Angiogenesis of prostate cancer and antiangiogenic therapy
Hisanori Uehara


Department of Molecular and Environmental Pathology, The University of Tokushima School of Medicine, Tokushima, Japan

Abstract: Tumor-associated angiogenesis refers to the growth of new vessels toward and within the tumor. Several studies have revealed that increasing intratumoral microvessel density, a major of tumor-associated angiogenesis, correlates with greater aggressiveness of prostate cancer. Angiogenesis consists of multiple, sequential, and interdependent steps dependent on the local balance of proangiogenic and antiangiogenic molecules. Many proangiogenic and antiangiogenic molecules have been demonstrated to regulate growth and metastasis of prostate cancer.
As tumor-associated angiogenesis is a crucial step in the process of prostate cancer development, inhibition of tumor neovascularization, and/or destruction of tumor vasculature (antiangiogenic therapy) may maintain the tumors in a dormant state or, perhaps in combination with cytotoxic therapies, potentiate shrinkage of tumors. Recently, therapeutic agents targeting the receptors of proangiogenic molecules and their signal transduction cascade have been developed.
In this article, the role of angiogenic molecules in prostate cancer biology, and the application of angiogenesis inhibition to therapeutics for prostate cancer are reviewed.
J. Med. Invest. 50:146-153, 2003

Keywords:angiogenesis, prostate cancer, metastasis, antiangiogenic therapy

INTRODUCTION
Prostate cancer is the most common cancer and the second leading cause of death in men in North America (1). In Japan, age - standardized mortality has increased by 150% over the 25 years up to 1997 (2). The major cause of death from this disease is metastasis of hormone-refractory cancer cells. The metastases are commonly found in lymph nodes or bones (1, 3), and the specific organ microenvironment can influence the biological behavior of metastatic cells, including their response to systemic therapy (4). Stephen Paget proposed that some tissues may provide a better environment than other tissues for the growth of certain tumor cells (the seed) are compatible with a particular organ tissue (the soil). Metastasis only resulted when the seed and soil were compatible (5). A prime example of this principle is the contribution of angiogenesis to the growth and metastasis of cancer. The growth and spread of prostate cancer are dependent on the formation of adequate vasculature, i.e., angiogenesis regulated by angiogenic factors (6 - 8).

TUMOR-ASSOCIATED ANGIOGENESIS
Angiogenesis consists of multiple, sequential, and interdependent steps dependent on the local balance of positive and negative regulatory factors. The major pro-angiogenic factors include vascular endothelial growth factor/vascular permeability factor (VEGF/VPF), inter-leukin-8 (IL-8), fibroblast growth factor 2 (FGF 2), epidermal growth factor (EGF) and platelet - derived growth factor (PDGF), and the major antiangiogenic factors include interferon (IFN), endostatin, angiostatin, and thrombospondin (8 -14).
Tumor-associated angiogenesis refers to the growth of new vessels toward and within the tumor. Tumors can not grow no longer than 2 - 4 mm in diameter until they are vascularized (9, 15). Compelling data implicate tumor-associated angiogenesis as a central pathologic step in the process of tumor growth, invasion, and metastasis. Several studies have revealed that increasing intratumoral microvessel density, a major of tumor-associated angiogenesis, correlates with greater tumor aggressiveness, such as a higher frequency of metastases and/or decreased survival in prostate cancer and other solid tumors (15 -17).

PROANGIOGENIC MOLECULES
1) Vascular endothelial growth factor (VEGF)
VEGF is one of the most potent facilitators of angiogenesis with affect on endothelial cell proliferation, motility, and vascular permeability. VEGF binds with high-affinity to the tyrosine kinase receptors Flt-1(VEGFR-1) and Flk-1/KDR (VEGFR-2) expressed by endothelial cells (18 -22). VEGF expression has been demonstrated in prostate cancer specimens (23) and in LNCaP, PC3, and DU145prostate cancer cell lines(24 -26). Kuniyasu et al. evaluated the expression level of VEGF/VPF in archival prostatectomy specimens from prostate cancer patients using a rapid colorimetric in situ hybridization technique. The relationship between advancing pathological stage and expression of VEGF/VPF gene was highly significant. Increased expression of VEGF/VPF was associated with the Gleason score of the tumors (27). Monoclonal antibodies that neutralize VEGF inhibit both the growth and metastatic spread of DU 145 prostate cancer xenografts in severe combined immune - deficient mice and decrease the growth of LNCaP tumors in nude mice, suggesting that VEGF is a critical factor for the progression of prostate cancer (28, 29).
Additionally, expression of VEGFR-1 and R-2 in prostate cancer, prostatic intraepithelial neoplasia, and the basal cells of normal glands, has been reported (23, 30). In comparison with normal glands, the expression of VEGFR-1 and R-2 increases in prostatic intraepithelial neoplasia and well to moderately differentiated prostate cancer. These observations suggest that VEGF plays a role on tumor cell activation (autocrine), in addition to paracrine actions whereby it regulates endothelial cell functions and subsequent neovascular development (23).
Recently, vascular endothelial growth factor C (VEGF-C) which belongs to the platelet-derived growth factor (PDGF)/VEGF family of growth factors was identified as a ligand for the endothelial-specific receptor tyrosine kinases VEGFR-3 and VEGFR-2. The expression of VEGFR-3was found to be highly restricted to the lymphatic endothelial cells (31). VEGF-C expression in prostate cancer cells is implicated in the lymph node metastasis(32).
2) Interleukin-8(IL-8)
IL- 8, which belongs to the superfamily of CXC chemokines, has a wide range of proinflammmatory effects and is produced by various cells, including lymphocytes, monocytes, endothelial cells, fibroblasts, hepatocyte, keratinocyte, and various tumor cells including prostate cancer cells (33, 34). It has been shown that IL- 8 enhances production and secretion of collagenase type IV by tumor cells, suggesting that it can modulate invasiveness, and/or extracellular matrix remodeling in the tumor environment. As cell proliferation, angiogenesis, migration, and invasion are important component of the metastatic process, IL - 8 expression by tumor cells can influence their metastatic capabilities (35). Indeed, the expression of IL - 8 has been shown to correlate with angiogenesis and the metastatic potential of human prostate cancer cells (36 -38). When low and high IL - 8 - producing clones isolated from the heterogeneous PC3 human prostate cancer cell line were injected into the prostate of nude mice, PC3 cells expressing high levels of IL- 8 were highly tumorigenic, producing rapidly growing prostate tumors. On the other hand, low IL - 8 - expressing PC -3 cells were less tumorigenic, producing slower growing tumors (Table 1). Additionally, prostate tumors produced by high IL - 8 - expressing PC-3 cells showed higher vascularity with significantly higher incidence of metastasis than the tumors produced by low IL - 8 - expressing PC -3 cells(38).
The pleiotropic transcription factor NF-κB regulates the expression of multiple genes including IL-8and matrix metalloproteinase (MMP)-9in several types of cells (39 - 42), and is constitutively activated in prostate cancer cells(43). Blockade of NF-κB activity in human prostate cancer cells inhibits in vitro and in vivo expression of VEGF, IL- 8 and MMP-9, and hence decreases neoplastic angiogenesis (44).
3) Fibroblast growth factor2(FGF2)
FGF2 (i.e. bFGF) is synthesized mainly by stromal fibroblasts in prostate. When prostate cancer converts to an invasive phenotype, the cancer cells respond to FGF2through high-affinity FGFR 2?c receptor. Then, the cancer cells synthesize their own FGF 2 to propagate their own growth. In addition, secreted FGF 2 acts on the endothelial cells to promote tumor angiogenesis (45).Several studies have shown that the metastatic potential of prostate cancer cells directly correlated with the gene expression level of FGF 2 (46, 47). Metastatic variant of human prostate cancer cell line expresses higher FGF2mRNA than parental cell line (47).
4) Epidermal growth factor (EGF)
The interaction of EGF with its receptor EGF-R has been shown to play an important role in neoplastic angiogenesis (48 - 50). The expression of VEGF, major proangiogenic factor, is strongly induced by EGF and transforming growth factor- α (51, 52). The expression of EGF and EGF-R is observed in both benign prostatic hyperplasia and prostate cancer (53, 54). Several studies have shown that the metastatic potential of prostate cancer cells directly correlated with the expression level of angiogenesis - and metastasis-related genes including EGFR (19, 55).
5) Platelet-derived growth factor (PDGF)
PDGF is a dimer that consist of AA, BB and AB proteins, and a ligand of PDGF receptor (PDGF-R), a member of a family of protein tyrosine kinases, encoded by two genes (PDGF-Rα and PDGF-R β)(56). PDGF and PDGF-R are co - expressed in many human cancers including prostate cancer (57). The binding of PDGF to PDGF-R can stimulate cell division (58 - 60), cell migration (61), and angiogenesis (62).

ANTIANGIOGENIC MOLECULES
1) Interferon (IFN)
IFNs are multifunctional regulatory cytokines involved in control of cell function and replication. IFN-α and IFN-β directly inhibit the proliferation of tumor cells of different histological origins (63 - 66). IFN-α and IFN-β can also down-regulate the expression of proangiogenic molecules, such as FGF2 (67- 69) and IL - 8 (70, 71). The combined treatment with pegylated IFN- α and docetaxel inhibits neoplastic angiogenesis by inducing a decrease in the local production of proangiogenic molecules by human prostate cancer cells in nude mice, resulting in increased apoptosis of tumor-associated endothelial cells (72).
2) Endostatin
Endostatin is a 20 kDa C-terminal fragment of collagen XVIII. Endostatin specifically inhibits endothelial proliferation and potently inhibits angiogenesis (73). Endostatin treatment delays the onset of spontaneous mammary tumorigenesis in female transgenic mice, and prolonged survival time of male transgenic mice that develop prostate adenocarcinomas (74).

INHIBITION OF TUMOR-ASSOCIATED ANGIOGENESIS BY MOLECULAR TARGETING AGENTS
Therapeutic approaches targeting the receptors of proangiogenic molecules and their signal transduction cascade may result in small avascular tumors maintained in a dormant state or, perhaps in combination with cytotoxic therapies, they may potentiate shrinkage of tumors.
PDGF binding causes PDGF-R activation, which involves dimerization and autophosphorylation (i.e. activation) of specific tyrosines in the cytoplasmic domain of PDGF-R. Activation of PDGF-R has been shown to inhibit some apoptotic pathways in normal cells and in tumor cells (75, 76). We determined whether blockade of the PDGF-R signaling pathway by oral administration of STI571(Gleevec, Novartis Pharmaceuticals), PDGF-R tyrosine kinase inhibitor, inhibits the growth of PC-3MM2 human prostate cancer cells in the bone of nude mice. PC-3 MM2 induced lytic lesions in the bone and expanded into the surrounding muscle. Tumor cells adjacent to the bone expressed high levels of VEGF, IL - 8, FGF2, PDGF B, PDGF-R β, and activated PDGF-R β, compared with the tumor cells growing in the surrounding muscle (Fig.1). Treatment with STI571or STI571 plus paclitaxel inhibited tumor growth and angiogenesis, and it was more effective to the tumor cells adjacent to the bone (77).
Agents targeting epidermal growth factor-receptor (EGF-R) and its signal transduction cascade include 1)monoclonal antibodies, directed against the extracellular binding domain of the receptor, or binding to the HER2receptor;and2) low-molecular-weight inhibitors of the EGF-R tyrosine kinase.
Cetuximab (C225, ImClone Systems, Inc.) is a chimeric monoclonal antibody with specificity for the external ligand-binding domain of EGF-R. Both dihydrotestosterone and EGF can stimulate proliferation of androgen-responsive prostate cancer cell lines, MDA PCa 2 a and MDA Pca 2 b, and this proliferation is associated with stimulation of cyclin-dependent kinase (CDK)-2 activity and downregulation of the CDK inhibitor gene p27Kip1. Dual blockade of the EGF-R family with C225 and of androgen-receptor function resulted in significant growth inhibition (78). Treatment with C225 with or without paclitaxel inhibits growth, metastasis, and angiogenesis of PC-3M-LN4 prostate cancer cells implanted orthotopically in athymic nude mice(79).
ZD1839 (AstraZeneca), a low-molecular-weight anilinoquinazoline is a potent and specific inhibitor of EGFR tyrosine kinase activity. EGF-induced neovascularization of mice cornea is inhibited by ZD1839 treatment (80).Administration of ZD1839 and more so ZD1839 plus cytotoxic agents inhibit the growth of a wide range of human tumors grown as xenografts in nude mice, including prostate tumors (81). PKI 166 (Novartis Pharmaceuticals) is also a selective inhibitor of EGFR tyrosine kinase activity. Treatment with PKI 166 inhibits the growth and angiogenesis of PC-3 MM 2 human prostate cancer cells implanted in the bone of nude mice (82).
DC101(ImClone Systems, Inc.) is a neutralizing monoclonal antibody that binds to the murine VEGFR-2/flk-1 receptor with high affinity and blocks ligand-induced receptor activation. Tumorigenicity, metastasis, and neovascularization in orthotopic prostate cancer xenografts in nude mice are reduced by a treatment with DC101(83).

CONCLUDING REMARKS
Prostate cancer is the most common cancer in North America. The prostate cancer death rate is rapidly increasing in Japan. The major cause of death from prostate cancer is metastases that are resistant to therapy. In prostate cancer, same as other cancers, tumor-associated angiogenesis is a crucial step in the process of tumor growth, invasion, and metastasis, and depends on the local balance of proangiogenic and antiangiogenic factors. Therefore therapeutic agents and strategies are being devised either to interrupt or inhibit one or more of the pathogenic steps involved in the process of tumor neovascularization or to directly target and destroy the tumor vasculature. Antiangiogenic therapy may provide an additional novel prostate cancer treatment suitable for combination with standard therapies.

REFFERENCES
1.Landis SH, Murray T, Bolden Wingo PA : Cancer statics1999. Cancer49:8 -31, 1999
2.Nakata S, Takahashi H, Ohtake N, Takei T, Yamanaka H:Trends and characteristics in prostate cancer mortality in Japan. Int J Urol7:254-7, 2000
3.Soh S, Kattan MW, Berkman S, Wheeler TM, Scardino PT : Has there been a recent shift in the pathological features and prognosis of patients treated with radical prostatectomy ? J Urol 157 :2212-2218, 1997
4.Fidler IJ : Critical factors in the biology of human cancer metastasis : Twenty-eighth G. H. A. Clowes Memorial Award Lecture. Cancer Res 50 : 6130 -6138, 1990
5.Paget S:The distribution of secondary growth in cancer of the breast. Lancet1:571-573, 1889
6.Fidler IJ : Modulation of the organ microenvironment for the treatment of cancer metastasis. J Natl Cancer Inst87:1588 -1592, 1995
7.Folkman MJ : The role of angiogenesis in tumor growth. Semin Cancer Biol3:65 -71, 1992
8.Auerbach W, Auerbach R : Angiogenesis inhibition:a review. Pharmacol Ther63:265-311, 1994
9.Pluda JM : Tumor-associated angiogenesis : mechanisms, clinical implications, and therapeutic strategies. Semin Oncol24:203-218, 1997
10.Bouck N, Stellmach V, Hsu SC:How tumors become angiogenic. Adv Cancer Res69:135 -174,1996
11.Dvorak HF:Tumors:wound that do not heal. New Engl J Med315:1650 -1659, 1986
12.Fidler IJ:Angiogenic heterogeneity : regulation of neoplastic angiogenesis by the organ microenvironment. J Natl Cancer Inst93:1040 -1041, 2001
13.Folkman J:Clinical application of research on angiogenesis. New Engl J Med 333:1753 -1763, 1995
14.Hanahan D, Folkman J : Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell86:353 -364, 1996
15.Weidner N:Current pathologic methods for measuring intratumoral microvessel density within breast carcinoma and other solid tumors. Breast Cancer Res Treat36:169-180, 1995
16.Campbell SC : Advances in angiogenesis research : relevance to urological oncology. J Urol158:1663-1674, 1997
17.Bettencourt MC, Bauer JJ, Sesterhenn IA, Connelly RR, Moul JW:CD34 immunohistochemical assessment of angiogenesis as a prognostic marker for prostate cancer recurrence after radical prostatectomy. J Urol160:459 - 465, 1998
18.Gospodarowicz D, Abraham JA, Schilling J: Isolation and characterization of a vascular endothelial cell mitogen produced by pituitary-derived folliculo stellate cells. Proc Natl Acad Sci USA 86 : 7311-7315, 1989
19.Connolly DT, Heuvelman DM, Nelson R, Olander JV, Eppley BL, Delfino JJ, Siegel NR, Leimgruber RM, Feder J : Tumor vascular permeability factor stimulates endothelial cell growth and angiogenesis. J Clin Invest84:1470 -1478, 1989
20.Waltenberger J, Claesson Welsh L, Siegbahn A, Shibuya M, Heldin CH:Different signal transduction properties of KDR and Flt1,two receptors for vascular endothelial growth factor. J Biol Chem269:26988 -26995, 1994
21.De Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N, Williams LT : The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science255:989-991, 1992
22.Terman BI, Dougher Vermazen M, Carrion ME, Dimitrov D, Armellino DC, Gospodarowicz D, Bohlen P:Identification of the KDR tyrosine kinase as a receptor for vascular endothelial cell growth factor. Biochem Biophys Res Commun, 187:1579-1586, 1992
23.Jackson MW, Bentel JM, Tilley WD:Vascular endothelial growth factor (VEGF) expression in prostate cancer and benign prostatic hyperplasia. J Urol157:2323-2328, 1997
24.Harper ME, Glynne -Jones E, Goddard L , Thurston VJ, Griffiths K : Vascular endothelial growth factor (VEGF) expression in prostatic tumours and its relationship to neuroendocrine cells. Br J Cancer74:910-916, 1996
25.Ferrer FA, Miller LJ, Andrawis RI, Kurtzman SH, Albertsen PC, Laudone VP, Kreutzer DL : Vascular endothelial growth factor (VEGF) expression in human prostate cancer : in situ and in vitro expression of VEGF by human prostate cancer cells. J Urol157:2329 -2333, 1997
26.Levine AC, Liu XH, Greenberg PD, Eliashvili M, Schiff JD, Aaronson SA, Holland JF, Kirschenbaum A:Androgens induce the expression of vascular endothelial growth factor in human fetal prostatic fibroblasts. Endocrinology 139 : 4672 - 4678, 1998
27.Kuniyasu H, Troncoso P, Johnston D, Bucana CD, Tahara E, Fidler IJ, Pettaway CA : Relative expression of type IV collagenase, E- cadherin, and vascular endothelial growth factor/vascular permeability factor in prostatectomy specimens distinguishes organ-confined from pathologically advanced prostate cancers. Clin Cancer Res 6 : 2295 -2308, 2000
28.Kirschenbaum A, Wang JP, Ren M, Schiff JD, Aaronson SA, Droller MJ, Ferrara N, Holland JF, Levine AC: Inhibition of vascular endothelial cell growth factor suppresses the in vivo growth of human prostate tumors. Urol Oncol 3:3 -10, 1997
29.Melnyk O, Zimmerman M, Kim KJ, Shuman M : Neutralizing anti-vascular endothelial growth factor antibody inhibits further growth of established prostate cancer and metastases in a pre - clinical model. J Urol161:960-963, 1999
30.Ferrer FA, Miller LJ, Lindquist R, Kowalczyk P, Laudone VP, Albertsen PC, Kreutzer DL: Expression of vascular endothelial growth factor receptors in human prostate cancer. Urology54:567-572, 1999
31.Joukov V, Sorsa T, Kumar V, Jeltsch M, Claesson-Welsh L, Cao Y, Saksela O, Kalkkinen N, Alitalo K : Proteolytic processing regulates receptor specificity and activity of VEGF-C. EMBO J16 : 3898 -3911, 1997
32.Tsurusaki T, Kanda S, Sakai H, Kanetake H, Saito Y, Alitalo K, Koji T : Vascular endothelial growth factor-C expression in human prostatic carcinoma and its relationship to lymph node metastasis. Br J Cancer80:309 -313, 1999
33.Herbert CA, Baker JB: Interleukin-8:a review. Cancer invest11:743 -750, 1993
34.Fidler IJ: Critical determinants of human colon cancer metastasis. In : Tahara E ed. Molecular pathology of gastrointestinal cancer : Application to clinical practice. Springer-Verlag, Tokyo, 1997, pp.147-169
35.Singh RK, Gutman M, Radinsky R, Bucana CD, Fidler IJ: Expression of interleukin 8 correlates with the metastatic potential of human melanoma cells in nude mice. Cancer Res54:3242-3247, 1994
36.Greene GF, Kitadai Y, Pettaway CA, von Eschenbach AC, Bucana CD, Fidler IJ : Correlation of metastasis-related gene expression with metastatic potential in human prostate carcinoma cells implanted in nude mice using an in situ messenger RNA hybridization technique. Am J Patho l150 : 1571-1582, 1997
37.Ferrer FA, Miller LJ, Andrawis RI, Kurtzman SH, Albertsen PC, Laudone VP, Kreutzer DL: Angiogenesis and prostate cancer: in vivo and in vitro expression of angiogenesis factors by prostate cancer cells. Urology 51 : 161-167, 1998
38.Kim SJ, Uehara H, Karashima T, Mccarty M, Shih N, Fidler IJ: Expression of interleukin-8correlates with angiogenesis, tumorigenicity, and metastasis of human prostate cancer cells implanted orthotopically in nude mice. Neoplasia3:33-42, 2001
39.Mukaida N, Okamoto S, Ishikawa Y, Matsushima KJ:Molecular mechanism of interleukin-8 gene expression. Leukoc Biol56:554-558, 1994
40.Villarete LH, Remick DG:Transcriptional and post-transcriptional regulation of interleukin - 8. Am J Pathol149:1685 -1693, 1996
41.Yokoo T, Kitamura M : Dual regulation of IL-1beta-mediated matrix metalloproteinase-9 expression in mesangial cells by NF-kappa B and AP-1. Am J Physiol270:123 -130, 1996
42.Bond M, Fabunmi RP, Baker AH, Newby AC: Synergistic upregulation of metalloproteinase-9 (MMP-9) by growth factors and inflammatory cytokines: an absolute requirement for transcription factor NF-kappa B. FEBS Lett 435:29 -34, 1998
43.Pajonk F, Pajonk K, McBride WH : Inhibition of NF-kappaB, clonogenicity, and radiosensitivity of human cancer cells. J Natl Cancer Inst 91 : 1956 -1960, 1999
44.Huang S, Pettaway CA, Uehara H, Bucana CD, Fidler IJ:Blockade of NF-kappaB activity in human prostate cancer cells is associated with suppression of angiogenesis, invasion, and metastasis. Oncogene20:4188 -97, 2001
45.Dow JK, deVere White RW:Fibroblast growth factor2:its structure and property, paracrine function, tumor angiogenesis, and prostate - related mitogenic and oncogenic functions. Urology 55:800 - 806, 2000
46.Greene GF, Kitadai Y, Pettaway CA, von Eschenbach AC, Fidler IJ : In situ mRNA hybridization technique for analysis of metastasis-related genes in human prostate carcinoma cells. Am J Patho l150 : 1571-1582, 1997
47.Mydlo JH, Michaeli J, Heston WDW, Fair WR : Expression of basic fibroblast growth factor mRNA in benign prostatic hyperplasia and prostatic carcinoma. Prostate13:297-304, 1988
48.Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, Abbruzzese J, Traxler P, Buchdunger E, Radinsky R, Fidler IJ : Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of endothelial cells and therapy of human pancreatic carcinoma. Cancer Res60:2926 -2935, 2000
49.Solorzano CC, Baker CH, Tsan R, Traxler P, Cohen P, Buchdunger E, Killion JJ, Fidler IJ: Optimization for the blockade of the epidermal growth factor receptor signaling for therapy of human pancreatic carcinoma. Clin Cancer Res7:2563 -2572, 2001
50.Perrotte P, Matsumoto T, Inoue K, Kuniyasu H, Eve BY, Hicklin DJ, Radinsky R, Dinney CP : Antiepidermal growth factor receptor antibody C225inhibits angiogenesis in human transitional cell carcinoma growing orthotopically in nude mice. Clin Cancer Res5:257-265, 1999
51.Goldman CK, Kim J, Wong WL, King V, Brock T, Gillespie GY : Epidermal growth factor stimulates vascular endothelial growth factor production by human malignant glioma cells:a model of glioblastoma multiforme pathophysiology. Mol. Biol. Cell4:121-133, 1993
52.Schreiber AB, Winkler ME, Derynck R:Transforming growth factor-a : a more potent angiogenic mediator than epidermal growth factor. Science 232:1250 -1253, 1986
53.Myers RB, Kudlow JE, Grizzle WE: Expression of transforming growth factor-alpha, epidermal growth factor and the epidermal growth factor receptor in adenocarcinoma of the prostate and benign prostatic hyperplasia. Mod Pathol 6 : 733-737, 1993
54.Glynne -Jones E, Goddard L, Harper ME : Comparative analysis of mRNA and protein expression for epidermal growth factor receptor and ligands relative to the proliferative index in human prostate tissue. Hum Pathol27:688 - 694, 1996
55.Turkeri LN, Sakr WA, Wykes SM, Grignon DJ, Pontes JE, Macoska J A : Comparative analysis of epidermal growth factor receptor gene expression and protein product in benign, premalignant, and malignant prostate tissue. Prostate 25 :199 - 205, 1994
56.Ross P, Paines EW, Bowen-Pope DF : The biology of platelet-derived growth factor. Cell 46:155-159, 1986
57.Schiffer CA : Signal transduction inhibition : changing paradigms in cancer care. Semin Oncol 28 :34 -39, 2001
58.Xie J, Aszterbaum M, Zhang X, Bonifas JM, Zachary C, Epstein E, McCormick F:A role of PDGFR-alpha in basal cell carcinoma proliferation. Proc Natl Acad Sci USA98:9255 - 9259, 2001
59.Funa K, Papanicolaou V, Juhlin C, Rastad J, Akerstrom G, Heldin CH, Oberg K : Expression of platelet-derived growth factor beta-receptors on stromal tissue cells in human carcinoid tumors. Cancer Res50:748 - 53, 1990
60.Liu YC, Chen SC, Chang C, Leu CM, Hu CP: Platelet-derived growth factor is an autocrine stimulator for the growth and survival of human esophageal carcinoma cell lines. Exp Cell Res228:206 -211, 1996
61.Bornfeldt KE, Raines EW, Nakano T, Graves LM, Krebs EG, Ross R : Insulin-like growth factor-I and platelet - derived growth factor-BB induce directed migration of human arterial smooth muscle cells via signaling pathways that are distinct from those of proliferation. J Clin Invest 93 : 1266 -1274, 1994
62.Plate KH, Breier G, Farrell CL, Risau W: Platelet-derived growth factor receptor-beta is induced during tumor development and upregulated during tumor progression in endothelial cells in human gliomas. Lab Invest67:529 -34, 1992
63.Hertzog PJ, Hwang SY, Kola I : Role of interferons in the regulation of cell proliferation, differentiation, and development. Mol Reprod Dev 39 : 226 -232, 1994
64.Gutterman JU:Cytokine therapeutics:lessons from interferon-alpha. Proc Natl Acad Sci USA91:1198 -1205, 1994
65.Krown SE : Interferons in malignancy : biological products or biological response modifiers ? J Natl Cancer Inst80:306 -309, 1988
66.Thomas H, Balkwill FR:Effects of interferons and other cytokines on tumors in animals:a review. Pharmacol Ther52:307- 414, 1991
67.Singh RK, Bucana CD, Llansa N, Sanchez R, Fidler IJ:Cell density-dependent modulation of basic fibroblast growth factor expression by human interferon - β. Int J Oncol 8 : 649 - 656, 1996
68.Vermeulen PB, Diriz LY, Martin M, Lemmens J, Van Oosterom AT:Serum basic fibroblast growth factor and vascular endothelial growth factor in metastatic renal cell carcinoma treated with interferon alfa-2b. J Natl Cancer Inst89:1316 -1317, 1997
69.Slaton JW, Perrotte P, Inoue K, Dinney CP, Fidler IJ: Interferon-alfa-mediated down -regulation of angiogenesis-related genes and therapy of bladder cancer are dependent on optimization of biological dose and schedule. Clin Cancer Res 5 : 2726 -2734, 1999
70.Oliveiera IC, Sciavolino PJ, Lee TH, Vilcek J: Down-regulation of interleukin-8expression in human fibroblasts : unique mechanism of transcriptional inhibition of interferon. Proc Natl Acad Sci USA89:9049 - 9053, 1992
71.Singh RK, Gutman M, Llansa N, Fidler IJ: Interferon -β prevents the upregulation of interleukin-8expression in human melanoma. J Interferon Cytokine Res16:577-584, 1996
72.Huang SF, Kim SJ, Lee AT, Karashima T, Bucana C, Kedar D, Sweeney P, Mian B, Fan D, Shepherd D, Fidler IJ, Dinney CP, Killion JJ:Inhibition of growth and metastasis of orthotopic human prostate cancer in athymic mice by combination therapy with pegylated interferon-alpha-2b and docetaxel. Cancer Res 62:5720 - 5726, 2002
73.O'Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn E, Birkhead JR, Olsen BR, Folkman J : Endostatin : an endogenous inhibitor of angiogenesis and tumor growth. Cell 88 : 277-285, 1997
74.Yokoyama Y, Green JE, Sukhatme VP, Ramakrishnan S: Effect of endostatin on spontaneous tumorigenesis of mammary adenocarcinoma in a transgenic mouse model. Cancer Res60:4362-4365, 2000
75.Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME : Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell91:231-241, 1997
76.Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ :Serine phosphorylation of death agonist BAD in response to survival factor results in binding to14-3-3not BCL-X (L). Cell87:619 - 628, 1996
77.Uehara H, Kim SJ, Karashima T, Shepherd DL, Fan D, Tsan R, Killion JJ, Logothetis C, Mathew P, Fidler IJ : Effects of blocking platelet-derived growth factor-receptor signaling in a mouse model of experimental prostate cancer bone metastases. J Natl Cancer Inst95:458 -470, 2003
78.Ye D, Mendelsohn J, Fan Z:Androgen and epidermal growth factor down-regulate cyclin-dependent kinase inhibitor p27kip1 and costimulate proliferation of MDA PCa2a and MDA PCa2B prostate cancer cells. Clin Cancer Res5:2171-2177, 1999
79.Karashima T, Sweeney P, Slaton JW, Kim SJ, Kedar D, Izawa JI, Fan Z, Pettaway C, Hicklin DJ, Shuin T, Dinney CP: Inhibition of angiogenesis by the antiepidermal growth factor receptor antibody ImClone C225 in androgen - independent prostate cancer growing orthotopically in nude mice. Clin Cancer Res8:1253 -1264, 2002
80.Hirata A, Ogawa S, Kometani T, Kuwano T, Naito S, Kuwano M, Ono M : ZD 1839(Iressa) induces antiangiogenic effects through inhibition of epidermal growth factor receptor tyrosine kinase. Cancer Res62:2554 -2560, 2002
81.Sirotnak FM, Zakowski MF, Miller VA, Scher HI, Kris MG:Efficacy of cytotoxic agents against human tumor xenografts is markedly enhanced by coadministration of ZD 1839(Iressa), an inhibitor of EGFR tyrosine kinase. Clin Cancer Res6:4885 - 4892, 2000
82.Kim SJ, Uehara H, Karashima T, Shepherd DL, Killion JJ, Fidler IJ : Blockade of Epidermal Growth Factor Receptor Signaling in Tumor Cells and Tumor-associated Endothelial Cells for Therapy of Androgen-independent Human Prostate Cancer Growing in the Bone of Nude Mice. Clin Cancer Res 9 : 1200 -1210, 2003
83.Sweeney P, Karashima T, Kim SJ, Kedar D, Mian B, Huang S, Baker C, Fan Z, Hicklin DJ, Pettaway CA, Dinney CP : Anti-vascular endothelial growth factor receptor 2 antibody reduces tumorigenicity and metastasis in orthotopic prostate cancer xenografts via induction of endothelial cell apoptosis and reduction of endothelial cell matrix metalloproteinase type 9 production. Clin Cancer Res8:2714-2724, 2002