Skip to main content
Erschienen in: Molecular Cancer 1/2003

Open Access 01.12.2003 | Review

Pathways for aberrant angiogenesis in pancreatic cancer

verfasst von: M Korc

Erschienen in: Molecular Cancer | Ausgabe 1/2003

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease. Althoughthe specific mechanisms that dictate its biological aggressiveness are notclearly established, it is characterized by a variety of molecularalterations as well as by the overexpression of mitogenic and angiogenicgrowth factors and their receptors. PDACs also express high levels ofvascular endothelial growth factor (VEGF). Recent studies indicate thatsuppression of VEGF expression attenuates pancreatic cancer celltumorigenicity in a nude mouse model, and that VEGF can exert directmitogenic effects on some pancreatic cancer cells. These findings suggestthat cancer cell derived VEGF promotes pancreatic cancer growth in vivo viaa paracrine angiogenic pathway and an autocrine mitogenic pathway, andprovide novel opportunities for therapeutic intervention in this deadlydisease.

Carcinoma of the pancreas: An overview

Pancreatic ductal adenocarcinoma (PDAC) is responsible for over 20% of deaths due to gastrointestinal malignancies, making it the fourth most common cause of cancer related mortality in the United States and other industrialized countries. The prognosis of patients with PDAC is extremely poor, with overall 5-year survival rates that are less than 1% [1], one-year overall survival of 12%, and a median survival of 6 months [2]. Survival is often limited to patients who had surgical resection at an early stage of the disease. However, the diagnosis of PDAC is often established at an advanced stage, precluding patients from undergoing tumor resection in spite of limited results with other treatment modalities [3]. These dismal statistics are due to the tumor's propensity to metastasize when small and undetectable, the advanced stage at which many patients first develop symptoms, and the intrinsic resistance of pancreatic cancer cells to cytotoxic agents and radiotherapy [35]. PDAC may be an even more serious problem in the future since its incidence increases after age 50 and the general population world-wide is aging. There is, therefore, an urgent need for an improved understanding of the mechanisms that contribute to pancreatic tumor growth and metastasis, and for the design of therapies for this disorder that are more effective than current regimens. This review will cover in a brief manner the molecular biology of pancreatic cancer, and will then focus on various aspects of vascular endothelial growth factors in angiogenesis in general and in relation to PDAC in particular.

Molecular biology of pancreatic cancer

A plethora of genetic mutations have been described in the cancer cells of PDAC patients. The most frequent alterations (approximate frequency indicated in parenthesis) include mutations in the K-ras oncogene (90%), the p53 (85%) and Smad4 (50%) tumor suppressor genes, and the p16 (85% mutated and 15% silenced epigenetically) cell cycle inhibitory gene [6, 7]. Together, these alterations promote cellular proliferation, suppress apoptotic pathways, and facilitate tumor spread and metastasis. In addition, there is overexpression of multiple tyrosine kinase receptors and their ligands which enhances mitogenesis, and loss of responsiveness to the growth-inhibitory signals of members of the transforming growth factor beta (TGF-β) family [6, 7], which contribute in a significant manner to the biological aggressiveness of PDAC.
It is well established that human pancreatic cancer cell lines overexpress the epidermal growth factor (EGF) receptor (EGFR) and produce multiple ligands that bind directly to EGFR, including transforming growth factor-alpha (TGF-α, amphiregulin, heparin-binding EGF-like growth factor (HB-EGF), betacellulin and epiregulin [812]. These cell lines also express other growth factors such as fibroblast growth factors (FGFs) and platelet-derived growth factor (PDGF) B chain [1316]. However, expression of receptors and ligands in cell lines does not necessarily indicate parallel alterations in PDAC in vivo. Therefore, studies using human tissues have been of vital importance in this regard. Studies using immunohistochemistry, Northern blot analysis and in situ hybridization techniques, have demonstrated that PDAC tissue samples overexpress EGFR and six ligands that bind directly to EGFR (EGF, TGF-α, HB-EGF, betacellulin, epiregulin and amphiregulin), as well as c-erb-B2, c-erb-B3, and c-erb-B4 [10, 11, 1719]. These cancers also overexpress basic fibroblast growth factor (FGF-2), acidic FGF (FGF-1), keratinocyte growth factor (KGF), FGF-5, PDGF B chain (but not A chain), insulin-like growth factor-I (IGF-I), the EGF-like growth factor Cripto, hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), all 3 mammalian transforming growth factor beta (TGF-β) isoforms, bone morphogenetic protein-2 (BMP-2) and activin βA [14, 15, 2029]. Many, but not all of the corresponding receptors are concomitantly overexpressed. For example, there is overexpression of PDGF receptor α and β, the IGF-1 receptor, MET (the receptor that binds HGF), the 2 Ig-like form of type I FGF receptor (FGFR-1), and the type II TGF-β receptor (TβRII) but not the insulin receptor [16, 21, 23, 26, 3033]. IGF-II and insulin are not overexpressed in PDAC [21], whereas the type I TGF-β receptor (TβRI) is under-expressed [3133]. Thus, there is selective overexpression of specific receptors and their ligands in PDAC, and this concomitant overexpression leads to the creation of aberrant paracrine and autocrine pathways that confers a distinct growth advantage to pancreatic cancer cells.
The clinical importance of the above observations is underscored by numerous observations. For example, the concomitant presence in the cancer cells of EGFR and either EGF or TGF-α is associated with disease progression and decreased survival of PDAC patients [34]. Overexpression of c-erbB3 [19], FGF-2 [20] or TGF-β [35] is associated with decreased patient survival. The aberrant cytoplasmic localization of amphiregulin [36] is also associated with decreased patient survival. Dominant negative inhibition of either EGFR or FGFR-1 markedly attenuates pancreatic cancer cell growth [3739]. Expression of a cyclin D1 antisense construct in pancreatic cancer cells lowers cyclin D1 levels in these cells, attenuates their growth in vitro, and blocks their tumorigenicity in vivo [40]. EGFR blockade with an anti-EGFR antibody attenuates pancreatic tumor growth, and inhibition of EGFR tyrosine kinase activity suppresses pancreatic tumor angiogenesis [41, 42]. Together, these findings are among many that support the hypothesis that tyrosine kinase receptors and ligands have an important role in PDAC.

VEGF family of growth factors and their receptors

VEGF-A, also called "vascular permeability factor", is a homodimeric heparin-binding glycoprotein [4345]. Five major VEGF-A isoforms having 121, 145, 165, 189 and 206 amino acid residues, respectively, arise as a result of alternative splicing from a single gene [46, 47]. VEGF-A121 and VEGF-A145 are usually secreted while VEGF-A189 and VEGF-A206 are almost completely sequestered in the extracellular matrix [47]. VEGF-A165 is half secreted and half bound to the cell surface and the extracellular matrix [48]. All 5 isoforms are mitogenic toward vascular endothelial cells and induce vascular permeabilization. Additional VEGF isoforms and VEGF-related genes have been identified, including VEGF-B [49, 50], VEGF-C [51], VEGF-D [52], VEGF-E [53] and placenta growth factor [54]. Direct evidence for the role played by VEGF-A in embryonic vasculogenesis and angiogenesis was also demonstrated in VEGF-A gene knockout studies [55, 56], in which loss of a single VEGF-A allele in mice resulted in embryonic lethality between day 11 and 12. Angiogenesis and blood-island formation were impaired, resulting in severe developmental anomalies. This heterozygous lethal phenotype is indicative of the tight dose-dependent regulation of embryonic vessel development by VEGF-A [55, 56]. VEGF-A is also required for the cyclical blood vessel proliferation in the female reproductive tract and for longitudinal bone growth and endochondral bone formation in postnatal development [43]. Together, these observations indicate that VEGF-A has an important role in embryogenesis, development, and tissue remodeling.
VEGF-A stimulates endothelial cell proliferation through binding to two related tyrosine kinase receptors, VEGFR-1 (flt-1) VEGFR-2 (flk-1/KDR), on the surface of endothelial cells, with most of the mitogenic effects taken to occur via VEGFR-2 [5759] (57–59). A third high affinity VEGF receptor, termed VEGFR-3 (Flt4), is expressed in lymphatic vessels [60, 61]. It is activated by VEGF-C, which can be processed to a form that also binds to VEGFR-2 [5761]. Furthermore, placenta growth factor and VEGF-B bind only VEGFR-1, whereas VEGF-D, like VEGF-C, interacts with both VEGFR-2 and VEGFR-3 [5761]. However, VEGF-E binds only to VEGFR-2 [59]. All three VEGFRs are class III transmembrane protein tyrosine kinases that possess seven immunoglobulin-like sequences in their extracellular domains and a kinase insert in their intracellular domains [5761]. In addition, neuropilin-1 (Np-1), a neuronal guidance molecule for axons in the developing nervous system, also acts as a co-receptor for VEGF-A165 (but not for VEGF-A121), PlGF-2, VEGF-B and VEGF-E [62]. Np-1 is a non-tyrosine kinase transmembrane protein whose overexpression in transgenic mice is associated with various abnormalities, including excess capillary and blood vessel formation [63]. The closely related neuropilin-2 (Np-2) also binds VEGF-A165 (but not VEGF-A121), as well as VEGF-A145 and PlGF-2, strongly implying that both Np-1 and Np-2 in angiogenesis [6264].
Gene knockout studies have shown that both VEGFR-1-/- and VEGFR-2-/- mice die in utero between day 8.5 and 9.5 [65, 66]. In VEGFR-1-/- mice, endothelial cells developed in both embryonic and extra-embryonic sites but failed to organize into normal vascular channels [65]. In VEGFR-2-/- mice, hematopoietic precursors were severely reduced, yolk-sac blood islands were absent, organized blood vessels failed to develop throughout the embryo or the yolk sac [66]. Furthermore, double knockouts for Np-1 and Np-2 die in utero between day 8.5 and 9.5 [67]. They exhibit avascular yolk sacs, and mice that are deficient for Np-1 but heterozygous for Np-2, or deficient for Np-2 but heterozygous for Np-1, die at day 10 to 10.5 and exhibit diffuse vascular abnormalities that are more marked than either Np-1 or Np-2 single knockouts [67]. Together, these observations suggest that VEGFR-1 and VEGFR-2 are essential for embryonic vasculature development, whereas VEGFR-3 is essential for lymphangiogenesis, and that Np-1 and Np-2 are as important as the other components of the VEGF pathway in embryonic angiogenesis.

Angiogenesis in cancer

Tumor angiogenesis is often the consequence of an angiogenic imbalance in which pro-angiogenic factors predominate over anti-angiogenic factors [6871]. Furthermore, angiogenesis is essential for growth and metastasis of most solid malignancies, and VEGF-A is believed to be critical for tumor angiogenesis [72, 73]. Thus, secretion of bioactive VEGF-A by cancer cells may be directly involved in tumor progression [43]. For example, ovarian cancer cells secrete large amounts of bioactive VEGF-A that may play a crucial role in the genesis of ascitic fluid accumulation, angiogenesis and tumor induced immunosuppression in ovarian cancer patients [74]. In high grade gliomas, bioactive VEGF-A secreted by the glioma cells may account for the histopathological and clinical features of these tumors, including such characteristics as marked tumor angiogenesis and increased cerebral edema [75, 76].
VEGF-A is expression is induced by multiple mechanisms. These include mutant K-ras and mutant p53, the von Hippel Lindau gene product, growth factors such as FGF-2 and TGF-β, hypoxia, and transcription factors such as hypoxia inducible factor 1 alpha and SP1 [7781]. VEGF-A is up-regulated in many tumors including mammary, colorectal, renal, liver, ovarian and gastric carcinomas and gliomas [43], and its overexpression has been correlated with poor prognosis. For example, breast cancer patients with metastatic disease whose tumors exhibit increased angiogenesis have a worse prognosis than the corresponding patients whose tumors do not exhibit increased angiogenesis [82]. Furthermore, suppression of VEGF-A functions inhibits tumor growth in animal models as demonstrated with a dominant negative VEGFR-2, soluble VEGFR-1, neutralizing anti-VEGF-A antibody, VEGF-A anti-sense expression, anti-VEGFR-1 or anti-VEGFR-2 ribozymes, tyrosine kinase inhibitors of VEGFR-2, and anti-VEGFR-2 antibodies [8392].

Role of VEGF in pancreatic cancer angiogenesis

Although PDAC is not a grossly vascular tumor, this malignancy often exhibits enhanced foci of endothelial cell proliferation. Moreover, several [24, 93, 94], but not all [95] studies, have reported a positive correlation between blood vessel density, tumor VEGF-A levels, and disease progression in PDAC, raising the possibility that VEGF-A may have an important role in this disease. However, PDACs overexpress multiple additional mitogenic growth factors which are also angiogenic (Table I), such as EGF, TGF-α, HGF, FGFs such as FGF-1, FGF-2, and FGF-5, and PDGF-beta [6, 96]. Therefore, while VEGF-A is of crucial importance in promoting the growth and metastasis of pancreatic cancer cells in PDAC, other factors are most likely also involved in this process. Nonetheless, it has been demonstrated that pancreatic cancer cells secrete biologically active VEGF-A [25], and the cancer cells in PDAC as well as pancreatic cancer cell lines sometimes express VEGFR-1 and/or VEGFR-2 [97]. Moreover, some of these cells may be growth stimulated by VEGF-A in cell culture [97, 98], and the major angiogenic agent toward human dermal microvascular endothelial cells (HDMEC) that is produced by T3M4 and PANC-1 human pancreatic cancer cells is VEGF-A, since the mitogenic activity of conditioned medium from these cells can be nearly completely suppressed by neutralizing anti-VEGF-A antibodies [99]. Together, these observations suggest that by promoting angiogenesis VEGF-A enhances tumor spread and metastasis in this malignancy.
Table 1
Examples of Angiogenic Growth Factors that Are Overexpressed in Human Pancreatic Cancer and their Cognate Receptors
Growth Factors Activating Tyrosine Kinase Receptors
Receptor
VEGF-A
VEGFR-1 and VEGFR-2
VEGF-C
VEGFR-3
EGF, TGF-α, HB-EGF
EGF receptor
FGF-1, -2, -5
FGF receptors, types 1 and 2
PDGF B chain
PDGF receptors α and β
IGF-1
IGF-1 receptor
Hepatocyte growth factor
MET
Growth Factors that Activate Serine-Threonine Kinase Receptors
 
TGF-β1, -2, -3
Type II TGF-β receptor
Pro-Angiogenic Chemokines
 
IL-8
CXCR1 and CXCR2
Mip 3α
CCR6
In support of the above conclusion, it has been demonstrated that anti-angiogenic therapy is effective at suppressing tumor growth in animal models of PDAC. Thus, the anti-angiogenic agent TNP-470 reduces neoangiogenesis in tumors formed by pancreatic cancer cell lines, and decreases tumor growth and metastasis [99]. Suppression of VEGF-A expression with a VEGF-A antisense construct and with a VEGF directed ribozyme markedly attenuates tumorigenicity in nude mice and formation of hepatic metastases [25, 100]. VEGF-A fused to diphtheria toxin (DT-VEGF) internalizes in target cells via VEGFRs, inhibits protein synthesis, and suppresses the growth of HUVEC endothelial cells, thereby decreasing the volume and microvessel density in tumors formed by pancreatic cancer cells [101]. Adenoviral vectors carrying sequences encoding soluble VEGFR-1 and VEGFR-2 [102, 103], or the VEGFR tyrosine kinase inhibitor PTK 787 [104], also inhibit the growth of growth and/or metastasis of pancreatic cancers in mouse models. These findings underscore the importance of the angiogenic process in PDAC, support the hypothesis that VEGF-A exerts a crucial role in this regard, and raise the possibility that VEGF-A may exert direct effects on pancreatic cancer cells in vivo.
VEGF-A can also act as a survival factor for endothelial cells, rendering these cells more radioresistant [105]. It can also promote the survival of leukemic cells, certain tumor cells and hematopoietic stem cells [106108]. In addition, VEGF-C is also overexpressed in PDAC, and this overexpression has been correlated with enhanced lymph node metastasis [109]. Thus, various members of the VEGF family of ligands may contribute to the growth and metastasis of pancreatic cancer cells through a variety of mechanisms.

Additional mechanisms for promoting pancreatic cancer angiogenesis

Although VEGF appears to be of paramount importance for the angiogenic process in PDAC, these cancers express many other pro-angiogenic factors (Table I). As in the case of VEGF, some of these growth factors activate tyrosine kinase receptors that are expressed in endothelial cells within the pancreatic tumor mass, such as EGFR [17]. The importance of tyrosine kinase receptors other than VEGFR in pancreatic cancer angiogenesis is underscored by recent observations that inhibition of EGFR tyrosine kinase activity suppresses pancreatic tumor angiogenesis [42], and that NK4, an antagonist that is composed of the N-terminal hairpin and subsequent four-kringle domains of HGF, is a competitive antagonist for HGF that potently inhibits angiogenesis in tumors formed by SUIT-2 pancreatic cancer cells [110].
Other pro-angiogenic factors that are overexpressed in PDAC include certain chemokines such as Mip3α and interleukin-8 (IL-8), which activate G-protein coupled receptors [111113]. By contrast, TGF-βs activate serine-threonine kinase receptors [114]. The importance of TGF-βs are pro-angiogenic factors in PDAC is underscored by the recent observation that expression of a soluble TβRII in pancreatic cancer cells interferes with TGF-β actions, attenuates tumor growth and metastasis, and suppresses tumor angiogenesis [Rowland-Goldsmith, 2001 #905; Rowland-Goldsmith MA, 2002 #2548].
Often, there is evidence for cross-talk between the various angiogenic factors. For example, TGF-β1 and plasminogen activator inhibitor-1 (PAI-1) are overexpressed in PDAC [117, 118], TGF-β1 induces PAI-1 expression in pancreatic cancer cells [119], and both TGF-β1 and PAI-1 and can promote angiogenesis in vivo [120122]. TGF-βs are initially released as latent molecules that form complexes with latent binding protein (LTBP), and their biological effectiveness is dependent on their activation by such proteins as plasmin, uPA and its receptor, the insulin-like growth factor II (IGF-2) receptor, and tissue transglutaminase [123, 124]. The IGF-2 receptor, as well as uPA and its receptor are overexpressed in PDAC [125, 126], and pancreatic cancer cell lines express tissue transglutaminase [127]. Furthermore, uPA and its receptor, as well as tissue transglutaminase, have been implicated in the angiogenic process [128, 129], and the angiogenic potential of TGF-βs may be enhanced by the presence of Smad4 mutations [130], which are frequent in PDAC. uPA can transctivate EGFR [131], and EGFR activation can induce the expression of VEGF and the pro-angiogenic chemokine interleukin-8 [132, 133]. Taken together, these observations suggest that multiple pathways interact to enhance angiogenesis in PDAC.
The pancreatic microenvironment may also serve to promote tumor angiogenesis [134]. In addition, as a consequence of the existence of a continuous intra-pancreatic portal circulation, pancreatic cancer cells may be exposed to high levels of islet cell derived hormones such as insulin and growth factors such as TGF-βs [135]. High insulin levels bind and activate the IGF-1 receptor, which can then promote angiogenesis [136, 137]. Furthermore, islet cell derived TGF-βs may enhance matrix metalloprotease-9 (MMP-9) and VEGF expression in PDAC [31, 138], and suppress PTEN expression [139]. MMP-9 enhances tumor angiogenesis [140] whereas PTEN, a phosphatase with specificity for 3-phosphorylated inositol phospholipids, has been implicated in the suppression of tumor angiogenesis [141].

Conclusion

PDAC is a biologically aggressive malignancy that has a propensity to spread locally and metastasize distally. While not grossly vascular, these cancers exhibit foci of micro-angiogenesis and overexpress multiple pro-angiogenic factors. VEGF and related ligands represent a crucial component of this pro-angiogenic switch, as evidenced by the presence of high levels of VEGF in ascitic fluid of PDAC patients [142], the correlation between high serum VEGF levels and disease recurrence post-operatively [143], and the observation that high VEGFR-2 levels are associated with a worse prognosis in this disease [144]. Therefore, mechanisms that target VEGF and the various pathways that enhance the angiogenic process in PDAC [145] may ultimately be of great therapeutic benefit in patients with unresectable disease as well as following surgery to prevent disease recurrence.
Literatur
1.
Zurück zum Zitat Warshaw AL, Fernandez-del Castillo C: Pancreatic carcinoma. N Engl J Med. 1992, 326 (7): 455-65.PubMedCrossRef Warshaw AL, Fernandez-del Castillo C: Pancreatic carcinoma. N Engl J Med. 1992, 326 (7): 455-65.PubMedCrossRef
3.
Zurück zum Zitat Bramhall SR, Neoptolemos JP: Adjuvant chemotherapy in pancreatic cancer. Int J Pancreatol. 1997, 21 (1): 59-63.PubMed Bramhall SR, Neoptolemos JP: Adjuvant chemotherapy in pancreatic cancer. Int J Pancreatol. 1997, 21 (1): 59-63.PubMed
4.
Zurück zum Zitat Abrams RA: Role of radiation therapy in the management of the patient with pancreatic cancer. Oncology (Huntingt). 1996, 10 (9 Suppl): 13-7. Abrams RA: Role of radiation therapy in the management of the patient with pancreatic cancer. Oncology (Huntingt). 1996, 10 (9 Suppl): 13-7.
5.
Zurück zum Zitat Kuvshinoff BW, Bryer MP: Treatment of resectable and locally advanced pancreatic cancer. Cancer Control. 2000, 7 (5): 428-36.PubMed Kuvshinoff BW, Bryer MP: Treatment of resectable and locally advanced pancreatic cancer. Cancer Control. 2000, 7 (5): 428-36.PubMed
6.
Zurück zum Zitat Korc M: Role of growth factors in pancreatic cancer. Surg Oncol Clin N Am. 1998, 7 (1): 25-41.PubMed Korc M: Role of growth factors in pancreatic cancer. Surg Oncol Clin N Am. 1998, 7 (1): 25-41.PubMed
7.
Zurück zum Zitat Kern SE: Molecular genetic alterations in ductal pancreatic adenocarcinomas. Med Clin North Am. 2000, 84 (3): 691-5.PubMedCrossRef Kern SE: Molecular genetic alterations in ductal pancreatic adenocarcinomas. Med Clin North Am. 2000, 84 (3): 691-5.PubMedCrossRef
8.
Zurück zum Zitat Korc M, Meltzer P, Trent J: Enhanced expression of epidermal growth factor receptor correlates with alterations of chromosome 7 in human pancreatic cancer. Proc Natl Acad Sci U S A. 1986, 83 (14): 5141-4.PubMedCentralPubMedCrossRef Korc M, Meltzer P, Trent J: Enhanced expression of epidermal growth factor receptor correlates with alterations of chromosome 7 in human pancreatic cancer. Proc Natl Acad Sci U S A. 1986, 83 (14): 5141-4.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Smith JJ, Derynck R, Korc M: Production of transforming growth factor alpha in human pancreatic cancer cells: evidence for a superagonist autocrine cycle. Proc Natl Acad Sci U S A. 1987, 84 (21): 7567-70.PubMedCentralPubMedCrossRef Smith JJ, Derynck R, Korc M: Production of transforming growth factor alpha in human pancreatic cancer cells: evidence for a superagonist autocrine cycle. Proc Natl Acad Sci U S A. 1987, 84 (21): 7567-70.PubMedCentralPubMedCrossRef
10.
Zurück zum Zitat Ebert M: Induction and expression of amphiregulin in human pancreatic cancer. Cancer Res. 1994, 54 (15): 3959-62.PubMed Ebert M: Induction and expression of amphiregulin in human pancreatic cancer. Cancer Res. 1994, 54 (15): 3959-62.PubMed
11.
Zurück zum Zitat Kobrin MS: Induction and expression of heparin-binding EGF-like growth factor in human pancreatic cancer. Biochem Biophys Res Commun. 1994, 202 (3): 1705-9.PubMedCrossRef Kobrin MS: Induction and expression of heparin-binding EGF-like growth factor in human pancreatic cancer. Biochem Biophys Res Commun. 1994, 202 (3): 1705-9.PubMedCrossRef
12.
Zurück zum Zitat Yokoyama YFH, Kobrin MS, Ebert M, Friess H, Büchler MW, Korc M: Betacellulin, a member of the EGF family is overexpressed in human pancreatic cancer. Int J Oncol. 1995, 7: 825-829.PubMed Yokoyama YFH, Kobrin MS, Ebert M, Friess H, Büchler MW, Korc M: Betacellulin, a member of the EGF family is overexpressed in human pancreatic cancer. Int J Oncol. 1995, 7: 825-829.PubMed
13.
Zurück zum Zitat Kornmann MH, Beger G, Korc M: Role of fibroblast growth factors and their receptors in pancreatic cancer and chronic pancreatitis. Pancreas. 1998, 17 (2): 169-75.PubMedCrossRef Kornmann MH, Beger G, Korc M: Role of fibroblast growth factors and their receptors in pancreatic cancer and chronic pancreatitis. Pancreas. 1998, 17 (2): 169-75.PubMedCrossRef
14.
Zurück zum Zitat Kornmann M: Fibroblast growth factor-5 stimulates mitogenic signaling and is overexpressed in human pancreatic cancer: evidence for autocrine and paracrine actions. Oncogene. 1997, 15 (12): 1417-24.PubMedCrossRef Kornmann M: Fibroblast growth factor-5 stimulates mitogenic signaling and is overexpressed in human pancreatic cancer: evidence for autocrine and paracrine actions. Oncogene. 1997, 15 (12): 1417-24.PubMedCrossRef
15.
Zurück zum Zitat Siddiqi I: Increased expression of keratinocyte growth factor in human pancreatic cancer. Biochem Biophys Res Commun. 1995, 215 (1): 309-15.PubMedCrossRef Siddiqi I: Increased expression of keratinocyte growth factor in human pancreatic cancer. Biochem Biophys Res Commun. 1995, 215 (1): 309-15.PubMedCrossRef
16.
Zurück zum Zitat Ebert M: Induction of platelet-derived growth factor A and B chains and over-expression of their receptors in human pancreatic cancer. Int J Cancer. 1995, 62 (5): 529-35.PubMedCrossRef Ebert M: Induction of platelet-derived growth factor A and B chains and over-expression of their receptors in human pancreatic cancer. Int J Cancer. 1995, 62 (5): 529-35.PubMedCrossRef
17.
Zurück zum Zitat Korc M: Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. J Clin Invest. 1992, 90 (4): 1352-60.PubMedCentralPubMedCrossRef Korc M: Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha. J Clin Invest. 1992, 90 (4): 1352-60.PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Yamanaka Y: Overexpression of HER2/neu oncogene in human pancreatic carcinoma. Hum Pathol. 1993, 24 (10): 1127-34.PubMedCrossRef Yamanaka Y: Overexpression of HER2/neu oncogene in human pancreatic carcinoma. Hum Pathol. 1993, 24 (10): 1127-34.PubMedCrossRef
19.
Zurück zum Zitat Friess H: Enhanced erbB-3 expression in human pancreatic cancer correlates with tumor progression. Clin Cancer Res. 1995, 1 (11): 1413-20.PubMed Friess H: Enhanced erbB-3 expression in human pancreatic cancer correlates with tumor progression. Clin Cancer Res. 1995, 1 (11): 1413-20.PubMed
20.
Zurück zum Zitat Yamanaka Y: Overexpression of acidic and basic fibroblast growth factors in human pancreatic cancer correlates with advanced tumor stage. Cancer Res. 1993, 53 (21): 5289-96.PubMed Yamanaka Y: Overexpression of acidic and basic fibroblast growth factors in human pancreatic cancer correlates with advanced tumor stage. Cancer Res. 1993, 53 (21): 5289-96.PubMed
21.
Zurück zum Zitat Bergmann U: Insulin-like growth factor I overexpression in human pancreatic cancer: evidence for autocrine and paracrine roles. Cancer Res. 1995, 55 (10): 2007-11.PubMed Bergmann U: Insulin-like growth factor I overexpression in human pancreatic cancer: evidence for autocrine and paracrine roles. Cancer Res. 1995, 55 (10): 2007-11.PubMed
22.
Zurück zum Zitat Friess H: Cripto, a member of the epidermal growth factor family, is over-expressed in human pancreatic cancer and chronic pancreatitis. Int J Cancer. 1994, 56 (5): 668-74.PubMedCrossRef Friess H: Cripto, a member of the epidermal growth factor family, is over-expressed in human pancreatic cancer and chronic pancreatitis. Int J Cancer. 1994, 56 (5): 668-74.PubMedCrossRef
23.
Zurück zum Zitat Ebert M: Coexpression of the c-met proto-oncogene and hepatocyte growth factor in human pancreatic cancer. Cancer Res. 1994, 54 (22): 5775-8.PubMed Ebert M: Coexpression of the c-met proto-oncogene and hepatocyte growth factor in human pancreatic cancer. Cancer Res. 1994, 54 (22): 5775-8.PubMed
24.
Zurück zum Zitat Itakura J: Enhanced expression of vascular endothelial growth factor in human pancreatic cancer correlates with local disease progression. Clin Cancer Res. 1997, 3 (8): 1309-16.PubMed Itakura J: Enhanced expression of vascular endothelial growth factor in human pancreatic cancer correlates with local disease progression. Clin Cancer Res. 1997, 3 (8): 1309-16.PubMed
25.
Zurück zum Zitat Luo J: Pancreatic cancer cell-derived vascular endothelial growth factor is biologically active in vitro and enhances tumorigenicity in vivo. Int J Cancer. 2001, 92 (3): 361-9.PubMedCrossRef Luo J: Pancreatic cancer cell-derived vascular endothelial growth factor is biologically active in vitro and enhances tumorigenicity in vivo. Int J Cancer. 2001, 92 (3): 361-9.PubMedCrossRef
26.
Zurück zum Zitat Friess H: Enhanced expression of the type II transforming growth factor beta receptor in human pancreatic cancer cells without alteration of type III receptor expression. Cancer Res. 1993, 53 (12): 2704-7.PubMed Friess H: Enhanced expression of the type II transforming growth factor beta receptor in human pancreatic cancer cells without alteration of type III receptor expression. Cancer Res. 1993, 53 (12): 2704-7.PubMed
27.
Zurück zum Zitat Kleeff J: Bone morphogenetic protein 2 exerts diverse effects on cell growth in vitro and is expressed in human pancreatic cancer in vivo. Gastroenterology. 1999, 116 (5): 1202-16.PubMedCrossRef Kleeff J: Bone morphogenetic protein 2 exerts diverse effects on cell growth in vitro and is expressed in human pancreatic cancer in vivo. Gastroenterology. 1999, 116 (5): 1202-16.PubMedCrossRef
28.
Zurück zum Zitat Kleeff J: Concomitant over-expression of activin/inhibin beta subunits and their receptors in human pancreatic cancer. Int J Cancer. 1998, 77 (6): 860-8.PubMedCrossRef Kleeff J: Concomitant over-expression of activin/inhibin beta subunits and their receptors in human pancreatic cancer. Int J Cancer. 1998, 77 (6): 860-8.PubMedCrossRef
29.
Zurück zum Zitat Bergmann U: Increased expression of insulin receptor substrate-1 in human pancreatic cancer. Biochem Biophys Res Commun. 1996, 220 (3): 886-90.PubMedCrossRef Bergmann U: Increased expression of insulin receptor substrate-1 in human pancreatic cancer. Biochem Biophys Res Commun. 1996, 220 (3): 886-90.PubMedCrossRef
30.
Zurück zum Zitat Kobrin MS: Aberrant expression of type I fibroblast growth factor receptor in human pancreatic adenocarcinomas. Cancer Res. 1993, 53 (20): 4741-4.PubMed Kobrin MS: Aberrant expression of type I fibroblast growth factor receptor in human pancreatic adenocarcinomas. Cancer Res. 1993, 53 (20): 4741-4.PubMed
31.
Zurück zum Zitat Wagner M: Enhanced expression of the type II transforming growth factor-beta receptor is associated with decreased survival in human pancreatic cancer. Pancreas. 1999, 19 (4): 370-6.PubMedCrossRef Wagner M: Enhanced expression of the type II transforming growth factor-beta receptor is associated with decreased survival in human pancreatic cancer. Pancreas. 1999, 19 (4): 370-6.PubMedCrossRef
32.
Zurück zum Zitat Lu Z: Presence of two signaling TGF-beta receptors in human pancreatic cancer correlates with advanced tumor stage. Dig Dis Sci. 1997, 42 (10): 2054-63.PubMedCrossRef Lu Z: Presence of two signaling TGF-beta receptors in human pancreatic cancer correlates with advanced tumor stage. Dig Dis Sci. 1997, 42 (10): 2054-63.PubMedCrossRef
33.
Zurück zum Zitat Wagner M: Transfection of the type I TGF-beta receptor restores TGF-beta responsiveness in pancreatic cancer. Int J Cancer. 1998, 78 (2): 255-60.PubMedCrossRef Wagner M: Transfection of the type I TGF-beta receptor restores TGF-beta responsiveness in pancreatic cancer. Int J Cancer. 1998, 78 (2): 255-60.PubMedCrossRef
34.
Zurück zum Zitat Yamanaka Y: Coexpression of epidermal growth factor receptor and ligands in human pancreatic cancer is associated with enhanced tumor aggressiveness. Anticancer Res. 1993, 13 (3): 565-9.PubMed Yamanaka Y: Coexpression of epidermal growth factor receptor and ligands in human pancreatic cancer is associated with enhanced tumor aggressiveness. Anticancer Res. 1993, 13 (3): 565-9.PubMed
35.
Zurück zum Zitat Friess H: Enhanced expression of transforming growth factor beta isoforms in pancreatic cancer correlates with decreased survival. Gastroenterology. 1993, 105 (6): 1846-56.PubMed Friess H: Enhanced expression of transforming growth factor beta isoforms in pancreatic cancer correlates with decreased survival. Gastroenterology. 1993, 105 (6): 1846-56.PubMed
36.
Zurück zum Zitat Yokoyama MEM, Funatomi H, Friess H, Büchler MW, Johnson GR, Korc M: Amphiregulin is a potent mitogen in human pancreatic cancer cells: correlation with patient survival. Int J Oncol. 1995, 6: 625-631.PubMed Yokoyama MEM, Funatomi H, Friess H, Büchler MW, Johnson GR, Korc M: Amphiregulin is a potent mitogen in human pancreatic cancer cells: correlation with patient survival. Int J Oncol. 1995, 6: 625-631.PubMed
37.
Zurück zum Zitat Wagner M: Expression of a truncated EGF receptor is associated with inhibition of pancreatic cancer cell growth and enhanced sensitivity to cisplatinum. Int J Cancer. 1996, 68 (6): 782-7.PubMedCrossRef Wagner M: Expression of a truncated EGF receptor is associated with inhibition of pancreatic cancer cell growth and enhanced sensitivity to cisplatinum. Int J Cancer. 1996, 68 (6): 782-7.PubMedCrossRef
38.
Zurück zum Zitat Matsuda K: Multiple mitogenic pathways in pancreatic cancer cells are blocked by a truncated epidermal growth factor receptor. Cancer Res. 2002, 62 (19): 5611-7.PubMed Matsuda K: Multiple mitogenic pathways in pancreatic cancer cells are blocked by a truncated epidermal growth factor receptor. Cancer Res. 2002, 62 (19): 5611-7.PubMed
39.
Zurück zum Zitat Wagner M: Suppression of fibroblast growth factor receptor signaling inhibits pancreatic cancer growth in vitro and in vivo. Gastroenterology. 1998, 114 (4): 798-807.PubMedCrossRef Wagner M: Suppression of fibroblast growth factor receptor signaling inhibits pancreatic cancer growth in vitro and in vivo. Gastroenterology. 1998, 114 (4): 798-807.PubMedCrossRef
40.
Zurück zum Zitat Kornmann M, Arber N, Korc M: Inhibition of basal and mitogen-stimulated pancreatic cancer cell growth by cyclin D1 antisense is associated with loss of tumorigenicity and potentiation of cytotoxicity to cisplatinum. J Clin Invest. 1998, 101 (2): 344-52.PubMedCentralPubMedCrossRef Kornmann M, Arber N, Korc M: Inhibition of basal and mitogen-stimulated pancreatic cancer cell growth by cyclin D1 antisense is associated with loss of tumorigenicity and potentiation of cytotoxicity to cisplatinum. J Clin Invest. 1998, 101 (2): 344-52.PubMedCentralPubMedCrossRef
41.
Zurück zum Zitat Overholser JP: Epidermal growth factor receptor blockade by antibody IMC-C225 inhibits growth of a human pancreatic carcinoma xenograft in nude mice. Cancer. 2000, 89 (1): 74-82.PubMedCrossRef Overholser JP: Epidermal growth factor receptor blockade by antibody IMC-C225 inhibits growth of a human pancreatic carcinoma xenograft in nude mice. Cancer. 2000, 89 (1): 74-82.PubMedCrossRef
42.
Zurück zum Zitat Bruns CJ: 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 Res. 2000, 60 (11): 2926-35.PubMed Bruns CJ: 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 Res. 2000, 60 (11): 2926-35.PubMed
43.
Zurück zum Zitat Ferrara N: Molecular and biological properties of vascular endothelial growth factor. J Mol Med. 1999, 77 (7): 527-43.PubMedCrossRef Ferrara N: Molecular and biological properties of vascular endothelial growth factor. J Mol Med. 1999, 77 (7): 527-43.PubMedCrossRef
44.
Zurück zum Zitat Dvorak HF: Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol. 1999, 237: 97-132.PubMed Dvorak HF: Vascular permeability factor/vascular endothelial growth factor and the significance of microvascular hyperpermeability in angiogenesis. Curr Top Microbiol Immunol. 1999, 237: 97-132.PubMed
45.
Zurück zum Zitat Ortega N, Hutchings H, Plouet J: Signal relays in the VEGF system. Front Biosci. 1999, 4: D141-52.PubMedCrossRef Ortega N, Hutchings H, Plouet J: Signal relays in the VEGF system. Front Biosci. 1999, 4: D141-52.PubMedCrossRef
46.
Zurück zum Zitat Houck KA: The vascular endothelial growth factor family: identification of a fourth molecular species and characterization of alternative splicing of RNA. Mol Endocrinol. 1991, 5 (12): 1806-14.PubMedCrossRef Houck KA: The vascular endothelial growth factor family: identification of a fourth molecular species and characterization of alternative splicing of RNA. Mol Endocrinol. 1991, 5 (12): 1806-14.PubMedCrossRef
47.
Zurück zum Zitat Poltorak Z: VEGF145, a secreted vascular endothelial growth factor isoform that binds to extracellular matrix. J Biol Chem. 1997, 272 (11): 7151-8.PubMedCrossRef Poltorak Z: VEGF145, a secreted vascular endothelial growth factor isoform that binds to extracellular matrix. J Biol Chem. 1997, 272 (11): 7151-8.PubMedCrossRef
48.
Zurück zum Zitat Park JE, Keller GA, Ferrara N: The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol Biol Cell. 1993, 4 (12): 1317-26.PubMedCentralPubMedCrossRef Park JE, Keller GA, Ferrara N: The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol Biol Cell. 1993, 4 (12): 1317-26.PubMedCentralPubMedCrossRef
49.
50.
Zurück zum Zitat Grimmond S: Cloning and characterization of a novel human gene related to vascular endothelial growth factor. Genome Res. 1996, 6 (2): 124-31.PubMedCrossRef Grimmond S: Cloning and characterization of a novel human gene related to vascular endothelial growth factor. Genome Res. 1996, 6 (2): 124-31.PubMedCrossRef
51.
Zurück zum Zitat Joukov V: A novel vascular endothelial growth factor, VEGF-C, is a ligand for the Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine kinases. Embo J. 1996, 15 (7): 1751-PubMedCentralPubMed Joukov V: A novel vascular endothelial growth factor, VEGF-C, is a ligand for the Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine kinases. Embo J. 1996, 15 (7): 1751-PubMedCentralPubMed
52.
Zurück zum Zitat Yamada Y: Molecular cloning of a novel vascular endothelial growth factor, VEGF-D. Genomics. 1997, 42 (3): 483-8.PubMedCrossRef Yamada Y: Molecular cloning of a novel vascular endothelial growth factor, VEGF-D. Genomics. 1997, 42 (3): 483-8.PubMedCrossRef
53.
Zurück zum Zitat Meyer M: A novel vascular endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signalling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine kinases. Embo J. 1999, 18 (2): 363-74.PubMedCentralPubMedCrossRef Meyer M: A novel vascular endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signalling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine kinases. Embo J. 1999, 18 (2): 363-74.PubMedCentralPubMedCrossRef
54.
Zurück zum Zitat Maglione D: Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc Natl Acad Sci U S A. 1991, 88 (20): 9267-71.PubMedCentralPubMedCrossRef Maglione D: Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc Natl Acad Sci U S A. 1991, 88 (20): 9267-71.PubMedCentralPubMedCrossRef
55.
Zurück zum Zitat Ferrara N: Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature. 1996, 380 (6573): 439-42.PubMedCrossRef Ferrara N: Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature. 1996, 380 (6573): 439-42.PubMedCrossRef
56.
Zurück zum Zitat Carmeliet P: Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996, 380 (6573): 435-9.PubMedCrossRef Carmeliet P: Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996, 380 (6573): 435-9.PubMedCrossRef
57.
Zurück zum Zitat Shibuya M: Structure and function of VEGF/VEGF-receptor system involved in angiogenesis. Cell Struct Funct. 2001, 26 (1): 25-35.PubMedCrossRef Shibuya M: Structure and function of VEGF/VEGF-receptor system involved in angiogenesis. Cell Struct Funct. 2001, 26 (1): 25-35.PubMedCrossRef
58.
Zurück zum Zitat Veikkola T: Regulation of angiogenesis via vascular endothelial growth factor receptors. Cancer Res. 2000, 60 (2): 203-12.PubMed Veikkola T: Regulation of angiogenesis via vascular endothelial growth factor receptors. Cancer Res. 2000, 60 (2): 203-12.PubMed
59.
Zurück zum Zitat Neufeld G: Vascular endothelial growth factor (VEGF) and its receptors. Faseb J. 1999, 13 (1): 9-22.PubMed Neufeld G: Vascular endothelial growth factor (VEGF) and its receptors. Faseb J. 1999, 13 (1): 9-22.PubMed
60.
Zurück zum Zitat Kukk E: VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests a role in lymphatic vascular development. Development. 1996, 122 (12): 3829-37.PubMed Kukk E: VEGF-C receptor binding and pattern of expression with VEGFR-3 suggests a role in lymphatic vascular development. Development. 1996, 122 (12): 3829-37.PubMed
61.
Zurück zum Zitat Iljin K: VEGFR3 gene structure, regulatory region, and sequence polymorphisms. Faseb J. 2001, 15 (6): 1028-36.PubMedCrossRef Iljin K: VEGFR3 gene structure, regulatory region, and sequence polymorphisms. Faseb J. 2001, 15 (6): 1028-36.PubMedCrossRef
62.
Zurück zum Zitat Soker S: Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell. 1998, 92 (6): 735-45.PubMedCrossRef Soker S: Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell. 1998, 92 (6): 735-45.PubMedCrossRef
63.
Zurück zum Zitat Kitsukawa T: Overexpression of a membrane protein, neuropilin, in chimeric mice causes anomalies in the cardiovascular system, nervous system and limbs. Development. 1995, 121 (12): 4309-18.PubMed Kitsukawa T: Overexpression of a membrane protein, neuropilin, in chimeric mice causes anomalies in the cardiovascular system, nervous system and limbs. Development. 1995, 121 (12): 4309-18.PubMed
64.
Zurück zum Zitat Gluzman-Poltorak Z: Neuropilin-2 is a receptor for the vascular endothelial growth factor (VEGF) forms VEGF-145 and VEGF-165. J Biol Chem. 2000, 275 (38): 29922- Gluzman-Poltorak Z: Neuropilin-2 is a receptor for the vascular endothelial growth factor (VEGF) forms VEGF-145 and VEGF-165. J Biol Chem. 2000, 275 (38): 29922-
65.
Zurück zum Zitat Fong GH: Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature. 1995, 376 (6535): 66-70.PubMedCrossRef Fong GH: Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature. 1995, 376 (6535): 66-70.PubMedCrossRef
66.
Zurück zum Zitat Shalaby F: Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995, 376 (6535): 62-6.PubMedCrossRef Shalaby F: Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995, 376 (6535): 62-6.PubMedCrossRef
67.
Zurück zum Zitat Takashima S: Targeting of both mouse neuropilin-1 and neuropilin-2 genes severely impairs developmental yolk sac and embryonic angiogenesis. Proc Natl Acad Sci U S A. 2002, 99 (6): 3657-62.PubMedCentralPubMedCrossRef Takashima S: Targeting of both mouse neuropilin-1 and neuropilin-2 genes severely impairs developmental yolk sac and embryonic angiogenesis. Proc Natl Acad Sci U S A. 2002, 99 (6): 3657-62.PubMedCentralPubMedCrossRef
68.
Zurück zum Zitat Hanahan D, Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996, 86 (3): 353-64.PubMedCrossRef Hanahan D, Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996, 86 (3): 353-64.PubMedCrossRef
69.
Zurück zum Zitat Fang J: HIF-1alpha-mediated up-regulation of vascular endothelial growth factor, independent of basic fibroblast growth factor, is important in the switch to the angiogenic phenotype during early tumorigenesis. Cancer Res. 2001, 61 (15): 5731-5.PubMed Fang J: HIF-1alpha-mediated up-regulation of vascular endothelial growth factor, independent of basic fibroblast growth factor, is important in the switch to the angiogenic phenotype during early tumorigenesis. Cancer Res. 2001, 61 (15): 5731-5.PubMed
70.
Zurück zum Zitat Giordano FJ, RS Johnson: Angiogenesis: the role of the microenvironment in flipping the switch. Curr Opin Genet Dev. 2001, 11 (1): 35-40.PubMedCrossRef Giordano FJ, RS Johnson: Angiogenesis: the role of the microenvironment in flipping the switch. Curr Opin Genet Dev. 2001, 11 (1): 35-40.PubMedCrossRef
71.
Zurück zum Zitat Udagawa T: Persistence of microscopic human cancers in mice: alterations in the angiogenic balance accompanies loss of tumor dormancy. Faseb J. 2002, 16 (11): 1361-70.PubMedCrossRef Udagawa T: Persistence of microscopic human cancers in mice: alterations in the angiogenic balance accompanies loss of tumor dormancy. Faseb J. 2002, 16 (11): 1361-70.PubMedCrossRef
72.
Zurück zum Zitat Folkman J: What is the evidence that tumors are angiogenesis dependent?. J Natl Cancer Inst. 1990, 82 (1): 4-6.PubMedCrossRef Folkman J: What is the evidence that tumors are angiogenesis dependent?. J Natl Cancer Inst. 1990, 82 (1): 4-6.PubMedCrossRef
73.
Zurück zum Zitat Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995, 1 (1): 27-31.PubMedCrossRef Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995, 1 (1): 27-31.PubMedCrossRef
74.
Zurück zum Zitat Santin AD: Secretion of vascular endothelial growth factor in ovarian cancer. Eur J Gynaecol Oncol. 1999, 20 (3): 177-81.PubMed Santin AD: Secretion of vascular endothelial growth factor in ovarian cancer. Eur J Gynaecol Oncol. 1999, 20 (3): 177-81.PubMed
75.
Zurück zum Zitat Plate KH: Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature. 1992, 359 (6398): 845-8.PubMedCrossRef Plate KH: Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature. 1992, 359 (6398): 845-8.PubMedCrossRef
76.
Zurück zum Zitat Goldman CK: Brain edema in meningiomas is associated with increased vascular endothelial growth factor expression. Neurosurgery. 1997, 40 (6): 1269-77.PubMedCrossRef Goldman CK: Brain edema in meningiomas is associated with increased vascular endothelial growth factor expression. Neurosurgery. 1997, 40 (6): 1269-77.PubMedCrossRef
77.
Zurück zum Zitat Okada F: Impact of oncogenes in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc Natl Acad Sci U S A. 1998, 95 (7): 3609-14.PubMedCentralPubMedCrossRef Okada F: Impact of oncogenes in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells. Proc Natl Acad Sci U S A. 1998, 95 (7): 3609-14.PubMedCentralPubMedCrossRef
78.
Zurück zum Zitat Meadows KN, Bryant P, Pumiglia K: Vascular endothelial growth factor induction of the angiogenic phenotype requires Ras activation. J Biol Chem. 2001, 276 (52): 49289-98.PubMedCrossRef Meadows KN, Bryant P, Pumiglia K: Vascular endothelial growth factor induction of the angiogenic phenotype requires Ras activation. J Biol Chem. 2001, 276 (52): 49289-98.PubMedCrossRef
79.
Zurück zum Zitat Blancher C: Effects of ras and von Hippel-Lindau (VHL) gene mutations on hypoxia-inducible factor (HIF)-1alpha, HIF-2alpha, and vascular endothelial growth factor expression and their regulation by the phosphatidylinositol 3'-kinase/Akt signaling pathway. Cancer Res. 2001, 61 (19): 7349-55.PubMed Blancher C: Effects of ras and von Hippel-Lindau (VHL) gene mutations on hypoxia-inducible factor (HIF)-1alpha, HIF-2alpha, and vascular endothelial growth factor expression and their regulation by the phosphatidylinositol 3'-kinase/Akt signaling pathway. Cancer Res. 2001, 61 (19): 7349-55.PubMed
80.
Zurück zum Zitat Yu JL: Effect of p53 status on tumor response to antiangiogenic therapy. Science. 2002, 295 (5559): 1526-8.PubMedCrossRef Yu JL: Effect of p53 status on tumor response to antiangiogenic therapy. Science. 2002, 295 (5559): 1526-8.PubMedCrossRef
81.
Zurück zum Zitat Shi Q: Constitutive Sp1 activity is essential for differential constitutive expression of vascular endothelial growth factor in human pancreatic adenocarcinoma. Cancer Res. 2001, 61 (10): 4143-54.PubMed Shi Q: Constitutive Sp1 activity is essential for differential constitutive expression of vascular endothelial growth factor in human pancreatic adenocarcinoma. Cancer Res. 2001, 61 (10): 4143-54.PubMed
82.
Zurück zum Zitat Arora R: Angiogenesis as an independent prognostic indicator in node-negative breast cancer. Anal Quant Cytol Histol. 2002, 24 (4): 228-33.PubMed Arora R: Angiogenesis as an independent prognostic indicator in node-negative breast cancer. Anal Quant Cytol Histol. 2002, 24 (4): 228-33.PubMed
83.
Zurück zum Zitat Sledge GW: Vascular endothelial growth factor in breast cancer: biologic and therapeutic aspects. Semin Oncol. 2002, 29 (3 Suppl 11): 104-10.PubMedCrossRef Sledge GW: Vascular endothelial growth factor in breast cancer: biologic and therapeutic aspects. Semin Oncol. 2002, 29 (3 Suppl 11): 104-10.PubMedCrossRef
84.
Zurück zum Zitat Millauer B: Glioblastoma growth inhibited in vivo by a dominant-negative Flk-1 mutant. Nature. 1994, 367 (6463): 576-9.PubMedCrossRef Millauer B: Glioblastoma growth inhibited in vivo by a dominant-negative Flk-1 mutant. Nature. 1994, 367 (6463): 576-9.PubMedCrossRef
85.
Zurück zum Zitat Kong HL: Regional suppression of tumor growth by in vivo transfer of a cDNA encoding a secreted form of the extracellular domain of the flt-1 vascular endothelial growth factor receptor. Hum Gene Ther. 1998, 9 (6): 823-33.PubMedCrossRef Kong HL: Regional suppression of tumor growth by in vivo transfer of a cDNA encoding a secreted form of the extracellular domain of the flt-1 vascular endothelial growth factor receptor. Hum Gene Ther. 1998, 9 (6): 823-33.PubMedCrossRef
86.
Zurück zum Zitat Goldman CK: Paracrine expression of a native soluble vascular endothelial growth factor receptor inhibits tumor growth, metastasis, and mortality rate. Proc Natl Acad Sci U S A. 1998, 95 (15): 8795-800.PubMedCentralPubMedCrossRef Goldman CK: Paracrine expression of a native soluble vascular endothelial growth factor receptor inhibits tumor growth, metastasis, and mortality rate. Proc Natl Acad Sci U S A. 1998, 95 (15): 8795-800.PubMedCentralPubMedCrossRef
87.
Zurück zum Zitat Kim KJ: Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature. 1993, 362 (6423): 841-4.PubMedCrossRef Kim KJ: Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature. 1993, 362 (6423): 841-4.PubMedCrossRef
88.
Zurück zum Zitat Cheng SY: Suppression of glioblastoma angiogenicity and tumorigenicity by inhibition of endogenous expression of vascular endothelial growth factor. Proc Natl Acad Sci U S A. 1996, 93 (16): 8502-7.PubMedCentralPubMedCrossRef Cheng SY: Suppression of glioblastoma angiogenicity and tumorigenicity by inhibition of endogenous expression of vascular endothelial growth factor. Proc Natl Acad Sci U S A. 1996, 93 (16): 8502-7.PubMedCentralPubMedCrossRef
89.
Zurück zum Zitat Saleh M, Stacker SA, Wilks AF: Inhibition of growth of C6 glioma cells in vivo by expression of antisense vascular endothelial growth factor sequence. Cancer Res. 1996, 56 (2): 393-401.PubMed Saleh M, Stacker SA, Wilks AF: Inhibition of growth of C6 glioma cells in vivo by expression of antisense vascular endothelial growth factor sequence. Cancer Res. 1996, 56 (2): 393-401.PubMed
90.
Zurück zum Zitat Pavco PA: Antitumor and antimetastatic activity of ribozymes targeting the messenger RNA of vascular endothelial growth factor receptors. Clin Cancer Res. 2000, 6 (5): 2094-103.PubMed Pavco PA: Antitumor and antimetastatic activity of ribozymes targeting the messenger RNA of vascular endothelial growth factor receptors. Clin Cancer Res. 2000, 6 (5): 2094-103.PubMed
91.
Zurück zum Zitat Fong TA: SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types. Cancer Res. 1999, 59 (1): 99-106.PubMed Fong TA: SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types. Cancer Res. 1999, 59 (1): 99-106.PubMed
92.
Zurück zum Zitat Witte L: Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev. 1998, 17 (2): 155-61.PubMedCrossRef Witte L: Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev. 1998, 17 (2): 155-61.PubMedCrossRef
93.
Zurück zum Zitat Seo Y: High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer. 2000, 88 (10): 2239-45.PubMedCrossRef Seo Y: High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer. 2000, 88 (10): 2239-45.PubMedCrossRef
94.
Zurück zum Zitat Ikeda N: Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer. 1999, 79: 9-10.CrossRef Ikeda N: Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer. 1999, 79: 9-10.CrossRef
95.
Zurück zum Zitat Ellis LM: Vessel counts and vascular endothelial growth factor expression in pancreatic adenocarcinoma. Eur J Cancer. 1998, 34 (3): 337-40.PubMedCrossRef Ellis LM: Vessel counts and vascular endothelial growth factor expression in pancreatic adenocarcinoma. Eur J Cancer. 1998, 34 (3): 337-40.PubMedCrossRef
96.
Zurück zum Zitat Balaz P, Friess H, Büchler MW: Growth factors in pancreatic health and disease. Pancreatology. 2001, 1 (4): 343-55.PubMedCrossRef Balaz P, Friess H, Büchler MW: Growth factors in pancreatic health and disease. Pancreatology. 2001, 1 (4): 343-55.PubMedCrossRef
97.
Zurück zum Zitat Itakura J: Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int J Cancer. 2000, 85 (1): 27-34.PubMedCrossRef Itakura J: Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int J Cancer. 2000, 85 (1): 27-34.PubMedCrossRef
98.
Zurück zum Zitat von Marschall Z: De novo expression of vascular endothelial growth factor in human pancreatic cancer: evidence for an autocrine mitogenic loop. Gastroenterology. 2000, 119 (5): 1358-72.PubMedCrossRef von Marschall Z: De novo expression of vascular endothelial growth factor in human pancreatic cancer: evidence for an autocrine mitogenic loop. Gastroenterology. 2000, 119 (5): 1358-72.PubMedCrossRef
99.
Zurück zum Zitat Hotz HG: Angiogenesis inhibitor TNP-470 reduces human pancreatic cancer growth. J Gastrointest Surg. 2001, 5 (2): 131-8.PubMedCrossRef Hotz HG: Angiogenesis inhibitor TNP-470 reduces human pancreatic cancer growth. J Gastrointest Surg. 2001, 5 (2): 131-8.PubMedCrossRef
100.
Zurück zum Zitat Tokunaga T: Ribozyme mediated cleavage of cell-associated isoform of vascular endothelial growth factor inhibits liver metastasis of a pancreatic cancer cell line. Int J Oncol. 2002, 21 (5): 1027-32.PubMed Tokunaga T: Ribozyme mediated cleavage of cell-associated isoform of vascular endothelial growth factor inhibits liver metastasis of a pancreatic cancer cell line. Int J Oncol. 2002, 21 (5): 1027-32.PubMed
101.
Zurück zum Zitat Hotz HG: Specific targeting of tumor vasculature by diphtheria toxin-vascular endothelial growth factor fusion protein reduces angiogenesis and growth of pancreatic cancer. J Gastrointest Surg. 2002, 6 (2): 159-66.PubMedCrossRef Hotz HG: Specific targeting of tumor vasculature by diphtheria toxin-vascular endothelial growth factor fusion protein reduces angiogenesis and growth of pancreatic cancer. J Gastrointest Surg. 2002, 6 (2): 159-66.PubMedCrossRef
102.
Zurück zum Zitat Hoshida T: Gene therapy for pancreatic cancer using an adenovirus vector encoding soluble flt-1 vascular endothelial growth factor receptor. Pancreas. 2002, 25 (2): 111-21.PubMedCrossRef Hoshida T: Gene therapy for pancreatic cancer using an adenovirus vector encoding soluble flt-1 vascular endothelial growth factor receptor. Pancreas. 2002, 25 (2): 111-21.PubMedCrossRef
103.
Zurück zum Zitat Ogawa T: Anti-tumor angiogenesis therapy using soluble receptors: enhanced inhibition of tumor growth when soluble fibroblast growth factor receptor-1 is used with soluble vascular endothelial growth factor receptor. Cancer Gene Ther. 2002, 9 (8): 633-40.PubMedCrossRef Ogawa T: Anti-tumor angiogenesis therapy using soluble receptors: enhanced inhibition of tumor growth when soluble fibroblast growth factor receptor-1 is used with soluble vascular endothelial growth factor receptor. Cancer Gene Ther. 2002, 9 (8): 633-40.PubMedCrossRef
104.
Zurück zum Zitat Solorzano CC: Inhibition of growth and metastasis of human pancreatic cancer growing in nude mice by PTK 787/ZK22 an inhibitor of the vascular endothelial growth factor receptor tyrosine kinases. Cancer Biother Radiopharm. 2584, 16 (5): 359-70.CrossRef Solorzano CC: Inhibition of growth and metastasis of human pancreatic cancer growing in nude mice by PTK 787/ZK22 an inhibitor of the vascular endothelial growth factor receptor tyrosine kinases. Cancer Biother Radiopharm. 2584, 16 (5): 359-70.CrossRef
105.
Zurück zum Zitat Gupta VK: Vascular endothelial growth factor enhances endothelial cell survival and tumor radioresistance. Cancer J. 2002, 8 (1): 47-54.PubMedCrossRef Gupta VK: Vascular endothelial growth factor enhances endothelial cell survival and tumor radioresistance. Cancer J. 2002, 8 (1): 47-54.PubMedCrossRef
106.
Zurück zum Zitat Harmey JH, Bouchier-Hayes D: Vascular endothelial growth factor (VEGF), a survival factor for tumour cells: implications for anti-angiogenic therapy. Bioessays. 2002, 24 (3): 280-3.PubMedCrossRef Harmey JH, Bouchier-Hayes D: Vascular endothelial growth factor (VEGF), a survival factor for tumour cells: implications for anti-angiogenic therapy. Bioessays. 2002, 24 (3): 280-3.PubMedCrossRef
107.
Zurück zum Zitat Dias S: VEGF(165) promotes survival of leukemic cells by Hsp90-mediated induction of Bcl-2 expression and apoptosis inhibition. Blood. 2002, 99 (7): 2532-40.PubMedCrossRef Dias S: VEGF(165) promotes survival of leukemic cells by Hsp90-mediated induction of Bcl-2 expression and apoptosis inhibition. Blood. 2002, 99 (7): 2532-40.PubMedCrossRef
108.
Zurück zum Zitat Gerber HP: VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature. 2002, 417 (6892): 954-8.PubMedCrossRef Gerber HP: VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature. 2002, 417 (6892): 954-8.PubMedCrossRef
109.
Zurück zum Zitat Tang RF: Overexpression of lymphangiogenic growth factor VEGF-C in human pancreatic cancer. Pancreas. 2001, 22 (3): 285-92.PubMedCrossRef Tang RF: Overexpression of lymphangiogenic growth factor VEGF-C in human pancreatic cancer. Pancreas. 2001, 22 (3): 285-92.PubMedCrossRef
110.
Zurück zum Zitat Saimura M: Tumor suppression through angiogenesis inhibition by SUIT-2 pancreatic cancer cells genetically engineered to secrete NK4. Clin Cancer Res. 2002, 8 (10): 3243-9.PubMed Saimura M: Tumor suppression through angiogenesis inhibition by SUIT-2 pancreatic cancer cells genetically engineered to secrete NK4. Clin Cancer Res. 2002, 8 (10): 3243-9.PubMed
111.
Zurück zum Zitat Kleeff J: Detection and localization of Mip-3alpha/LARC/Exodus, a macrophage proinflammatory chemokine, and its CCR6 receptor in human pancreatic cancer. Int J Cancer. 1999, 81 (4): 650-7.PubMedCrossRef Kleeff J: Detection and localization of Mip-3alpha/LARC/Exodus, a macrophage proinflammatory chemokine, and its CCR6 receptor in human pancreatic cancer. Int J Cancer. 1999, 81 (4): 650-7.PubMedCrossRef
112.
Zurück zum Zitat Le X: Molecular regulation of constitutive expression of interleukin-8 in human pancreatic adenocarcinoma. J Interferon Cytokine Res. 2000, 20 (11): 935-46.PubMedCrossRef Le X: Molecular regulation of constitutive expression of interleukin-8 in human pancreatic adenocarcinoma. J Interferon Cytokine Res. 2000, 20 (11): 935-46.PubMedCrossRef
113.
Zurück zum Zitat Shi Q: Constitutive and inducible interleukin 8 expression by hypoxia and acidosis renders human pancreatic cancer cells more tumorigenic and metastatic. Clin Cancer Res. 1999, 5 (11): 3711-21.PubMed Shi Q: Constitutive and inducible interleukin 8 expression by hypoxia and acidosis renders human pancreatic cancer cells more tumorigenic and metastatic. Clin Cancer Res. 1999, 5 (11): 3711-21.PubMed
115.
Zurück zum Zitat Rowland-Goldsmith MA: Soluble type II transforming growth factor-beta (TGF-beta) receptor inhibits TGF-beta signaling in COLO-357 pancreatic cancer cells in vitro and attenuates tumor formation. Clin Cancer Res. 2001, 7 (9): 2931-40.PubMed Rowland-Goldsmith MA: Soluble type II transforming growth factor-beta (TGF-beta) receptor inhibits TGF-beta signaling in COLO-357 pancreatic cancer cells in vitro and attenuates tumor formation. Clin Cancer Res. 2001, 7 (9): 2931-40.PubMed
116.
Zurück zum Zitat Rowland-Goldsmith MA MH, Matsuda K, Idezawa T, Ralli M, Ralli S, Korc M: Soluble type II transforming growth factor-b receptor attenuates expression of metastasis-associated genes and suppresses pancreatic cancer cell metastasis. Mol Cancer Therapeutics. 2002, 1: 161-167. Rowland-Goldsmith MA MH, Matsuda K, Idezawa T, Ralli M, Ralli S, Korc M: Soluble type II transforming growth factor-b receptor attenuates expression of metastasis-associated genes and suppresses pancreatic cancer cell metastasis. Mol Cancer Therapeutics. 2002, 1: 161-167.
117.
Zurück zum Zitat Takeuchi Y: Expression of plasminogen activators and their inhibitors in human pancreatic carcinoma: immunohistochemical study. Am J Gastroenterol. 1993, 88 (11): 1928-33.PubMed Takeuchi Y: Expression of plasminogen activators and their inhibitors in human pancreatic carcinoma: immunohistochemical study. Am J Gastroenterol. 1993, 88 (11): 1928-33.PubMed
118.
Zurück zum Zitat Kleeff J: Overexpression of Smad2 and colocalization with TGF-beta1 in human pancreatic cancer. Dig Dis Sci. 1999, 44 (9): 1793-802.PubMedCrossRef Kleeff J: Overexpression of Smad2 and colocalization with TGF-beta1 in human pancreatic cancer. Dig Dis Sci. 1999, 44 (9): 1793-802.PubMedCrossRef
119.
Zurück zum Zitat Kleeff J, Korc M: Up-regulation of transforming growth factor (TGF)-beta receptors by TGF-beta1 in COLO-357 cells. J Biol Chem. 1998, 273 (13): 7495-500.PubMedCrossRef Kleeff J, Korc M: Up-regulation of transforming growth factor (TGF)-beta receptors by TGF-beta1 in COLO-357 cells. J Biol Chem. 1998, 273 (13): 7495-500.PubMedCrossRef
120.
Zurück zum Zitat Yang EY, Moses HL: Transforming growth factor beta 1-induced changes in cell migration, proliferation, and angiogenesis in the chicken chorioallantoic membrane. J Cell Biol. 1990, 111 (2): 731-41.PubMedCrossRef Yang EY, Moses HL: Transforming growth factor beta 1-induced changes in cell migration, proliferation, and angiogenesis in the chicken chorioallantoic membrane. J Cell Biol. 1990, 111 (2): 731-41.PubMedCrossRef
121.
Zurück zum Zitat Lambert V: Influence of plasminogen activator inhibitor type 1 on choroidal neovascularization. Faseb J. 2001, 15 (6): 1021-7.PubMedCrossRef Lambert V: Influence of plasminogen activator inhibitor type 1 on choroidal neovascularization. Faseb J. 2001, 15 (6): 1021-7.PubMedCrossRef
122.
Zurück zum Zitat Andreasen PA, Egelund R, Petersen HH: The plasminogen activation system in tumor growth, invasion, and metastasis. Cell Mol Life Sci. 2000, 57 (1): 25-40.PubMedCrossRef Andreasen PA, Egelund R, Petersen HH: The plasminogen activation system in tumor growth, invasion, and metastasis. Cell Mol Life Sci. 2000, 57 (1): 25-40.PubMedCrossRef
123.
Zurück zum Zitat Munger JS: Latent transforming growth factor-beta: structural features and mechanisms of activation. Kidney Int. 1997, 51 (5): 1376-82.PubMedCrossRef Munger JS: Latent transforming growth factor-beta: structural features and mechanisms of activation. Kidney Int. 1997, 51 (5): 1376-82.PubMedCrossRef
124.
Zurück zum Zitat Kojima S, Nara K, Rifkin DB: Requirement for transglutaminase in the activation of latent transforming growth factor-beta in bovine endothelial cells. J Cell Biol. 1993, 121 (2): 439-48.PubMedCrossRef Kojima S, Nara K, Rifkin DB: Requirement for transglutaminase in the activation of latent transforming growth factor-beta in bovine endothelial cells. J Cell Biol. 1993, 121 (2): 439-48.PubMedCrossRef
125.
126.
Zurück zum Zitat Ishiwata T: Altered expression of insulin-like growth factor II receptor in human pancreatic cancer. Pancreas. 1997, 15 (4): 367-73.PubMedCrossRef Ishiwata T: Altered expression of insulin-like growth factor II receptor in human pancreatic cancer. Pancreas. 1997, 15 (4): 367-73.PubMedCrossRef
127.
Zurück zum Zitat Elsasser HP: Characterization of a transglutaminase expressed in human pancreatic adenocarcinoma cells. Eur J Cell Biol. 1993, 61 (2): 321-8.PubMed Elsasser HP: Characterization of a transglutaminase expressed in human pancreatic adenocarcinoma cells. Eur J Cell Biol. 1993, 61 (2): 321-8.PubMed
128.
Zurück zum Zitat Haroon ZA: Tissue transglutaminase is expressed, active, and directly involved in rat dermal wound healing and angiogenesis. Faseb J. 1999, 13 (13): 1787-95.PubMed Haroon ZA: Tissue transglutaminase is expressed, active, and directly involved in rat dermal wound healing and angiogenesis. Faseb J. 1999, 13 (13): 1787-95.PubMed
129.
Zurück zum Zitat Mishima K: A peptide derived from the non-receptor-binding region of urokinase plasminogen activator inhibits glioblastoma growth and angiogenesis in vivo in combination with cisplatin. Proc Natl Acad Sci U S A. 2000, 97 (15): 8484-9.PubMedCentralPubMedCrossRef Mishima K: A peptide derived from the non-receptor-binding region of urokinase plasminogen activator inhibits glioblastoma growth and angiogenesis in vivo in combination with cisplatin. Proc Natl Acad Sci U S A. 2000, 97 (15): 8484-9.PubMedCentralPubMedCrossRef
130.
131.
Zurück zum Zitat Liu D: EGFR is a transducer of the urokinase receptor initiated signal that is required for in vivo growth of a human carcinoma. Cancer Cell. 2002, 1 (5): 445-57.PubMedCrossRef Liu D: EGFR is a transducer of the urokinase receptor initiated signal that is required for in vivo growth of a human carcinoma. Cancer Cell. 2002, 1 (5): 445-57.PubMedCrossRef
132.
Zurück zum Zitat Bancroft CC: Effects of pharmacologic antagonists of epidermal growth factor receptor, PI3K and MEK signal kinases on NF-kappaB and AP-1 activation and IL-8 and VEGF expression in human head and neck squamous cell carcinoma lines. Int J Cancer. 2002, 99 (4): 538-48.PubMedCrossRef Bancroft CC: Effects of pharmacologic antagonists of epidermal growth factor receptor, PI3K and MEK signal kinases on NF-kappaB and AP-1 activation and IL-8 and VEGF expression in human head and neck squamous cell carcinoma lines. Int J Cancer. 2002, 99 (4): 538-48.PubMedCrossRef
133.
Zurück zum Zitat Hirata A: ZD1839 (Iressa) induces antiangiogenic effects through inhibition of epidermal growth factor receptor tyrosine kinase. Cancer Res. 2002, 62 (9): 2554-60.PubMed Hirata A: ZD1839 (Iressa) induces antiangiogenic effects through inhibition of epidermal growth factor receptor tyrosine kinase. Cancer Res. 2002, 62 (9): 2554-60.PubMed
134.
Zurück zum Zitat Tsuzuki Y: Pancreas microenvironment promotes VEGF expression and tumor growth: novel window models for pancreatic tumor angiogenesis and microcirculation. Lab Invest. 2001, 81 (10): 1439-51.PubMedCrossRef Tsuzuki Y: Pancreas microenvironment promotes VEGF expression and tumor growth: novel window models for pancreatic tumor angiogenesis and microcirculation. Lab Invest. 2001, 81 (10): 1439-51.PubMedCrossRef
135.
Zurück zum Zitat Yamanaka Y: Synthesis and expression of transforming growth factor beta-1, beta-2, and beta-3 in the endocrine and exocrine pancreas. Diabetes. 1993, 42 (5): 746-56.PubMedCrossRef Yamanaka Y: Synthesis and expression of transforming growth factor beta-1, beta-2, and beta-3 in the endocrine and exocrine pancreas. Diabetes. 1993, 42 (5): 746-56.PubMedCrossRef
136.
Zurück zum Zitat Reinmuth N: Impact of insulin-like growth factor receptor-I function on angiogenesis, growth, and metastasis of colon cancer. Lab Invest. 2002, 82 (10): 1377-89.PubMedCrossRef Reinmuth N: Impact of insulin-like growth factor receptor-I function on angiogenesis, growth, and metastasis of colon cancer. Lab Invest. 2002, 82 (10): 1377-89.PubMedCrossRef
137.
Zurück zum Zitat Reinmuth N: Blockade of insulin-like growth factor I receptor function inhibits growth and angiogenesis of colon cancer. Clin Cancer Res. 2002, 8 (10): 3259-69.PubMed Reinmuth N: Blockade of insulin-like growth factor I receptor function inhibits growth and angiogenesis of colon cancer. Clin Cancer Res. 2002, 8 (10): 3259-69.PubMed
138.
Zurück zum Zitat Teraoka H: Enhanced VEGF production and decreased immunogenicity induced by TGF-beta 1 promote liver metastasis of pancreatic cancer. Br J Cancer. 2001, 85 (4): 612-7.PubMedCentralPubMedCrossRef Teraoka H: Enhanced VEGF production and decreased immunogenicity induced by TGF-beta 1 promote liver metastasis of pancreatic cancer. Br J Cancer. 2001, 85 (4): 612-7.PubMedCentralPubMedCrossRef
139.
142.
Zurück zum Zitat Liu CD: Vascular endothelial growth factor is increased in ascites from metastatic pancreatic cancer. J Surg Res. 2002, 102 (1): 31-4.PubMedCrossRef Liu CD: Vascular endothelial growth factor is increased in ascites from metastatic pancreatic cancer. J Surg Res. 2002, 102 (1): 31-4.PubMedCrossRef
143.
Zurück zum Zitat Niedergethmann M: High expression of vascular endothelial growth factor predicts early recurrence and poor prognosis after curative resection for ductal adenocarcinoma of the pancreas. Pancreas. 2002, 25 (2): 122-9.PubMedCrossRef Niedergethmann M: High expression of vascular endothelial growth factor predicts early recurrence and poor prognosis after curative resection for ductal adenocarcinoma of the pancreas. Pancreas. 2002, 25 (2): 122-9.PubMedCrossRef
145.
Zurück zum Zitat Baker CH, Solorzano CC, Fidler IJ: Angiogenesis and cancer metastasis: antiangiogenic therapy of human pancreatic adenocarcinoma. Int J Clin Oncol. 2001, 6 (2): 59-65.PubMedCrossRef Baker CH, Solorzano CC, Fidler IJ: Angiogenesis and cancer metastasis: antiangiogenic therapy of human pancreatic adenocarcinoma. Int J Clin Oncol. 2001, 6 (2): 59-65.PubMedCrossRef
Metadaten
Titel
Pathways for aberrant angiogenesis in pancreatic cancer
verfasst von
M Korc
Publikationsdatum
01.12.2003
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2003
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-2-8

Weitere Artikel der Ausgabe 1/2003

Molecular Cancer 1/2003 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.