391
Views
38
CrossRef citations to date
0
Altmetric
Reviews

PDGFRα: a new therapeutic target in the treatment of hepatocellular carcinoma?

&
Pages 443-454 | Published online: 31 Mar 2009

Bibliography

  • Ferlay J, Bray F, Pisani P, et al. GLOBOCAN 2002: Cancer Incidence, Mortality and Prevalence Worldwide. IARC CancerBase. Lyon, IARC Press;2002
  • El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 2007;132:2557-76
  • Bonilla Guerrero R and Roberts LR. The role of hepatitis B virus integrations in the pathogenesis of human hepatocellular carcinoma. J Hepatol 2005;42:760-77
  • Sarpel U, Schwartz M. Liver transplantation for hepatocellular carcinoma. Hepatol Res 2007;37(Suppl 2):S264-6
  • Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378-90
  • Roberts LR. Sorafenib in liver cancer-just the beginning. N Engl J Med 2008;359:420-2
  • Sandhu D, Tharayil V, Lai J, et al. Treatment options for hepatocellular carcinoma. Expert Rev Gastroenterol Hepatol 2008;2:81-92
  • Roberts LR, Gores GJ. Emerging drugs for hepatocellular carcinoma. Expert Opin Emerg Drugs 2006;11:469-87
  • Lorusso PM, Eder JP. Therapeutic potential of novel selective-spectrum kinase inhibitors in oncology. Expert Opin Investig Drugs 2008;17:1013-28
  • Fredriksson L, Li H, Eriksson U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev 2004;15:197-204
  • McDonald NQ, Hendrickson WA. A structural superfamily of growth factors containing a cystine knot motif. Cell 1993;73:421-4
  • Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev 2008;22:1276-312
  • Betsholtz C, Johnsson A, Heldin CH, et al. cDNA sequence and chromosomal localization of human platelet-derived growth factor A-chain and its expression in tumour cell lines. Nature 1986;320:695-9
  • Uutela M, Lauren J, Bergsten E, et al. Chromosomal location, exon structure, and vascular expression patterns of the human PDGFC and PDGFC genes. Circulation 2001;103:2242-7
  • Claesson-Welch L. PDGF receptors: structure and mechanism of action, Basel: Karger 1993
  • Heldin CH, Eriksson U and Ostman A. New members of the platelet-derived growth factor family of mitogens. Arch Biochem Biophys 2002;398:284-90
  • Pietras K, Sjoblom T, Rubin K, et al. PDGF receptors as cancer drug targets. Cancer Cell 2003;3:439-43
  • Ostman A. PDGF receptors–mediators of autocrine tumor growth and regulators of tumor vasculature and stroma. Cytokine Growth Factor Rev 2004;15:275-86
  • Pietras K, Pahler J, Bergers G, et al. Functions of paracrine PDGF signaling in the proangiogenic tumor stroma revealed by pharmacological targeting. PLoS Med 2008;5:e19, published online 29 January 2008,doi:10.1371/journal.pmed.0050019
  • Fleming TP, Saxena A, Clark WC, et al. Amplification and/or overexpression of platelet-derived growth factor receptors and epidermal growth factor receptor in human glial tumors. Cancer Res 1992;52:4550-3
  • Hermanson M, Funa K, Koopmann J, et al. Association of loss of heterozygosity on chromosome 17p with high platelet-derived growth factor α receptor expression in human malignant gliomas. Cancer Res 1996;56:164-71
  • Cools J, DeAngelo DJ, Gotlib J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med 2003;348:1201-14
  • Griffin JH, Leung J, Bruner RJ, et al. Discovery of a fusion kinase in EOL-1 cells and idiopathic hypereosinophilic syndrome. Proc Natl Acad Sci USA 2003;100:7830-5
  • Stover EH, Chen J, Folens C, et al. Activation of FIP1L1-PDGFRα requires disruption of the juxtamembrane domain of PDGFRα and is FIP1L1-independent. Proc Natl Acad Sci USA 2006;103:8078-83
  • Heldin NE, Gustavsson B, Claesson-Welsh L, et al. Aberrant expression of receptors for platelet-derived growth factor in an anaplastic thyroid carcinoma cell line. Proc Natl Acad Sci USA 1988;85:9302-6
  • Jechlinger M, Sommer A, Moriggl R, et al. Autocrine PDGFR signaling promotes mammary cancer metastasis. J Clin Invest 2006;116:1561-70
  • Seymour L, Dajee D, Bezwoda WR. Tissue platelet derived-growth factor (PDGF) predicts for shortened survival and treatment failure in advanced breast cancer. Breast Cancer Res Treat 1993;26:247-52
  • Romanque P, Piguet AC, Dufour JF. Targeting vessels to treat hepatocellular carcinoma. Clin Sci (Lond) 2008;114:467-77
  • Beckermann BM, Kallifatidis G, Groth A, et al. VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br J Cancer 2008;99:622-31
  • Lange S, Heger J, Euler G, et al. Platelet-derived growth factor BB stimulates vasculogenesis of embryonic stem cell-derived endothelial cells by calcium-mediated generation of reactive oxygen species. Cardiovasc Res 2009;81:159-68
  • Ball SG, Shuttleworth CA, Kielty CM. Vascular endothelial growth factor can signal through platelet-derived growth factor receptors. J Cell Biol 2007;177:489-500
  • Zeng W, Gouw AS, van den Heuvel MC, et al. The angiogenic makeup of human hepatocellular carcinoma does not favor vascular endothelial growth factor/angiopoietin-driven sprouting neovascularization. Hepatology 2008;48:1517-27
  • Risau W, Drexler H, Mironov V, et al. Platelet-derived growth factor is angiogenic in vivo. Growth Factors 1992;7:261-6
  • Cao R, Brakenhielm E, Li X, et al. Angiogenesis stimulated by PDGF-CC, a novel member in the PDGF family, involves activation of PDGFR-αα and -αβ receptors. FASEB J 2002;16:1575-83
  • Hermanson M, Funa K, Hartman M, et al. Platelet-derived growth factor and its receptors in human glioma tissue: expression of messenger RNA and protein suggests the presence of autocrine and paracrine loops. Cancer Res 1992;52:3213-9
  • Lindmark G, Sundberg C, Glimelius B, et al. Stromal expression of platelet-derived growth factor β-receptor and platelet-derived growth factor B-chain in colorectal cancer. Lab Invest 1993;69:682-9
  • Li H, Fredriksson L, Li X, et al. PDGF-D is a potent transforming and angiogenic growth factor. Oncogene 2003;22:1501-10
  • Tejada ML, Yu L, Dong J, et al. Tumor-driven paracrine platelet-derived growth factor receptor α signaling is a key determinant of stromal cell recruitment in a model of human lung carcinoma. Clin Cancer Res 2006;12:2676-88
  • Li J, Hoyle GW. Overexpression of PDGF-A in the lung epithelium of transgenic mice produces a lethal phenotype associated with hyperplasia of mesenchymal cells. Dev Biol 2001;239:338-49
  • Ostman A, Heldin CH. Involvement of platelet-derived growth factor in disease: development of specific antagonists. Adv Cancer Res 2001;80:1-38
  • Jones A, Cross NC. Oncogenic derivatives of platelet-derived growth factor receptors. Cell Mol Life Sci 2004;61:2912-23
  • Henriksen R, Funa K, Wilander E, et al. Expression and prognostic significance of platelet-derived growth factor and its receptors in epithelial ovarian neoplasms. Cancer Res 1993;53:4550-4
  • Apte SM, Bucana CD, Killion JJ, et al. Expression of platelet-derived growth factor and activated receptor in clinical specimens of epithelial ovarian cancer and ovarian carcinoma cell lines. Gynecol Oncol 2004;93:78-86
  • Fudge K, Wang CY, Stearns ME. Immunohistochemistry analysis of platelet-derived growth factor A and B chains and platelet-derived growth factor α and β receptor expression in benign prostatic hyperplasias and Gleason-graded human prostate adenocarcinomas. Mod Pathol 1994;7:549-54
  • de Jong JS, van Diest PJ, van der Valk P et al. Expression of growth factors, growth inhibiting factors, and their receptors in invasive breast cancer. I: An inventory in search of autocrine and paracrine loops. J Pathol 1998;184:44-52
  • Zhang P, Gao WY, Turner S, et al. Gleevec (STI-571) inhibits lung cancer cell growth (A549) and potentiates the cisplatin effect in vitro. Mol Cancer 2003;2:1, published online 3 January 2003,doi:10.1186/1476-4598-2-1
  • Barnhill RL, Xiao M, Graves D, et al. Expression of platelet-derived growth factor (PDGF)-A, PDGF-B and the PDGF-α receptor, but not the PDGF-β receptor, in human malignant melanoma in vivo. Br J Dermatol 1996;135:898-904
  • Sulzbacher I, Traxler M, Mosberger I, et al. Platelet-derived growth factor-AA and -α receptor expression suggests an autocrine and/or paracrine loop in osteosarcoma. Mod Pathol 2000;13:632-7
  • Sulzbacher I, Birner P, Traxler M, et al. Expression of platelet-derived growth factor-αα receptor is associated with tumor progression in clear cell renal cell carcinoma. Am J Clin Pathol 2003;120:107-12
  • Carvalho I, Milanezi F, Martins A, et al. Overexpression of platelet-derived growth factor receptor α in breast cancer is associated with tumour progression. Breast Cancer Res 2005;7:R788-95
  • Galmiche MC, Koteliansky VE, Briere J, et al. Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway. Blood 1993;82:66-76
  • Kashiwakura Y, Katoh Y, Tamayose K, et al. Isolation of bone marrow stromal cell-derived smooth muscle cells by a human SM22α promoter: in vitro differentiation of putative smooth muscle progenitor cells of bone marrow. Circulation 2003;107:2078-81
  • George D. Targeting PDGF receptors in cancer–rationales and proof of concept clinical trials. Adv Exp Med Biol 2003;532:141-51
  • Loizos N, Xu Y, Huber J, et al. Targeting the platelet-derived growth factor receptor α with a neutralizing human monoclonal antibody inhibits the growth of tumor xenografts: Implications as a potential therapeutic target. Mol Cancer Ther 2005;4:369-79
  • Youssoufian H, Amato R, Sweeney C, et al. Phase 1 study of IMC-3G2, an IgG1 monoclonal antibody targeting PDGFRα in patients with advanced solid malignancies, J Clin Oncol 200826:636s
  • Stock P, Monga D, Tan X, et al. Platelet-derived growth factor receptor-α: a novel therapeutic target in human hepatocellular cancer. Mol Cancer Ther 2007;6:1932-41
  • Zhang T, Sun HC, Xu Y, et al. Overexpression of platelet-derived growth factor receptor α in endothelial cells of hepatocellular carcinoma associated with high metastatic potential. Clin Cancer Res 2005;11:8557-63
  • Tsutsumi N, Yonemitsu Y, Shikada Y, et al. Essential role of PDGFRα-p70S6K signaling in mesenchymal cells during therapeutic and tumor angiogenesis in vivo: role of PDGFRα during angiogenesis. Circ Res 2004;94:1186-94
  • Campbell JS, Hughes SD, Gilbertson DG, et al. Platelet-derived growth factor C induces liver fibrosis, steatosis and hepatocellular carcinoma. Proc Natl Acad Sci USA 2005;102:3389-94
  • Campbell JS, Johnson MM, Bauer RL, et al. Targeting stromal cells for the treatment of platelet-derived growth factor C-induced hepatocellular carcinogenesis. Differentiation 2007;75:843-52
  • LaRochelle WJ, Jensen RA, Heidaran MA, et al. Inhibition of platelet-derived growth factor autocrine growth stimulation by a monoclonal antibody to the human α platelet-derived growth factor receptor. Cell Growth Differ 1993;4:547-53
  • Iizuka N, Oka M, Yamada-Okabe H, et al. Oligonucleotide microarray for prediction of early intrahepatic recurrence of hepatocellular carcinoma after curative resection. Lancet 2003;361:923-9
  • Tackels-Horne D, Goodman MD, Williams AJ, et al. Identification of differentially expressed genes in hepatocellular carcinoma and metastatic liver tumors by oligonucleotide expression profiling. Cancer 2001;92:395-405
  • Pang RW, Poon RT. From molecular biology to targeted therapies for hepatocellular carcinoma: the future is now. Oncology 2007;72(Suppl 1):30-44
  • Pietras K, Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose “chemo-switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol 2005;23:939-52
  • Treiber G, Wex T, Malfertheiner P. Impact of different anticancer regimens on biomarkers of angiogenesis in patients with advanced hepatocellular cancer. J Cancer Res Clin Oncol 2009;135:271-81
  • Capdeville R, Buchdunger E, Zimmermann J, et al. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 2002;1:493-502
  • Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med 2001;344:1031-7
  • van Oosterom T, Judson I, Verweij J, et al. Safety and efficacy of imatinib (STI571) in metastatic gastrointestinal stromal tumours: a phase I study. Lancet 2001;358:1421-3
  • Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 2002;347:472-80
  • Andre C, Martin E, Cornu F, et al. Genomic organization of the human c-kit gene: evolution of the receptor tyrosine kinase subclass III. Oncogene 1992;7:685-91
  • Zwick E, Bange J, Ullrich A. Receptor tyrosine kinases as targets for anticancer drugs. Trends Mol Med 2002;8:17-23
  • Heinrich MC, Corless CL, Demetri GD, et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol 2003;21:4342-9
  • Debiec-rychter M, Dumez H, Judson I, et al. Use of c-KIT/PDGFRA mutational analysis to predict the clinical response to imatinib in patients with advanced gastrointestinal stromal tumours entered on phase I and II studies of the EORTC Soft Tissue and Bone Sarcoma Group. Eur J Cancer 2004;40:689-95
  • Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003;299:708-10
  • Hirota S, Ohashi A, Nishida T, et al. Gain-of-function mutations of platelet-derived growth factor receptor α gene in gastrointestinal stromal tumors. Gastroenterology 2003;125:660-7
  • Medeiros F, Corless CL, Duensing A, et al. KIT-negative gastrointestinal stromal tumors: proof of concept and therapeutic implications. Am J Surg Pathol 2004;28:889-94
  • Becker G, Schmitt-graeff A, Ertelt V, et al. CD117 (c-kit) expression in human hepatocellular carcinoma. Clin Oncol (R Coll Radiol) 2007;19:204-8
  • Ostman A, Heldin CH. PDGF receptors as targets in tumor treatment. Adv Cancer Res 2007;97:247-74
  • Ramadori G, Fuzesi L, Grabbe E, et al. Successful treatment of hepatocellular carcinoma with the tyrosine kinase inhibitor imatinib in a patient with liver cirrhosis. Anticancer Drugs 2004;15:405-9
  • Fujio K, Hu Z, Evarts RP, et al. Coexpression of stem cell factor and c-kit in embryonic and adult liver. Exp Cell Res 1996;224:243-50
  • Braun L, Mikumo R, Fausto N. Production of hepatocellular carcinoma by oval cells: cell cycle expression of c-myc and p53 at different stages of oval cell transformation. Cancer Res 1989;49:1554-61
  • Dumble ML, Croager EJ, Yeoh GC, et al. Generation and characterization of p53 null transformed hepatic progenitor cells: oval cells give rise to hepatocellular carcinoma. Carcinogenesis 2002;23:435-45
  • Burger H, den Bakker MA, Kros JM et al. Activating mutations in c-KIT and PDGFRα are exclusively found in gastrointestinal stromal tumors and not in other tumors overexpressing these imatinib mesylate target genes. Cancer Biol Ther 2005;4:1270-4
  • Sihto H, Sarlomo-Rikala M, Tynninen O, et al. KIT and platelet-derived growth factor receptor alpha tyrosine kinase gene mutations and KIT amplifications in human solid tumors. J Clin Oncol 2005;23:49-57
  • Lin AY, Fisher GA, So S, et al. Phase II study of imatinib in unresectable hepatocellular carcinoma. Am J Clin Oncol 2008;31:84-8
  • Eckel F, von Delius S, Mayr M, et al. Pharmacokinetic and clinical Phase II trial of imatinib in patients with impaired liver function and advanced hepatocellular carcinoma. Oncology 2005;69:363-71
  • Treiber G, Wex T, Schleyer E, et al. Imatinib for hepatocellular cancer-focus on pharmacokinetic/pharmacodynamic modelling and liver function. Cancer Lett 2008;260:146-54
  • Wilhelm SM, Carter C, Tang L, et al. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 2004;64:7099-109
  • Bergers G, Song S, Meyer-Morse N, et al. Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J Clin Invest 2003;111:1287-95
  • Abrams TJ, Murray LJ, Pesenti E, et al. Preclinical evaluation of the tyrosine kinase inhibitor SU11248 as a single agent and in combination with “standard of care” therapeutic agents for the treatment of breast cancer. Mol Cancer Ther 2003;2:1011-21
  • Murray LJ, Abrams TJ, Long KR, et al. SU11248 inhibits tumor growth and CSF-1R-dependent osteolysis in an experimental breast cancer bone metastasis model. Clin Exp Metastasis 2003;20:757-66
  • Carey DJ. Syndecans: multifunctional cell-surface co-receptors. Biochem J 1997;327( Pt 1):1-16
  • Bernfield M, Kokenyesi R, Kato M, et al. Biology of the syndecans: a family of transmembrane heparan sulfate proteoglycans. Annu Rev Cell Biol 1992;8:365-93
  • Bernfield M, Gotte M, Park PW, et al. Functions of cell surface heparan sulfate proteoglycans. Annu Rev Biochem 1999;68:729-77
  • Lustig F, Hoebeke J, Simonson C, et al. Processing of PDGF gene products determines interactions with glycosaminoglycans. J Mol Recognit 1999;12:112-20
  • Ostrovsky O, Berman B, Gallagher J, et al. Differential effects of heparin saccharides on the formation of specific fibroblast growth factor (FGF) and FGF receptor complexes. J Biol Chem 2002;277:2444-53
  • Lai J, Chien J, Staub J, et al. Loss of HSulf-1 up-regulates heparin-binding growth factor signaling in cancer. J Biol Chem 2003;278:23107-17
  • Lai JP, Chien JR, Moser DR, et al. HSulf-1 Sulfatase promotes apoptosis of hepatocellular cancer cells by decreasing heparin-binding growth factor signaling. Gastroenterology 2004;126:231-48
  • Narita K, Staub J, Chien J, et al. HSulf-1 inhibits angiogenesis and tumorigenesis in vivo. Cancer Res 2006;66:6025-32
  • Uchimura K, Morimoto-Tomita M, Bistrup A, et al. HSulf-2, an extracellular endoglucosamine-6-sulfatase, selectively mobilizes heparin-bound growth factors and chemokines: effects on VEGF, FGF-1, and SDF-1. BMC Biochem 2006;7:2 published online 17 January 2006,doi:10.1186/1471-2091-7-2.
  • Nawroth R, van Zante A, Cervantes S, et al. Extracellular sulfatases, elements of the Wnt signaling pathway, positively regulate growth and tumorigenicity of human pancreatic cancer cells. PLoS ONE 2007;2:e392, published online 25 April 2007,doi:10.1371/journal.pone.0000392
  • Lai JP, Sandhu DS, Yu C, et al. Sulfatase 2 up-regulates glypican 3, promotes fibroblast growth factor signaling, and decreases survival in hepatocellular carcinoma. Hepatology 2008;47:1211-22
  • Ding Q, Xia W, Liu JC, et al. Erk associates with and primes GSK-3β for its inactivation resulting in upregulation of β-catenin. Mol Cell 2005;19:159-70
  • Beurel E, Kornprobst M, Blivet-Van Eggelpoel MJ, et al. GSK-3β reactivation with LY294002 sensitizes hepatoma cells to chemotherapy-induced apoptosis. Int J Oncol 2005;27:215-22
  • Street A, Macdonald A, McCormick C, et al. Hepatitis C virus NS5A-mediated activation of phosphoinositide 3-kinase results in stabilization of cellular β-catenin and stimulation of β-catenin-responsive transcription. J Virol 2005;79:5006-16
  • Hasumi Y, Klosowska-Wardega A, Furuhashi M, et al. Identification of a subset of pericytes that respond to combination therapy targeting PDGF and VEGF signaling. Int J Cancer 2007;121:2606-14
  • Homsi J, Daud AI. Spectrum of activity and mechanism of action of VEGF/PDGF inhibitors. Cancer Control 2007;14:285-94

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.