Review
Fibroblast Growth Factor Signaling in Non–Small-Cell Lung Cancer Thomas J. Semrad,1,2 Philip C. Mack1 Abstract Despite recent progress in the treatment on non–small cell lung cancer (NSCLC), outcomes remain suboptimal. Treatment advances that target the epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) signaling pathways highlight the need to understand the multiple convergent growth factor signaling pathways involved in the pathogenesis of NSCLC. Signaling through fibroblast growth factors (FGF), long recognized for its pro-angiogenic activity, has recently emerged as a contributing factor in the pathogenesis and progression of NSCLC through an autocrine signaling loop. In addition, this pathway may function as a mechanism of resistance to anti-EGFR and anti-VEGF treatment. Clinical experience with FGF receptor (FGFR) inhibitors is mounting, and more specific inhibitors of this signaling pathway are in development. This review describes the structure of the FGF signaling pathway, delineates its dual roles in angiogenesis and proliferation in NSCLC, evaluates FGF ligand and receptor expression as prognostic biomarkers in NSCLC, and discusses the development of FGF pathway inhibitors for the treatment of lung malignancies. Clinical Lung Cancer, Vol. 13, No. 2, 90-5 Published by Elsevier Inc. Keywords: Angiogenesis, Cellular proliferation, Fibroblast growth factors, Fibroblast growth factor receptor, Non–small-cell lung
Introduction In the United States, lung cancer is the leading cause of cancerrelated death for both men and women, and accounted for 157,300 deaths in 2010.1 The majority of lung cancer cases (approximately 85%) are classified as non–small cell lung cancer (NSCLC) for which survival has minimally improved in the last several decades.2 Recently, novel treatments that target the epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) have emerged from an improved understanding of growth factor signaling in NSCLC. Although EGFR inhibitors exhibit some benefit in unselected patients, activating mutations in the EGFR identify a subset of tumors that are exquisitely sensitive to EGFR tyrosine kinase inhibitors.3,4 Similarly, anti-VEGF treatment produces an improved outcome for certain patients with NSCLC,5 although defining pre-
1 Division of Hematology/Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, CA 2 Section of Hematology/Oncology, The Department of Veterans Affairs Northern California Health Care System, Sacramento, CA
Submitted: May 12, 2011; Revised: Jul 27, 2011; Accepted: Aug 01, 2011 Address for correspondence: Thomas J. Semrad, MD, Division of Hematology/ Oncology, Department of Internal Medicine, University of California, Davis, 4501 X Street, Suite 3016, Sacramento, CA 95817 Tel: 916-734-3771; fax: 916-734-7946; e-mail contact: thomas.semrad@ ucdmc.ucdavis.edu
90
Clinical Lung Cancer March 2012
dictive biomarkers for anti-VEGF therapeutic efficacy has been less successful.6 The experience with EGFR and VEGF inhibitors in NSCLC highlights the need to define other signaling pathways involved in tumor progression, angiogenesis, and resistance to currently available treatments. Fibroblast growth factors (FGF) comprise a complex family of signaling molecules that have been implicated in angiogenesis and inflammation in a wide variety of human disorders.7 Signaling of FGFs through the FGF receptors (FGFR) has been implicated as an autocrine signaling loop that leads to tumor proliferation and angiogenesis in a variety of NSCLC cell lines8 and is potentially a mechanism of resistance to both anti-VEGF and anti-EGFR therapies.9,10 In this review, we provide an overview of FGF signaling, consider its role in NSCLC, and describe potential therapeutic strategies for the ongoing development of FGF pathway inhibitors.
FGF Signaling The mammalian FGF family of growth factors consists of 18 distinct members in 6 subfamilies involved in multiple physiologic processes, including angiogenesis, organogenesis, tissue development, and endocrine signaling.7 The FGFR tyrosine kinases are coded by 4 genes (FGFR1, FGFR2, FGFR3, and FGFR4) but exist in numerous isoforms due to alternative messenger RNA splicing.7,11 The extracellular domain of FGFRs is composed of 2 or 3 immunoglobulin (Ig)-like binding loops.12 Importantly, tissue-specific alternative
1525-7304/$ - see frontmatter Published by Elsevier Inc. doi: 10.1016/j.cllc.2011.08.001
Figure 1 Overview of Fibroblast Growth Factor Signaling. Schematic Representation of Signal Transduction Pathways Activated by Signaling of FGFs Through FGFRs, HSPGs, and Integrins
FGF FGFRs
Integrins
HSPGs Transmembrane FRS2
PIP2 PLCγ1
GRB2 FAK
c-Src
SOS
PIP2 Raf
Ras
DAG+IP3 Ca2+
MAPKK PKC MAPK
Activation of Target Genes
Nucleus
Abbreviations: DAG ⫽ diacylglycerol; FAK ⫽ focal adhesion kinase; FGF ⫽ fibroblast growth factor; FGFR ⫽ fibroblast growth factor receptor; FRS2 ⫽ fibroblast growth factor receptor substrate 2; GRB2 ⫽ growth factor receptor bound protein 2; HSPG ⫽ heparan sulfate proteoglycans; IP3 ⫽ inositol 1,4,5-trisphosphate; MAPK ⫽ mitogen-activated protein kinase; MAPKK ⫽ mitogen-activated protein kinase kinase; PIP2 ⫽ phosphatidylinositol 4,5-bisphosphate; PKC ⫽ protein kinase C; PLC␥1 ⫽ phospholipase C-gamma 1; Raf ⫽ v-raf 1 murine leukemia viral oncogene homolog 1; Ras ⫽ retrovirus-associated DNA sequences; Src ⫽ v-src sarcoma viral oncogene homolog; SOS ⫽ son of sevenless.
splicing in FGFR1-3 of the invariant exon IIIa with exon IIIb tends to occur in epithelial cells, whereas splicing of exon IIIa with exon IIIc is preferentially seen in mesenchymal cells.13 Local paracrine loops can be generated by the epithelial-specific expression of ligands cognate for the mesenchymal isoform or, conversely, by expression of ligands for the epithelial isoform in adjacent mesenchymal cells. The binding of heparin sulfate glycosaminoglycan (HSGAG) to the FGF/ FGFR complex is required for dimerization, autophosphorylation, and activating of intracellular signaling.7 In addition, the binding of FGF to HSGAG traps FGF at the cell surface and protects the ligand from degradation (Figure 1).14 Signaling through FGFRs is mediated by direct recruitment of signaling intermediates to autophosphorylation sites on the activated receptor and by phosphorylation of fibroblast growth factor receptor substrate (FRS) docking proteins.15 Autophosphorylation of the receptor leads to binding and activation of phospholipase C-gamma, which results in the generation of the second messengers diacylglycerol and inositol triphosphate, that ultimately leads to activation of protein kinase Cs. In addition, activated FGFR phosphorylates the FRS2␣ and FRS2 docking proteins that recruit a multiprotein adaptor complex, including growth factor receptor bound protein 2 and son of sevenless. This complex ultimately activates both the
phosphatidylinositol-3-kinase and Ras/mitogen-activated protein kinase (MAPK) signaling cascades. Signaling through FGF/FGFR pathways is regulated by multiple feedback systems. Members of the Sprouty, Sprouty-related protein with EVH-1 domain, and similar expression to FGF families negatively regulate FGFR-induced MAPK signaling by binding to growth factor receptor bound protein 2, son of sevenless, and Raf1.16 MAPK phosphorylation of FRS2␣ reduces its binding to the FGFRs and thus reduces FGFR signaling.17 Moreover, interactions of FGF and FGFR with syndecans, integrins (especially ␣v3), Ncadherin, and neural cell adhesion molecule may modulate the intensity of signaling in different biologic contexts.18 Finally, there is emerging evidence that FGFs can induce signaling independent of the FGFR, although these signals largely seem to converge on the same intracellular pathways.18
Implications of FGF Signaling in NSCLC Signaling through FGFs has been implicated in cell proliferation, motility, and angiogenesis in a variety of human malignancies, including NSCLC.12,19 Mutations in FGFRs have only been identified in a small minority of NSCLCs.20,21 However, a case-control study of 274 Italian patients with surgically treated lung adenocarcinoma
Clinical Lung Cancer March 2012
91
FGF Signaling in NSCLC compared with 401 controls suggested that a Gly388Arg polymorphism in the transmembrane domain of FGFR4 may impart a higher risk of recurrence.22 This polymorphism is associated with altered tumor cell motility in a breast cancer cell line.23 Further investigation in an expanded cohort of Italian patients with NSCLC associated the Gly388Arg polymorphism with an increased risk of node positivity (hazard ratio, 1.8 [95% confidence interval, 1.3-2.6]) and poorer survival (hazard ratio, 1.5 [95% confidence interval, 1.1-1.9]); however, the same associations were not seen in a smaller cohort of Norwegian patients in the same analysis.24 In addition, no impact of the Gly388Arg polymorphism was observed in 619 patients with lung cancer from the United Kingdom , although only 164 of these patients had adenocarcinoma.25 In a retrospective study of 387 Japanese patients with surgically treated NSCLC, poorer survival with the FGFR4 Gly388Arg polymorphism was not evident in the entire cohort (P ⫽ .4889).26 However, in the subset of patients who were node positive (n ⫽ 118), the Gly388Arg polymorphism was associated with a significantly (P ⫽ .0397) inferior overall survival. Current experimental evidence suggests that FGF signaling may play a major role in neoangiogenesis.27 FGFs induce endothelial cell proliferation in vitro28 and facilitate the degradation and reorganization of the extracellular matrix through upregulation of the urokinase-type plasminogen activator and matrix metalloproteinase systems in endothelial cells.29,30 In addition, activation of FGFR2 stimulates chemotaxis, which leads to capillary-like structures and endothelial cell reorganization when cultured on 3-dimensional matrices.31–33 Furthermore, FGFR2 regulates the expression of certain cadherins and integrins that contribute to the organization and maturation of new blood vessels.34 –36 There is emerging evidence that a subset of NSCLC relies on the FGF pathway for cellular proliferation through autocrine or paracrine signaling loops. Results of several studies have demonstrated the coexpression of specific FGFs, particularly FGF2 and FGF9, along with FGFR1 and FGFR2 in human lung cancers.37 FGF2 and FGF9 specifically bind the FGFR-IIIc splice variants of FGFR1 and FGFR2, which are expressed in NSCLC cell lines.8,9,38 Furthermore, inhibition of FGFR signaling through antisense RNA, RNA interference, naturalizing FGF2 antibodies, or FGFR tyrosine kinase inhibitors leads to inhibition of cellular proliferation and tumor growth in vitro.8 In addition, NSCLC cell line data suggest that transcriptional derepression of FGFR2 and FGFR3 expression is induced by EGFR inhibitors and that these reactivated receptors provide proliferation signals through the extracellular signal-regulated kinase (ERK) pathway.39 Moreover, FGFR1 amplification has recently been observed in approximately 20% of squamous cell NSCLC. These FGFR1 amplified tumors are exquisitely sensitive to FGFR inhibition in vitro, which suggests that FGFR1 may be a critical target in this subset of NSCLC.40 Together, these studies demonstrate multiple potential mechanisms for the induction of autocrine and paracrine signaling loops through FGFs and FGFRs that are necessary for tumor proliferation and survival in a subset of NSCLC, and suggest a subset of tumors that may have heightened sensitivity to FGFR inhibitors. Finally, the FGFR signaling pathway also has been implicated in the epithelial to mesenchymal transition (EMT), which is necessary for invasion and metastases of tumor cells, and has been associated
92
Clinical Lung Cancer March 2012
with resistance to EGFR agents.41 FGF2 stimulates an EMT phenotype in vitro.42 In addition, the expression of both the plateletderived growth factor receptor (PDGFR) and FGFR1 are elevated in a NSCLC cell line (H358) induced into EMT by exogenous transforming growth factor .43 Furthermore, FGFR1 expression is higher in NSCLC cell lines with a mesenchymal phenotype compared with those with an epithelial phenotype.43 Inhibition of FGFR reduces proliferation in these cell lines, which are relatively resistant to EGFR inhibition. Thus, FGFR signaling may stimulate the development of EMT and maintain cellular proliferation in the mesenchymal state.
FGF2 and FGFR1 Expression As Prognostic Biomarkers in NSCLC There are conflicting data on the prognostic impact of FGF2 expression in NSCLC. In a recent study of 335 patients with resected NSCLC, high tumoral FGF2 expression levels by immunohistochemistry (IHC) were associated with a poorer 5-year survival (59% vs. 37%; P ⫽ .015); however, no such impact was seen for a high expression of FGFR1 (P ⫽ .15).44 Interestingly, high stromal expression of FGF2 was associated with an improved 5-year survival (70% vs. 53%; P ⫽ .024), an effect that remained significant in multivariate analyses.44 Separate analysis of FGF2 expression by IHC in 111 resected stage I to III NSCLCs associated higher expression with poor prognosis (P ⫽ .0173),45 and a similar analysis of FGF2 IHC expression in 143 lung adenocarcinoma corroborated these findings (P ⫽ .0089).46 In addition, a poor prognosis was associated with higher levels of FGF2 measured by enzyme-linked immunosorbent assay (ELISA) on frozen tumor specimens in 71 patients with surgically resected NSCLC (P ⫽ .0059).47 These results were not corroborated by 2 separate studies that evaluated tumoral IHC expression levels and that did not find a significant prognostic value of FGF2 in 132 patients with resected stage I NSCLC48 or 206 patients with stage I to III NSCLC.49 However, the latter study did find a significant association with high levels of FGFR1 expression and poor prognosis (P ⫽ .025). Similarly discrepant results have been observed in studies that evaluated the prognostic impact of serum FGF2 levels in patients with NSCLC.50 FGF2 levels measured by ELISA have been associated with a better prognosis in 2 studies,51,52 and a poor prognosis in at least 3 others.53–55
FGF Signaling Inhibitors A variety of FGFR tyrosine kinase inhibitors and FGFR-targeted monoclonal antibodies are in development. Because of the considerable similarity among the tyrosine kinase domains of FGFRs, PDGFRs, and VEGFRs, a number of small molecules that developed primarily as VEGFR antagonists have anti-FGFR activity as well.9 The considerable overlap in activity of these agents for VEGFR-2 and PDGFR clouds the ascertainment of activity that is dependent on FGFR inhibition. Indeed, it is quite likely that the toxicity induced by inhibition of alternative kinases such as VEGFR-2 precludes dosing of many of these agents to sufficient levels for therapeutic inhibition of FGFR. Published half maximal inhibitory concentration (IC50) values of selected multikinase inhibitors for FGFR1, VEGFR-2, and PDGFR are listed in Table 1. Cediranib (Recentin; AstraZeneca, London, U.K.) is an oral small molecule tyrosine inhibitor that is in advanced stages of clinical de-
Thomas J. Semrad, Philip C. Mack Table 1 Selected Tyrosine Kinase Inhibitors With FGFR1, VEGFR-2, and PDGFR Activity IC50 (nM) FGFR1
VEGFR-2
PDGFR
8
13
27
XL999
8.2
2.6
1.5
E-381066
17.5
25
525
PD17307467
22
100-200
18
Cediranib (AZD2171)68
26
⬍1
5
Dovitinib (TKI258)64 65
57
BIBF 1120
69
21
65
Pazopanib69
140
30
84
Brivanib (BMS-540215)70
148
25
⬎6,000
Abbreviations: FGFR ⫽ fibroblast growth factor receptor; IC50 ⫽ half maximal inhibitory concentration; PDGFR ⫽ platelet-derived growth factor receptor; VEGFR ⫽ vascular endothelial growth factor receptor.
velopment. Although it principally inhibits VEGFRs, PDGFRs, and stem cell factor receptor (c-kit), FGFR1 is also a putative target. Interim analysis of a phase II-III study of cediranib vs. placebo in combination with carboplatin and paclitaxel (n ⫽ 251) as initial therapy for NSCLC suggested improved responses in the cediranib arm (38% vs. 16%; P ⬍ .001), although this study was halted due to excess toxicities, including higher rates of hypertension, hypothyroidism, hand-foot syndrome, and gastrointestinal toxicity on the cediranib arm.56 A similar study has been initiated with a lower dose of cediranib (NCT00795340). BIBF 1120 (Boehringer Ingelheim, Ingelheim am Rhein, Germany) is an oral small molecule inhibitor of FGFRs, PDGFRs, and VEGFRs as well as fms-like tyrosine kinase 3 and v-src sarcoma viral oncogene homolog.57,58 In a randomized phase II trial conducted in advanced NSCLC (n ⫽ 73) after failure of platinum-based chemotherapy, 48% of patients treated with BIBF 1120 had stable disease as the best tumor response and the median progression-free survival was 6.9 weeks.59 Toxicities were manageable, with grade 3/4 enzyme elevations, diarrhea, nausea, vomiting, and abdominal pain being the most common. In addition, 2 phase III trials that compared BIBF 1120 to placebo in addition to either docetaxel (LUME-Lung 1; NCT00805194) or pemetrexed (LUME-Lung 2; NCT00806819) as second-line treatments are underway. Pazopanib (Votrient; GlaxoSmithKline, Middlesex, U.K.) is an oral small molecule inhibitor of the VEGFRs, PDGFRs, and c-kit that has activity against FGFRs. Pazopanib is approved by the U.S. Food and Drug Administration for the treatment of advanced renal cell carcinoma. In a multicenter, open-label, phase II “window of opportunity” trial of 2 to 6 weeks of treatment with pazopanib in clinical stage I-II NSCLC (n ⫽ 26), 86% of patients had some tumor reduction, with 3 response evaluation criteria in solid tumors partial responses.60 Pazopanib was well tolerated in this study, with the most common adverse events consisting of grade 2 hypertension, diarrhea, and fatigue. Phase II/III clinical trials of pazopanib in NSCLC are ongoing (NCT01208064, NCT00775307). Additional kinase inhibitors of FGFR that are in various phases of clinical development include brivanib (BMS-540215; Bristol-Myers
Squibb, New York, NY), XL999 (Symphony Evolution, Inc, Rockville, MD), dovitinib (TKI-258; Novartis, East Hanover, NJ), E-3810 (Ethical Oncology Science, Milano, Italy), AZD4547 (AstraZeneca), and BGJ398 (Novartis). Brivanib is being tested in NSCLC as part of a randomized discontinuation trial in multiple tumor types (NCT00633789). A phase I trial of XL999 in NSCLC was terminated due to safety concerns (NCT00491699). Dovitinib is being studied in metastatic breast, urothelial, and prostate cancers, although no trials in NSCLC are currently ongoing. Single-agent phase I trials of E-3810 (NCT01283945), AZD4547 (NCT00979134 and NCT01213160), and BGJ398 (NCT01004224) are in process. Interestingly, the BGJ398 trial is limited to those patients with amplification of FGFR1 or FGFR2 or mutation of FGFR3. The development of monoclonal antibodies that inhibit FGFR has lagged somewhat behind the development of tyrosine kinase inhibitors, although the potential advantage of this approach is the ability to selectively inhibit specific FGFR isoforms.9 Fully human antibodies for FGFR1-IIIb and FGFR1-IIIc have been developed.61 In addition, soluble FGFR1 ligand traps have been developed that exhibit potent antitumor activity in human NSCLC xenografts.62 These agents remain in early clinical development, although a phase I trial of a FGF ligand trap (FP-1039; Five Prime Therapeutics, Inc, South San Francisco, CA) has completed enrollment (NCT00687505).
Discussion The FGF signaling pathway is aberrantly activated in at least a subset of NSCLC, which leads to tumor proliferation and/or angiogenesis. Emerging data suggest that some NSCLCs may rely on FGF signaling through autocrine or paracrine loops for proliferation and survival.8,40 In addition, the FGF pathway may serve as an angiogenic growth factor pathway that allows tumor escape from VEGF inhibition.10 Furthermore, the FGF signaling pathway has been implicated as a mechanism of resistance to anti-EGFR treatment.9 To date, the predominant evidence for a proliferative dependency on FGFR signaling in NSCLC is derived from squamous and large cell lung cancer, subtypes that are frequently intrinsically resistant to EGFR inhibitors.8,39,40 Because the elucidation of driver mutations in NSCLC (eg, EGFR and EML4-ALK) has been largely confined to the adenocarcinoma subtype, the discovery of FGFR1 amplification may represent a major breakthrough in the search for a therapeutic target in squamous NSCLC. Inhibitors of the FGF signaling pathway should be further explored in tumors with evidence for FGFR signaling dependence as well as in those that are resistant to antiVEGF and anti-EGFR treatment. An improved understanding of the role of FGF signaling in angiogenesis, proliferation, and resistance is required to optimize investigations into FGF pathway inhibition in NSCLC. Subsets of NSCLC addicted to FGFR mutations in an analogous manner to those with activating EGFR mutations have not been identified. The relative binding affinities of multitargeted agents for FGFR vs. other targets, for example, VEGFR, must also be taken into consideration. Because currently available agents have lower specificity for FGFR, it is difficult to ascertain the relative importance of FGFR inhibition to their therapeutic effect or whether FGFR is even being inhibited at doses used. Data derived from agents with increased relative potency
Clinical Lung Cancer March 2012
93
FGF Signaling in NSCLC toward FGFR compared with other tyrosine kinases will be instructive. It will be important to identify the level of cross-talk and redundancy between these different signaling pathways to design combination strategies that tip the therapeutic balance toward cell death.63 Current preclinical evidence suggests that FGFR signaling may be an important proliferative pathway for squamous and large cell NSCLC, which are generally refractory to EGFR inhibition. In addition, cell line studies imply a potential for synergism of FGFR and EGFR inhibition in tumors that are susceptible to EGFR inhibition.38 These preclinical observations need to be prospectively validated in clinical trials.
Conclusions The FGF signaling pathway is an alternative growth factor signaling pathway associated with normal development and tumor angiogenesis. Preclinical evidence suggests that it may form an autocrine loop in a subset of NSCLC. Further evidence suggests that activation of the FGF signaling pathway may provide a mechanism of resistance to anti-VEGF and anti-EGFR therapy. FGFR inhibitors may have multiple potential therapeutic roles in NSCLC, including (1) a direct effect on tumor growth and survival through interference with FGFR autocrine signaling dependency, (2) an anti-angiogenic effect on tumors when using FGF as a pro-angiogenic stimulant, and (3) a means of overcoming FGF-mediated acquired resistance to angiogenic and tyrosine kinase inhibitors. A growing number of small molecule inhibitors of the FGFRs are in development, with varying degrees of selectivity and specificity. Further research is needed to clarify the contribution of FGF signaling to NSCLC pathogenesis and resistance to therapy to define a role of FGF pathway inhibitors in the clinical management of NSCLC.
Acknowledgments This work was supported by Boehringer Ingelheim Pharmaceuticals, Inc (BIPI). Editorial assistance was provided by Alyssa Tippens, PhD, of MedErgy, who was contracted by BIPI for these services. The authors met criteria for authorship as recommended by the International Committee of Medical Journal Editors and were fully responsible for all content and editorial decisions, and were involved at all stages of manuscript development. The authors received no compensation related to the development of the manuscript.
Disclosure All authors have no conflicts of interest.
References 1. Jemal A, Siegel R, Xu J, et al. Cancer statistics, 2010. CA Cancer J Clin 2010; 60:277-300. 2. American Cancer Society. Cancer Facts & Figures, 2010. Atlanta, GA: American Cancer Society; 2010. 3. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004; 350:2129-39. 4. Shepherd FA, Rodrigues PJ, Ciuleanu T, et al. Erlotinib in previously treated nonsmall-cell lung cancer. N Engl J Med 2005; 353:123-32. 5. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006; 355:2542-50. 6. Ramalingam SS, Belani CP. Antiangiogenic agents in the treatment of nonsmall cell lung cancer: reality and hope. Curr Opin Oncol 2010; 22:79-85. 7. Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov 2009; 8:235-53. 8. Marek L, Ware KE, Fritzsche A, et al. Fibroblast growth factor (FGF) and FGF receptor-mediated autocrine signaling in non-small-cell lung cancer cells. Mol Pharmacol 2009; 75:196-207.
94
Clinical Lung Cancer March 2012
9. Kono SA, Marshall ME, Ware KE, et al. The fibroblast growth factor receptor signaling pathway as a mediator of intrinsic resistance to EGFR-specific tyrosine kinase inhibitors in non-small cell lung cancer. Drug Resist Updat 2009; 12:95-102. 10. Ellis LM, Hicklin DJ. Pathways mediating resistance to vascular endothelial growth factor-targeted therapy. Clin Cancer Res 2008; 14:6371-5. 11. Johnson DE, Williams LT. Structural and functional diversity in the FGF receptor multigene family. Adv Cancer Res 1993; 60:1-41. 12. Grose R, Dickson C. Fibroblast growth factor signaling in tumorigenesis. Cytokine Growth Factor Rev 2005; 16:179-86. 13. Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol 2001; 2:REVIEWS3005. 14. Rusnati M, Presta M. Fibroblast growth factors/fibroblast growth factor receptors as targets for the development of anti-angiogenesis strategies. Curr Pharm Des 2007; 13:2025-44. 15. Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev 2005; 16:139-49. 16. Murphy T, Hori S, Sewell J, et al. Expression and functional role of negative signalling regulators in tumour development and progression. Int J Cancer 2010; 127:2491-9. 17. Lax I, Wong A, Lamothe B, et al. The docking protein FRS2alpha controls a MAP kinase-mediated negative feedback mechanism for signaling by FGF receptors. Mol Cell 2002; 10:709-19. 18. Murakami M, Elfenbein A, Simons M. Non-canonical fibroblast growth factor signalling in angiogenesis. Cardiovasc Res 2008; 78:223-31. 19. Korc M, Friesel RE. The role of fibroblast growth factors in tumor growth. Curr Cancer Drug Targets 2009; 9:639-51. 20. Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008; 455:1069-75. 21. Marks JL, McLellan MD, Zakowski MF, et al. Mutational analysis of EGFR and related signaling pathway genes in lung adenocarcinomas identifies a novel somatic kinase domain mutation in FGFR4. PLoS One 2007; 2:e426. 22. Spinola M, Leoni V, Pignatiello C, et al. Functional FGFR4 Gly388Arg polymorphism predicts prognosis in lung adenocarcinoma patients. J Clin Oncol 2005; 23:7307-11. 23. Bange J, Prechtl D, Cheburkin Y, et al. Cancer progression and tumor cell motility are associated with the FGFR4 Arg(388) allele. Cancer Res 2002; 62:840-7. 24. Falvella FS, Frullanti E, Galvan A, et al. FGFR4 Gly388Arg polymorphism may affect the clinical stage of patients with lung cancer by modulating the transcriptional profile of normal lung. Int J Cancer 2009; 124:2880-5. 25. Matakidou A, El Galta R, Rudd MF, et al. Further observations on the relationship between the FGFR4 Gly388Arg polymorphism and lung cancer prognosis. Br J Cancer 2007; 96:1904-7. 26. Sasaki H, Okuda K, Kawano O, et al. Fibroblast growth factor receptor 4 mutation and polymorphism in Japanese lung cancer. Oncol Rep 2008; 20:1125-30. 27. Presta M, Dell’Era P, Mitola S, et al. Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev 2005; 16: 159-78. 28. Cross MJ, Claesson-Welsh L. FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends Pharmacol Sci 2001; 22:201-7. 29. Rusnati M, Dell’Era P, Urbinati C, et al. A distinct basic fibroblast growth factor (FGF-2)/FGF receptor interaction distinguishes urokinase-type plasminogen activator induction from mitogenicity in endothelial cells. Mol Biol Cell 1996; 7:369-81. 30. Taraboletti G, D’Ascenzo S, Borsotti P, et al. Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells. Am J Pathol 2002; 160:673-80. 31. Kumar R, Yoneda J, Bucana CD, et al. Regulation of distinct steps of angiogenesis by different angiogenic molecules. Int J Oncol 1998; 12:749-57. 32. Montesano R, Vassalli JD, Baird A, et al. Basic fibroblast growth factor induces angiogenesis in vitro. Proc Natl Acad Sci U S A 1986; 83:7297-301. 33. Terranova VP, DiFlorio R, Lyall RM, et al. Human endothelial cells are chemotactic to endothelial cell growth factor and heparin. J Cell Biol 1985; 101:2330-4. 34. Gerritsen ME, Soriano R, Yang S, et al. Branching out: a molecular fingerprint of endothelial differentiation into tube-like structures generated by Affymetrix oligonucleotide arrays. Microcirculation 2003; 10:63-81. 35. Klein S, Giancotti FG, Presta M, et al. Basic fibroblast growth factor modulates integrin expression in microvascular endothelial cells. Mol Biol Cell 1993; 4:973-82. 36. Underwood PA, Bean PA, Gamble JR. Rate of endothelial expansion is controlled by cell:cell adhesion. Int J Biochem Cell Biol 2002; 34:55-69. 37. Behrens C, Lin HY, Lee JJ, et al. Immunohistochemical expression of basic fibroblast growth factor and fibroblast growth factor receptors 1 and 2 in the pathogenesis of lung cancer. Clin Cancer Res 2008; 14:6014-22. 38. Fischer H, Taylor N, Allerstorfer S, et al. Fibroblast growth factor receptor-mediated signals contribute to the malignant phenotype of non-small cell lung cancer cells: therapeutic implications and synergism with epidermal growth factor receptor inhibition. Mol Cancer Ther 2008; 7:3408-19. 39. Ware KE, Marshall ME, Heasley LR, et al. Rapidly acquired resistance to EGFR tyrosine kinase inhibitors in NSCLC cell lines through de-repression of FGFR2 and FGFR3 expression. PLoS One 2010; 5:e14117. 40. Weiss J, Sos ML, Seidel D, et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med 2010; 2:62ra93.
Thomas J. Semrad, Philip C. Mack 41. Yauch RL, Januario T, Eberhard DA, et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res 2005; 11:8686-98. 42. Strutz F, Zeisberg M, Ziyadeh FN, et al. Role of basic fibroblast growth factor-2 in epithelial-mesenchymal transformation. Kidney Int 2002; 61:1714-28. 43. Thomson S, Petti F, Sujka-Kwok I, et al. Kinase switching in mesenchymal-like non-small cell lung cancer lines contributes to EGFR inhibitor resistance through pathway redundancy. Clin Exp Metastasis 2008; 25:843-54. 44. Donnem T, Al-Shibli K, Al-Saad S, et al. Prognostic impact of fibroblast growth factor 2 in non-small cell lung cancer: coexpression with VEGFR-3 and PDGF-B predicts poor survival. J Thorac Oncol 2009; 4:578-85. 45. Shou Y, Hirano T, Gong Y, et al. Influence of angiogenetic factors and matrix metalloproteinases upon tumour progression in non-small-cell lung cancer. Br J Cancer 2001; 85:1706-12. 46. Takanami I, Tanaka F, Hashizume T, et al. The basic fibroblast growth factor and its receptor in pulmonary adenocarcinomas: an investigation of their expression as prognostic markers. Eur J Cancer 1996; 32A:1504-9. 47. Iwasaki A, Kuwahara M, Yoshinaga Y, et al. Basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) levels, as prognostic indicators in NSCLC. Eur J Cardiothorac Surg 2004; 25:443-8. 48. Kojima H, Shijubo N, Abe S. Thymidine phosphorylase and vascular endothelial growth factor in patients with stage I lung adenocarcinoma. Cancer 2002; 94:1083-93. 49. Volm M, Koomagi R, Mattern J, et al. Prognostic value of basic fibroblast growth factor and its receptor (FGFR-1) in patients with non-small cell lung carcinomas. Eur J Cancer 1997; 33:691-3. 50. Bremnes RM, Camps C, Sirera R. Angiogenesis in non-small cell lung cancer: the prognostic impact of neoangiogenesis and the cytokines VEGF and bFGF in tumours and blood. Lung Cancer 2006; 51:143-58. 51. Brattstrom D, Bergqvist M, Larsson A, et al. Basic fibroblast growth factor and vascular endothelial growth factor in sera from non-small cell lung cancer patients. Anticancer Res 1998; 18:1123-7. 52. Isa S, Kawaguchi T, Teramukai S, et al. Serum osteopontin levels are highly prognostic for survival in advanced non-small cell lung cancer: results from JMTO LC 0004. J Thorac Oncol 2009; 4:1104-10. 53. Brattstrom D, Bergqvist M, Hesselius P, et al. Elevated preoperative serum levels of angiogenic cytokines correlate to larger primary tumours and poorer survival in non-small cell lung cancer patients. Lung Cancer 2002; 37:57-63. 54. Brattstrom D, Bergqvist M, Hesselius P, et al. Serum VEGF and bFGF adds prognostic information in patients with normal platelet counts when sampled before, during and after treatment for locally advanced non-small cell lung cancer. Lung Cancer 2004; 43:55-62. 55. Joensuu H, Anttonen A, Eriksson M, et al. Soluble syndecan-1 and serum basic fibroblast growth factor are new prognostic factors in lung cancer. Cancer Res 2002; 62:5210-7. 56. Goss GD, Arnold A, Shepherd FA, et al. Randomized, double-blind trial of carboplatin and paclitaxel with either daily oral cediranib or placebo in advanced non-
57. 58.
59.
60. 61. 62.
63. 64. 65. 66. 67. 68. 69.
70.
small-cell lung cancer: NCIC Clinical Trials Group BR24 study. J Clin Oncol 2010; 28:49-55. Hilberg F, Roth GJ, Krssak M, et al. BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 2008; 68: 4774-82. Roth GJ, Heckel A, Colbatzky F, et al. Design, synthesis, and evaluation of indolinones as triple angiokinase inhibitors and the discovery of a highly specific 6-methoxycarbonyl-substituted indolinone (BIBF 1120). J Med Chem 2009; 52:4466-80. Reck M, Kaiser R, Eschbach C, et al. A phase II double-blind study to investigate efficacy and safety of two doses of the triple angiokinase inhibitor BIBF 1120 in patients with relapsed advanced non-small-cell lung cancer. Ann Oncol 2011; 22: 1374-81. Altorki N, Lane ME, Bauer T, et al. Phase II proof-of-concept study of pazopanib monotherapy in treatment-naive patients with stage I/II resectable non-small-cell lung cancer. J Clin Oncol 2010; 28:3131-7. Sun HD, Malabunga M, Tonra JR, et al. Monoclonal antibody antagonists of hypothalamic FGFR1 cause potent but reversible hypophagia and weight loss in rodents and monkeys. Am J Physiol Endocrinol Metab 2007; 292:E964-76. Ogawa T, Takayama K, Takakura N, et al. 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; 9:633-40. Xavier JB, Sander C. Principle of system balance for drug interactions. N Engl J Med 2010; 362:1339-40. Lee SH, Lopes de Menezes D, Vora J, et al. In vivo target modulation and biological activity of CHIR-258, a multitargeted growth factor receptor kinase inhibitor, in colon cancer models. Clin Cancer Res 2005; 11:3633-41. You WK, Sennino B, Williamson CW, et al. VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer. Cancer Res 2011; 71;4758-68. Bello E, Colella G, Scarlato V, et al. E-3810 is a potent dual inhibitor of VEGFR and FGFR that exerts antitumor activity in multiple preclinical models. Cancer Res 2011; 71:1396-405. Mohammadi M, Froum S, Hamby JM, et al. Crystal structure of an angiogenesis inhibitor bound to the FGF receptor tyrosine kinase domain. EMBO J 1998; 17: 5896-904. Wedge SR, Kendrew J, Hennequin LF, et al. AZD2171: a highly potent, orally bioavailable, vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res 2005; 65:4389-400. Kumar R, Knick VB, Rudolph SK, et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol Cancer Ther 2007; 6:2012-21. Bhide RS, Cai ZW, Zhang YZ, et al. Discovery and preclinical studies of (R)-1(4-(4-fluoro-2-methyl-1H-indol-5-yloxy)-5-methylpyrrolo[2,1-f][1,2,4]triazin-6yloxy)propan- 2-ol (BMS-540215), an in vivo active potent VEGFR-2 inhibitor. J Med Chem 2006; 49:2143-6.
Clinical Lung Cancer March 2012
95