<<

Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Clinical Cancer Review Research

Beyond VEGF: Inhibition of the Pathway and Antiangiogenesis

Christopher Lieu1, John Heymach2, Michael Overman1, Hai Tran2, and Scott Kopetz1

Abstract Fibroblast growth factor (FGF) signaling regulates , differentiation, survival, angiogen- esis, and . Compelling evidence for deregulated FGF signaling in tumorigenesis continues to emerge, and a growing body of research suggests that FGF may also play an integral role in the resistance to anti-VEGF therapy. Although agents targeting FGF signaling are early in development, the potential to target both the VEGF and FGF pathways may translate into improvements in the clinical care of cancer patients. Clin Cancer Res; 17(19); 1–10. 2011 AACR.

Introduction FGFs in the development of resistance to anti-VEGF ther- apy. Finally, FGF as a potential therapeutic target is Fibroblast growth factors (FGF) belong to a structurally reviewed, including the current state of drug development related family of 22 molecules that interact with high- in this field. affinity FGF receptors (FGFR) to regulate multiple fundamental pathways and cellular behaviors (1). Fibroblast Growth Factor Family FGF signaling regulates events in embryonal development, including mesenchymal–epithelial signaling and the de- FGFs were first isolated as from pituitary velopment of multiple organ systems (2, 3). FGF signaling extracts and bovine brain tissue in the 1970s (9, 10). Since also regulates cell proliferation, differentiation, and surviv- initial discovery, 22 members of the FGF family have been al, as well as and wound healing (4). In some identified, all of which are structurally related signaling situations, FGFs can act as a negative regulator of prolifer- molecules ranging from 17 to 34 kDa in size (11). Some ation and positive regulator of differentiation (5). Given FGFs seem to be expressed exclusively during embryonic the role of FGF in multiple cellular processes, compelling development (FGF-3, 4, 8, 15, 17, and 19), and others are evidence for deregulated FGF signaling in tumorigenesis expressed in both embryonic and adult tissues (FGF-1, 2, continues to emerge (6). Studies with animal models have 5–7, 9–14, 16, 18, and 20–23; ref. 11). suggested that aberrant FGF signaling can promote tumor FGFs are structurally similar to one another, with 28 development through increased cell proliferation and sur- highly conserved and 6 identical amino acid residues (12). vival, as well as increased tumor angiogenesis (7, 8). This Most FGFs are secreted glycoproteins (FGF-3–8, 10, 15, 17– body of research indicates that FGF signaling is an impor- 19, and 21–23), but FGF-1 and FGF-2 are exported from tant pathway in tumorigenesis and tumor angiogenesis and cells by mechanisms that remain unclear. FGF-1 and FGF-2 that appropriate targeting of the FGFR tyrosine kinases may may be released from damaged cells or by exocytosis that is be an effective therapeutic intervention for cancer. independent of the endoplasmic reticulum–Golgi pathway This review describes the FGF family of growth factors (13). FGFs have strong affinities for the glycosaminoglycan and their associated receptors, as well as their association side chains of cell surface proteoglycans, which help to with tumor development, growth, and metastasis. We also sequester FGFs on the surface of the secreting cell or on describe the effect of FGFs on tumor angiogenesis, includ- nearby cells, and in the binding of FGF to the FGFR. ing key preclinical and clinical evidence about the role of FGFRs are transmembrane tyrosine kinases that contain 2 or 3 extracellular immunoglobulin-like domains and an extracellular -binding sequence (14–16). The 2 Authors' Affiliations: Departments of 1Gastrointestinal Medical Oncology proximal immunoglobulin-like extracellular domains bind 2 and Thoracic/Head and Neck Medical Oncology, The University of Texas the FGF , whereas the heparin-binding sequence MD Anderson Cancer Center, Houston, Texas binds a glycosaminoglycan moiety, resulting in the forma- Corresponding Author: Scott Kopetz, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, tion of a complex containing 2 FGFs, 2 FGFRs, and the 1515 Holcombe Boulevard, Unit 0426, Houston, TX 77030. Phone: 713- glycosaminoglycan moiety (17). Only 4 FGFRs are known 792-2828; Fax: 713-563-6764; E-mail: [email protected] to exist, designated FGFR-1 through FGFR-4 (Table 1). doi: 10.1158/1078-0432.CCR-11-0659 Their specificity for various ligands is altered by processes 2011 American Association for Cancer Research. that create multiple isoforms. Alternative mRNA splicing of

www.aacrjournals.org OF1

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Lieu et al.

Translational Relevance Fibroblast Growth Factor and Tumor Growth

Angiogenic inhibitors have shown promise in the Aberrant FGF and/or FGFR signaling has a wide range of treatment of multiple tumor types and have improved effects and can involve both tumor cells and the surround- outcomes in patients with metastatic disease. However, ing stroma. These effects include cellular proliferation; many patients will not respond to antiangiogenic ther- resistance to cell death; increased motility and invasiveness; apy, and for those that do, resistance develops quickly. increased angiogenesis; enhanced metastasis; and resis- Fibroblast growth factor (FGF) seems to play an integral tance to chemotherapy (27). Deregulation of FGF signaling role in the resistance to antiangiogenic therapy, and in tumorigenesis can result from activating mutations, agents that specifically target the FGF pathway are being FGFR gene amplifications, and chromosomal transloca- developed and tested in clinical trials. It is critical that tions, which lead to increased autocrine and paracrine investigators review the existing literature on FGF and signaling (28–30). Because aberrant signaling can promote tumor angiogenesis to understand the importance of tumorigenesis by affecting major downstream biologic this target in various malignancies, as well as to design processes, FGFs have been implicated in multiple tumor rational clinical trials that can expedite a potentially types, including prostate, astrocytoma, breast, , blad- meaningful therapy in patients previously treated with der, hepatocellular, and colon cancers (31–41). antiangiogenic agents. Alternative splicing of the FGFR gene has also been implicated in carcinogenesis. Carcinomas have been ob- served to switch expression of FGFR to the mesenchymal the FGFR gene significantly alters the ligand specificity by isoforms, enabling cells to receive signals usually restricted changing the sequence of the carboxy-terminal half of to the connective tissue, such as ectopic expression of immunoglobulin-domain III. This sequence change results typically stroma-localized FGFR1-IIIc (42, 43). The IIIc in 2 isoforms termed IIIb and IIIc (18, 19). In general, IIIb splice variant has been detected in colon carcinoma but isoforms are expressed on epithelial cells, and IIIc isoforms not in adenoma-derived cell lines, suggesting that it is are expressed on mesenchymal cells, where they mediate upregulated during tumor progression (44). During pros- tissue interactions in normal development and wound tate cancer progression, alternative splicing of FGFR2 can healing (20–22). occur in epithelial cells, leading to isoform changes from FGF signaling can also be modulated by several mem- FGFR2-IIIb into the more ligand-promiscuous FGFR2-IIIc brane , including cell adhesion molecules of the (Fig. 1; refs. 8, 43). Thus, this aberrant alternative splicing cadherin and immunoglobulin superfamilies (23). The of FGFR potentially destroys the balanced interdependence best characterized of these is neural cell adhesion molecule between stroma and (8, 43, 45). (NCAM). NCAM has been shown to be a major regulator of Clinical studies of FGF have suggested a critical role of FGF-FGFR interaction in several cell types, including non- FGF signaling in clinical tumor progression. In 1 study of neural tissues (24). NCAM has been shown to act as a advanced serous ovarian adenocarcinomas, FGF-1 mRNA nonconventional ligand that can induce a specific FGFR1- copy number was found to be significantly correlated with mediated cellular migration that is different from the DNA copy number and expression levels (46). Both response elicited by FGF-2 (25). Furthermore, NCAM ex- FGF-1 mRNA and protein levels were associated with worse pression has been shown to reduce FGF-stimulated extra- overall survival, possibly secondary to an increase in tumor cellular signal regulated kinase (ERK)1/2 activation, cell angiogenesis and autocrine stimulation of cancer cells. In proliferation, and cell-matrix adhesion in fibroblast cell another study of 100 resected colorectal cancer surgical lines, suggesting that NCAM acts as a novel control mech- specimens, high FGF-2 expression was associated with anism for FGFR signaling (26). increased grade, stage, lymphovascular invasion, and intra- tumoral microvessel density (47). Significantly elevated levels of FGF-2 in circulation were found in a small cohort of patients with metastatic colorectal cancer, relative to Table 1. Receptors for the FGFR family median levels in control subjects (48). In other studies, although increased levels of FGF-2 were found in plasma Receptor Specific For from patients with different types of cancer, it was unclear whether the higher levels resulted from tumor invasion and FGFR-1b FGF-1, 2, 3, and 10 degradation of the (ECM), liberating FGFR-1c FGF-1, 2, 4, 5, and 6 FGF to function as a paracrine growth factor, or whether FGFR-2b FGF-1, 3, 7, and 10 tumor cells induced FGF2 release from stromal inflamma- FGFR-2c FGF-1, 2, 4, 6, and 9 tory infiltrate (49, 50). FGFR-3b FGF-1 and 9 Loss of expression of NCAM has also been implicated in FGFR-3c FGF-1, 2, 4, 8, and 9 the progression of tumor metastasis. NCAM has been FGFR-4 FGF-1, 2, 4, 6, 8, and 9 shown to modulate neurite outgrowth and matrix adhesion b Adapted from Itoh and Ornitz (1). of cells from the Rip1-Tag2 transgenic mouse model of pancreatic neuroendocrine tumors by assembling a FGFR-4

OF2 Clin Cancer Res; 17(19) October 1, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Inhibition of the FGF Pathway and Antiangiogenic Therapy

Stroma Normal Epithelium Stroma Cancer Epithelium FGFR ECM FGFR ECM lllb lllc FGFR lllc lllc

PDGF

FGF

FGF FGF-BP FGF FGF VEGF Ang2

Blood Blood FGFR lllc NRP1 PDGFR vessel vessel FGFR lllc

© 2011 American Association for Cancer Research

Figure 1. Changes in FGF and FGFR in the microenvironment and epithelium in normal tissue and cancer. Compared with normal tissue, alternative splicing of the FGFR gene can switch expression of FGFR from the IIIb isoform to the ligand promiscuous IIIc isoforms. Furthermore, release of FGF from the tumor and stroma may initiate tumor angiogenesis. FGF-BP–mediated reduction of the affinity of FGF-2 to heparin can induce its release from the ECM. FGF-BP, FGF binding protein; PDGF, -derived growth factor; PDGFR, platelet-derived . signaling complex that leads to the modulation of b1- lagen gel, VEGF and FGF-2 were able to induce cells to –mediated cell-matrix adhesion (51). Ablation of invade the underlying matrix to form capillary-like tubules the expression of NCAM in this mouse model resulted in (60). Interestingly, FGF-2 was found to be twice as potent the disruption of the tumor tissue architecture, tumor- as VEGF in this model. The critical role of FGF in tumor associated lymphangiogenesis, and lymph node metastasis angiogenesis was shown through the interference of FGF (51, 52). function with a recombinant adenovirus expressing soluble Though a full review of FGF signaling and tumor growth FGFR (AdsFGFR). AdsFGFR seemed to impair the mainte- is beyond the scope of this article, please see the compre- nance of tumor angiogenesis, in contrast to inhibition of hensive review from Turner and Grose for additional the VEGF pathway that predominantly affected the initia- information (41). tion of tumor angiogenesis (61). Injection of AdsFGFR in the Rip1-Tag2 murine model of pancreatic neuroendocrine Fibroblast Growth Factors and Angiogenesis tumor resulted in repression of tumor growth, with a significant decrease in tumor vessel density. FGFs exert Angiogenesis, the formation of new blood vessels from their effects by modulating proliferation and migration the of the existing vasculature, plays a pivotal of endothelial cells, production of proteases, and promo- role in tumor growth, progression, and metastasis (53, 54). tion of integrin and cadherin receptor expression (62). Along with embryogenesis, angiogenesis was one of the In particular, FGF-1 and FGF-2 directly affect tumor first areas where FGF signaling was shown to be important angiogenesis by promoting the cellular proliferation of (55, 56). In particular, FGF-1 and FGF-2 were found to be endothelial cells. FGF-2 has been shown to induce revas- important for new growth at the site of an cularization in various experimental models in several excision wound and have been shown to stimulate angio- species, including rabbit, chicken, and mouse (63–65). genesis in various assays (57–59). FGF-2 is a very potent In a murine model, FGF-1 knockout mice and FGF-1/ inducer of angiogenesis. In a preclinical model of micro- FGF-2 double-knockout mice had poor wound healing vascular endothelial cells grown on a 3-dimensional col- compared with normal control mice (58). FGFR-1 is most

www.aacrjournals.org Clin Cancer Res; 17(19) October 1, 2011 OF3

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Lieu et al.

commonly expressed on endothelial cells, although FGFR- esis than does FGF (50). In 1 study, the angiogenic response 2 has been shown to be present under certain circum- occurred more quickly when VEGF and FGF-2 were added stances (50, 66). The presence of FGFR-3 and FGFR-4 simultaneously to microvascular endothelial cells on 3- has not been reported in the endothelium. dimensional collagen gels than the response to either cyto- FGFs can exert their angiogenic mechanism of action via kine alone, suggesting synergy between the 2 (60). through their release by tumor and It has also been shown that induction of FGF-2–induced stromal cells, or through their mobilization from the angiogenesis is partly dependent on the activation of VEGF ECM (50). FGF-1, in particular, may stimulate endothelial and the presence of endogenous VEGF and VEGF-C (76). cell growth and the motility of endothelial cells through Evidence for synergy between VEGF and FGF-2 has also been paracrine signaling, which results in increased prolifera- shown in xenograft models expressing FGF-2 and VEGF in tion, survival, and motility of endothelial cells (46). FGF-2 tumor cell transfectants. Simultaneous expression of FGF-2 has been studied in xenograft models in which liposome- and VEGF resulted in fast-growing lesions with high blood mediated gene transfer was used to deliver episomal vectors vessel density and permeability (77). Interestingly, inhibi- containing antisense FGF-2 or FGFR-1 cDNAs into human tion of FGF-2 production caused a significant decrease in melanomas grown in nude mice. In contrast to the stim- tumor burden with a concomitant decrease in blood vessel ulation seen during deregulated FGF signaling, tumors density and size. This differed from the effects of VEGF injected with these antisense constructs showed arrested inhibition, which caused a decrease in pericyte organization, tumor growth or reduced tumor density because of blocked vessel patency, and permeability. This finding suggests that intratumoral angiogenesis (67). There is also evidence that FGF-2 and VEGF stimulate angiogenesis synergistically but FGF-2 increases the expression of -2, a potent with different effects on vessel size and function. regulator of vascular branching and angiogenesis, as well as FGF-2 may also have indirect effects on VEGF pathways. VEGF in cultured human and endothelial cells (68). In a breast cancer cell line (T47D), FGF-2 was found to FGFs may also exert their effects through autocrine activate hypoxia-induced VEGF release through augmenta- signaling in endothelial cells, stimulating the proliferation tion of the phosphoinositide 3-kinase pathway, as well as of new capillary endothelial cells (66). This induces a hypoxia-inducible factor-1a expression (78). FGF-2 has proangiogenic state that creates an environment favorable also been shown to indirectly enhance the effect of VEGF for vascular growth. Evidence for this growth is seen in a through upregulation of neurolipin-1 (NRP-1), a corecep- particular subtype of Kaposi sarcoma (KS) in which FGF-2, tor for VEGF. FGF-2 increased NRP-1 levels and enhanced produced by the KS cells, has been found to synergize with the migration of human vascular smooth muscle cells in human immunodeficiency virus type 1 Tat protein to response to VEGF. enhance endothelial cell growth and type IV collagenase There is also evidence of cross-talk between FGF-2 and expression (66, 69). These tumors were found to have platelet-derived growth factor (PDGF)–BB. FGF-2 and increased aggressiveness and frequency compared with PDGF-BB together have been shown to synergistically other forms of KS. promote tumor angiogenesis and pulmonary metastasis, Preclinical models of tumor angiogenesis have suggested as well as formation of high-density primitive vascular that FGF–binding protein (FGF-BP) serves as an angiogenic plexuses coated with pericytes and vascular smooth muscle switch molecule and can be rate limiting for tumor growth cells (79). Pericyte recruitment to coat nascent vessels (70). FGF-2 is present in most tissues under normal phys- during angiogenesis is essential for the stabilization and iologic conditions but is inactivated by being tightly bound further establishment of a tumor’s vascular network. FGF-2 to the ECM. FGF-BP binds to FGF-2 in a dose-dependent seems to upregulate PDGF receptor (PDGFR) expression, manner, which results in a marked reduction of the affinity which causes increased responsiveness to PDGF-BB, and of FGF-2 to heparin and release from the ECM (Fig. 1; refs. PDGF-BB–treated vascular smooth muscle cells become 71, 72). Exogenously administered FGF-BP has been shown responsive to FGF-2 signaling through upregulation of to stimulate FGF-2–dependent growth of both adrenal FGFR-1. Interestingly, overexpression of PDGF-BB alone carcinoma and prostate carcinoma cells. In addition, down- in tumor cells was shown to result in decreased recruitment regulation of FGF-BP expression has been shown to inhibit of pericytes and dissociation of vascular smooth muscle angiogenesis in squamous cell carcinoma xenografts (73). cells. In tumor cells subjected to prior upregulation of FGF-2 caused by VEGF receptor 2 (VEGFR2) inhibition, Fibroblast Growth Factor–2 Cross-Talk with PDGFR-b inhibition also attenuated the expression of Angiogenic Factors FGF-2 (80). In a mouse model of cervical carcinogenesis, pharmacologic blockade of PDGFR signaling with VEGF plays an integral role in the development of new slowed progression of premalignant cervical lesions and blood vessels in tumor formation, and angiogenesis inhi- impaired the growth of preexisting invasive carcinomas bitors targeting the VEGF pathway have proved to be effi- through suppression of the expression of FGF-2 and cacious in preclinical cancer models and in clinical trials (74, FGF-7, which resulted in decreased proliferation and an- 75). A significant amount of cross-talk is thought to exist giogenesis within the lesions (81). Treatment with an FGF between members of the VEGF family and FGF-2 during trap also impaired the angiogenic phenotype similar to the angiogenesis, with VEGF appearing earlier during angiogen- blockade of the PDGFR with imatinib.

OF4 Clin Cancer Res; 17(19) October 1, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Inhibition of the FGF Pathway and Antiangiogenic Therapy

Fibroblast Growth Factor Signaling and baseline levels of VEGF. In 1 xenograft model of renal cell Resistance to Anti-VEGF Therapy carcinoma, tumors unresponsive to treatment were sensitive to a dual inhibitor of the VEGF and FGFRs Emerging evidence has suggested that upregulation of (85). This inhibitor seemed to reduce blood vessel density FGF and FGFR may serve as a mechanism of resistance to through inhibition of FGF-2–induced angiogenesis. anti-VEGF therapy. Preclinical trials of anti-VEGF therapy Clinical evidence in colon cancer also supports the role in a murine pancreatic neuroendocrine tumor model of FGF-2 in resistance to -containing regi- (PNET), Rip1-Tag2, showed that tumors with an initial mens. To determine the dynamic changes in circulating response to VEGFR2 blockade expressed higher levels of factors during the emergence of therapeutic resistance, FGF-2 at the time of progression compared with stable profiles of cytokines and angiogenic factors were assessed tumors (82). When the tumors were first treated with a in 40 patients receiving a regimen of 5-fluorouracil (5-FU), VEGFR inhibitor alone and subsequently treated at the leucovorin, and irinotecan (FOLFIRI) plus bevacizumab. peak of response with an FGF trap, the combination Levels were assessed after a single dose of bevacizumab and attenuated revascularization and slowed tumor growth FOLFIRI plus bevacizumab, at the time of progression, and (Fig. 2). The dual tyrosine kinase inhibitor (TKI) of VEGFR at the nadir of radiographic response (86, 87). The inves- and FGFR, brivanib, has also been studied in this murine tigators showed that FGF-2 levels were elevated immedi- model and has proved to be efficacious following the ately prior to progression compared with baseline levels. failure of 2 VEGFR inhibitors (83). It was even more Strikingly, 30% of patients had an increase in FGF-2 to 10- effective when used in the frontline setting, suggesting that fold the upper limit of normal, suggesting significant dual inhibition of VEGFR and FGFR was able to delay heterogeneity in the FGF-2 response. In 1 study of neoad- induction of evasive resistance. Further evidence for the juvant bevacizumab with 5-FU and radiation in rectal role of alternative angiogenic factors in tumor escape from cancer, the administration of a short regimen of bevacizu- angiogenesis inhibitors has been shown in tumors in which mab in combination with chemotherapy had no significant growth was impaired by ectopic expression of the endog- effect on plasma FGF-2 levels, suggesting that elevations in enous angiogenesis inhibitors thrombospondin-1, tumsta- FGF-2 are instead a later event temporally associated with tin, and endostatin (84). In that study, renal carcinoma the development of resistance to the bevacizumab combi- cells transfected with thrombospondin-1 resisted angio- nation regimen (88). genesis inhibition by upregulating the proangiogenic fac- Elevated FGF-2 levels have also been shown in glioblas- tors FGF-2, angiopoietin 2, and PDGF-A, despite high toma patients treated with a VEGFR small molecule

160

140

120 Figure 2. VEGFR2 blockade

) transiently stops tumor growth 3 * and decreases vascularity, 100 followed by tumor progression, reinduction of angiogenesis, and reestablishment of the tumor 80 vasculature. Quantification of overall tumor burden plotted as average and SD per animal at the 60 time of initiation of treatment

Tumor burden (mm burden Tumor (t ¼ 0; purple bar), after treatment for 10 days (orange bars), and at 40 4 weeks of age (teal and green bars) for the different treatment arms. Averages from cohorts of 8 20 to 10 mice per group are plotted with their respective SD bars. (Adapted from Cancer Cell, with 0 permission from Elsevier; ref. 82.) Control Control Anti-R2 Anti-R2 Anti-R2 FGF-trap t = 0 10 days 4 weeks

www.aacrjournals.org Clin Cancer Res; 17(19) October 1, 2011 OF5

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Lieu et al.

inhibitor (89). In patients who experienced tumor progres- from binding, and it had in vitro activity against lung and sion while receiving treatment, increased tumor enhance- renalcellcarcinoma(92). ment volume was associated with significant increases in The safety of FGFR inhibitors currently under develop- plasma levels of FGF-2 and stromal cell–derived factor-1. A ment seems acceptable. Preclinical toxicity studies with statistically significant positive correlation was observed these agents have not shown a reproducible class effect, between FGF-2 levels and tumor vessel size measured by aside from hypertension, which is difficult to attribute to MRI. This work provided further clinical evidence that FGF- the VEGFR or FGFR inhibitory effects of many of the agents 2 may play a role in tumor relapse in patients treated with (93, 94). Monoclonal antibodies directed against FGFR1- anti-VEGF agents (89). IIIc resulted in anorexia in mice and monkeys, which was Unlike preclinical models, clinical studies investigating caused by an FGF signaling blockade in the hypothalamus, levels of FGF can only show an association of elevated and a similar toxicity was seen in 1 phase I study (95, 96). FGF and/or FGFR with the resistance to anti-VEGF ther- Encouragingly, the concerns based on preclinical studies apy; they cannot show causation or an actual resistance about phosphate and vitamin D metabolism have not been mechanism. Furthermore, because most studies are con- reflected in the reported toxicities in the early-phase studies ducted in combination with cytotoxic chemotherapy, it is (97). difficult to discern whether the patient and tumor have Although a separate development strategy is being pur- become resistant to the chemotherapy, the antiangiogenic sued based on somatic mutation or amplification of FGFR therapy, or both. Therefore, high-quality preclinical mod- (reviewed in ref. 98), antiangiogenesis strategies targeting els will be essential for further investigation to provide a the pose additional drug devel- sound preclinical rationale for future clinical studies. opment strategies. Prior studies of anti-VEGF antibodies have shown clinical utility when the agents are used in Targeting Fibroblast Growth Factor combination with cytotoxic therapy and minimal activity as single agents with rare exceptions (75, 99). Similarly, Because FGFs have a clearly defined role in tumor FGF inhibition alone is unlikely to show a significant angiogenesis, as well as a possible role in resistance to benefit in various tumor types. Combination strategies current VEGF inhibitors, several FGF pathway inhibitors of FGF/FGFR inhibition with traditional cytotoxic therapy are currently under development. The inhibitors entering in tumor types in which proliferation and angiogenesis is the clinic are either TKIs or soluble antibody fragments FGF/FGFR–dependent will have the highest likelihood of that block ligand binding and receptor dimerization achieving a clinical benefit. (Table 2). Most of the TKIs have dual specificity for FGFR The timing of FGF/FGFR–directed therapy is also an area and VEGFR, both as a matter of design and because of the of emerging interest given the evolving evidence that the structural similarities in their kinase domains. However, FGF axis may serve as an adaptive resistance mechanism to the inhibitors vary in their relative potency for VEGFR and anti-VEGF therapy. Histopathologic evidence from a mu- FGFR, with many inhibitors having higher potency for rine PNET model suggests that the switch to an FGFR VEGFR than FGFR (90, 91). Targeting both kinases may inhibitor during treatment with a VEGFR inhibitor may also increase the toxicity associated with these com- be more effective at the time of early revascularization prior pounds, although biologically active doses have been to detectable tumor growth (83). Future clinical trials will shown with most of the compounds (41). The other class help determine whether dual inhibition of VEGFR and of compound in development, FGF ligand trap, has the FGFR will be more effective in the front-line setting or potential to block the interaction of multiple FGF ligands whether FGFR inhibition will be better instituted at the with the soluble FGFR fragments. In 1 study, such an time prior to or at progression after treatment with VEGF/ FGFR-1 antagonist was shown to prevent FGFR-1 ligands VEGFR inhibitors. However, determination of biologic

Table 2. Current FGF and FGFR-targeting agents

Drug Manufacturer Target(s) Clinical development stage

TKI258 (dovitinib; ref. 96) Novartis FGFR, PDGFR, VEGFR Phase II BIBF 1120 (Vargatef; ref. 104) Boehringer Ingelheim FGFR, PDGFR, VEGFR Phase III BMS-582664 (brivanib; ref. 105) Bristol-Myers Squibb FGFR and VEGFR Phase II and III E7080 (106) Eisai FGFR, PDGFR, VEGFR Phase I TSU-68 (SU6668; ref. 107) Taiho Pharmaceutical FGFR, PDGFR, VEGFR Phase I and II AZD4547 (108) AstraZeneca FGFR Phase I FP-1039 (109) Five Prime Therapeutics FGF ligand trap (multiple FGFs) Phase I BGJ398 (110) Novartis FGFR Phase I Astex FGFR inhibitor (111) Janssen Oncology FGFR Preclinical

OF6 Clin Cancer Res; 17(19) October 1, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Inhibition of the FGF Pathway and Antiangiogenic Therapy

resistance to VEGF/VEGFR inhibitors is difficult in the also play a large role in angiogenesis, which is a funda- clinic, as resistance to the cytotoxic component of the mental step in the transition from a dormant to a malig- treatment may be sufficient to induce tumor growth in nant state, and FGF acts synergistically with VEGF to the absence of single-agent anti-VEGF activity. Determining increase tumor blood vessel growth and maturation. Evi- resistance to these inhibitors is a high hurdle for develop- dence is also increasing that FGF may be part of the ment of such alternate antiangiogenic therapies, and sev- mechanism of resistance to anti-VEGF agents. eral strategies are being considered in the early clinical Although agents targeting FGF signaling are still early development, including randomized discontinuation stud- in development, the potential to target both the VEGF ies and enrichment designs. and FGF pathways is near, and the combination or Biomarkers under consideration for FGF inhibitors in- sequential inhibition of these 2 critical angiogenic path- clude plasma levels of FGF family members and increased ways may translate into improvements in the clinical care FGF-axis expression in the stroma; however, their use as of cancer patients. Successful strategies will depend on surrogates for FGF/FGFR–dependent tumors remains un- appropriate selection of tumors and patients in whom clear. Integration of imaging or biomarkers of antiangio- FGF inhibition is mostly likely to inhibit angiogenesis genesis therapy likewise has hurdles of cost reproducibility and proliferation. and lack of correlation with clinical efficacy, and these approaches are not routinely integrated into most clinical Disclosure of Potential Conflicts of Interest development strategies of these agents (100, 101). Based on the VEGF inhibitor experiences, radiographic endpoints S. Kopetz: commercial research grant, AstraZeneca. The other authors disclosed no potential conflicts of interest. with antiangiogenesis strategies will be difficult, and ulti- mately, the benefits of these agents will be measured by Grant Support their ability to improve overall survival (102, 103). NIH T32 CA-009666 (C. Lieu); National Research Service Award (NRSA) Summary Research Training Grant; Conquer Cancer Foundation Young Investigators Award (C. Lieu); NIH CA-136980 (S. Kopetz); NIH/National Cancer Institute (NCI) Cancer Center Support Grant (CCSG) Core Grant CA-106672. FGFs are a family of growth factors that are involved in many pathways that can contribute to carcinogenesis and Received March 18, 2011; revised July 20, 2011; accepted July 25, 2011; affect cellular proliferation, migration, and survival. They published OnlineFirst September 27, 2011.

References 1. Itoh N, Ornitz DM. Evolution of the Fgf and Fgfr gene families. Trends 12. Ornitz DM. FGFs, and FGFRs: complex interactions Genet 2004;20:563–9. essential for development. Bioessays 2000;22:108–12. 2. De Moerlooze L, Spencer-Dene B, Revest JM, Hajihosseini M, 13. Mignatti P, Morimoto T, Rifkin DB. Basic fibroblast growth factor, a Rosewell I, Dickson C. An important role for the IIIb isoform of protein devoid of secretory signal sequence, is released by cells via a fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithe- pathway independent of the endoplasmic reticulum-Golgi complex. lial signalling during mouse organogenesis. Development 2000; J Cell Physiol 1992;151:81–93. 127:483–92. 14. Lee PL, Johnson DE, Cousens LS, Fried VA, Williams LT. Purification 3. Yamaguchi TP, Harpal K, Henkemeyer M, Rossant J. fgfr-1 is and complementary DNA cloning of a receptor for basic fibroblast required for embryonic growth and mesodermal patterning during growth factor. Science 1989;245:57–60. mouse . Genes Dev 1994;8:3032–44. 15. Johnson DE, Williams LT. Structural and functional diversity in the 4. Baird A, Esch F, Mormede P, Ueno N, Ling N, Bohlen€ P, et al. FGF receptor multigene family. Adv Cancer Res 1993;60:1–41. Molecular characterization of fibroblast growth factor: distribution 16. McKeehan WL, Wang F, Kan M. The heparan sulfate-fibroblast and biological activities in various tissues. Recent Prog Horm Res growth factor family: diversity of structure and function. Prog Nucleic 1986;42:143–205. Acid Res Mol Biol 1998;59:135–76. 5. Colvin JS, Bohne BA, Harding GW, McEwen DG, Ornitz DM. Skeletal 17. Naski MC, Ornitz DM. FGF signaling in skeletal development. Front overgrowth and deafness in mice lacking fibroblast growth factor Biosci 1998;3:d781–94. receptor 3. Nat Genet 1996;12:390–7. 18. Miki T, Bottaro DP, Fleming TP, Smith CL, Burgess WH, Chan AM, 6. Grose R, Dickson C. Fibroblast growth factor signaling in tumori- et al. Determination of ligand-binding specificity by alternative splic- genesis. Growth Factor Rev 2005;16:179–86. ing: two distinct growth factor receptors encoded by a single gene. 7. Dickson C, Spencer-Dene B, Dillon C, Fantl V. Tyrosine kinase Proc Natl Acad Sci U S A 1992;89:246–50. signalling in breast cancer: fibroblast growth factors and their 19. Chellaiah AT, McEwen DG, Werner S, Xu J, Ornitz DM. Fibroblast receptors. Breast Cancer Res 2000;2:191–6. growth factor receptor (FGFR) 3. Alternative splicing in immunoglob- 8. Feng S, Wang F, Matsubara A, Kan M, McKeehan WL. Fibroblast ulin-like domain III creates a receptor highly specific for acidic FGF/ growth factor receptor 2 limits and receptor 1 accelerates tumori- FGF-1. J Biol Chem 1994;269:11620–7. genicity of prostate epithelial cells. Cancer Res 1997;57:5369–78. 20. Orr-Urtreger A, Bedford MT, Burakova T, Arman E, Zimmer Y, Yayon 9. Armelin HA. Pituitary extracts and steroid in the control of A, et al. Developmental localization of the splicing alternatives 3T3 cell growth. Proc Natl Acad Sci U S A 1973;70:2702–6. of fibroblast growth factor receptor-2 (FGFR2). Dev Biol 1993;158: 10. Gospodarowicz D. Localisation of a fibroblast growth factor and its 475–86. effect alone and with hydrocortisone on 3T3 cell growth. Nature 21. Murgue B, Tsunekawa S, Rosenberg I, deBeaumont M, Podolsky 1974;249:123–7. DK. Identification of a novel variant form of fibroblast growth factor 11. Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol 2001;2: receptor 3 (FGFR3 IIIb) in human colonic epithelium. Cancer Res reviews3005.1–3005.12. 1994;54:5206–11.

www.aacrjournals.org Clin Cancer Res; 17(19) October 1, 2011 OF7

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Lieu et al.

22. Scotet E, Houssaint E. The choice between alternative IIIb and IIIc growth factor (FGF)-FGF receptor genes in prostate epithelial cells exons of the FGFR-3 gene is not strictly tissue-specific. Biochim accompany stromal independence and malignancy. Mol Cell Biol Biophys Acta 1995;1264:238–42. 1993;13:4513–22. 23. Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins 43. Carstens RP, Eaton JV, Krigman HR, Walther PJ, Garcia-Blanco MA. and Ig-CAMs in cancer. Nat Rev Cancer 2004;4:118–32. Alternative splicing of fibroblast growth factor receptor 2 (FGF-R2) in 24. Hinsby AM, Berezin V, Bock E. Molecular mechanisms of NCAM human prostate cancer. Oncogene 1997;15:3059–65. function. Front Biosci 2004;9:2227–44. 44. Sonvilla G, Allerstorfer S, Heinzle C, Stattner€ S, Karner J, Klimpfinger 25. Francavilla C, Cattaneo P, Berezin V, Bock E, Ami D, de Marco A, M, et al. Fibroblast growth factor receptor 3-IIIc mediates colorectal et al. The binding of NCAM to FGFR1 induces a specific cellular cancer growth and migration. Br J Cancer 2010;102:1145–56. response mediated by receptor trafficking. J Cell Biol 2009;187: 45. Acevedo VD, Gangula RD, Freeman KW, Li R, Zhang Y, Wang F, et al. 1101–16. Inducible FGFR-1 activation leads to irreversible prostate adenocar- 26. Francavilla C, Loeffler S, Piccini D, Kren A, Christofori G, Cavallaro U. cinoma and an epithelial-to-mesenchymal transition. Cancer Cell Neural cell adhesion molecule regulates the cellular response to 2007;12:559–71. fibroblast growth factor. J Cell Sci 2007;120:4388–94. 46. Birrer MJ, Johnson ME, Hao K, Wong K-K, Park D-C, Bell A, et al. 27. Kwabi-Addo B, Ozen M, Ittmann M. The role of fibroblast growth Whole genome oligonucleotide-based array comparative genomic factors and their receptors in prostate cancer. Endocr Relat Cancer hybridization analysis identified fibroblast growth factor 1 as a 2004;11:709–24. prognostic marker for advanced-stage serous ovarian adenocarci- 28. Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell nomas. J Clin Oncol 2007;25:2281–7. G, et al. Patterns of somatic mutation in human cancer genomes. 47. Elagoz S, Egilmez R, Koyuncu A, Muslehiddinoglu A, Arici S. The Nature 2007;446:153–8. intratumoral microvessel density and expression of bFGF and nm23- 29. Kunii K, Davis L, Gorenstein J, Hatch H, Yashiro M, Di Bacco A, et al. H1 in colorectal cancer. Pathol Oncol Res 2006;12:21–7. FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 48. George ML, Tutton MG, Abulafi AM, Eccles SA, Swift RI. Plasma signaling for growth and survival. Cancer Res 2008;68:2340–8. basic fibroblast growth factor levels in colorectal cancer: a clinically 30. Yagasaki F, Wakao D, Yokoyama Y, Uchida Y, Murohashi I, Kayano useful assay? Clin Exp Metastasis 2002;19:735–8. H, et al. Fusion of ETV6 to fibroblast growth factor receptor 3 in 49. Poon RT-P, Fan S-T, Wong J. Clinical implications of circulating peripheral T-cell lymphoma with a t(4;12)(p16;p13) chromosomal angiogenic factors in cancer patients. J Clin Oncol 2001;19:1207–25. translocation. Cancer Res 2001;61:8371–4. 50. Presta M, Dell’Era P, Mitola S, Moroni E, Ronca R, Rusnati M. 31. Linder C, Bystrom€ P, Engel G, Auer G, Aspenblad U, Strander H, et al. Fibroblast growth factor/fibroblast growth factor receptor system Correlation between basic fibroblast growth factor immunostaining in angiogenesis. Cytokine Growth Factor Rev 2005;16:159–78. of stromal cells and stromelysin-3 mRNA expression in human 51. Cavallaro U, Niedermeyer J, Fuxa M, Christofori G. N-CAM mod- breast carcinoma. Br J Cancer 1998;77:941–5. ulates tumour-cell adhesion to matrix by inducing FGF-receptor 32. Halaban R. Growth factors and melanomas. Semin Oncol 1996; signalling. Nat Cell Biol 2001;3:650–7. 23:673–81. 52. Perl AK, Dahl U, Wilgenbus P, Cremer H, Semb H, Christofori G. 33. Bian XW, Du LL, Shi JQ, Cheng YS, Liu FX. Correlation of bFGF, Reduced expression of neural cell adhesion molecule induces met- FGFR-1 and VEGF expression with vascularity and malignancy of astatic dissemination of pancreatic beta tumor cells. Nat Med human astrocytomas. Anal Quant Cytol Histol 2000;22:267–74. 1999;5:286–91. 34. Relf M, LeJeune S, Scott PA, Fox S, Smith K, Leek R, et al. 53. Folkman J, Klagsbrun M, Sasse J, Wadzinski M, Ingber D, Vlodavsky Expression of the angiogenic factors vascular endothelial cell growth I. A heparin-binding angiogenic protein—basic fibroblast growth factor, acidic and basic fibroblast growth factor, tumor growth factor factor—is stored within basement membrane. Am J Pathol 1988; beta-1, platelet-derived endothelial cell growth factor, placenta 130:393–400. growth factor, and in human primary breast cancer 54. Ingber DE, Folkman J. Mechanochemical switching between growth and its relation to angiogenesis. Cancer Res 1997;57:963–9. and differentiation during fibroblast growth factor-stimulated angio- 35. Yamanaka Y, Friess H, Buchler M, Beger HG, Uchida E, Onda M, genesis in vitro: role of extracellular matrix. J Cell Biol 1989;109: et al. Overexpression of acidic and basic fibroblast growth factors in 317–30. human pancreatic cancer correlates with advanced tumor stage. 55. Werner S, Grose R. Regulation of wound healing by growth factors Cancer Res 1993;53:5289–96. and cytokines. Physiol Rev 2003;83:835–70. 36. Berger W, Setinek U, Mohr T, Kindas-Mugge€ I, Vetterlein M, Dekan 56. Abraham JA, Whang JL, Tumolo A, Mergia A, Friedman J, Gospo- G, et al. Evidence for a role of FGF-2 and FGF receptors in the darowicz D, et al. Human basic fibroblast growth factor: nucleotide proliferation of non-small cell lung cancer cells. Int J Cancer sequence and genomic organization. EMBO J 1986;5:2523–8. 1999;83:415–23. 57. Greenhalgh DG, Sprugel KH, Murray MJ, Ross R. PDGF and FGF 37. Gazzaniga P, Gandini O, Gradilone A, Silvestri I, Giuliani L, Magnanti stimulate wound healing in the genetically diabetic mouse. Am J M, et al. Detection of basic fibroblast growth factor mRNA in urinary Pathol 1990;136:1235–46. bladder cancer: correlation with local relapses. Int J Oncol 1999;14: 58. Broadley KN, Aquino AM, Woodward SC, Buckley-Sturrock A, Sato 1123–7. Y, Rifkin DB, et al. Monospecific antibodies implicate basic fibroblast 38. Dellacono FR, Spiro J, Eisma R, Kreutzer D. Expression of basic growth factor in normal wound repair. Lab Invest 1989;61:571–5. fibroblast growth factor and its receptors by head and neck squa- 59. Risau W. Angiogenic growth factors. Prog Growth Factor Res mous carcinoma tumor and vascular endothelial cells. Am J Surg 1990;2:71–9. 1997;174:540–4. 60. Pepper MS, Ferrara N, Orci L, Montesano R. Potent synergism 39. Huang X, Yu C, Jin C, Yang C, Xie R, Cao D, et al. Forced expression between vascular endothelial growth factor and basic fibroblast of hepatocyte-specific fibroblast growth factor 21 delays initiation of growth factor in the induction of angiogenesis in vitro. Biochem chemically induced hepatocarcinogenesis. Mol Carcinog 2006; Biophys Res Commun 1992;189:824–31. 45:934–42. 61. Compagni A, Wilgenbus P, Impagnatiello M-A, Cotten M, Christofori 40. Galdemard C, Brison O, Lavialle C. The proto-oncogene FGF-3 is G. Fibroblast growth factors are required for efficient tumor angio- constitutively expressed in tumorigenic, but not in non-tumorigenic, genesis. Cancer Res 2000;60:7163–9. clones of a human colon carcinoma cell line. Oncogene 1995;10: 62. Javerzat S, Auguste P, Bikfalvi A. The role of fibroblast growth factors 2331–42. in vascular development. Trends Mol Med 2002;8:483–9. 41. Turner N, Grose R. Fibroblast growth factor signalling: from devel- 63. Gualandris A, Urbinati C, Rusnati M, Ziche M, Presta M. Interaction of opment to cancer. Nat Rev Cancer 2010;10:116–29. high-molecular-weight basic fibroblast growth factor with endothe- 42. Yan G, Fukabori Y, McBride G, Nikolaropolous S, McKeehan WL. lium: biological activity and intracellular fate of human recombinant Exon switching and activation of stromal and embryonic fibroblast M(r) 24,000 bFGF. J Cell Physiol 1994;161:149–59.

OF8 Clin Cancer Res; 17(19) October 1, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Inhibition of the FGF Pathway and Antiangiogenic Therapy

64. Ribatti D, Urbinati C, Nico B, Rusnati M, Roncali L, Presta M. 83. Allen E, Walters I, Hanahan D. Brivanib, a dual FGF/VEGF Inhibitor, is Endogenous basic fibroblast growth factor is implicated in the active both first and second line against mouse pancreatic neuro- vascularization of the chick embryo chorioallantoic membrane. endocrine tumors developing adaptive/evasive resistance to VEGF Dev Biol 1995;170:39–49. inhibition. Clin Cancer Res 2011;17:5299–5310. 65. Passaniti A, Taylor RM, Pili R, Guo Y, Long PV, Haney JA, et al. A 84. Fernando NT, Koch M, Rothrock C, Gollogly LK, D’Amore PA, simple, quantitative method for assessing angiogenesis and anti- Ryeom S, et al. Tumor escape from endogenous, extracellular angiogenic agents using reconstituted basement membrane, hep- matrix-associated angiogenesis inhibitors by up-regulation of mul- arin, and fibroblast growth factor. Lab Invest 1992;67:519–28. tiple proangiogenic factors. Clin Cancer Res 2008;14:1529–39. 66. Ensoli B, Gendelman R, Markham P, Fiorelli V, Colombini S, Raffeld 85. Bello E, Colella G, Scarlato V, Oliva P, Berndt A, Valbusa G, et al. E- M, et al. Synergy between basic fibroblast growth factor and HIV-1 3810 is a potent dual inhibitor of VEGFR and FGFR that exerts Tat protein in induction of Kaposi's sarcoma. Nature 1994;371: antitumor activity in multiple preclinical models. Cancer Res 674–80. 2011;71:1396–405. 67. Wang Y, Becker D. Antisense targeting of basic fibroblast growth 86. Kopetz S, Hoff M, Eng C, Overman M, Glover K, Chang Z, et al. factor and fibroblast growth factor receptor-1 in human melanomas Levels of angiogenic cytokines prior to disease progression in blocks intratumoral angiogenesis and tumor growth. Nat Med metastatic colorectal cancer patients treated with bevacizumab. 1997;3:887–93. Gastrointestinal Cancers Symposium; 2009 Jan 15–17; San Fran- 68. Fujii T, Kuwano H. Regulation of the expression balance of angio- cisco, CA; 2009. poietin-1 and angiopoietin-2 by Shh and FGF-2. In Vitro Cell Dev Biol 87. Kopetz S, Hoff PM, Morris JS, Wolff RA, Eng C, Glover KY, et al. Anim 2010;46:487–91. Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab 69. Ensoli B, Nakamura S, Salahuddin SZ, Biberfeld P, Larsson L, Beaver for metastatic colorectal cancer: efficacy and circulating angiogenic B, et al. AIDS-Kaposi's sarcoma-derived cells express cytokines biomarkers associated with therapeutic resistance. J Clin Oncol with autocrine and paracrine growth effects. Science 1989;243: 2009;24:8252. 223–6. 88. Willett CG, Duda DG, di Tomaso E, Boucher Y, Ancukiewicz M, 70. Abuharbeid S, Czubayko F, Aigner A. The fibroblast growth factor- Sahani DV, et al. Efficacy, safety, and biomarkers of neoadjuvant binding protein FGF-BP. Int J Biochem Cell Biol 2006;38:1463–8. bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a 71. Tassi E, Al-Attar A, Aigner A, Swift MR, McDonnell K, Karavanov A, multidisciplinary phase II study. J Clin Oncol 2009;27:3020–6. et al. Enhancement of fibroblast growth factor (FGF) activity by an 89. Batchelor TT, Sorensen AG, di Tomaso E, Zhang W-T, Duda DG, FGF-binding protein. J Biol Chem 2001;276:40247–53. Cohen KS, et al. AZD2171, a pan-VEGF 72. Aigner A, Butscheid M, Kunkel P, Krause E, Lamszus K, Wellstein inhibitor, normalizes tumor vasculature and alleviates edema in A, et al. An FGF-binding protein (FGF-BP) exerts its biological glioblastoma patients. Cancer Cell 2007;11:83–95. function by parallel paracrine stimulation of tumor cell and endo- 90. Garrett CR, Siu LL, Giaccone G, El-Khoueiry A, Marshall J, LoRusso thelial cell proliferation through FGF-2 release. Int J Cancer 2001; P, et al. A phase I study of BMS-582664 (), an oral 92:510–7. dual inhibitor of VEGFR and FGFR tyrosine kinases, in combination 73. Czubayko F, Liaudet-Coopman EDE, Aigner A, Tuveson AT, with full-dose in patients with advanced gastrointestinal Berchem GJ, Wellstein A. A secreted FGF-binding protein can serve malignancies who failed prior therapy. J Clin Oncol 2007;25 as the angiogenic switch in human cancer. Nat Med 1997;3: (18S):14018. 1137–40. 91. Matsui J, Yamamoto Y, Funahashi Y, Tsuruoka A, Watanabe T, 74. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, Wakabayashi T, et al. E7080, a novel inhibitor that targets multiple et al. Antiangiogenic therapy elicits malignant progression of tumors kinases, has potent antitumor activities against to increased local invasion and distant metastasis. Cancer Cell producing human small cell lung cancer H146, based on angiogen- 2009;15:220–31. esis inhibition. Int J Cancer 2008;122:664–71. 75. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, 92. Zhang H, Masuoka L, Baker K, Sadra A, Bosch E, Brennan T, et al. Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leu- FP-1039 (FGFR1:Fc), a soluble FGFR1 receptor antagonist, inhibits covorin for metastatic colorectal cancer. N Engl J Med 2004; tumor growth and angiogenesis. AACR-NCI-EORTC International 350:2335–42. Conference; 2007 Oct 22–26; San Francisco, CA; 2007. 76. Tille JC, Wood J, Mandriota SJ, Schnell C, Ferrari S, Mestan J, et al. 93. Jonker DJ, Rosen LS, Sawyer MB, de Braud F, Wilding G, Sweeney Vascular endothelial growth factor (VEGF) receptor-2 antagonists CJ, et al. A phase I study to determine the safety, pharmacokinetics inhibit VEGF- and basic fibroblast growth factor-induced angiogen- and pharmacodynamics of a dual VEGFR and FGFR inhibitor, bri- esis in vivo and in vitro. J Pharmacol Exp Ther 2001;299:1073–85. vanib, in patients with advanced or metastatic solid tumors. Ann 77. Giavazzi R, Sennino B, Coltrini D, Garofalo A, Dossi R, Ronca R, et al. Oncol 2011;22:1413–9. Distinct role of fibroblast growth factor-2 and vascular endothelial 94. Okamoto I, Kaneda H, Satoh T, Okamoto W, Miyazaki M, Morinaga growth factor on tumor growth and angiogenesis. Am J Pathol R, et al. Phase I safety, pharmacokinetic, and biomarker study of bibf 2003;162:1913–26. 1120, an oral triple tyrosine kinase inhibitor in patients with advanced 78. Shi YH, Bingle L, Gong LH, Wang YX, Corke KP, Fang WG. Basic solid tumors. Mol Cancer Ther 2010;9:2825–33. FGF augments hypoxia induced HIF-1-alpha expression and VEGF 95. Sun HD, Malabunga M, Tonra JR, DiRenzo R, Carrick FE, Zheng release in T47D breast cancer cells. Pathology 2007;39:396–400. H, et al. Monoclonal antibody antagonists of hypothalamic FGFR1 79. Nissen LJ, Cao R, Hedlund E-M, Wang Z, Zhao X, Wetterskog D, cause potent but reversible hypophagia and weight loss in et al. Angiogenic factors FGF2 and PDGF-BB synergistically pro- rodents and monkeys. Am J Physiol Endocrinol Metab 2007; mote murine tumor and metastasis. J Clin Invest 292:E964–76. 2007;117:2766–77. 96. Sarker D, Molife R, Evans TR, Hardie M, Marriott C, Butzberger- 80. Shen J, Vil MD, Prewett M, Damoci C, Zhang H, Li H, et al. Devel- Zimmerli P, et al. A phase I pharmacokinetic and pharmacodynamic opment of a fully human anti-PDGFRbeta antibody that suppresses study of TKI258, an oral, multitargeted receptor tyrosine kinase growth of human tumor xenografts and enhances antitumor activity inhibitor in patients with advanced solid tumors. Clin Cancer Res of an anti-VEGFR2 antibody. Neoplasia 2009;11:594–604. 2008;14:2075–81. 81. Pietras K, Pahler J, Bergers G, Hanahan D. Functions of paracrine 97. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita PDGF signaling in the proangiogenic tumor stroma revealed by T, et al. Targeted ablation of Fgf23 demonstrates an essential pharmacological targeting. PLoS Med 2008;5:e19. physiological role of FGF23 in phosphate and vitamin D metabolism. 82. Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by J Clin Invest 2004;113:561–8. evasion of antiangiogenic targeting of VEGF signaling in late-stage 98. Greulich H, Pollock PM. Targeting mutant fibroblast growth factor pancreatic islet tumors. Cancer Cell 2005;8:299–309. receptors in cancer. Trends Mol Med 2011;17:283–92.

www.aacrjournals.org Clin Cancer Res; 17(19) October 1, 2011 OF9

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Lieu et al.

99. Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, 106. Yamada K, Yamamoto N, Yamada Y, Nokihara H, Fujiwara Y, Hirata et al. Bevacizumab in combination with oxaliplatin-based T, et al. Phase I dose-escalation study and biomarker analysis of chemotherapy as first-line therapy in metastatic colorectal cancer: E7080 in patients with advanced solid tumors. Clin Cancer Res a randomized phase III study. J Clin Oncol 2008;26:2013–9. 2011;17:2528–37. 100. Jain RK, Duda DG, Willett CG, Sahani DV, Zhu AX, Loeffler JS, et al. 107. Ueda Y, Shimoyama T, Murakami H, Yamamoto N, Yamada Y, Biomarkers of response and resistance to antiangiogenic therapy. Arioka H, et al. Phase I and pharmacokinetic study of TSU-68, a Nat Rev Clin Oncol 2009;6:327–38. novel multiple receptor tyrosine kinase inhibitor, by twice daily oral 101. Murukesh N, Dive C, Jayson GC. Biomarkers of angiogenesis and administration between meals in patients with advanced solid their role in the development of VEGF inhibitors. Br J Cancer tumors. Cancer Chemother Pharmacol 2011;67:1101–9. 2010;102:8–18. 108. Gavine P, Mooney L, Kilgour E, Thomas A, Al-Kadhimi K, Beck S, 102. Chun YS, Vauthey JN, Boonsirikamchai P, Maru DM, Kopetz S, et al. Characterization of AZD4547: An orally bioavailable, potent and Palavecino M, et al. Association of computed tomography morpho- selective inhibitor of FGFR tyrosine kinases 1, 2, and 3. 102nd AACR logic criteria with pathologic response and survival in patients Annual Meeting; 2011 Apr 2–6; Orlando, FL; 2011. treated with bevacizumab for colorectal metastases. JAMA 109. Harding T, Palencia S, Long L, Finer J, Keer H, Baker K, et al. 2009;302:2338–44. Preclinical efficacy of FP-1039 (FGFR1:Fc) in endometrial carcinoma 103. Ocaña A, Amir E, Vera F, Eisenhauer EA, Tannock IF. Addition of models with activating mutations in FGFR2. American AACR Annual bevacizumab to chemotherapy for treatment of solid tumors: Meeting; 2010 Apr 17–21; Washington, DC; 2010. similar results but different conclusions. J Clin Oncol 2011;29: 110. ClinicalTrials.gov. Bethesda (MD): NIH; 2011. Novartis. A dose 254–6. escalation study in adult patients with advanced solid malignancies. 104. Santos ES, Gomez JE, Raez LE. Targeting angiogenesis from mul- NCT01004224. Available from: http://clinicaltrials.gov/ct2/show/ tiple pathways simultaneously: BIBF 1120, an investigational novel NCT01004224. triple angiokinase inhibitor. Invest New Drugs 2011. [Epub ahead of 111. Saxty G, Akkari R, Angibaud P, Arts J, Benderitter P, Berdini V, et al. print] 25 Feb 2011. Fragment based drug discovery of selective inhibitors of fibroblast 105. Dempke WCM, Zippel R. Brivanib, a novel dual VEGF-R2/bFGF-R growth factor receptor (FGFR). 102nd AACR Annual Meeting; 2011 inhibitor. Anticancer Res 2010;30:4477–83. Apr 2–6; Orlando, FL; 2011.

OF10 Clin Cancer Res; 17(19) October 1, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst September 27, 2011; DOI: 10.1158/1078-0432.CCR-11-0659

Beyond VEGF: Inhibition of the Fibroblast Growth Factor Pathway and Antiangiogenesis

Christopher Lieu, John Heymach, Michael Overman, et al.

Clin Cancer Res Published OnlineFirst September 27, 2011.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-11-0659

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2011/09/23/1078-0432.CCR-11-0659. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 25, 2021. © 2011 American Association for Cancer Research.