201
|
Kong J, Diao Z, Deng X. Inhibition of IGF receptor signaling and hepatoma cell growth by an antibody to ligand oligopeptide of receptor. Growth Factors 2008; 26:35-43. [PMID: 18365877 DOI: 10.1080/08977190801987315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Research on insulin-like growth factor (IGF) system have shown it to be potent mitogen for hepatoma cells and made it an attractive therapeutic target. But little strategy has been reported to date on targeting and sequestrating IGF against hepatoma. This study is based on the capability of ligand oligopeptide (LOP) to recognize IGF receptor with high efficiency, which is over-expressed on some hepatoma cells. We have been hypothesizing that antibody to LOP would mimic the extracellular ligand-binding domain of IGF receptor and inhibit receptor signaling and cell proliferation. Gene encoding for LOP [E5 (EPFRSPDLALETYG)] of IGF receptor was inserted into HBc carrier for expression in Escherichia coli. The monoclonal antibody (mAb) specific LOP potently inhibited signal transduction mediated by the IGF-IR interaction with IGF-I. Furthermore, it exhibited 47% inhibitory rate of soft agar colony formation and also induced apoptosis. These results indicate an anti-hepatoma potential of the mAb to an LOP of IGF receptor could block the activation of receptor and downstream signaling pathways, and suppress the biological effects mediated by receptor.
Collapse
Affiliation(s)
- Jing Kong
- School of Life Science and Chemical Engineering, Huaiyin Institute of Technology, Huaian, People's Republic of China.
| | | | | |
Collapse
|
202
|
Sun H, Baserga R. The role of insulin receptor substrate-1 in transformation by v-src. J Cell Physiol 2008; 215:725-32. [PMID: 18064649 DOI: 10.1002/jcp.21352] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Hongzhi Sun
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
203
|
Nagase H, Bryson S, Fee F, Balmain A. Multigenic control of skin tumour development in mice. CIBA FOUNDATION SYMPOSIUM 2007; 197:156-68; discussion 168-80. [PMID: 8827373 DOI: 10.1002/9780470514887.ch9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Different inbred mouse strains vary greatly in their susceptibility to tumour development in a variety of tissues. Intraspecific and interspecific crosses can, therefore, be used to map the loci that control this predisposition. Crosses of Mus musculus with Mus spretus are highly resistant to tumour development in the skin, liver, lung and lymphoid system. M. spretus, therefore, has dominantly acting resistance loci, which we have attempted to map. More than 350 interspecific backcross mice were followed for 18 months to assess susceptibility to development of chemically induced papillomas and carcinomas. The results were analysed using a combination of MAPMAKER/QTL analysis and multiple regression analysis for the determination of linkage in multigenic quantitative traits. The results showed clearly that at least three genes on chromosomes 5 and 7 control resistance to tumour development. Importantly, some genes confer resistance to benign tumours but they have relatively little effect on malignant progression. This suggests the existence of different classes of benign tumours: those that are capable of tumour progression and those that have only a very low probability of becoming malignant. Identification of these genes will improve our understanding of mechanisms of carcinogenesis and may provide a novel route to the identification of "low-penetrance' human tumour susceptibility genes.
Collapse
Affiliation(s)
- H Nagase
- CRC Beatson Laboratories, Department of Medical Oncology, University of Glasgow, UK
| | | | | | | |
Collapse
|
204
|
Maor S, Yosepovich A, Papa MZ, Yarden RI, Mayer D, Friedman E, Werner H. Elevated insulin-like growth factor-I receptor (IGF-IR) levels in primary breast tumors associated with BRCA1 mutations. Cancer Lett 2007; 257:236-43. [PMID: 17766039 DOI: 10.1016/j.canlet.2007.07.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 07/25/2007] [Accepted: 07/26/2007] [Indexed: 11/28/2022]
Abstract
The insulin-like growth factors (IGFs) play a pivotal role in breast cancer. Inherited predisposition to breast and ovarian cancer is associated with germline BRCA1/BRCA2 mutations. To evaluate the impact of BRCA1 mutations on IGF-IR gene expression, we performed an immunohistochemical analysis of IGF-IR in primary breast tumors from BRCA1 mutation carriers and non-carriers. Results obtained revealed a significant elevation in IGF-IR levels in tumors from BRCA1 mutation carriers compared with non-carriers. To assess the potential inhibitory role of BRCA1 on IGF-IR levels, we infected the BRCA1-deficient HCC1937 cell line with a BRCA1-encoding adenoviral vector. Results of Western blots showed that BRCA1 induced a large reduction in endogenous IGF-IR levels. Furthermore, results of chromatin immunoprecipitation assays indicated that the mechanism of action of BRCA1 involves interaction with Sp1, a potent transactivator of the IGF-IR gene. In conclusion, our data suggests that the IGF-IR gene is a physiologically relevant downstream target for BRCA1 action.
Collapse
Affiliation(s)
- Sharon Maor
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | | | | | |
Collapse
|
205
|
Wu A, Chen J, Baserga R. Nuclear insulin receptor substrate-1 activates promoters of cell cycle progression genes. Oncogene 2007; 27:397-403. [PMID: 17700539 DOI: 10.1038/sj.onc.1210636] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The insulin receptor substrate-1 (IRS-1) is a docking protein of the insulin-like growth factor-1 (IGF-1) receptor and of the insulin receptor. IRS-1 sends a strong mitogenic, anti-apoptotic signal and plays an important role in cell transformation and cancer. IRS-1 translocates to nuclei of cells, where it increases the activity of the rDNA, c-myc and cyclin D1 promoters. We show, by chromatin immunoprecipitation, occupancy by IRS-1 of the same promoters. Both promoter activation and promoter occupancy are IGF-1-dependent. In cells that respond to IGF-1 but in which IRS-1 does not translocate to nuclei, promoter occupancy is absent and promoter activation is absent or much reduced. Transcriptional activation of c-myc and cyclin D1 promoters by nuclear IRS-1 does not occur with a mutant, inactive IRS-1 protein (deletion of the phosphotyrosine-binding domain, PTB) and does not require PI3-kinase activity. Taken together, these results indicate a novel mechanism by which nuclear IRS-1 activates cell cycle genes.
Collapse
Affiliation(s)
- A Wu
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | |
Collapse
|
206
|
Omazic B, Näsman-Björk I, Permert J, Lundkvist I. Insulin-like growth factor-1 receptor RNA expression in hematopoietic stem cell transplanted patients does not correlate with graft-versus-host disease. Immunol Invest 2007; 36:493-506. [PMID: 17691028 DOI: 10.1080/08820130701266526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In vivo activated T lymphocytes exhibit altered expression of insulin-like growth factor-1 receptor (IGF-1R) compared to the naïve T lymphocyte pool. The objective of this study was to investigate the expression of IGF-1R RNA in CD4 and CD8 positive cells after allogeneic hematopoietic stem cell transplantation (HSCT) in patients with and without GVHD. For this purpose we isolated RNA from CD4 and CD8 positive cells, sorted with immunomagnetic beads. We used real-time PCR for RNA quantification. We demonstrate a significantly decreased expression of IGF-1R RNA in both CD4 and CD8 positive cells up to 12 months after HSCT. We could not demonstrate a correlation between the IGF-1R RNA expression and T cell activating processes like GVHD, expansion of CD4 or CD8 populations or virus infections.
Collapse
Affiliation(s)
- Brigitta Omazic
- Arvid Wretlind Laboratory at the Center for Surgical Sciences, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| | | | | | | |
Collapse
|
207
|
Hartog H, Wesseling J, Boezen HM, van der Graaf WTA. The insulin-like growth factor 1 receptor in cancer: old focus, new future. Eur J Cancer 2007; 43:1895-904. [PMID: 17624760 DOI: 10.1016/j.ejca.2007.05.021] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Accepted: 05/25/2007] [Indexed: 12/16/2022]
Abstract
The importance of insulin-like growth factor 1 receptor (IGF-1R) signalling in malignant behaviour of tumour cells is well established. Currently, development of drugs targeting the IGF-1R as anticancer treatment is emerging. Several IGF-1R targeting strategies are being investigated in phases I and II clinical trials. Interactions of IGF-1R with insulin receptor, however, might complicate efficiency and tolerability of such drugs. This review describes mechanisms, recent developments and potential limitations of IGF-1R antibodies and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Hermien Hartog
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
208
|
Morgillo F, Bareschino MA, Bianco R, Tortora G, Ciardiello F. Primary and acquired resistance to anti-EGFR targeted drugs in cancer therapy. Differentiation 2007; 75:788-99. [PMID: 17608727 DOI: 10.1111/j.1432-0436.2007.00200.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In recent years, the epidermal growth factor receptor (EGFR) has been recognized as a central player and regulator of cancer cell proliferation, apoptosis and angiogenesis and, therefore, as a potentially relevant therapeutic target. Several strategies for EGFR targeting have been developed, the most succesful being represented by monoclonal antibodies, that directly interfere with ligand-receptor binding and small molecule tyrosine kinase inhibitors, that interfere with activation/phosphorylation of EGFR. These agents have been authorized in advanced chemorefractory cancers, including colorectal cancer, non-small-cell lung cancer and head and neck cancer. However, evidence of resistance to these drugs has been described and extensive studies have been performed to investigate whether resistance to EGFR-targeted therapy is primary or secondary. Cellular levels of EGFR do not always correlate with response to the EGFR inhibitors. Indeed, in spite of the over expression and efficient inhibition of EGFR, resistance to EGFR inhibitors may occur. Moreover, given the genetic instability of cancer cells, genetic modifications could enable them to acquire a resistant phenotype to anti-EGFR therapies. Taken together, these findings support the importance of understanding the molecular mechanisms affecting cancer cell sensitivity or resistance to such inhibitors. This review will focus on the most relevant mechanisms contributing to the acquisition of sensitivity/resistance to EGFR inhibitors.
Collapse
Affiliation(s)
- Floriana Morgillo
- Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale F. Magrassi e A. Lanzara, Seconda Università degli Studi di Napoli, Via Pansini 5 80131 Napoli, Italy.
| | | | | | | | | |
Collapse
|
209
|
Li M, He Z, Ermakova S, Zheng D, Tang F, Cho YY, Zhu F, Ma WY, Sham Y, Rogozin EA, Bode AM, Cao Y, Dong Z. Direct inhibition of insulin-like growth factor-I receptor kinase activity by (-)-epigallocatechin-3-gallate regulates cell transformation. Cancer Epidemiol Biomarkers Prev 2007; 16:598-605. [PMID: 17372258 DOI: 10.1158/1055-9965.epi-06-0892] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Insulin-like growth factor-I receptor (IGF-IR) has been implicated in cancer pathophysiology. Furthermore, impairment of IGF-IR signaling in various cancer cell lines caused inhibition of the transformed phenotype as determined by the inhibition of colony formation in soft agar and the inhibition of tumor formation in athymic nude mice. Thus, the IGF-IR might be an attractive target for cancer prevention. We showed that the tea polyphenol, (-)-epigallocatechin-3-gallate (EGCG), is a small-molecule inhibitor of IGF-IR activity (IC50 of 14 micromol/L). EGCG abrogated anchorage-independent growth induced by IGF-IR overexpression and also prevented human breast and cervical cancer cell phenotype expression through inhibition of IGF-IR downstream signaling. Our findings are the first to show that the IGF-IR is a novel binding protein of EGCG and thus may help explain the chemopreventive effect of EGCG on cancer development.
Collapse
Affiliation(s)
- Ming Li
- Hormel Institute, University of Minnesota, 801 16th Avenue Northeast, Austin, MN 55912, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Alexia C, Bras M, Fallot G, Vadrot N, Daniel F, Lasfer M, Tamouza H, Groyer A. Pleiotropic effects of PI-3' kinase/Akt signaling in human hepatoma cell proliferation and drug-induced apoptosis. Ann N Y Acad Sci 2007; 1090:1-17. [PMID: 17384242 DOI: 10.1196/annals.1378.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IGF-II and type I-IGF receptor (IGF-IR) gene expression is increased in primary liver tumors, and transgenic mice overexpressing IGF-II in the liver develop hepatocellular carcinoma (HCC) spontaneously, suggesting that alterations of IGF-IR signaling in vivo may play a role in the auto/paracrine control of hepatocarcinogenesis. We have addressed the contribution of PI-3'K/Akt signaling on the proliferation of HepG2 human hepatoma cells and on their protection against doxorubicin-induced apoptosis. Both basal HepG2 cell DNA replication and that stimulated by IGF-IR signaling were inhibited by the specific PI-3'K inhibitor Ly294002 (Ly). In the former case, PI-3'K signaling overcame cell cycle arrest in G1 via increased cyclin D1 protein and decreased p27kip1 gene expression. Doxorubicin treatment induced apoptosis in HepG2 cells and was concomitant with the proteolytic cleavage of Akt-1 and -2. Drug-induced apoptosis was reversed by IGF-I and this effect was (i) dependent on Akt-1 and -2 phosphorylation and (ii) accompanied by the inhibition of initiator caspase-9 activity, suggesting that IGF-IR signaling interferes with mitochondria-dependent apoptosis. Accordingly, Ly enhanced doxorubicin-induced apoptosis and suppressed its reversal by IGF-I. Altogether, the data emphasize the crucial role of PI-3'K/Akt signaling (i) in basal as well as IGF-IR-stimulated HepG2 cell proliferation and (ii) in controlling both doxorubicin-induced apoptosis (e.g., drug-induced cleavage of Akt) and its reversal by IGF-I (protection against apoptosis parallels the extent of Akt phosphorylation). They suggest that targeting Akt activity or downstream Akt effectors (e.g., GSK3-beta, FOXO transcription factors) may help define novel therapeutic strategies of increased efficacy in the treatment of HCC-bearing patients.
Collapse
Affiliation(s)
- Catherine Alexia
- INSERM U.481, Faculté de Médecine Xavier Bichat, 16, rue Henri Huchard, BP 416, 75870-Paris Cédex 18, France
| | | | | | | | | | | | | | | |
Collapse
|
211
|
Lee KW, Cobb LJ, Paharkova-Vatchkova V, Liu B, Milbrandt J, Cohen P. Contribution of the orphan nuclear receptor Nur77 to the apoptotic action of IGFBP-3. Carcinogenesis 2007; 28:1653-8. [PMID: 17434920 DOI: 10.1093/carcin/bgm088] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Tumor suppression by insulin-like growth factor-binding protein-3 (IGFBP-3) has been demonstrated to occur via insulin-like growth factor-dependent and -independent mechanisms in vitro and in vivo. We have recently described IGFBP-3-induced mitochondrial translocation of the nuclear receptors RXRalpha/Nur77 in the induction of prostate cancer (CaP) cell apoptosis. Herein, we demonstrate that IGFBP-3 and Nur77 associate in the cytoplasmic compartment in 22RV1 CaP cells. Nur77 is a major component of IGFBP-3-induced apoptosis as shown by utilizing mouse embryonic fibroblasts (MEFs) derived from Nur77 wild-type and knockout (KO) mice. However, dose-response experiments revealed that a small component of IGFBP-3-induced apoptosis is Nur77 independent. Reintroduction of Nur77 into Nur77 KO MEFs restores full responsiveness to IGFBP-3. IGFBP-3 induces phosphorylation of Jun N-terminal kinase and inhibition of Akt phosphorylation and activity, which have been associated with Nur77 translocation. Finally, IGFBP-3 administration to CaP xenografts on SCID mice induced apoptosis and translocated Nur77 out of the nucleus. Taken together, our results verify an important role for the orphan nuclear receptor Nur77 in the apoptotic actions of IGFBP-3.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Cell Line, Tumor
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/physiology
- Fibroblasts/cytology
- Fibroblasts/enzymology
- Fibroblasts/metabolism
- Humans
- Insulin-Like Growth Factor Binding Protein 3
- Insulin-Like Growth Factor Binding Proteins/metabolism
- Insulin-Like Growth Factor Binding Proteins/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Protein Binding/physiology
- Protein Transport/physiology
- Receptors, Cytoplasmic and Nuclear/deficiency
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Cytoplasmic and Nuclear/physiology
- Receptors, Steroid/deficiency
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Receptors, Steroid/physiology
- Subcellular Fractions/metabolism
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Kuk-Wha Lee
- Division of Pediatric Endocrinology, Mattel Children's Hospital at University of California at Los Angeles, David Geffen School of Medicine, 10833 Le Conte Avenue, MDCC 22-315, Los Angeles, CA 90095, USA.
| | | | | | | | | | | |
Collapse
|
212
|
Velaparthi U, Wittman M, Liu P, Stoffan K, Zimmermann K, Sang X, Carboni J, Li A, Attar R, Gottardis M, Greer A, Chang CY, Jacobsen BL, Sack JS, Sun Y, Langley DR, Balasubramanian B, Vyas D. Discovery and initial SAR of 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-ones as inhibitors of insulin-like growth factor 1-receptor (IGF-1R). Bioorg Med Chem Lett 2007; 17:2317-21. [PMID: 17317169 DOI: 10.1016/j.bmcl.2007.01.102] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 01/12/2007] [Accepted: 01/18/2007] [Indexed: 11/29/2022]
Abstract
The discovery and synthesis of 3-(1H-benzo[d]imidazol-2-yl)pyridin-2(1H)-one inhibitors of insulin-like growth factor 1-receptor (IGF-1R) are presented. Installing amine containing side chains at the 4-position of pyridone ring significantly improved the enzyme potency. SAR and biological activity of these compounds is presented.
Collapse
Affiliation(s)
- Upender Velaparthi
- Department of Discovery Chemistry, Bristol-Myers Squibb Pharmaceutical Research Institute, 5 Research Parkway, Wallingford, CT 06492, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Takaoka M, Kim SH, Okawa T, Michaylira CZ, Stairs DB, Johnstone CN, Andl CD, Rhoades B, Lee JJ, Klein-Szanto AJ, El-Deiry WS, Nakagawa H. IGFBP-3 regulates esophageal tumor growth through IGF-dependent and independent mechanisms. Cancer Biol Ther 2007; 6:534-40. [PMID: 17457048 PMCID: PMC2993006 DOI: 10.4161/cbt.6.4.3832] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Insulin-like growth factor binding protein (IGFBP)-3 exerts either proapoptotic or growth stimulatory effects depending upon the cellular context. IGFBP-3 is overexpressed frequently in esophageal cancer. Yet, the role of IGFBP-3 in esophageal tumor biology remains elusive. To delineate the functional consequences of IGFBP-3 overexpression, we stably transduced Ha-Ras(V12)-transformed human esophageal cells with either wild-type or mutant IGFBP-3, the latter incapable of binding Insulin-like growth factor (IGFs) as a result of substitution of amino-terminal Ile56, Leu80, and Leu81 residues with Glycine residues. Wild-type, but not mutant, IGFBP-3 prevented IGF-1 from activating the IGF-1 receptor and AKT, and suppressed anchorage-independent cell growth. When xenografted in nude mice, in vivo bioluminescence imaging demonstrated that wild-type, but not mutant IGFBP-3, abrogated tumor formation by the Ras-transformed cells with concurrent induction of apoptosis, implying a prosurvival effect of IGF in cancer cell adaptation to the microenvironment. Moreover, there was more aggressive tumor growth by mutant IGFBP-3 overexpressing cells than control cell tumors, without detectable caspase-3 cleavage in tumor tissues, indicating an IGF-independent growth stimulatory effect of mutant IGFBP-3. In aggregate, these data suggest that IGFBP-3 contributes to esophageal tumor development and progression through IGF-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Munenori Takaoka
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Seok-Hyun Kim
- Hematology/Oncology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Laboratory of Molecular Oncology and Cell Cycle Regulation, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Genetics, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Pharmacology, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Takaomi Okawa
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Carmen Z. Michaylira
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Douglas B. Stairs
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Cameron N. Johnstone
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Claudia D. Andl
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Ben Rhoades
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - James J. Lee
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | | | - Wafik S. El-Deiry
- Hematology/Oncology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Laboratory of Molecular Oncology and Cell Cycle Regulation, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Genetics, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Pharmacology, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| | - Hiroshi Nakagawa
- Gastroengerology Division, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Department of Medicine, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
- Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine; Philadelphia, Pennsylvania USA
| |
Collapse
|
214
|
Fischer B, Marinov M, Arcaro A. Targeting receptor tyrosine kinase signalling in small cell lung cancer (SCLC): what have we learned so far? Cancer Treat Rev 2007; 33:391-406. [PMID: 17368733 DOI: 10.1016/j.ctrv.2007.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 01/15/2007] [Accepted: 01/23/2007] [Indexed: 11/16/2022]
Abstract
Small cell lung cancer (SCLC) is an aggressive form of lung cancer, which represents 13% of all cases and is strongly associated with cigarette smoking. The survival of SCLC patients is dismal and has not greatly improved in the last 20 years, despite advances in chemotherapy regimens and a better understanding of SCLC biology. The development of resistance to chemotherapy and metastasis are commonly recognized as important causes of poor clinical outcome in SCLC. Targeting receptor tyrosine kinase (RTK) signalling represents an attractive approach to develop new drugs for SCLC, in view of the accumulating data demonstrating that polypeptide growth factors play a key role in driving SCLC cell proliferation, chemoresistance and metastasis. The insulin-like growth factor-I receptor (IGF-IR), c-Kit, vascular endothelial growth factor receptor (VEGFR) and epidermal growth factor receptor (EGFR) have been identified as potential drug targets in SCLC. Moreover, downstream signalling mediators of RTKs, such as phosphoinositide 3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) may also represent attractive candidate molecules for anti-cancer therapies in SCLC. Here we will review the available data concerning results with RTK inhibitors in SCLC and the clinical trials undertaken to investigate the potential of these compounds as anti-tumour agents in SCLC.
Collapse
Affiliation(s)
- Barbara Fischer
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland.
| | | | | |
Collapse
|
215
|
Insulin-like growth factor type I biology and targeting in malignant gliomas. Neuroscience 2007; 145:795-811. [DOI: 10.1016/j.neuroscience.2007.01.021] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 01/04/2007] [Accepted: 01/05/2007] [Indexed: 11/20/2022]
|
216
|
Prince SN, Foulstone EJ, Zaccheo OJ, Williams C, Hassan AB. Functional evaluation of novel soluble insulin-like growth factor (IGF)-II–specific ligand traps based on modified domain 11 of the human IGF2 receptor. Mol Cancer Ther 2007; 6:607-17. [PMID: 17308058 DOI: 10.1158/1535-7163.mct-06-0509] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ligands transported by the mannose 6-phosphate/insulin-like growth factor (IGF)-II receptor (IGF2R) include IGF-II- and mannose 6-phosphate-modified proteins. Increased extracellular supply of IGF-II, either secondary to loss of the clearance function of IGF2R, loss of IGF binding protein function, or increased IGF2 gene expression, can lead to embryonic overgrowth and cancer promotion. Reduced supply of IGF-II is detrimental to tumor growth, and this suggests that gain of function of IGF-II is a molecular target for human cancer therapy. Domain 11 of IGF2R binds IGF-II with high specificity and affinity. Mutagenesis studies have shown that substitution of glutamic acid for lysine at residue 1554 results in a 6-fold higher affinity for IGF-II (20.5 nmol/L) than native domain 11 (119 nmol/L). Here, we generate a novel high-affinity IGF-II ligand trap by fusion of mutated human 11(E1554K) to a COOH-terminal human IgG1 Fc domain (11(E1554K)-Fc). The resulting homodimer has a significantly increased affinity for IGF-II (1.79 nmol/L) when measured by surface plasmon resonance. IGF-II signaling via the IGF-I receptor and the proliferative effect of IGF-II were specifically inhibited by 11(E1554K)-Fc in both HaCaT and Igf2(-/-) mouse embryonic fibroblast cells. These data confirm that a novel engineered and soluble IGF2R-11(E1554K)-Fc protein functions as an IGF-II-specific and high-affinity ligand trap in vitro and that this protein has potential application as an IGF-II antagonist for cancer therapy following in vivo experimental evaluation.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Proliferation
- Drosophila melanogaster/growth & development
- Drosophila melanogaster/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Embryo, Nonmammalian
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Genetic Vectors
- Humans
- Insulin-Like Growth Factor II/genetics
- Insulin-Like Growth Factor II/physiology
- Keratinocytes/metabolism
- Ligands
- Mice
- Mice, Knockout
- Pichia/chemistry
- Pichia/metabolism
- Protein Binding
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, IGF Type 2/genetics
- Receptor, IGF Type 2/metabolism
- Receptors, Fc/genetics
- Receptors, Fc/metabolism
- Surface Plasmon Resonance
- Thymidine/metabolism
Collapse
Affiliation(s)
- Stuart N Prince
- Weatherall Institute for Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | | | | | | | | |
Collapse
|
217
|
Keyhanfar M, Booker G, Whittaker J, Wallace J, Forbes B. Precise mapping of an IGF-I-binding site on the IGF-1R. Biochem J 2007; 401:269-77. [PMID: 16981855 PMCID: PMC1698672 DOI: 10.1042/bj20060890] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The IGF-1R [type 1 IGF (insulin-like growth factor) receptor] is activated upon binding to IGF-I and IGF-II leading to cell growth, survival and migration of both normal and cancerous cells. We have characterized the binding interaction between the IGF-1R and its ligands using two high-affinity mouse anti-IGF-1R mAbs (monoclonal antibodies), 7C2 and 9E11. These mAbs both block IGF-I binding to the IGF-1R but have no effect on IGF-II binding. Epitope mapping using chimaeras of the IGF-1R and insulin receptor revealed that the mAbs bind to the CR (cysteine-rich) domain of IGF-1R. The epitope was finely mapped using single point mutations in the IGF-1R. Mutation of Phe241, Phe251 or Phe266 completely abolished 7C2 and 9E11 binding. The three-dimensional structure showed that these residues cluster on the surface of the CR-domain. BIAcore analyses revealed that IGF-I and a chimaeric IGF-II with the IGF-I C-domain competed for the binding of both mAbs with the IGF-1R, whereas neither IGF-II nor a chimaeric IGF-I with the IGF-II C-domain affected antibody binding. We therefore conclude the IGF-I C-domain interacts with the CR (cysteine-rich) domain of the receptor at the cluster of residues Phe241, Phe251 and Phe266. These results allow precise orientation of IGF-I within the IGF-I-IGF-1R complex involving the IGF-I C-domain binding to the IGF-1R CR domain. In addition, mAbs 7C2 and 9E11 inhibited both IGF-I- and IGF-II-induced cancer cell proliferation, migration and IGF-1R down-regulation, demonstrating that targeting the IGF-1R is an effective strategy for inhibition of cancer cell growth.
Collapse
Affiliation(s)
- Mehrnaz Keyhanfar
- *School of Molecular and Biomedical Science, The University of Adelaide, Adelaide 5005, South Australia, Australia
- †Bu-Ali Sina University, Hamedan, Iran
| | - Grant W. Booker
- *School of Molecular and Biomedical Science, The University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Jonathan Whittaker
- ‡Departments of Nutrition and Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106-4906, U.S.A
| | - John C. Wallace
- *School of Molecular and Biomedical Science, The University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Briony E. Forbes
- *School of Molecular and Biomedical Science, The University of Adelaide, Adelaide 5005, South Australia, Australia
- To whom correspondence should be addressed (email )
| |
Collapse
|
218
|
Ayllón V, O'connor R. PBK/TOPK promotes tumour cell proliferation through p38 MAPK activity and regulation of the DNA damage response. Oncogene 2006; 26:3451-61. [PMID: 17160018 DOI: 10.1038/sj.onc.1210142] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The contribution of the insulin-like growth-factor-I receptor (IGF-IR) to tumour progression is well documented. To identify new mediators of IGF-IR function in cancer, we recently isolated genes differentially expressed in cells overexpressing the IGF-IR. Among these was the serine/threonine kinase PBK/TOPK (PDZ-binding kinase/T-LAK cell-originated protein kinase), previously associated with highly proliferative cells and tissues. Here, we show that PBK is expressed at high levels in tumour cell lines compared with non-transformed cells. IGF-I could induce PBK expression only in transformed cells, whereas epidermal growth factor could induce PBK in non-transformed MCF-10A breast epithelial cells. Suppression of PBK expression using small interfering RNA did not prevent progression through the cell cycle, but caused decreased proliferation over time in culture, and reduced clonogenic growth in soft agarose. PBK knockdown impaired p38 activation after long-term stimulation with different growth factors and reduced DU145 cells motility. Suppressed PBK expression also resulted in an impaired response to DNA damage that was evident by the decreased generation of gamma-H2AX, increased DNA damage and decreased cell survival. Taken together, the data indicate that PBK is necessary for appropriate activation and function of the p38 pathway by growth factors. Thus, enhanced expression of PBK may facilitate tumour growth by mediating p38 activation and by helping cells to overcome DNA damage.
Collapse
Affiliation(s)
- V Ayllón
- Cell Biology Laboratory, Department of Biochemistry, Biosciences Institute, National University of Ireland, Cork, Ireland
| | | |
Collapse
|
219
|
Adachi Y, Yamamoto H, Imsumran A, Oka T, Oki M, Nosho K, Min Y, Shinomura Y, Lee C, Carbone DP, Imai K. INSULIN‐LIKE GROWTH FACTOR‐I RECEPTOR AS A CANDIDATE FOR A NOVEL MOLECULAR TARGET IN GASTROINTESTINAL CANCERS. Dig Endosc 2006; 18:245-251. [DOI: 10.1111/j.1443-1661.2006.00657.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Abnormal activation of growth factor receptors and their signal pathways are required for neoplastic transformation and tumor progression. The concept of targeting specific tumorigenic receptors has been validated by successful clinical application of multiple new drugs, such as those acting against HER2/neu, epidermal growth factor receptor 1, and c‐Kit. In this review, we focus on the next promising therapeutic molecular target of insulin‐like growth factor (IGF)‐I receptor (IGF‐Ir). The IGF/IGF‐Ir system is an important modifier of cancer cell proliferation, survival, growth, and treatment sensitivity in a number of neoplastic diseases, including human gastrointestinal carcinomas. Preclinical studies demonstrated that downregulation of IGF‐Ir signals reversed the neoplastic phenotype and sensitized cells to antitumor treatments. We summarize a variety of ways to disrupt IGF‐Ir function. Then, we introduce our strategy of adenoviruses expressing dominant negative of IGF‐Ir (IGF‐Ir/dn) against gastrointestinal cancers, including stomach, colon, and pancreas. IGF‐Ir/dn suppresses tumorigenicity both in vitro and in vivo and increases stressor‐induced apoptosis. IGF‐Ir/dn expression upregulates chemotherapy‐induced apoptosis and these combination therapies with chemotherapy are very effective against tumors in mice. Some drugs blocking IGF‐Ir function are now entering clinical trial, thus IGF‐Ir might be a candidate for a therapeutic target in several gastrointestinal malignancies.
Collapse
|
220
|
Abstract
The insulin-like growth factor (IGF) axis has been implicated in malignant transformation and in tumor cell biology. Human population studies have demonstrated that high levels of circulating IGF-I are associated with an increased risk of certain malignancies. Many model systems show that IGFs stimulate tumor cell proliferation, survival and metastasis. In a new era of anticancer treatments aimed at tumor-specific targets, efforts are in progress for the development of novel anti-IGF therapies. Disrupting type I IGF-receptor (IGF-IR) function in vitro and in vivo results in tumor growth inhibition in several model systems. Antireceptor therapies in particular have provided encouraging results leading to the approval of the first Phase I human clinical trial targeting IGF-IR. Additional methods to decrease levels of circulating IGF-I and II have also been developed. In principle, a benefit of targeted therapies could be their relative lack of toxicity compared with conventional chemotherapy. Anti-IGF-IR therapies, however, raise theoretical concerns for the development of serious side effects, including diabetes. As targeted therapies against the IGF axis continue to be developed, efforts will need to be made to minimize the side effects that result from blocking normal ligand and receptor-induced functions.
Collapse
Affiliation(s)
- Tufia Haddad
- Department of Medicine, Hematology, Oncology and Transplantation, University of Minnesota Cancer Center, MMC 806, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
221
|
Rosengren L, Vasilcanu D, Vasilcanu R, Fickenscher S, Sehat B, Natalishvili N, Naughton S, Yin S, Girnita A, Girnita L, Axelson M, Larsson O. IGF-1R tyrosine kinase expression and dependency in clones of IGF-1R knockout cells (R−). Biochem Biophys Res Commun 2006; 347:1059-66. [PMID: 16857168 DOI: 10.1016/j.bbrc.2006.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Accepted: 07/01/2006] [Indexed: 12/31/2022]
Abstract
Insulin-like growth factor 1 receptor (IGF-1R) plays many crucial roles in cancer, like anti-apoptotic activity and necessity for transformation. IGF-1R knockout cells (R-) represent a useful tool for molecular mapping of biological properties of the receptor. R- cells have been shown to be refractory to transformation by viral and cellular oncogenes, highlighting the necessity of this receptor for transformation. Surprisingly, more recent studies have shown that these cells can undergo spontaneous transformation. This observation raises the question as whether R- cells over the years have acquired some properties mimicking those of IGF-1R. Using an IGF-1R inhibitor (cyclolignan PPP) we have identified clones of R- (R-s) that are sensitive to this compound. Since, PPP is closely related to podophyllotoxin, which is an efficient microtubule inhibitor, we first investigated if such a mechanism could explain the sensitivity to PPP. However, highly purified PPP showed no or very slight tubulin binding. Further analysis of R-s revealed expression of a 90 kDa protein being reactive to IGF-1R beta-subunit antibodies. This protein was weakly but constitutively tyrosine phosphorylated and was downregulated by siRNA targeting IGF-1R. This downregulation was paralleled by decreased R-s survival. Taken together, our study suggests that clones of R- express IGF-1R activity and dependency, which in turn may explain that R- can undergo spontaneous transformation.
Collapse
Affiliation(s)
- Linda Rosengren
- Department of Oncology and Pathology, Division of Cellular and Molecular Tumor Pathology, CCK, R8:04, Karolinska University Hospital Solna, SE-171 76 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Werner H, Maor S. The insulin-like growth factor-I receptor gene: a downstream target for oncogene and tumor suppressor action. Trends Endocrinol Metab 2006; 17:236-42. [PMID: 16815029 DOI: 10.1016/j.tem.2006.06.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2006] [Revised: 06/06/2006] [Accepted: 06/20/2006] [Indexed: 11/20/2022]
Abstract
The insulin-like growth factor-I receptor (IGF-IR) mediates the biological actions of both IGF-I and IGF-II. The IGF-IR is expressed in most transformed cells, where it displays potent antiapoptotic and cell-survival activities. IGF-IR levels are tightly regulated by the concerted action of secreted (e.g. peptide and steroid hormones, growth factors and cytokines) and cellular (e.g. transcription factors, oncogenes and tumor suppressors) factors. The mode of action of many tumor suppressors involves transcriptional suppression of the IGF-IR gene, with a subsequent reduction in cell-surface IGF-IR concentrations and IGF-I action. Loss-of-function mutation of tumor suppressors, a common theme in human cancer, can lead to aberrant regulation of IGF-IR gene expression.
Collapse
Affiliation(s)
- Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | | |
Collapse
|
223
|
Keyhanfar M, Forbes BE, Cosgrove LJ, Wallace JC, Booker GW. Production and Characterization of Monoclonal Antibodies Against Insulin-Like Growth Factor Type 1 Receptor. Hybridoma (Larchmt) 2006; 25:230-7. [PMID: 16934020 DOI: 10.1089/hyb.2006.25.230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The type 1 insulin-like growth factor receptor (IGF-1R) has been extensively reported to play an important role in cancer. Activation of the IGF-1R by IGF-I and IGF-II binding to the extracellular domains of the receptor induces mitogenic and anti-apoptotic effects, which are important events in tumor growth and survival. Several cancer cell types overexpress IGF-1R, suggesting a possible use of monoclonal antibodies (MAbs) against IGF-1R as diagnostic reagents. Here, we report the production and characterization of two independent MAbs, namely 7C2 and 9E11, generated by immunizing mice with the soluble extracellular part of this receptor (amino acids 1-906). Both MAbs bind to membrane bound IGF-1R and do not cross-react with insulin receptor isoforms, IR-A and IR-B expressed on IGF-1R() cells. MAbs 7C2 and 9E11 stained the IGF- 1R on frozen or paraffin-embedded tissue sections or frozen cells. The MAbs 7C2 and 9E11 immunoprecipitated the IGF-1R from P6 cell lysates (cells overexpressing human IGF-1R) and could detect non-reduced intact IGF-1R on immunoblots. However, the MAbs were not able to detect reduced and denatured receptor alpha and beta chains. Sequencing of the heavy- and light-chain variable regions revealed that the 7C2 and 9E11 CDR amino acid sequences are different but result in antibodies with similar properties. MAbs 7C2 and 9E11 are therefore potentially useful diagnostic tools and could be of therapeutic use for humans in the future.
Collapse
Affiliation(s)
- Mehrnaz Keyhanfar
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia.
| | | | | | | | | |
Collapse
|
224
|
Sarfstein R, Maor S, Reizner N, Abramovitch S, Werner H. Transcriptional regulation of the insulin-like growth factor-I receptor gene in breast cancer. Mol Cell Endocrinol 2006; 252:241-6. [PMID: 16647191 DOI: 10.1016/j.mce.2006.03.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The insulin-like growth factor-I receptor (IGF-IR) has an important role in normal mammary gland growth and morphogenesis. In addition, the IGF-IR has been implicated in the initiation and progression of breast cancer. Previous studies have indicated that acquisition of the malignant phenotype in breast cancer is initially IGF-IR dependent. Most breast cancer-derived cell lines and primary tumors express high levels of IGF-IR mRNA and protein, whereas metastatic stages are usually associated with a decrease in IGF-IR levels. Transcription of the IGF-IR gene is controlled by complex interactions involving DNA-binding and non DNA-binding transcription factors. This review highlights selected examples of tumor suppressors, including BRCA1, p53, and WT1, whose mechanism of action involves regulation of IGF-IR gene expression.
Collapse
Affiliation(s)
- Rive Sarfstein
- Department of Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | |
Collapse
|
225
|
Spence SL, Shaffer AL, Staudt LM, Amde S, Manney S, Terry C, Weisz K, Nissley P. Transformation of late passage insulin-like growth factor-I receptor null mouse embryo fibroblasts by SV40 T antigen. Cancer Res 2006; 66:4233-9. [PMID: 16618746 DOI: 10.1158/0008-5472.can-05-2257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is evidence that the insulin-like growth factor-I (IGF-I) receptor is required for transformation by a variety of viral and cellular oncogenes in a mouse embryo fibroblast model. To further investigate the IGF-I receptor signaling pathways that are required for the permissive effect of the receptor on transformation by SV40 T antigen, we established three independent fibroblast cell lines each from wild-type and IGF-I receptor null embryos (R-). We transfected the wild-type and R- cell lines with an SV40 T antigen plasmid and selected three clones from each cell line that expressed T antigen. As in previous reports, none of the cloned R- cell lines expressing T antigen were transformed as measured by the ability to form large colonies in soft agar. However, with further passage, all three T antigen-expressing clones from one of the R- cell lines (R(-)3) formed large colonies in soft agar and the transformation of these T antigen-expressing clones was confirmed by tumorigenesis experiments in immunodeficient mice. DNA microarray analysis comparing gene expression between early passage and late passage R(-)3/T antigen clones showed, among other changes, an increase in the expression of ErbB-3 mRNA in the late passage clones. Also, the expression of ErbB-3 protein was dramatically increased in the late passage R(-)3/T antigen clones. We conclude that late passage IGF-I receptor null mouse embryo fibroblasts can be transformed by SV40 T antigen, and that ErbB-3 may play a role in permitting transformation by T antigen.
Collapse
MESH Headings
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/physiology
- Cell Growth Processes/physiology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- DNA/biosynthesis
- Embryo, Mammalian
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Fibroblasts/physiology
- Focal Adhesion Kinase 1/metabolism
- GRB2 Adaptor Protein/biosynthesis
- GRB2 Adaptor Protein/genetics
- Genotype
- Insulin Receptor Substrate Proteins
- Ligands
- Mice
- Mice, Knockout
- Phosphoproteins/biosynthesis
- Phosphoproteins/genetics
- Phosphorylation
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptor, ErbB-3/biosynthesis
- Receptor, ErbB-3/genetics
- Receptor, IGF Type 1/deficiency
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Transfection
Collapse
Affiliation(s)
- Susan L Spence
- Metabolism Branch, National Cancer Institute, NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Robinson SA, Rosenzweig SA. Paradoxical effects of the phage display-derived peptide antagonist IGF-F1-1 on insulin-like growth factor-1 receptor signaling. Biochem Pharmacol 2006; 72:53-61. [PMID: 16684509 DOI: 10.1016/j.bcp.2006.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 03/30/2006] [Accepted: 03/30/2006] [Indexed: 10/24/2022]
Abstract
The insulin-like growth factor binding proteins (IGFBPs) represent a unique class of IGF antagonists regulating the bioavailability of the IGFs extracellularly. Accordingly, they represent an important class of proteins for cancer therapeutics and chemoprevention. IGF-F1-1 is a cyclic hexadecapeptide identified by high throughput phage display that binds to the IGFBP-binding domain on IGF-1. It acts as an IGFBP-mimetic, capable of inhibiting IGF-1 binding to the IGFBPs. To further examine the utility of IGF-F1-1 as an IGF-1 antagonist we tested its ability to inhibit IGFBP-2 and IGFBP-3 binding to IGF-1, (125)I-IGF-1 binding to IGF-1Rs and to block IGF-1 induced Akt activation, cell cycle changes and [(3)H]thymidine incorporation in MCF-7 cells. These biological activities were inhibited by treatment with IGFBP-2, wortmannin or the IGF-1R tyrosine kinase inhibitor, NVP-AEW541, but not by IGF-F1-1. Our findings confirm previous studies indicating that IGF-F1-1 is a weak antagonist of IGF-1 binding to the IGFBPs and the IGF-1R and suggest that it does not effectively inhibit downstream events stimulated by IGF-1. We further demonstrated that IGF-F1-1 treatment of MCF-7 cells results in the paradoxical activation of Akt, S-phase transition and [(3)H]thymidine incorporation. These results suggest that IGF-F1-1 is a weak agonist, exhibiting mitogenic actions. IGF-F1-1 may act in conjunction with IGF-1 at the IGF-1R or independently of IGF-1 at the IGF-1R or another receptor.
Collapse
Affiliation(s)
- Stephanie A Robinson
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | |
Collapse
|
227
|
Schillaci R, Salatino M, Cassataro J, Proietti CJ, Giambartolomei GH, Rivas MA, Carnevale RP, Charreau EH, Elizalde PV. Immunization with murine breast cancer cells treated with antisense oligodeoxynucleotides to type I insulin-like growth factor receptor induced an antitumoral effect mediated by a CD8+ response involving Fas/Fas ligand cytotoxic pathway. THE JOURNAL OF IMMUNOLOGY 2006; 176:3426-37. [PMID: 16517711 DOI: 10.4049/jimmunol.176.6.3426] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have demonstrated that in vivo administration of phosphorothioate antisense oligodeoxynucleotides (AS[S]ODNs) to type I insulin-like growth factor receptor (IGF-IR) mRNA resulted in inhibition of C4HD breast cancer growth in BALB/c mice. The present study focused on whether in vivo administration of C4HD tumor cells pretreated with IGF-IR AS[S]ODN and irradiated could provide protection against C4HD wild-type tumor challenge and also on elucidating the mechanism mediating this effect. Our results showed that mice immunized with IGF-IR AS[S]ODN-treated C4HD cells experienced a growth inhibition of 53.4%, 61.6%, and 60.2% when compared with PBS-treated mice, wild-type C4HD cell-injected mice, or phosphorothioate sense oligodeoxynucleotide-treated C4HD cell-injected mice, respectively. The protective effect was C4HD-specific, because no cross-protection was observed against other syngeneic mammary tumor lines. The lack of protection against tumor formation in nude mice indicated that T cells were involved in the antitumoral response. Furthermore, cytotoxicity and splenocyte proliferation assays demonstrated that a cellular CD8(+)-dependent immune response, acting through the Fas/Fas ligand death pathway, could be mediating the antitumor effect induced by immunization with AS[S]ODN-treated cells. Immunization also induced splenocytes to produce Ag-dependent IFN-gamma, indicating the presence of a type 1 response. We demonstrated for the first time that IGF-IR AS[S]ODN treatment of breast cancer cells induced expression of CD86 and heat shock protein 70 molecules, both involved in the induction of the immunogenic phenotype. Immunization with these tumor immunogens imparted protection against parental tumor growth through activation of a specific immune response.
Collapse
Affiliation(s)
- Roxana Schillaci
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Huang H, Reis R, Yonekawa Y, Lopes JM, Kleihues P, Ohgaki H. Identification in human brain tumors of DNA sequences specific for SV40 large T antigen. Brain Pathol 2006; 9:33-42. [PMID: 9989448 PMCID: PMC8098629 DOI: 10.1111/j.1750-3639.1999.tb00207.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Simian virus 40 (SV40) sequences have recently been identified in a variety of human neoplasms, including mesothelioma, osteosarcoma, and brain tumors, but significant discrepancies exist regarding the frequency at which this occurs. The SV40 genome is 70% homologous to JC and BK, two related polyomaviruses that are highly prevalent in humans and which may cause in immune-compromised patients progressive multifocal leukoencephalopathy (PML) and cystitis, respectively. We have established a specific and sensitive method to identify SV40 sequence in DNA extracted from histological sections, using PCR followed by Southern hybridization to probes specific to the large T region. We found SV40 large T antigen sequences in all brain tumor types investigated. High frequencies were found in low-grade astrocytomas, anaplastic astrocytomas and secondary glioblastomas derived thereof (13/22, 59%) while somewhat lower frequencies were found in gemistocytic astrocytomas (9/28, 32%) and oligodendrogliomas (3/12, 25%). Primary glioblastomas, giant cell glioblastomas, and gliosarcomas, which clinically develop de novo, contained SV40 sequences in 11-25% of cases. Presence of viral DNA was also observed in pediatric brain tumors, including ependymomas (9/16, 56%), choroid plexus papillomas (6/16, 38%), and medulloblastomas (5/17, 29%). In 8 tumor biopsies with SV40 sequences, the adjacent normal brain tissue was also analyzed but was devoid of viral DNA in all but one case. BK and JC virus sequences were rarely detected, the overall frequencies being 3% and 2%, respectively. It remains to be shown whether the presence of SV40 contributes significantly to malignant transformation or whether certain human neoplasms provide a microenvironment that favors viral replication in humans with latent SV40 infection.
Collapse
Affiliation(s)
- Huatao Huang
- International Agency for Research on Cancer, 69372 Lyon, France
| | - Rui Reis
- International Agency for Research on Cancer, 69372 Lyon, France
- Institute of Pathology and Molecular Immunology, IPATIMUP and Medical Faculty of University of Porto, 4200 Porto, Portugal
| | - Yasuhiro Yonekawa
- Department of Neurosurgery, University of Zurich, 8091 Zurich, Switzerland
| | - Jose Manuel Lopes
- Institute of Pathology and Molecular Immunology, IPATIMUP and Medical Faculty of University of Porto, 4200 Porto, Portugal
| | - Paul Kleihues
- International Agency for Research on Cancer, 69372 Lyon, France
- Department of Neuropathology, University of Zurich, 8091 Zurich, Switzerland
| | - Hiroko Ohgaki
- International Agency for Research on Cancer, 69372 Lyon, France
| |
Collapse
|
229
|
Alexia C, Fourmatgeat P, Delautier D, Groyer A. Insulin-like growth factor-I stimulates H4II rat hepatoma cell proliferation: Dominant role of PI-3′K/Akt signaling. Exp Cell Res 2006; 312:1142-52. [PMID: 16487514 DOI: 10.1016/j.yexcr.2006.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2004] [Revised: 11/23/2005] [Accepted: 01/10/2006] [Indexed: 01/26/2023]
Abstract
Although hepatocytes are the primary source of endocrine IGF-I and -II in mammals, their autocrine/paracrine role in the dysregulation of proliferation and apoptosis during hepatocarcinogenesis and in hepatocarcinomas (HCC) remains to be elucidated. Indeed, IGF-II and type-I IGF receptors are overexpressed in HCC cells, and IGF-I is synthesized in adjacent non-tumoral liver tissue. In the present study, we have investigated the effects of type-I IGF receptor signaling on H4II rat hepatoma cell proliferation, as estimated by 3H-thymidine incorporation into DNA. IGF-I stimulated the rate of DNA synthesis of serum-deprived H4II cells, stimulation being maximal 3 h after the onset of IGF-I treatment and remaining elevated until at least 6 h. The IGF-I-induced increase in DNA replication was abolished by LY294002 and only partially inhibited by PD98059, suggesting that phosphoinositol-3' kinase (PI-3'K) and to a lesser extent MEK/Erk signaling were involved. Furthermore, the 3- to 19-fold activation of the Erks in the presence of LY294002 suggested a down-regulation of the MEK/Erk cascade by PI-3'K signaling. Finally, the effect of IGF-I on DNA replication was almost completely abolished in clones of H4II cells expressing a dominant-negative form of Akt but was unaltered by rapamycin treatment of wild-type H4II cells. Altogether, these data support the notion that the stimulation of H4II rat hepatoma cell proliferation by IGF-I is especially dependent on Akt activation but independent on the Akt/mTOR signaling.
Collapse
Affiliation(s)
- Catherine Alexia
- Inserm U.481, Faculté de Médecine Xavier Bichat, 16, rue Henri Huchard, BP416, 75870 Paris Cedex 18, France
| | | | | | | |
Collapse
|
230
|
Sachdev D, Singh R, Fujita-Yamaguchi Y, Yee D. Down-regulation of Insulin Receptor by Antibodies against the Type I Insulin-Like Growth Factor Receptor: Implications for Anti–Insulin-Like Growth Factor Therapy in Breast Cancer. Cancer Res 2006; 66:2391-402. [PMID: 16489046 DOI: 10.1158/0008-5472.can-05-3126] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin-like growth factor-I (IGF-I), IGF-II, and insulin have all been implicated in regulating several aspects of the malignant phenotype via the type I IGF receptor (IGF1R) and insulin receptor (IR). We have previously shown that a chimeric single-chain antibody against IGF1R (scFv-Fc) and a murine antibody EM164 down-regulate IGF1R, making breast cancer cells unresponsive to IGF-I. To determine if IR signaling is affected, we examined regulation of IR in MCF-7 cells after exposure to these antibodies. Surprisingly, both scFv-Fc and EM164 resulted in decreased levels of IR in vitro and in vivo despite their lack of reactivity against IR. Twenty-four-hour pretreatment with EM164 also inhibited insulin-mediated phosphorylation of IR and insulin-stimulated proliferation of MCF-7 cells. Neither scFv-Fc nor EM164 caused down-regulation of IR in cells that express very low levels of IGF1R or no IGF1R. Expression of IGF1R was required for IR down-regulation, which was specific as neither antibody caused down-regulation of beta1 integrin or epidermal growth factor receptor. Reagents that disrupt lipid rafts inhibited IR down-regulation by the antibodies, suggesting that IR in close physical proximity to IGF1R in lipid rafts was being endocytosed. Our data show that down-regulation of IR by monoclonal antibodies against IGF1R requires the coexpression of IGF1R and may be due to endocytosis of hybrid IR/IGF1R or holo-IR. Thus, antibodies against IGF1R provide inhibition of both IGF and insulin signaling in cancer cells.
Collapse
Affiliation(s)
- Deepali Sachdev
- University of Minnesota Cancer Center, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
231
|
Gennigens C, Menetrier-Caux C, Droz JP. Insulin-Like Growth Factor (IGF) family and prostate cancer. Crit Rev Oncol Hematol 2006; 58:124-45. [PMID: 16387509 DOI: 10.1016/j.critrevonc.2005.10.003] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/30/2005] [Accepted: 10/07/2005] [Indexed: 11/28/2022] Open
Abstract
There is abundant in vitro, animal and epidemiologic evidence to suggest that the Insulin-Like Growth Factor (IGF) family is a multi-component network of molecules which is involved in the regulation of both physiological and pathological growth processes in prostate. The IGF family plays a key role in cellular metabolism, differentiation, proliferation, transformation and apoptosis, during normal development and malignant growth. This family also seem essential in prostate cancer bone metastases, angiogenesis and androgen-independent progression. Therapeutic alternatives in men with progressive prostate cancer after androgen ablation are very limited. More effective therapies are needed for these patients. Pharmacologic interventions targeting the IGF family are being devised. Such strategies include reduction of IGF-I levels (growth hormone-releasing hormone antagonists, somatostatin analogs), reduction of functional IGF-I receptor levels (antisense oligonucleotides, small interfering RNA), inhibition of IGF-IR and its signalling (monoclonal antibodies, small-molecule tyrosine kinase inhibitors) and Insulin-Like Growth Factor Binding Proteins.
Collapse
Affiliation(s)
- C Gennigens
- Department of Medecine, Division of Hematology/Oncology, University Hospital of Liege, Belgium.
| | | | | |
Collapse
|
232
|
Sachdev D, Yee D. Inhibitors of insulin-like growth factor signaling: a therapeutic approach for breast cancer. J Mammary Gland Biol Neoplasia 2006; 11:27-39. [PMID: 16947084 DOI: 10.1007/s10911-006-9010-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The peptide growth factors IGF-I and IGF-II not only play a role in the development of the mammary gland but are also implicated in breast cancer. Several reagents disrupting IGF signaling have been developed and clinical trials validating IGF signaling as a target in cancer therapy are underway. This review highlights the approaches to inhibiting IGF signaling in breast cancer.
Collapse
Affiliation(s)
- Deepali Sachdev
- Department of Medicine and Cancer Center, University of Minnesota, Mayo Mail Code 806, 420 Delaware St, SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
233
|
Trojanek J, Croul S, Ho T, Wang JY, Darbinyan A, Nowicki M, Del Valle L, Skorski T, Khalili K, Reiss K. T-antigen of the human polyomavirus JC attenuates faithful DNA repair by forcing nuclear interaction between IRS-1 and Rad51. J Cell Physiol 2006; 206:35-46. [PMID: 15965906 DOI: 10.1002/jcp.20425] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
JC polyomavirus (JCV), which infects 90% of the human population, is detectable in human tumors. Its early protein, JCV T-antigen, transforms cells in vitro and is tumorigenic in experimental animals. Although T-antigen-mediated transformation involves genetic alterations of the affected cells, the mechanism underlying this genomic instability is not known. We show that JCV T-antigen inhibits homologous recombination DNA repair (HRR), which results in an accumulation of mutations. T-antigen does not operate directly but utilizes a cytosolic molecule, insulin receptor substrate 1 (IRS-1). Following T-antigen-mediated nuclear translocation, IRS-1 binds Rad51 at the site of damaged DNA. This T-antigen-mediated inhibition of HRR does not function in cells lacking IRS-1, and can be reproduced in the absence of T-antigen by IRS-1 with artificial nuclear localization signal. Our observations define a new mechanism by which viral protein utilizes cytosolic molecule to inhibit faithful DNA repair, and suggest how polyomaviruses could compromise stability of the genome. (c) 2005 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Joanna Trojanek
- Center for Neurovirology and Cancer Biology, College of Science and Technology, Temple University, Biology Life Science Building, Philadelphia, Pennsylvania 19122, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Affiliation(s)
- Adda Grimberg
- The Children's Hospital of Philadelphia, PA 19104-4318, USA
| |
Collapse
|
235
|
Yakar S, Leroith D, Brodt P. The role of the growth hormone/insulin-like growth factor axis in tumor growth and progression: Lessons from animal models. Cytokine Growth Factor Rev 2005; 16:407-20. [PMID: 15886048 DOI: 10.1016/j.cytogfr.2005.01.010] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Accepted: 01/20/2005] [Indexed: 02/08/2023]
Abstract
Over the past two decades it has become widely appreciated that a relationship exists between the insulin-like growth factors (IGFs) and cancer. Many cancers have been shown to overexpress the IGF-I receptor and produce the ligands (IGF-I or IGF-II) and some combinations of the six IGF-binding proteins. With the recent demonstration by epidemiological studies that an elevated serum IGF-I level is associated with an increased relative risk of developing a number of epithelial cancers, interest has been sparked in this area of research with the possibility of targeting the IGF-I receptor in cancer treatment protocols. This review highlights many of the most relevant studies in this exciting area of research, focusing in particular on lessons learned from animal models of cancer.
Collapse
Affiliation(s)
- Shoshana Yakar
- Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1758, USA.
| | | | | |
Collapse
|
236
|
Goel HL, Breen M, Zhang J, Das I, Aznavoorian-Cheshire S, Greenberg NM, Elgavish A, Languino LR. beta1A integrin expression is required for type 1 insulin-like growth factor receptor mitogenic and transforming activities and localization to focal contacts. Cancer Res 2005; 65:6692-700. [PMID: 16061650 DOI: 10.1158/0008-5472.can-04-4315] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cells' ability to proliferate in response to growth factor stimulation is significantly altered during cancer progression. To investigate the mechanisms underlying these alterations in prostate cancer, the role and expression of beta1A integrin and type 1 insulin-like growth factor receptor (IGF-IR), known to contribute to cell proliferation and transformation, were analyzed. Using small interfering RNA oligonucleotides to down-regulate beta1A, we show that beta1A expression is required for IGF-IR-mediated prostate cancer cell proliferation and anchorage-independent growth. In vivo, using age-matched transgenic adenocarcinoma of mouse prostate (TRAMP) mice at different stages of prostate cancer [prostatic intraepithelial neoplasia, PIN; well-differentiated adenocarcinoma, WD; and poorly differentiated adenocarcinoma, PD], the expression of beta1A and of IGF-IR was studied. beta1A and IGF-IR expression levels were concurrently up-regulated in high PIN and WD, whereas their expression did not correlate in late-stage PD. In contrast to the up-regulated expression of beta1A, the levels of beta1C, a beta1 cytoplasmic variant that inhibits cell proliferation, were down-regulated in all stages of prostate cancer. A similar expression pattern was observed for a beta1C downstream effector, Grb2-associated binder-1 (Gab1) which is known to inhibit IGF-IR phosphorylation. To analyze in vitro the mechanistic implications of beta1A, beta1C, and Gab1 deregulation in prostate cancer, we investigated whether expression of either beta1 variant in beta1-null cells affected IGF-IR localization. We found that IGF-IR and beta1A were colocalized in highly specialized integrin signaling compartments, designated focal contacts. However, in the presence of beta1C, IGF-IR remained diffuse on the cell surface and did not localize to focal contacts. The findings that beta1 integrins and IGF-IR are concurrently deregulated and that expression of beta1 integrins is necessary to achieve appropriate IGF-IR intracellular distribution point to the important role that the cross-talk between these receptors may have during prostate cancer progression and will be helpful in formulating new therapeutic strategies.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology and the Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Morgillo F, Lee HY. Resistance to epidermal growth factor receptor-targeted therapy. Drug Resist Updat 2005; 8:298-310. [PMID: 16172017 DOI: 10.1016/j.drup.2005.08.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Revised: 08/10/2005] [Accepted: 08/11/2005] [Indexed: 10/25/2022]
Abstract
The epidermal growth factor receptor (EGFR) has been a major target of molecular anticancer therapy. Two approaches have been developed, involving monoclonal antibodies and receptor tyrosine kinase inhibitors, and both have demonstrated benefit in clinical trials. However, evidence of resistance to these drugs has been described. Cellular levels of EGFR do not always correlate with response to the EGFR tyrosine kinase inhibitors, indicating acquired resistance to these drugs. Since EGFR antagonists interfere with the activation of several intracellular pathways that control cell proliferation, survival, apoptosis, angiogenesis, invasion and metastasis, acquired resistance can occur as a result of several different molecular mechanisms: autocrine/paracrine production of ligand, receptor mutation, constitutive activation of the downstream pathway and activation of alternative pathways. We will describe here potential mechanisms that can cause resistance to EGFR-targeted drugs. Combinations of EGFR antagonists with inhibitors targeting different signaling mechanism(s) - such as insulin-like growth factor receptor and vascular endothelial growth factor receptor - that share the same downstream mediator (e.g., phosphatidylinositol 3-kinase/Akt, mitogen-activated protein kinase), may circumvent or delay the development of resistance to EGFR antagonists resulting in enhanced antitumor activities.
Collapse
Affiliation(s)
- Floriana Morgillo
- Department of Thoracic/Head and Neck Medical Oncology, Unit 432, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | |
Collapse
|
238
|
DeAngelis T, Chen J, Wu A, Prisco M, Baserga R. Transformation by the simian virus 40 T antigen is regulated by IGF-I receptor and IRS-1 signaling. Oncogene 2005; 25:32-42. [PMID: 16170362 DOI: 10.1038/sj.onc.1209013] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Previous work has shown that the Simian Virus 40 T antigen (T antigen) cannot transform mouse embryo fibroblasts (MEFs) that do not express the type 1 insulin-like growth factor receptor (IGF-IR). We have now investigated the mechanism(s) by which the transforming activity of T antigen is affected by IGF-IR signaling. We demonstrate that transformation by T antigen of MEFs and several other cell lines requires an insulin receptor substrate-1 (IRS-1) phosphorylated on tyrosines. If IRS-1 is not expressed, or is serine phosphorylated or otherwise inactive, T antigen fails to transform cells in culture. For instance, while T antigen cannot transform 32D myeloid cells (that do not express IRS-1), its transforming activity is restored by the expression of a wild-type IRS-1, but not of an IRS-1 mutated at the PI3K binding sites. The importance of IRS-1 activation of PI3K in T-antigen transformation is supported by the finding that a constitutively activated p110 subunit of PI3K, a target of IRS-1, overcomes the inability of T antigen to transform MEFs with a serine phosphorylated IRS-1. Taken together, these results indicate that the IRS-1/PI3K signaling is one of the mechanisms regulating transformation by the SV40 T antigen. We propose that the requirement for a tyrosyl-phosphorylated IRS-1 provides a mechanism to explain the failure of T antigen to transform MEFs with deleted IGF-IR genes.
Collapse
MESH Headings
- Agar/chemistry
- Animals
- Antigens, Polyomavirus Transforming/chemistry
- Antigens, Polyomavirus Transforming/metabolism
- Antigens, Viral, Tumor/chemistry
- Binding Sites
- Blotting, Western
- Breast Neoplasms/metabolism
- Cell Line
- Cell Line, Transformed
- Cell Survival
- Cell Transformation, Neoplastic
- Cells, Cultured
- Fibroblasts/metabolism
- Gene Deletion
- Insulin Receptor Substrate Proteins
- Mice
- Mutation
- Neurons/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoproteins/metabolism
- Phosphorylation
- Pol1 Transcription Initiation Complex Proteins/metabolism
- RNA/chemistry
- RNA, Ribosomal/chemistry
- Receptor, IGF Type 1/metabolism
- Ribosomes/metabolism
- Serine/chemistry
- Signal Transduction
- Time Factors
- Transfection
- Tyrosine/chemistry
Collapse
Affiliation(s)
- T DeAngelis
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | |
Collapse
|
239
|
Jiang Y, Wang L, Gong W, Wei D, Le X, Yao J, Ajani J, Abbruzzese JL, Huang S, Xie K. A high expression level of insulin-like growth factor I receptor is associated with increased expression of transcription factor Sp1 and regional lymph node metastasis of human gastric cancer. Clin Exp Metastasis 2005; 21:755-64. [PMID: 16035620 DOI: 10.1007/s10585-005-1198-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor I receptor (IGF-IR) is critical to cell survival and growth and altered IGF-IR expression is found in many human cancers. However, its expression and potential role in gastric cancer development and progression has not been explored. The IGF-IR expression level was determined via immunohistochemistry in primary tumor and lymph node metastasis of 86 cases of resected gastric cancer. Relationships of IGF-IR expression with transcription factor Spl expression and clinicopathological features were analyzed. The impact of altered Sp1 expression on IGF-IR expression and gastric cancer biology was further determined using small inhibitory RNA for Sp1 mRNA. We found that IGF-IR was overexpressed in 62% of the tumor samples when compared with adjacent tumor-free gastric mucosa. Patients with lymph node metastases had strong expression of IGF-IR in both primary and metastatic tumor cells. IGF-IR overexpression in the primary tumor correlated with increased lymph node metastasis. Furthermore, the level of IGF-IR expression directly correlated with that of Spl, an important transcription factor for IGF-IR regulation. Knocking-down of Spl expression by small inhibitory RNA led to decreased IGF-IR expression and attenuated growth and metastasis of gastric cancer cells. Therefore, dysregulated expression of IGF-IR and/or Sp1 may contribute to the growth and metastasis of gastric cancer and potentially can be a target of therapeutic intervention.
Collapse
Affiliation(s)
- Yixing Jiang
- Department of Gastrointestinal Medical Oncology, The University of Texas, MD, Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Abstract
This review examines the rationale for targeting the insulin-like growth factor (IGF)-I receptor in the therapy of human tumours and their metastases. The rationale is based on two crucial findings: 1) in experimental animals, normal cells are only partially affected by the deletion of the IGF-I receptor, whereas tumour cells undergo apoptosis when the IGF-I receptor is downregulated; and 2) cells with a deleted IGF-I receptor are refractory to transformation by viral and cellular oncogenes. This review focuses on the mechanisms underlying the experimental findings, and discusses the possibility of extrapolating the results obtained in animals to the cure of human tumours.
Collapse
Affiliation(s)
- Renato Baserga
- Thomas Jefferson University, Kimmel Cancer Center, 233 S. 10th Street, 624 BLSB, Philadelphia, PA 19107, USA.
| |
Collapse
|
241
|
Reiss K, Khalili K, Giordano A, Trojanek J. JC virus large T-antigen and IGF-I signaling system merge to affect DNA repair and genomic integrity. J Cell Physiol 2005; 206:295-300. [PMID: 15991250 DOI: 10.1002/jcp.20455] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The progression of cancer is often associated with genomic instability, which may develop as a result of compromised defense mechanisms responsible for the maintenance of chromosomal integrity. These include defects in telomere preservation, chromosomal segregation, and DNA repair. In this review, we discuss molecular interactions between viral and cellular signaling components, which interfere with DNA repair mechanisms, and possibly contribute to the development of a mutagenic phenotype. Our studies indicate that large T-antigen from the human polyomavirus JC (JCV T-antigen) inhibits homologous recombination directed DNA repair (HRR)-causing accumulation of mutations in the affected cells (JCP 2005, in press). Surprisingly, T-antigen does not operate directly, but utilizes insulin receptor substrate 1 (IRS-1), which is the major signaling molecule for insulin-like growth factor I receptor (IGF-IR). Following T-antigen-mediated nuclear translocation, IRS-1 binds Rad51 at the site of damaged DNA. This T-antigen-mediated inhibition of HRR does not function in cells lacking IRS-1, and can be reproduced in the absence of T-antigen by IRS-1 with an artificial nuclear localization signal. The interplay described between the IGF-IR signaling system and JCV T-antigen in the process of DNA repair could be relevant, since nearly 90% of the human population is seropositive for JC virus, JCV T-antigen transforms cells in vitro, is tumorigenic in experimental animals, and the presence of JC virus has been shown in an increasing number of biopsies of human cancer.
Collapse
Affiliation(s)
- Krzysztof Reiss
- Center for Neurovirology and Cancer Biology, Temple University, 1900 North 12th Street, Biology Life Science Building, Philadelphia, Pennsylvania 19122, USA.
| | | | | | | |
Collapse
|
242
|
Chen J, Wu A, Sun H, Drakas R, Garofalo C, Cascio S, Surmacz E, Baserga R. Functional significance of type 1 insulin-like growth factor-mediated nuclear translocation of the insulin receptor substrate-1 and beta-catenin. J Biol Chem 2005; 280:29912-20. [PMID: 15967802 DOI: 10.1074/jbc.m504516200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous work has shown that the transcriptional regulator beta-catenin can translocate to the nuclei when cells are stimulated with the type 1 insulin-like growth factor (IGF-1). We show by immunocoprecipitation and by confocal microscopy that beta-catenin binds to and co-localizes with the insulin receptor substrate-1 (IRS-1), a docking protein for both the insulin and the IGF-1 receptors. IRS-1 is required for IGF-1-mediated nuclear translocation of beta-catenin, resulting in the activation of the beta-catenin target genes. IGF-1-mediated nuclear translocation of beta-catenin is facilitated by the nuclear translocation of IRS-1. Both IRS-1 and beta-catenin are recruited to the cyclin D1 promoter, an established target for beta-catenin, but only IRS-1 is recruited to the ribosomal DNA (rDNA) promoter. UBF proteins (known to interact with both IRS-1 and beta-catenin) are also detectable in the cyclin D1 and rDNA promoters. These results indicate that IRS-1 (activated by the IGF-1 receptor) is one of several proteins that regulate the subcellular localization and activity of beta-catenin. The ability of IRS-1 to localize to both RNA polymerase II (with beta-catenin) and RNA polymerase I-regulated promoters suggest an explanation for the effect of IRS-1 on both cell growth in size and cell proliferation. This possibility is supported by the demonstration that enforced nuclear localization of IRS-1 causes nuclear translocation of beta-catenin and transformation of normal mouse embryo fibroblasts (colony formation in soft agar).
Collapse
Affiliation(s)
- Jia Chen
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
243
|
Alexia C, Lasfer M, Groyer A. Role of constitutively activated and insulin-like growth factor-stimulated ERK1/2 signaling in human hepatoma cell proliferation and apoptosis: evidence for heterogeneity of tumor cell lines. Ann N Y Acad Sci 2005; 1030:219-29. [PMID: 15659801 DOI: 10.1196/annals.1329.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Enhanced insulin-like growth factor II (IGF-II) and type I IGF receptor (IGF-IR) gene expression in liver tumors and the development of liver tumors in transgenic mice overexpressing IGF-II in the liver suggest that the IGFs and underlying signaling cascades may play auto/paracrine roles in the control of hepatocarcinoma (HCC) cell proliferation and in their protection against apoptosis. We have focused on the role of mitogen-activated protein kinase (ERK1/2) signaling on human HepG2 and Huh-7 hepatoma cell proliferation and on the protection of these cells against drug-induced apoptosis. Physiological concentrations of IGF-I stimulated DNA replication in HepG2 cells (1.5-fold) but not in Huh-7 cells, and this effect was abolished by PD98059 (MEK-1 inhibitor). Doxorubicin or cisplatin treatment induced apoptosis (caspase-dependent poly[ADP-ribose]polymerase cleavage) in both cell lines, but dose-dependent reversion of drug-induced apoptosis (57-84%) by IGF-I was only observed in HepG2 cells. The very low level of IGF-IR at the plasma membrane of Huh-7 cells may account for their unresponsiveness to IGF-I. We have shown that drug treatment enhanced (17-fold) or did not modify constitutive ERK1/2 activity in cultured HepG2 or Huh-7 cells, respectively. In both cell lines, inhibition of constitutive and drug-induced ERK1/2 activity by PD98059 yielded a complete inhibition of drug-induced apoptosis. Altogether, our data demonstrate the heterogeneous response of human hepatoma cells to an IGF stimulus and suggest (1) that auto/paracrine effects of IGF-I/-II might contribute to the proliferation of HCC cells and to their protection against apoptosis in vivo and (2) that drug-induced activation of ERK1/2 plays a role in drug-induced apoptosis in human hepatoma cells.
Collapse
Affiliation(s)
- Catherine Alexia
- Institut National de la Sané et de la Recherche Médicale U481, Faculté de Médecine Xavier Bichat, 16 rue Henri Huchard, BP 416, 75870 Paris Cédex 18, France
| | | | | |
Collapse
|
244
|
Loughran G, Huigsloot M, Kiely PA, Smith LM, Floyd S, Ayllon V, O'Connor R. Gene expression profiles in cells transformed by overexpression of the IGF-I receptor. Oncogene 2005; 24:6185-93. [PMID: 15940254 DOI: 10.1038/sj.onc.1208772] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To identify genes associated with insulin-like growth factor-I receptor (IGF-IR)-mediated cellular transformation, we isolated genes that are differentially expressed in R- cells (derived from the IGF-IR knockout mouse) and R+ cells (R- cells that overexpress the IGF-IR). From these, 45 genes of known function were expressed at higher levels in R+ cells and 22 were expressed at higher levels in R- cells. Differential expression was confirmed by Northern blot analysis of R+ and R- cells. Genes expressed more abundantly in R+ cells are associated with (1) tumour growth and metastasis including, betaigH3, mts1, igfbp5 protease, and mystique; (2) cell division, including cyclin A1 and cdk1; (3) signal transduction, including pkcdeltabp and lmw-ptp; and (4) metabolism including ATPase H+ transporter and ferritin. In MCF-7 cells IGF-I induced expression of two genes, lasp-1 and mystique, which could contribute to metastasis. Lasp-1 expression required activity of the PI3-kinase signalling pathway. Mystique was highly expressed in metastatic but not in androgen-dependent prostate cancer cell lines and Mystique overexpression in MCF-7 cells promoted cell migration and invasion. We conclude that genes identified in this screen may mediate IGF-IR function in cancer progression.
Collapse
Affiliation(s)
- Gary Loughran
- Cell Biology Laboratory, Department of Biochemistry, BioSciences Institute, National University of Ireland, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
245
|
Hao S, Bi X, Su L, Dong W, Moyana T, Xiang J. Molecular and immunophenotypical characterization of progressive and regressive leukemia cell lines. Cancer Biother Radiopharm 2005; 20:290-299. [PMID: 15989474 DOI: 10.1089/cbr.2005.20.290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The P815 and P198 cell lines are clonally related mouse mastocytoma cell lines. They differ in their biologic behavior in that P815 is a progressive tumor cell line, whereas P198 is a regressive one. These cell lines have been extensively used as models for the study of tumor-host relationships and tumor immunology. Although some of their biological properties have been well documented, the molecular mechanisms underlying tumor progression or regression have not been completely elucidated. In this study, we characterized the growth behavior and immunophenotype of these two cell lines, and analyzed their gene profiles using a complementary deoxynucleic acid (cDNA) microarray composed of 514 immunologically relevant genes. Our data showed that the two cell lines exhibited quite dissimilar and contrasting growth characteristics when inoculated into syngeneic mice. P815 tumors grew unremittingly, while P198 tumors gradually regressed. From a molecular viewpoint, P815 cells showed a higher expression of genes promoting tumor growth, such as IGF-1, IL-8R, FGFR1, VEGF-A, and VEGF-B. On the other hand, P198 tumor cells expressed CD11b and CD80, which favor the recruitment of lymphocytes and antigen-presenting cells (APCs), as well as the elicitation of antitumor immunity. P198 tumor cells also depicted a higher expression of genes inhibiting tumor growth, such as TNF-alpha, SOCS-1, CIS1, 4-1BB, and GDF-10. In conclusion, our results contribute further information in the understanding of the molecular mechanisms associated with the regression and progression of P815 and P198 tumor cells.
Collapse
Affiliation(s)
- Siguo Hao
- Research Unit, Saskatchewan Cancer Agency, Department of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | | | | | | | | | | |
Collapse
|
246
|
Meng Z, King PH, Nabors LB, Jackson NL, Chen CY, Emanuel PD, Blume SW. The ELAV RNA-stability factor HuR binds the 5'-untranslated region of the human IGF-IR transcript and differentially represses cap-dependent and IRES-mediated translation. Nucleic Acids Res 2005; 33:2962-79. [PMID: 15914670 PMCID: PMC1140080 DOI: 10.1093/nar/gki603] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The type I insulin-like growth factor receptor (IGF-IR) is an integral component in the control of cell proliferation, differentiation and apoptosis. The IGF-IR mRNA contains an extraordinarily long (1038 nt) 5'-untranslated region (5'-UTR), and we have characterized a diverse series of proteins interacting with this RNA sequence which may provide for intricate regulation of IGF-IR gene expression at the translational level. Here, we report the purification and identification of one of these IGF-IR 5'-UTR-binding proteins as HuR, using a novel RNA crosslinking/RNase elution strategy. Because HuR has been predominantly characterized as a 3'-UTR-binding protein, enhancing mRNA stability and generally increasing gene expression, we sought to determine whether HuR might serve a different function in the context of its binding the IGF-IR 5'-UTR. We found that HuR consistently repressed translation initiation through the IGF-IR 5'-UTR. The inhibition of translation by HuR was concentration dependent, and could be reversed in trans by addition of a fragment of the IGF-IR 5'-UTR containing the HuR binding sites as a specific competitor, or abrogated by deletion of the third RNA recognition motif of HuR. We determined that HuR repressed translation initiation through the IGF-IR 5'-UTR in cells as well, and that siRNA knockdown of HuR markedly increased IGF-IR protein levels. Interestingly, we also found that HuR potently inhibited IGF-IR translation mediated through internal ribosome entry. Kinetic assays were performed to investigate the mechanism of translation repression by HuR and the dynamic interplay between HuR and the translation apparatus. We found that HuR, occupying a cap-distal position, significantly delayed translation initiation mediated by cap-dependent scanning, but was eventually displaced from its binding site, directly or indirectly, as a consequence of ribosomal scanning. However, HuR perpetually blocked the activity of the IGF-IR IRES, apparently arresting the IRES-associated translation pre-initiation complex in an inactive state. This function of HuR as a 5'-UTR-binding protein and dual-purpose translation repressor may be critical for the precise regulation of IGF-IR expression essential to normal cellular homeostasis.
Collapse
Affiliation(s)
- Zheng Meng
- Department of Biochemistry and Molecular Genetics, University of Alabama at BirminghamBirmingham, AL, USA
| | - Peter H. King
- Department of Neurology, University of Alabama at BirminghamBirmingham, AL, USA
- Birmingham Veterans Affairs Medical CenterBirmingham, AL 35294, USA
| | - L. Burt Nabors
- Department of Neurology, University of Alabama at BirminghamBirmingham, AL, USA
| | - Nateka L. Jackson
- Department of Medicine, University of Alabama at BirminghamBirmingham, AL, USA
| | - Ching-Yi Chen
- Department of Biochemistry and Molecular Genetics, University of Alabama at BirminghamBirmingham, AL, USA
| | - Peter D. Emanuel
- Department of Biochemistry and Molecular Genetics, University of Alabama at BirminghamBirmingham, AL, USA
- Department of Medicine, University of Alabama at BirminghamBirmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at BirminghamBirmingham, AL, USA
| | - Scott W. Blume
- Department of Biochemistry and Molecular Genetics, University of Alabama at BirminghamBirmingham, AL, USA
- Department of Medicine, University of Alabama at BirminghamBirmingham, AL, USA
- Comprehensive Cancer Center, University of Alabama at BirminghamBirmingham, AL, USA
- To whom correspondence should be addressed at 1824 6th Avenue South, Wallace Tumor Institute, Room 508, University of Alabama at Birmingham, Birmingham, AL 35294, USA. Tel: +1 205 975 2409; Fax: +1 205 975 6911;
| |
Collapse
|
247
|
Min Y, Adachi Y, Yamamoto H, Imsumran A, Arimura Y, Endo T, Hinoda Y, Lee CT, Nadaf S, Carbone DP, Imai K. Insulin-like growth factor I receptor blockade enhances chemotherapy and radiation responses and inhibits tumour growth in human gastric cancer xenografts. Gut 2005; 54:591-600. [PMID: 15831900 PMCID: PMC1774471 DOI: 10.1136/gut.2004.048926] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 12/08/2004] [Accepted: 12/21/2004] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Insulin-like growth factor (IGF) I receptor (IGF-Ir) signalling is required for carcinogenicity and proliferation of many tumours but this pathway has not been studied in detail in gastric cancer. We have previously shown successful therapy for colorectal, pancreatic, and lung cancer using recombinant adenoviruses expressing dominant negative (dn) IGF-Ir. In this study, we sought to better dissect the role of IGF-Ir on progression of gastric cancer and determine whether IGF-Ir targeted adenoviruses represent potentially effective therapeutics for human gastric cancer. METHODS We assessed the effect of IGF-Ir ligands on proliferation and survival in gastric cancer cells in culture. Then, recombinant adenoviruses expressing truncated IGF-Ir (482 and 950 amino acids long, IGF-Ir/dn) that function as dn inhibitors were studied in the treatment of human gastric cancer xenografts. We characterised the effects of IGF-Ir/dn on signalling blockade, growth, apoptosis induction, and in vivo therapeutic efficacy. RESULTS IGF-Ir signalling promoted tumour growth and survival in gastric cancer. IGF-Ir/dn expression suppressed tumorigenicity both in vitro and in vivo and upregulated stressor induced apoptosis. IGF-Ir/dn blocked Akt-1 activation induced by IGF-I, IGF-II, and des(1-3)IGF-I, but not by insulin. IGF-Ir/dn expression increased radiation and chemotherapy induced apoptosis and the combination of IGF-Ir/dn and chemotherapy was very effective against tumours in mice. In an intraperitoneal model, IGF-Ir/dn therapy also suppressed peritoneal dissemination. CONCLUSIONS IGF-Ir is involved in the regulation of survival and cell growth in human gastric cancer and may be a good molecular therapeutic target. Adenovirus-IGF-Ir/dn may thus have therapeutic use in gastric cancer.
Collapse
Affiliation(s)
- Y Min
- First Department of Internal Medicine, Sapporo Medical University, S-1, W-16, Chuo-ku, Sapporo 060-8543, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Lee KW, Ma L, Yan X, Liu B, Zhang XK, Cohen P. Rapid Apoptosis Induction by IGFBP-3 Involves an Insulin-like Growth Factor-independent Nucleomitochondrial Translocation of RXRα/Nur77. J Biol Chem 2005; 280:16942-8. [PMID: 15731112 DOI: 10.1074/jbc.m412757200] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Insulin-like growth factor-binding protein-3 (IGFBP-3) induces apoptosis by its ability to bind insulin-like growth factors (IGFs) as well as its IGF-independent effects involving binding to other molecules including the retinoid X receptor-alpha (RXRalpha). Here we describe that in response to IGFBP-3, the RXRalpha binding partner nuclear receptor Nur77 rapidly undergoes translocation from the nucleus to the mitochondria, initiating an apoptotic cascade resulting in caspase activation within 6 h. This translocation is a type 1 IGF receptor-signaling independent event as IGFBP-3 induces Nur77 translocation in R-cells. IGFBP-3 and Nur77 are additive in inducing apoptosis. GFP-Nur77 transfection into RXRalpha wild-type and knock-out mouse embryonic fibroblasts and subsequent treatment with IGFBP-3 show that RXRalpha is required for IGFBP-3-induced Nur77 translocation and apoptosis. Addition of IGFBP-3 to 22RV1 cell lysates enhanced the ability of GST-RXRalpha to "pull down" Nur77, and overexpression of IGFBP-3 enhanced the accumulation of mitochondrial RXRalpha. This unique nongenotropic nuclear pathway supports an emerging role for IGFBP-3 as a novel, multicompartmental signaling molecule involved in induction of apoptosis in malignant cells.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Apoptosis
- Blotting, Western
- Caspases/metabolism
- Cell Nucleus/metabolism
- Cells, Cultured
- Cytoplasm/metabolism
- DNA-Binding Proteins/metabolism
- Densitometry
- Dimerization
- Enzyme Activation
- Enzyme-Linked Immunosorbent Assay
- Fibroblasts/metabolism
- Fluorescent Antibody Technique, Indirect
- Glutathione Transferase/metabolism
- Insulin-Like Growth Factor Binding Protein 3/metabolism
- Insulin-Like Growth Factor Binding Protein 3/physiology
- Mice
- Microscopy, Fluorescence
- Mitochondria/metabolism
- Mutagenesis, Site-Directed
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Protein Transport
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Steroid/metabolism
- Retinoid X Receptor alpha/metabolism
- Signal Transduction
- Somatomedins/metabolism
- Time Factors
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- Kuk-Wha Lee
- Division of Pediatric Endocrinology, Mattel Children's Hospital at UCLA, David Geffen School of Medicine, Los Angeles, California 90095, USA
| | | | | | | | | | | |
Collapse
|
249
|
Foulstone E, Prince S, Zaccheo O, Burns JL, Harper J, Jacobs C, Church D, Hassan AB. Insulin-like growth factor ligands, receptors, and binding proteins in cancer. J Pathol 2005; 205:145-53. [PMID: 15641016 DOI: 10.1002/path.1712] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This review aims to summarize experimental evidence supporting the role of the insulin-like growth factor (IGF) signalling system in the progression, maintenance, and treatment of cancer. These data implicate the IGF system as an important modifier of cancer cell proliferation, survival, growth, and treatment sensitivity. The role of the IGF system in cancer should be examined in the context of the extra-cellular and intra-cellular signalling networks, in particular: phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt/PKB), mammalian target of rapamycin (mTOR), and forkhead transcription factors (FOXO). This review highlights evidence derived from molecular structure and functional genetics with respect to how the extra-cellular components of the IGF system function normally, and their subsequent modifications in cancer. The therapeutic relevance of the research evidence described is also addressed, as the challenge is to apply this knowledge to human health.
Collapse
Affiliation(s)
- E Foulstone
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | | | | | | | |
Collapse
|
250
|
Goetsch L, Gonzalez A, Leger O, Beck A, Pauwels PJ, Haeuw JF, Corvaia N. A recombinant humanized anti-insulin-like growth factor receptor type I antibody (h7C10) enhances the antitumor activity of vinorelbine and anti-epidermal growth factor receptor therapy against human cancer xenografts. Int J Cancer 2005; 113:316-28. [PMID: 15386423 DOI: 10.1002/ijc.20543] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interaction of insulin-like growth factor receptor I (IGF-IR) with its ligands has been reported to induce cell proliferation, transformation and blockade of cell apoptotic functions. IGF-IR is overexpressed on numerous tumor cell types and its blockade could be of importance for anti-cancer therapy. We have generated a humanized anti-IGF-IR antibody h7C10 that blocks in vitro IGF-I and IGF-II-induced cell proliferation of MCF-7 breast cancer cells. Analysis of the IGF-I transduction cascade demonstrated that the humanized anti-IGF-IR antibody and its murine parental form block IGF-I-induced tyrosine phosphorylation, both its beta-chain and IRS-1 tyrosine phosphorylation. This presumably leads to cell cycle arrest and, consequently, growth inhibition. Treatment of nude mice bearing either human breast cancer cells (MCF-7) or non small lung cancer cells (A549) with h7C10, or its murine parental form 7C10, inhibited significantly tumor growth. An almost complete inhibition of A549 tumor growth was observed when mice were treated with the anti-IGF-IR antibody combined with either a chemotherapeutic agent, Vinorelbine or an anti-epidermal growth factor receptor (EGFR) antibody, 225. Combined therapy prolonged significantly the life span of mice in an orthotopic in vivo model of A549; the combination of the anti-IGF-IR antibody with an anti-EGFR antibody was superior to the Vinorelbine combination. The present results indicate that the humanized anti-IGF-IR antibody h7C10 has a great potential for cancer therapy when combined with either a chemotherapeutic agent or an antibody that targets other growth factor receptors, such as the epidermal growth factor receptor.
Collapse
Affiliation(s)
- Liliane Goetsch
- Centre d'Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160, St. Julien en Genevois, France.
| | | | | | | | | | | | | |
Collapse
|