant phenotype. Although no specific ligand for this receptor has been positively identified, ErbB-2 was shown to play a central role in a network of interactions with the related ErbB-1, ErbB-3 and ErbB-4 receptors. We have selected new peptides binding to ErbB-2 extracellular domain protein (ECD) by screening 2 newly developed constrained and unconstrained random hexapeptide phage libraries. Out of 37 phage clones, which bound specifically to ErbB-2 ECD, we found 6 constrained and 10 linear different hexapeptide sequences. Among the latter, 5 consensus motifs, all with a common methionine and a positively charged residue at positions 1 and 3, respectively, were identified. Furthermore, 3 representative hexapeptides were fused to a coiled-coil pentameric recombinant protein to form the so-called peptabodies recently developed in our laboratory. The 3 peptabodies bound specifically to the ErbB-2 ECD, as determined by enzyme-linked immunosorbent assay and BIAcore analysis and to tumor cells overexpressing ErbB-2, as shown by flow cytometry. Interestingly, one of the free selected linear peptides and all 3 peptabodies inhibited the proliferation of tumor cells overexpressing ErbB-2. In conclusion, a novel type of ErbB-2-specific ligand is described that might complement presently available monoclonal antibodies.
Collapse
Affiliation(s)
- M Houimel
- Institute of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | | | | | | |
Collapse
|
25
|
Abstract
Human carcinomas frequently express high levels of receptors in the EGF receptor family, and overexpression of at least two of these receptors, the EGF receptor (EGFr) and closely related ErbB2, has been associated with a more aggressive clinical behavior. Further, transfection or activation of high levels of these two receptors in nonmalignant cell lines can lead to a transformed phenotype. For these reasons therapies directed at preventing the function of these receptors have the potential to be useful anti-cancer treatments. In the last two decades monoclonal antibodies (MAbs) which block activation of the EGFr and ErbB2 have been developed. These MAbs have shown promising preclinical activity and 'chimeric' and 'humanized' MAbs have been produced in order to obviate the problem of host immune reactions. Clinical activity with these antibodies has been documented: trastuzumab, a humanized anti-ErbB2 MAb, is active and was recently approved in combination with paclitaxel for the therapy of patients with metastatic ErbB2-overexpressing breast cancer; IMC-C225, a chimeric anti-EGFr MAb, has shown impressive activity when combined with radiation therapy and reverses resistance to chemotherapy. In addition to antibodies, compounds that directly inhibit receptor tyrosine kinases have shown preclinical activity and early clinical activity has been reported. A series of phase III studies with these antibodies and direct tyrosine kinase inhibitors are ongoing or planned, and will further address the role of these active anti-receptor agents in the treatment of patients with cancer.
Collapse
Affiliation(s)
- J Mendelsohn
- Department of Medicine, The University of Texas, MD Anderson Cancer Center, Houston, 77030-4009, USA
| | | |
Collapse
|
26
|
Lim SJ, Lopez-Berestein G, Hung MC, Lupu R, Tari AM. Grb2 downregulation leads to Akt inactivation in heregulin-stimulated and ErbB2-overexpressing breast cancer cells. Oncogene 2000; 19:6271-6. [PMID: 11175341 DOI: 10.1038/sj.onc.1204014] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
ErbB2 can be activated by its own overexpression or be transactivated by the heregulin polypeptide growth factor. Activation of ErbB2 leads to breast cancer cell proliferation, presumably by inducing the activation of extracellular signal-regulated kinases 1,2 (Erk1,2) and Akt. We have previously reported that the growth factor receptor bound protein-2 (Grb2) is required for the proliferation of ErbB2-overexpressing breast cancer cells. We investigated here whether Grb2 protein plays a role in heregulin-stimulated proliferation. Grb2 protein inhibition led to growth inhibition of heregulin-stimulated breast cancer cells, but not Erk1,2 inactivation. These findings are similar to our earlier observations in ErbB2-overexpressing cells. Since Akt can also be activated by heregulin, the effects of Grb2 inhibition on Akt were examined. Akt was inactivated following Grb2 downregulation in heregulin-stimulated breast cancer cells. We then examined the effects of Grb2 downregulation on Akt in ErbB2-overexpressing cells in the absence of heregulin. Similar to heregulin-stimulated cells, Grb2 inhibition also led to Akt inactivation in ErbB2-overexpressing breast cancer cells. Our results indicate that the activation of ErbB2 by heregulin or by its overexpression requires Grb2 to stimulate the Akt pathway to propagate mitogenic signals.
Collapse
Affiliation(s)
- S J Lim
- Department of Bioimmunotherapy, Section of Immunobiology and Drug Carriers, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
27
|
Nagy P, Jenei A, Damjanovich S, Jovin TM, Szölôsi J. Complexity of signal transduction mediated by ErbB2: clues to the potential of receptor-targeted cancer therapy. Pathol Oncol Res 2000; 5:255-71. [PMID: 10607920 DOI: 10.1053/paor.1999.0255] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The erbB2 oncogene belongs to the type I trans-membrane tyrosine kinase family of receptors. Its medical importance stems from its widespread over-expression in breast cancer. This review will focus on the signal transduction through this protein, and explains how the overexpression of erbB2 may result in poor prognosis of breast cancer, and finally it will summerize our current understanding about the therapeutic potential of receptor-targeted therapy in breast cancer. ErbB2 does not have any known ligand which is able to bind to it with high affinity. However the kinase activity of erbB2 can be activated without any ligand, if it is overexpressed, and by heteroassociation with other members of the erbB family (erbB1 or epidermal growth factor receptor, erbB3 and erbB4). This interaction substantially increases the efficiency and diversity of signal transduction through these receptor complexes. In addition, erbB2 forms large scale receptor clusters containing hundreds of proteins. These receptor islands may take part in recruiting cytosolic factors which relay the signal towards the nucleus or the cytoplasm. Overexpression of erbB2 was linked to higher transforming activity, increased metastatic potential, angiogenesis and drug resistence of breast tumor in laboratory experiments. As a corollary of these properties, erbB2 amplification is generally thought to be associated with a poor prognosis in breast cancer patients. These early findings lead to the development of antibodies that down-regulate erbB2. Such a therapeutic approach has already been found effective in experimental tumor models and in clinical trials as well. Further understanding of the importance of erbB2 and growth factor receptors in the transformation of normal cells to malignant ones may once give us a chance to cure erbB2 over-expressing breast cancer.
Collapse
Affiliation(s)
- P Nagy
- Hungarian Academy of Sciences, Biophysical Workgroup, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
28
|
Abstract
A number of genes have been implicated in breast cancer development, yet few have been demonstrated to play causative roles in mammary tumor formation. The advent of transgenic mouse and embryonic stem cell technologies now permits manipulation of the mouse genome in such a way as to temporally and spatially control a gene product's expression. Thus, the basic researcher now can directly assess the involvement of particular genes in tumorigenesis and disease progression and, in the process, to develop mouse models of human genetic disease. The utility of such technologies is emphasized in transgenic mice expressing genes thought to play important roles in the initiation and progression of mammary carcinomas. As these transgenic strains have been the subject of several reviews, here we focus on two mouse mammary tumor models, Polyomavirus middle T antigen and the Neu/ErbB-2 receptor tyrosine kinase, which are most amenable to study specific signaling pathways in process of mammary tumorigenesis.
Collapse
Affiliation(s)
- D L Dankort
- Department of Biology, Institute for Molecular Biology and Biotechnology, McMaster University, 1280 Main St. West, Hamilton, Ontario, Canada, L8S 4K1
| | | |
Collapse
|
29
|
Abstract
The frequency of c-erb-B2 expression, clinical correlates and treatment outcome was investigated in 92 patients with metastatic breast cancer. Positive c-erb-B2 immunostaining was found in 24/92 (26%) of tumours. There was a statistically significant inverse correlation between c-erb-B2 expression and ER status. There was also a significant inverse correlation between c-erb-B2 expression and tumour free interval. c-erb-B2 expression had no influence on response to treatment with tamoxifen among patients co-expressing both c-erb-B2 and estrogen receptor (ER). Following chemotherapy (CAF) treatment there was a trend to a lower response rate among c-erb-B2+ as compared to c-erb-B2- patients. However, more c-erb-B2+ patients had received prior adjuvant chemotherapy, and when this factor was included in a multivariate analysis only prior adjuvant chemotherapy treatment predicted for response to CAF chemotherapy for metastatic disease. Time to treatment failure (TTF) was significantly shorter among cerb B2+ as compared to c-erb-B2- patients. The current study suggests that the c-erb-B2 expression is found at similar frequency in metastatic as in primary breast cancer. Although c-erb-B2 expression did not predict for response to either hormonal therapy or to chemotherapy for metastatic disease, patients with c-erb-B2+ metastatic lesions appear to have a more aggressive clinical course.
Collapse
Affiliation(s)
- W R Bezwoda
- Division of Clinical Haematology and Medical Oncology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
30
|
Aguilar Z, Akita RW, Finn RS, Ramos BL, Pegram MD, Kabbinavar FF, Pietras RJ, Pisacane P, Sliwkowski MX, Slamon DJ. Biologic effects of heregulin/neu differentiation factor on normal and malignant human breast and ovarian epithelial cells. Oncogene 1999; 18:6050-62. [PMID: 10557094 DOI: 10.1038/sj.onc.1202993] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The heregulins are a family of ligands with ability to induce phosphorylation of the p185HER-2/neu receptor. Various investigators have reported a variety of responses of mouse and human breast and ovarian cells to this family of ligands including growth stimulation, growth inhibition, apoptosis and induction of differentiation in cells expressing the HER-2/neu receptor. Some of the disparity in the literature has been attributed to variations in the cell lines studied, ligand dose applied, methodologies utilized or model system evaluated (i.e. in vitro or in vivo). To evaluate the effects of heregulin on normal and malignant human breast and ovarian epithelial cells expressing known levels of the HER-2/neu receptor, this report presents the use of several different assays, performed both in vitro and in vivo, in vitro proliferation assays, direct cell counts, clonogenicity under anchorage-dependent and anchorage-independent conditions, as well as the in vivo effects of heregulin on human cells growing in nude mice to address heregulin activity. Using a total of five different biologic assays in nine different cell lines, across two different epithelia and over a one log heregulin dose range, we obtained results that clearly indicate a growth-stimulatory role for this ligand in human breast and ovarian epithelial cells. We find no evidence that heregulin has any growth-inhibitory effects in human epithelial cells. We also quantitated the amount of each member of the type I receptor tyrosine kinase family (RTK I, i.e. HER-1, HER-2, HER-3 and HER-4) in the cell lines employed and correlated this to their respective heregulin responses. These data demonstrate that HER-2/neu overexpression itself affects the expression of other RTK I members and that cells expressing the highest levels of HER-2/neu have the greatest response to HRG.
Collapse
Affiliation(s)
- Z Aguilar
- Division of Hematology and Oncology, Department of Medicine, UCLA School of Medicine, Los Angeles, California, CA 90095 USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Breast carcinomas express high levels of ErbB receptors and their ligands, and their overexpression has been associated with a more aggressive clinical behavior. For these reasons therapies directed at these receptors have the potential to be useful anti-cancer treatments. A series of monoclonal antibodies (MAbs)3 directed against the EGF (ErbB1) receptor and the closely related HER2/Neu (ErbB2) receptor are currently under evaluation. These MAbs have shown promising preclinical activity and "chimeric" and "humanized" MAbs have been produced in order to obviate the problem of host immune reactions. These antibodies are currently being tested in clinical trials either alone or in combination with chemotherapeutic agents. Clinical activity with one of these antibodies, trastuzumab, a humanized anti-ErbB2 MAb, has been documented in patients with breast cancer in a series of clinical trials and has recently been approved for the therapy of patients with metastatic ErbB2 overexpressing breast cancer. In addition to antibodies, compounds that inhibit receptor tyrosine kinases have shown significant preclinical activity and are currently being evaluated in the clinic.
Collapse
Affiliation(s)
- J Albanell
- Medical Oncology Service, Hospital Universitari Vall d' Hebron, Barcelona, Spain
| | | |
Collapse
|
32
|
Fernandes AM, Hamburger AW, Gerwin BI. Production of epidermal growth factor related ligands in tumorigenic and benign human lung epithelial cells. Cancer Lett 1999; 142:55-63. [PMID: 10424781 DOI: 10.1016/s0304-3835(99)00166-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We recently demonstrated that human lung epithelial cells, overexpressing ErbB-2, formed tumors in nude mice only when high levels of transforming growth factor alpha (TGFalpha) were produced. Cells transfected with a TGFalpha antisense vector failed to form tumors in nude mice. In order to further evaluate the importance, for tumorigenicity, of TGFalpha and its stimulation of ErbB family signalling, the production of other EGF family growth factors by these human lung epithelial cells was studied. We demonstrate for the first time that both tumorigenic and non-tumorigenic human lung epithelial cells produced, in addition to TGFalpha, amphiregulin, betacellulin, heparin-binding EGF and heregulin. These data suggest that human lung epithelial cells have the potential for multifactorial modulation of ErbB receptor family signalling through control of ligand as well as receptor production. In this system, the probable importance of TGFalpha-stimulated signaling for tumorigenicity is supported by its 13-fold higher production in tumorigenic as compared with non-tumorigenic cells and the 2-fold or lower differences observed in production of the other epidermal growth factor (EGF) family ligands.
Collapse
Affiliation(s)
- A M Fernandes
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
33
|
Braun S, Pantel K. Prognostic significance of micrometastatic bone marrow involvement. Breast Cancer Res Treat 1999; 52:201-16. [PMID: 10066083 DOI: 10.1023/a:1006164914610] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The present review focuses on the methodology and clinical significance of new diagnostic approaches to identify micrometastatic breast cancer cells present in bone marrow (BM), as a frequent site of overt metastases. Using monoclonal antibodies (mAbs) to epithelial cytokeratins (CK) or tumor-associated cell membrane glycoproteins, individual carcinoma cells can be detected on cytologic BM preparations at frequencies of 10(-5) to 10(-6). Prospective clinical studies have shown that the presence of these immunostained cells is prognostically relevant with regard to relapse-free and overall survival. The current interest in autologous bone marrow transplantation in patients with solid tumors further underlines the need for screening methods that allow the detection of minute numbers of residual tumor cells in the transplant. Although the development of new molecular detection methods based on the amplification of a marker mRNA species by the polymerase chain reaction technique is a very exciting area of research, the clinical significance of this approach needs to be demonstrated in prospective studies. The immunocytochemical assays may be, therefore, used to improve tumor staging with potential consequences for adjuvant therapy. Another promising clinical application is monitoring the response of micrometastatic cells to adjuvant therapies, which, at present, can only be assessed retrospectively after an extended period of clinical follow-up. The extremely low frequency of BM tumor cells greatly hampers approaches to obtain more specific information on their biological properties. The available data indicate that these cells represent a selected population of cancer cells which, however, still express a considerable degree of heterogeneity with regard to the expression of MHC class I antigens, adhesion molecules (EpCAM), growth factor receptors (EGF receptor, erb-B2, transferrin receptor), or proliferation-associated markers (Ki-67, p120). Regardless of the detection technique applied, there is an urgent demand for large multicentre trials, in which standardized methods are related to specified clinical outcomes.
Collapse
Affiliation(s)
- S Braun
- I. Frauenklinik, Klinikum Innenstadt, and Institut für Immunologie, Ludwig-Maximilians-Universität, München, Germany
| | | |
Collapse
|
34
|
Xie W, Duan R, Safe S. Estrogen induces adenosine deaminase gene expression in MCF-7 human breast cancer cells: role of estrogen receptor-Sp1 interactions. Endocrinology 1999; 140:219-227. [PMID: 9886828 DOI: 10.1210/endo.140.1.6394] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adenosine deaminase (ADA) gene expression is induced by 17beta-estradiol (E2) in MCF-7 human breast cancer cells, whereas the antiestrogens 4'-hydroxytamoxifen and ICI 182,780 exhibit partial estrogen receptor (ER) agonist/antagonist and antagonist activities, respectively. Previous studies have shown that the -211 to +11 region of the ADA gene promoter contains six GC-rich sites (I-VI) that bind Sp1 protein, and these elements are required for high basal expression. In transient transfection studies with pADA211, which contains the -211 to +11 ADA gene promoter linked to a bacterial chloramphenicol acetyl transferase (CAT) reporter gene, E2 and tamoxifen (but not ICI 182,780) induced CAT activity. Ligand-induced transactivation was observed only in cells cotransfected with expression plasmids for wild-type ER or HE11, which does not contain the DNA-binding domain of the ER. Cotransfection with HE15 and HE19, which contain the DNA-binding domain and activation function-1 (AF-1) and AF-2 of the ER, respectively, did not result in E2-induced activity. Subsequent deletion analysis of the ADA gene promoter showed that Sp1 binding site IV (-79 to -73) was primarily responsible for hormone responsiveness. ER activation of ADA gene expression is another example of an E2-induced gene that is dependent on ER/Sp1 interactions with a site-specific GC-rich motif.
Collapse
Affiliation(s)
- W Xie
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station 77843-4466, USA
| | | | | |
Collapse
|
35
|
Klapper LN, Kirschbaum MH, Seta M, Yarden Y. Biochemical and Clinical Implications of the ErbB/HER Signaling Network of Growth Factor Receptors. Adv Cancer Res 1999. [DOI: 10.1016/s0065-230x(08)60784-8] [Citation(s) in RCA: 349] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
36
|
Boente MP, Berchuck A, Whitaker RS, Kalén A, Xu FJ, Clarke-Pearson DL, Bell RM, Bast RC. Suppression of diacylglycerol levels by antibodies reactive with the c-erbB-2 (HER-2/neu) gene product p185c-erbB-2 in breast and ovarian cancer cell lines. Gynecol Oncol 1998; 70:49-55. [PMID: 9698473 DOI: 10.1006/gyno.1998.5050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Seven of 10 murine monoclonal antibodies reactive with the extracellular domain of p185c-erbB-2 inhibited the anchorage independent growth of the SKBr3 breast cancer cell line that overexpressed p185c-erbB-2. Significant inhibition (56-72%) of diacylglycerol (DAG) levels (P < 0.0001) was observed with the 10 antibodies that inhibited SKBr3 growth (RC1, NB3, RC6, PB3, 741F8, DB5, ID5), whereas the 3 antibodies (TA1, 520C9, 454C11) that failed to inhibit SKBr3 growth also failed to affect DAG levels. Thus, DAG levels correlated with antibody-mediated growth regulation for each of the 10 monoclonal reagents. Antibody-induced inhibition of anchorage-independent growth of SKBr3 could be reversed by incubation with phorbol myristate acetate. The ID5 antibody inhibited growth of the SKBr3, SKOv3, and OVCA 432 tumor cell lines, but not of OVCA 420, OVCA 429, and OVCA 433. DAG levels were significantly decreased after ID5 treatment of the SKBr3 and SKOv3 cell lines, but not the OVCA 420, OVCA 429, and OVCA 433 lines. In the 432 line, there was a decrease which did not reach significance. Consequently, changes in DAG levels correlated with growth regulation in 5 of 6 breast and ovarian carcinoma cell lines tested with a trend toward correlation in the sixth. Decreases in DAG may be one mediator of the growth regulatory signals produced by anti-p185c-erbB-2 antibodies.
Collapse
Affiliation(s)
- M P Boente
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Yoo JY, Hamburger AW. Changes in heregulin beta1 (HRGbeta1) signaling after inhibition of ErbB-2 expression in a human breast cancer cell line. Mol Cell Endocrinol 1998; 138:163-71. [PMID: 9685225 DOI: 10.1016/s0303-7207(98)00004-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Specific biological responses to the erbB3-erbB4 ligand heregulin (HRG) have been postulated to be due to the formation of heterodimers of those receptors with erbB2. To test the role that erbB2 plays in the response to HRG in a human breast carcinoma cell line, antisense oriented erbB2 was stably transfected into AU565 cells. In the absence of HRG, inhibition of erbB2 expression slowed cell growth, leading to accumulation of cells in the G2/M phase, and suppressed colony growth in soft agar. Low concentrations of HRG induced cell proliferation in both the erbB2-nonexpressing cells and the parental AU565 cells. In contrast, high concentrations of HRG failed to induce differentiation of the erbB2-nonexpressing cells as compared with the parental cells. ErbB3 expression was significantly decreased in the erbB2 nonexpressing cells. ErbB3 was constitutively tyrosine phosphorylated in both the parental AU565 cells and in the erbB2 nonexpressing cells. HRG further increased tyrosine phosphorylation of erbB3 with a maximum response at 1 ng/ml of HRG in erbB2 nonexpressing cells, as compared with 10 ng/ml of HRG in AU565 cells. This finding suggested that the biochemical responsiveness of erbB3 to HRG was changed, but not abrogated, by inhibition of erbB2 expression. These results suggest that inhibition of erbB2 expression modulates, but does not abolish, HRG mediated signal transduction pathways in a human breast cancer cell line.
Collapse
Affiliation(s)
- J Y Yoo
- Molecular and Cellular Biology Program, University of Maryland, School of Medicine, Baltimore 21201, USA
| | | |
Collapse
|
38
|
Tzahar E, Yarden Y. The ErbB-2/HER2 oncogenic receptor of adenocarcinomas: from orphanhood to multiple stromal ligands. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1377:M25-37. [PMID: 9540810 DOI: 10.1016/s0304-419x(97)00032-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Extensive clinical and biochemical evidence implicates ErbB-2, a transmembrane tyrosine kinase related to growth factor receptors, in the development, metastasis, and resistance to therapy of multiple, common human carcinomas. Previous attempts to uncover an ErbB-2-specific ligand led to isolation of the neuregulin (NRG) family, but these ligands, like all other growth factors with an EGF-like motif, only indirectly active ErbB-2. On the other hand, biochemical and genetic evidence suggest a non-autonomous function of ErbB-2 in an interactive ErbB signaling network. Accordingly, the oncoprotein acts as a shared signaling subunit of primary growth factor receptors. By stabilizing heterodimers with other ErbB proteins, ErbB-2 prolongs and enhances signal transduction by a large group of stroma-derived growth factors. Furthermore, we have proposed a model in which all ErbB-2 ligands are bivalent and bind to ErbB-2 with low affinity, following high affinity binding to a primary receptor with which ErbB-2 is heterodimerized. Thus the presence of ErbB-2 in relevant ErbB heterodimeric structures on the surfaces of certain epithelial tumor cells can amplify signals arising from the binding of stromal ErbB ligands. This effect, in turn, may promote the growth of carcinoma cells.
Collapse
Affiliation(s)
- E Tzahar
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| | | |
Collapse
|
39
|
Chan HS, Ling V. Anti-P-glycoprotein antibody C219 cross-reactivity with c-erbB2 protein: diagnostic and clinical implications. J Natl Cancer Inst 1997; 89:1473-6. [PMID: 9337338 DOI: 10.1093/jnci/89.20.1473] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
40
|
Kiyokawa N, Lee EK, Karunagaran D, Lin SY, Hung MC. Mitosis-specific negative regulation of epidermal growth factor receptor, triggered by a decrease in ligand binding and dimerization, can be overcome by overexpression of receptor. J Biol Chem 1997; 272:18656-65. [PMID: 9228035 DOI: 10.1074/jbc.272.30.18656] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The function of epidermal growth factor receptor (EGFR) was found to be negatively regulated in M phase in which it showed less phosphotyrosine content and reduced intrinsic kinase activity accompanied by retarded electrophoretic mobility owing to total hyperphosphorylation. Ligand-induced autophosphorylation and downstream signaling of EGFR were tightly suppressed in M phase due to a decrease in ligand binding affinity and the inability of epidermal growth factor (EGF) to induce receptor dimerization. There was no change in the number of surface-exposed EGF receptors between G0/G1 and M phases of the cell cycle. Hyperphosphorylation (due to serine and/or threonine phosphorylation) correlates with the unresponsiveness of cells to EGF-mediated stimulation of tyrosine phosphorylation in cells that express the normal or basal level of EGFR. This M phase-specific negative regulation was overcome by overexpression of EGFR, which was responsive to ligand throughout the cell cycle and revealed ligand-induced signaling in the M phase. These findings indicate that EGFR does not respond to ligand stimulation in M phase and suggest that a negative regulation of ligand-receptor interactions in M phase may control the normal function of receptor tyrosine kinase and that receptor overexpression will disrupt this cell cycle-dependent regulation of receptor tyrosine kinases.
Collapse
Affiliation(s)
- N Kiyokawa
- Department of Tumor Biology, Breast Cancer Basic Research Program, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Advances in molecular biology have facilitated the recent investigation of gynecological malignancies. The presence of certain oncogenes within gynecological tumors indicates that transformation may be associated with genetic alteration of normal regulatory processes. This paper reviews several oncogenes that have been implicated in the transformation of gynecological tissues.
Collapse
Affiliation(s)
- G L Maxwell
- Department of Obstetrics and Gynecology, William Beaumont Army Medical Center, El Paso, Texas, USA
| | | |
Collapse
|
42
|
Kuhn PE, Miller MW. c-neu oncoprotein in developing rostral cerebral cortex: relationship to epidermal growth factor receptor. J Comp Neurol 1996; 372:189-203. [PMID: 8863125 DOI: 10.1002/(sici)1096-9861(19960819)372:2<189::aid-cne3>3.0.co;2-#] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The c-neu oncoprotein, p185c-neu, is a transmembrane tyrosine kinase that shares structural similarities with the receptor for epidermal growth factor (EGFr). We used immunoblots, immunoprecipitation, and immunohistochemistry 1) to test the hypothesis that p185c-neu and EGFr are coordinately expressed in central nervous system tissue and 2) to assess the spatiotemporal expression of both the c-neu oncoprotein and EGFr in the rostral cerebral cortex. In nondenaturing gels, anti-c-neu antibody identified high molecular weight proteins (about 300-400 kDa) that were reduced by EDTA to a molecular weight of 180-200 kDa. Sodium dodecylsulfate polyacrylamide gel electrophoresis broke down this protein into an array of smaller peptides, which were expressed prenatally, transiently during the first three postnatal weeks, or in the adult. Perinatally, c-neu immunoreactivity was evident in subplate neurons, ascending processes of neurons in the cortical plate, and ventricular zone cells. During the second postnatal week, cells throughout cortex expressed somatodendritic immunostaining, but, in the adult, c-neu immunoreactivity was expressed only by pyramidal neurons in layer V and by glia in the white matter and ependyma. EGFr-positive proteins behaved in the nondenaturing gels as did c-neu-positive oncoproteins, suggesting that both proteins naturally formed dimers. This contention was supported by the EGFr-or c-neu immunolabeling of tissue that was previously immunoprecipitated with anti-c-neu or anti-EGFr, respectively. The pattern of EGFr immunolabeling in the developing and mature cortex was virtually identical to that described for c-neu immunoreactivity. Cortical neurons express the c-neu oncoprotein and EGFr, probably as heterodimers. The specific immunolabeling of layer V neurons in the adult cortex with anti-c-neu and anti-EGFr suggests that the p185c-neu ligand and EGF regulate the activity of corticofugal systems. The expression of different c-neu- and EGFr-positive peptides is developmentally defined and may be related to specific ontogenetic events.
Collapse
Affiliation(s)
- P E Kuhn
- Program in Cell and Developmental Biology, Rutgers University, Piscataway, New Jersey 08854-1059, USA
| | | |
Collapse
|
43
|
Jinno H, Ueda M, Enomoto K, Ikeda T, Kyriakos P, Kitajima M. Effectiveness of an adriamycin immunoconjugate that recognizes the C-erbB-2 product on breast cancer cell lines. Surg Today 1996; 26:501-7. [PMID: 8840431 DOI: 10.1007/bf00311556] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Adriamycin (ADM) was chemically conjugated to a murine monoclonal antibody, A0011, which recognizes the c-erbB-2 product, via a disulfide bond using N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP) and 2-iminothiolane (2-IT). The molar ratio of ADM to the monoclonal antibody ranged from 15:1 to 25:1 and enzyme-linked immunosorbent assay (ELISA) showed that the binding activity of the conjugate was almost retained. We compared the efficacy of A0011 alone, ADM alone, the A0011-ADM conjugate, against the human breast cancer cell lines SK-BR-3, MDA-MB-361, MCF-7, and BT-20. The A0011-ADM conjugate was observed to be ten times more cytotoxic to the cell lines overexpressing the c-erbB-2 product, namely, SK-BR-3 and MDA-MB-361, than free ADM, but it showed weak cytotoxicity against the cell lines with a low level of c-erbB-2 product expression, namely, MCF-7 and BT-20. However, free A0011 and nonspecific murine IgM-ADM conjugate showed no cytotoxicity toward any of the four cell lines, while the addition of a tenfold molar excess of A0011 inhibited conjugate cytotoxicity. These data suggest that conjugate cytotoxicity is antibody-mediated. Moreover, conjugate cytotoxicity at 10(-6)M was correlated with antigen volume, and the data were fitted to the regression equation y = -11.63logX + 116.38 where the correlation coefficient = 0.950. Our results indicate that targeting therapy aiming at the c-erbB-2 product may be useful in the treatment of breast cancers overexpressing the c-erbB-2 product.
Collapse
Affiliation(s)
- H Jinno
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Karunagaran D, Tzahar E, Beerli RR, Chen X, Graus-Porta D, Ratzkin BJ, Seger R, Hynes NE, Yarden Y. ErbB-2 is a common auxiliary subunit of NDF and EGF receptors: implications for breast cancer. EMBO J 1996. [PMID: 8617201 DOI: 10.1002/j.1460-2075.1996.tb00356.x] [Citation(s) in RCA: 424] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Overexpression of the erbB-2 gene contributes to aggressive behavior of various human adenocarcinomas, including breast cancer, through an unknown molecular mechanism. The erbB-2-encoded protein is a member of the ErbB family of growth factor receptors, but no direct ligand of ErbB-2 has been reported. We show that in various cells ErbB-2 can form heterodimers with both EGF receptor (ErbB-1) and NDF receptors (ErbB-3 and ErbB-4), suggesting that it may affect the action of heterologous ligands without the involvement of a direct ErbB-2 ligand. This possibility was addressed in breast cancer cells through either overexpression of ErbB-2 or by blocking its delivery to the cell surface by means of an endoplasmic reticulum-trapped antibody. We report that ErbB-2 overexpression enhanced binding affinities to both EGF and NDF, through deceleration of ligand dissociation rates. Likewise, removal of ErbB-2 from the cell surface almost completely abolished ligand binding by accelerating dissociation of both growth factors. The kinetic effects resulted in enhancement and prolongation of the stimulation of two major cytoplasmic signaling pathways, namely: MAP kinase (ERK) and c-Jun kinase (SAPK), by either ligand. Our results imply that ErbB-2 is a pan-ErbB subunit of the high affinity heterodimeric receptors for NDF and EGF. Therefore, the oncogenic action of ErbB-2 in human cancers may be due to its ability to potentiate in trans growth factor signaling.
Collapse
Affiliation(s)
- D Karunagaran
- Departement of Chemical Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Haapasalo H, Hyytinen E, Sallinen P, Helin H, Kallioniemi OP, Isola J. c-erbB-2 in astrocytomas: infrequent overexpression by immunohistochemistry and absence of gene amplification by fluorescence in situ hybridization. Br J Cancer 1996; 73:620-3. [PMID: 8605096 PMCID: PMC2074346 DOI: 10.1038/bjc.1996.107] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Recent studies suggest that aberrations of c-erbB-2 may be involved in astrocytic brain tumours. We screened immunohistochemically c-erbB2 protein (p185) expression in 94 astrocytic grade 1-4 neoplasms of the brain. The amplification of the c-erbB-2 oncogene was investigated in protein overexpression cases by dual colour fluorescence in situ hybridisation (FISH). p185 overexpression was correlated with p53 and epidermal growth factor receptor (EGFR) expression, as well as with clinicopathological features. Only two anaplastic (grade 3) astrocytomas and one glioblastoma (grade 4) showed overexpression of p185 protein by immunohistochemistry (monoclonal MAb1 antibody TA250), whereas none of the grade 1-2 astrocytomas was positive. Interestingly, the expression of p185 was confined solely to the cytoplasm of neoplastic astrocytic cells and not to the cell membranes as found in malignancies with amplification of the c-erbB-2 oncogene. Two of the three overexpression cases were also positive by EGFR. No amplification of the c-erbB-2 gene was observed by FISH in the three tumours with immunohistochemical p185 overexpression or seven weakly positive/negative tumours. In conclusion, our results suggest that p185 overexpression is infrequent in astrocytomas, that it is of no important diagnostic or prognostic value and that c-erbB-2 oncogene amplification is not seen in the few cases in which there is overexpression.
Collapse
Affiliation(s)
- H Haapasalo
- Department of Pathology, Tampere University Hospital, Finland
| | | | | | | | | | | |
Collapse
|
46
|
Cohen BD, Green JM, Foy L, Fell HP. HER4-mediated biological and biochemical properties in NIH 3T3 cells. Evidence for HER1-HER4 heterodimers. J Biol Chem 1996; 271:4813-8. [PMID: 8617750 DOI: 10.1074/jbc.271.9.4813] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The EGF receptor family of tyrosine kinase growth factor receptors is expressed in a variety of cell types and has been implicated in the progression of certain human adenocarcinomas. The most recent addition to this family of receptors, HER4, was expressed in NIH 3T3 cells to determine its biological and biochemical characteristics. Cells expressing HER4 were responsive to heregulin beta2 as demonstrated by an increase in HER4 tyrosine phosphorylation and ability to form foci on a cell monolayer. HER4 exhibited in vitro kinase activity and was able to phosphorylate the regulatory subunit of phosphatidylinositol 3-kinase and SHC. Peptide competition studies identified tyrosine 1056 of HER4 as the phosphatidylinositol 3-kinase binding site and tyrosines 1188 and 1242 as two potential SHC binding sites. Interestingly, transfection of HER4 into NIH 3T3 cells conferred responsiveness to EGF with respect to colony formation in soft agar. It was also found that in response to heregulin beta2, endogenous murine HER1 or transfected human HER1 became phosphorylated when HER4 was present. This demonstrates that HER1 and HER4 can exist in a heterodimer complex and likely activate each other by transphosphorylation.
Collapse
Affiliation(s)
- B D Cohen
- Molecular Immunology Department, Bristol-Myers Squibb, Pharmaceutical Research Institute, Seattle, Washington 98121, USA
| | | | | | | |
Collapse
|
47
|
Bacus SS, Chin D, Yarden Y, Zelnick CR, Stern DF. Type 1 receptor tyrosine kinases are differentially phosphorylated in mammary carcinoma and differentially associated with steroid receptors. THE AMERICAN JOURNAL OF PATHOLOGY 1996; 148:549-58. [PMID: 8579117 PMCID: PMC1861670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The neu/erbB-2/HER-2 proto-oncogene is amplified and/or overexpressed in up to 30% of mammary carcinomas and has been variably correlated with poor prognosis. The signaling activity of the encoded receptor tyrosine kinase is regulated by interactions with other type 1 receptors and their ligands. We have used a novel approach, phosphorylation-sensitive anti-Neu antibodies, to quantify signaling by Neu and epidermal growth factor receptor in a panel of frozen sections of mammary carcinoma specimens. We also determined the relationship of Neu, phosphorylated Neu (and epidermal growth factor receptor), and phosphotyrosine to the expression of Neu-related receptors (epidermal growth factor receptor, HER-3, and HER-4) and to prognostic factors (estrogen and progesterone receptor). We found that tyrosine phosphorylation of Neu (and hence signaling activity) is highly variable among mammary carcinomas. Neu and HER-4 were associated with divergent correlates, suggesting that they have profoundly different biological activities. These results have implications for etiology of mammary carcinoma for clinical evaluation of mammary carcinoma patients, and for development of Neu-targeted therapeutic strategies.
Collapse
Affiliation(s)
- S S Bacus
- Advanced Cellular Diagnostics, Inc., Elmhurst, Illinois, USA
| | | | | | | | | |
Collapse
|
48
|
Lupu R, Cardillo M, Cho C, Harris L, Hijazi M, Perez C, Rosenberg K, Yang D, Tang C. The significance of heregulin in breast cancer tumor progression and drug resistance. Breast Cancer Res Treat 1996; 38:57-66. [PMID: 8825123 DOI: 10.1007/bf01803784] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The erbB-2 receptor plays an important role in the prognosis of breast cancer and is expressed at high levels in nearly 30% of tumors in breast cancer patients. While evidence accumulates to support the relationship between erbB-2 overexpression and poor overall survival in human breast cancer, understanding of the biological consequence(s) of erbB-2 overexpression remains elusive. The discovery of heregulin has allowed us to identify a number of related but distinct biological endpoints which appear responsive to signal transduction through the erbB-2/4 receptor. These endpoints of growth, invasiveness, and differentiation have clear implications for the emergence, maintenance, and/or control of malignancy, and represent established endpoints in the assessment of malignant progression in human breast cancer. Preliminary studies in vitro have shown that heregulin induces a biphasic growth effect on cells with erbB-2 overexpression. Interestingly, we observed that expression of heregulin correlates with a more aggressive/invasive, vimentin-positive phenotype in breast cancer cells lines. Therefore, we have postulated that heregulin is involved in breast cancer tumor progression. We have shown that heregulin induces in vitro chemoinvasion and chemotaxis of breast cancer cells as well as growth in an anchorage dependent and independent manner. Interestingly, a heregulin neutralizing antibody inhibits chemotaxis and results in cell growth inhibition and blockade of the invasive phenotype. Strikingly, genetically engineered cells which constitutively express heregulin demonstrate critical phenotypic changes that are associated with a more aggressive phenotype. Specifically, these cells are no longer dependent on estrogen for growth and are resistant to tamoxifen in vitro and in vivo, and moreover these cells metastasize to lymph nodes in athymic nude mice. These tumors appear to have lost bcl-2 expression as compared with the control tumors. In addition, presumably by activation/regulation of topoisomerase II, the heregulin-transfected cells become exquisitely sensitive to doxorubicin and VP-16. Clearly, mechanistic aspects of the erbB-2/4 and heregulin interaction need to be understood from a therapeutic standpoint which could provide additional insights into synergistic treatments for certain patients, or improve treatment regimens for a large number of women. The study of heregulin and its co-expression with erbB-2/4 receptor and the assessment of its involvement in the progression from the in situ stage of breast tumors to the invasive one will additionally increase the relevance of heregulin as a prognostic/diagnostic factor. We believe that our studies provide new insights into breast cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- R Lupu
- Vincent T. Lombardi Cancer Research Center, Georgetown University Medical Center, Washington DC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ethier SP. Human breast cancer cell lines as models of growth regulation and disease progression. J Mammary Gland Biol Neoplasia 1996; 1:111-21. [PMID: 10887484 DOI: 10.1007/bf02096306] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The routine isolation and culture of human breast cancer cells from patient samples has been a goal of breast cancer cell biologists for over 30 years. Despite extensive work in this area and the development of many human breast cancer cell lines, the proportion of patient samples that give rise to immortalized breast cancer cell lines is still disappointingly low. The majority of human breast cancer cell lines that have been established were isolated many years ago and have been grown continuously under poorly defined culture conditions. These cell lines have been useful for studies of the estrogen receptor biology in human breast cancer cells, in identifying growth factors synthesized by breast cancer cells, and for the characterization of genetic alterations in oncogenes and tumor suppressor genes present in these cells. More recently, tissue culture methods have improved, resulting in the ability to culture routinely normal human mammary epithelial cells of specific lineages and this has resulted in the development of new human breast cancer cell lines. The ability to isolate and culture normal and neoplastic human mammary epithelial cells under similar culture conditions has improved these models dramatically and has resulted in the identification of altered cellular phenotypes of human breast cancer cells.
Collapse
Affiliation(s)
- S P Ethier
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor 48109-0582, USA.
| |
Collapse
|
50
|
Pantel K, Braun S, Passlick B, Schlimok G. Minimal residual epithelial cancer: diagnostic approaches and prognostic relevance. PROGRESS IN HISTOCHEMISTRY AND CYTOCHEMISTRY 1996; 30:1-60. [PMID: 8724405 DOI: 10.1016/s0079-6336(96)80013-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- K Pantel
- Institut für Immunologie, Ludwig-Maximilians-Universität, München, Germany
| | | | | | | |
Collapse
|