Review
EphB4: A promising target for upper aerodigestive malignancies

https://doi.org/10.1016/j.bbcan.2018.01.003Get rights and content

Abstract

The erythropoietin-producing hepatocellular carcinoma (Eph) receptors are the largest family of receptor tyrosine kinases (RTKs) that include two major subclasses, EphA and EphB. They form an important cell communication system with critical and diverse roles in a variety of biological processes during embryonic development. However, dysregulation of the Eph/ephrin interactions is implicated in cancer contributing to tumour growth, metastasis, and angiogenesis. Here, we focus on EphB4 and review recent developments in elucidating its role in upper aerodigestive malignancies to include lung cancer, head and neck cancer, and mesothelioma. In particular, we summarize information regarding EphB4 structure/function and role in disease pathobiology. We also review the data supporting EphB4 as a potential pharmacological and immunotherapy target and finally, progress in the development of new therapeutic strategies including small molecule inhibitors of its activity is discussed. The emerging picture suggests that EphB4 is a valuable and attractive therapeutic target for upper aerodigestive malignancies.

Introduction

The erythropoietin-producing hepatoma (Eph) receptors represent the largest class of receptor tyrosine kinases (RTKs). They are type I transmembrane proteins that interact with their membrane-bound ligands the ephrins and facilitate cell-to-cell contacts resulting in bidirectional intracellular signaling [1,2]. Ephrin stimulation causes receptor dimerization and autophosphorylation of the Eph receptor on the juxtamembrane regions and cytoplasmic tails driving the subsequent recruitment of down-stream signaling molecules [3]. These include SH2 and SH3 adapter proteins, Src family kinases, PI3K, MAP kinases, small GTPases, guanine nucleotide exchange factors, and phosphatases, each of which contribute to the complex cell repulsion and adhesion pathways that modulate cell shape, motility and attachment [4].

Based on sequence homologies and their binding affinities and characteristics, the Eph receptors are grouped into subclasses namely, Eph A and Eph B receptors, although there is significant redundancy and cross talk between subclasses [[5], [6], [7], [8]]. Furthermore, while the members of the ephrin-A subclass (ephrin-A1 to -A5) are tethered to the membrane via a glycosylphosphatidylinositol (GPI) anchor, the members of the ephrin-B subclass (ephrin-B1 to -B3) are transmembrane proteins and have a highly conserved cytoplasmic domain [9].

The EphB subfamily, the largest of receptor tyrosine kinases, comprises of six members (EphB1 to 6). Five of them are catalytically competent, while the more divergent EphB6 is catalytically dead [10]. Unlike many of the other members in this family that exhibit promiscuity [4], the EphB4 receptor has distinctive specificity for a single ligand, ephrinB2; however, EphB4 also binds to ephrinB1 and ephrinB3 albeit weakly [11]. Physiologically, EphB4 plays important roles in vascular remodeling of primitive capillary networks into distinct arteries and veins [12,13]. On the other hand, EphB4 is also known to regulate vascularization in malignant tumours [14], and is up-regulated in numerous cancer types including upper aerodigestive cancers (reviewed, [8,15,16]). Nonetheless, since Eph-ephrin interactions have been shown to either promote or inhibit tumour growth [8,17], the role of these receptors in cancer remains a matter for debate. However, crosstalk between elevated Eph receptors and other oncogenes, such as the ErbB family of receptor tyrosine kinases is thought to result in enhanced cell proliferation and tumorigenesis, presumably independent of ephrin stimulation (reviewed, [18]) (Fig. 1). Here, we review the function of EphB4, its potential as a therapeutic target, the developments to date on small molecule inhibitors, and EphB4 immunotherapy in upper aerodigestive malignancies.

EphB4, like all other Eph receptors, is a type I transmembrane protein that has a prototypical RTK topology. Thus, the protein is characterized by an N-terminal multidomain extracellular region, a single transmembrane segment, and a cytoplasmic region that contains the kinase domain in the C-terminus [19]. The extracellular domain includes a globular ligand-binding domain, a cysteine-rich region, composed of an epidermal growth factor (EGF)-like motif, and two fibronectin type III repeats [11]. The ephrin-binding domain has a high- affinity binding site that mediates the biochemical communication between EphB4 and the ephrinB2 in adjacent cells. The cytoplasmic region contains a tyrosine kinase domain and protein-protein interacting modules, including a sterile-α-motif (SAM) and a PDZ-binding motif [1]. The SAM domain is a protein-interacting domain that promotes homo-dimerization and the oligomerization of receptors. The PDZ-binding motif mediates the organization of protein complexes at the plasma membrane [20] (Fig. 2).

The crystal structures of the EphB4 receptor in complex with the extracellular domain of ephrinB2 complex [21] and the EphB4 receptor in complex with antagonistic peptide TNYL-RAW (TNYLFSPNGPIARAW) that binds with high-affinity (15 nM) [22] have been solved. The EphB4-ephrinB2 structural analysis suggested that L95 plays a particularly important role in defining ligand selectivity of EphB4. Indeed, all other members of this receptor family that promiscuously bind many ephrins have a conserved R at the position corresponding to L95 of EphB4. Furthermore, amino acid changes in the EphB4 ligand binding cavity when designed based on comparison with the crystal structure of the more promiscuous EphB2 receptor, yielded EphB4 variants with altered binding affinity for ephrinB2 and TNYL-RAW. Finally, binding studies employing isothermal titration calorimetry with the EphB4 L95R mutant confirmed the importance of this amino acid in conferring high affinity binding to both ephrinB2 and TNYL-RAW [22].

More recently, Dai et al. [23] by simulating conformational ensembles and recognition energy landscapes starting from separated Eph and ephrin molecules and proceeding up to the formation of receptor-ligand complexes, demonstrated that two pairs of dynamic salt bridges are unique to EphB4, resulting in ephrinB2 recognition and conformational selection. Together these structural biology and computational studies provide tremendous insight on the specificities for the interaction between EphB4 and ephrinB2 and how this knowledge could be applied to drug discovery in designing novel compounds that recapitulate the critical contacts of the peptide and EphB4 with good pharmacokinetic properties.

Since the receptor and the ligand typically reside on different cells, a rather unique feature of the Eph/ephrin axis is that the complexes emanate signals bidirectionally. Thus, forward signals that depend on Eph kinase activity propagate in the receptor-expressing cell, and reverse signals that depend on Src family kinases propagate in the ligand (ephrin)-expressing cell (Fig. 3). Eph signaling controls cell morphology, adhesion, migration and invasion by modifying the organization of the actin cytoskeleton and influencing the activities of integrins and intercellular adhesion molecules [1,4]. Additionally, recent work has also uncovered Eph effects on cell proliferation and survival as well as specialized cellular functions such as synaptic plasticity, insulin secretion, bone remodeling and immune function [1].

With regard to forward signaling by EphB4 in cancer cells, while the receptor is upregulated in many cancers, its response to ephrin is muted and, in some cases, ephrin-dependent Eph forward signaling may even be detrimental to tumour progression. For example, EphB4 receptors that are activated by ephrins acquire the remarkable ability to inhibit oncogenic signaling pathways, including Abl-Crk, PI3K-Akt and HRAS-Erk [24]. In contrast, some cancer cells are stimulated by activation [25] and in others, overexpression of the active EphB4 enhanced anchorage-independent growth, migration and invasion, all characteristics associated with an aggressive phenotype [26] suggesting that the overexpression of EphB4 facilitates tumour progression by forward signaling. However, overexpression of EphB4 or that of an inactive mutant EphB4 kinase in esophageal squamous cell carcinoma cells promoted cell growth and migration, suggesting EphB4 promoted cell growth and migration independently of its kinase activity and that repressed tumourigenesis [27].

Insofar as reverse signaling is concerned, since B ephrins do not possess intrinsic catalytic activity, they rely on the recruitment of signaling molecules such as Src family kinases, which phosphorylate specific tyrosine residues in the intra-cytoplasmic domain of B ephrins, resulting in receptor engagement and clustering [28]. Furthermore, similar to the situation with forward signaling, reverse signaling has also been observed to result in both tumour suppression as well as progression. For example, tumour cells expressing dominant negative EphB4 are incapable of forward signaling, but remain able to stimulate ephrinB2 reverse signaling, that in turn promotes cell invasion and proliferation. The overexpression of EphrinB2 in cultured cancer cells enhances integrin mediated attachment, migration and invasion [29,30]. Considered together, these observations on bidirectional signaling suggest that the role of EphB4/ephrinB2 is context-dependent and can vary from one cancer type to the other. Furthermore, mutations dysregulating Eph function likely also play a role in cancer progression.

Many receptor tyrosine kinases (RTKs), including several members of the Eph family, are frequently altered in lung cancer where they are believed to play important roles [31]. For example, a mutation in EphA2 (G391R) causes constitutive kinase activation in lung squamous cell carcinoma (SCC) where it promotes increased cell survival, cell invasion, focal adhesions, and activation of mammalian target of rapamycin [32]. Three potentially relevant EphB6 mutations were identified in NSCLC patients (3.8%) that included two point mutations and an in-frame deletion mutation (del915–917). Although catalytically inactive, the point mutations in EphB6 lead to instable proteins while the in-frame deletion enhanced migration and accelerated wound healing in vitro. Furthermore, the del915–917 mutation also increased the metastatic capability of NSCLC cells in an in vivo mouse model suggesting that EphB6 mutations promote metastasis in a subset of patients with non-small cell lung cancer (NSCLC) cells [33]. Similarly, it has been reported that EphA3 and EphA5 are frequently altered in NSCLC. While mutations in EphA3 appear to have pro-tumourigenic effects thereby transforming tumour suppressor function of the WT protein in the lung [34], the functional significance of these alterations in EphA5 is unknown [35].

Comparatively, there is a lot more information on EphB4, particularly in lung cancer. For example, it has been reported that the gene is overexpressed and amplified in several lung cancer subtypes and is necessary for the growth of lung adenocarcinoma xenografts in mice [36]. Several non-synonymous mutations in EphB4 have also been identified, in human tumour tissues and cell lines. Thus, a mutation resulting in an R564K substitution occurring in the intracellular juxtamembrane (JM) domain was detected in one multiple myeloma cell line [37], and an R889W substitution was detected in one gastric carcinoma tissue sample [38]. A detailed genetic analysis of small cell lung cancer (SCLC) also identified several mutations in the ephrin receptor family including EphB4 [86].

Another comprehensive study by Ferguson et al. [39] revealed EphB4 can be mutated in lung cancer and that they lead to putative structural alterations as well as increased cellular proliferation and motility. Notably, the authors detected eight NS EphB4 mutations with one (A230V) in an extracellular linker region, two (A371V and P381S) in the first extracellular fibronectin III repeat, two (W534* and E536K) in the extracellular juxtamembrane domain, two (G723S and A742V) in the tyrosine kinase domain, and one (P881S) in an intracellular linker region just C-terminal to the tyrosine kinase domain. Three of these (A230V, A371V, and P381S) occurred in adenocarcinoma, one (A742V) occurred in SCC, and four (W534*, E536K, G723S, and P881S) occurred in SCLC. Seven of these eight mutations (all except A371V) had not been previously detected (Fig. 4). Overall, the non-synonymous mutations occurred in 7% of samples, with non-synonymous mutation rates of 9% in adenocarcinoma, 9% in SCLC, and 3% in SCC. These observations together with several others [[40], [41], [42]] provide a panoramic view of the EphB4 mutational landscape in lung cancer. Interestingly, of the 16 sites with EphB4 mutations in adenocarcinoma tissues identified by Ferguson et al. [39], 12.5% were located in the kinase domain underscoring the functional implications. Furthermore, a bioinformatics analysis of these mutations revealed that a) they are mutually exclusive from other common RTK variants in lung cancer, b) they correspond to analogous sites of other RTK's variations in cancers, c) they are predicted to be oncogenic based on biochemical, evolutionary, and domain-function constraints, and d) EPHB4 mutations can induce broad changes in the kinome signature of lung cancer cells [39]. Taken together, these data illuminate the role of EphB4 in lung cancer and further identify EphB4 as a potentially important therapeutic target. However, thus far, no non-synonymous EphB4 mutations have been reported in HNSCC or pleural mesothelioma tissues.

Section snippets

Lung cancer

Ferguson et al. [36] demonstrated that EphB4 is overexpressed 3-fold in lung tumours compared to paired normal tissues and frequently exhibits gene copy number increases in lung cancer. The authors also showed that overexpression of EphB4 promotes cellular proliferation, colony formation, and motility, while EphB4 inhibition reduces cellular viability in vitro, halts the growth of established tumours in mouse xenograft models when used as a single target strategy, and causes near complete

Kinase inhibitors

Numerous small molecule compounds and their derivatives that bind the ATPase domain and other molecules including proteins and antibodies that target receptor interaction other than the kinase domain have been reported. For excellent reviews that provide a comprehensive report of the various compounds, from discovery to validation, mode of action, and the indications that they are being evaluated both at the preclinical and clinical testing stages, the reader is referred to Boyd et al. [52],

Biomarker potential

Comparatively speaking, very little is known about the potential of EphB4 as a biomarker, especially in upper aerodigestive malignancies. A recent study by Yu et al. [75] who evaluated cell-free RNA content in the peripheral blood as a potential biomarker for detecting CTCs in NSCLC patients, found that the expression of EphB4 in PBMCs correlated with histopathological type. A survival analysis showed that the patients with enhanced expression of EphB4 along 3 other mRNAs in PBMCs had poorer

Concluding remarks

Notwithstanding the fact that both forward and reverse signaling by the EphB4/ephrinB2 interaction is context-dependent and can vary from one cancer type to another, the data reviewed here underscore the oncogenic, rather than tumour suppressor, role for EphB4 in upper aerodigestive malignancies including lung cancer, HNSCC and MM. Consistent with its oncogenic potential in these cancers, several small molecule inhibitors that inhibit the kinase activity, immunotherapy strategies, and other

Transparency document

Transparency document.

Acknowledgements

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under award number P30CA033572 (RS). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

References (86)

  • M. Imielinski et al.

    Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing

    Cell

    (2012)
  • A. Unzue et al.

    Three stories on Eph kinase inhibitors: from in silico discovery to in vivo validation

    Eur. J. Med. Chem.

    (2016)
  • M.C. Pietanza et al.

    Phase II study of the multitargeted tyrosine kinase inhibitor XL647 in patients with non-small-cell lung cancer

    J. Thorac. Oncol.

    (2012)
  • M. Koolpe et al.

    EphB receptor-binding peptides identified by phage display enable design of an antagonist with ephrin-like affinity

    J. Biol. Chem.

    (2005)
  • S. Duggineni et al.

    Design, synthesis and characterization of novel small molecular inhibitors of ephrin-B2 binding to EphB4

    Biochem. Pharmacol.

    (2013)
  • V. Krasnoperov et al.

    Novel EphB4 monoclonal antibodies modulate angiogenesis and inhibit tumour growth

    Am. J. Pathol.

    (2010)
  • N. Kertesz et al.

    The soluble extracellular domain of EphB4 (sEphB4) antagonizes EphB4 EphrinB2 interaction, modulates angiogenesis, and inhibits tumour growth

    Blood

    (2006)
  • S. Shi et al.

    Biophysical characterization and stabilization of the recombinant albumin fusion protein sEphB4-HAS

    J. Pharm. Sci.

    (2012)
  • J.S. Scehnet et al.

    The role of Ephs, Ephrins, and growth factors in Kaposi sarcoma and implications of EphrinB2 blockade

    Blood

    (2009)
  • T.J. Kao et al.

    Ephrin-mediated cis-attenuation of Eph receptor signaling is essential for spinal motor axon guidance

    Neuron

    (2011)
  • M. Zeltsman et al.

    CAR T-cell therapy for lung cancer and malignant pleural mesothelioma

    Transl. Res.

    (2017)
  • C. Bardelle et al.

    Inhibitors of the tyrosine kinase EphB4. Part 1:S-based design and optimization of a series of 2,4-bis-anilinopyrimidines

    Bioorg. Med. Chem. Lett.

    (2008)
  • C. Bardelle et al.

    Inhibitors of the tyrosine kinase EphB4. Part 2: structure-based discovery and optimisation of 3, 5-bis substituted anilinopyrimidines

    Bioorg. Med. Chem. Lett.

    (2008)
  • Y. Miyazaki et al.

    Design and effective synthesis of novel templates, 3, 7-diphenyl-4-amino-thieno and furo-[3,2-c]pyridines as protein kinase inhibitors and in vitro evaluation targeting angiogenetic kinases

    Bioorg. Med. Chem. Lett.

    (2007)
  • B. Barlaam et al.

    Inhibitors of the tyrosine kinase EphB4. Part 4: discovery and optimization of a benzylic alcoholseries

    Bioorg. Med. Chem. Lett.

    (2011)
  • S.R. Kumar et al.

    Receptor tyrosine kinase EphB4 is a survival factor in breast cancer

    Am. J. Pathol.

    (2006)
  • A. Das et al.

    Ephrin B2/EphB4 pathway in hepatic stellate cells stimulates Erk-dependent VEGF production and sinusoidal endothelial cell recruitment

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2010)
  • K. Kullander et al.

    Mechanisms and functions of Eph and ephrin signalling

    Nat. Rev. Mol. Cell Biol.

    (2002)
  • E.B. Pasquale

    Eph receptor signalling casts a wide net on cell behaviour

    Nat. Rev. Mol. Cell Biol.

    (2005)
  • M. Takemoto et al.

    Ephrin-B3-EphA4 interactions regulate the growth of specific thalamocortical axon populations in vitro

    Eur. J. Neurosci.

    (2002)
  • J.P. Himanen et al.

    Repelling class discrimination: ephrin-A5 binds to and activates EphB2 receptor signaling

    Nat. Neurosci.

    (2004)
  • E.B. Pasquale

    Eph receptors and ephrins in cancer: bidirectional signalling and beyond

    Nat. Rev. Cancer

    (2010)
  • K. Hamada et al.

    Distinct roles of ephrin-B2 forward and EphB4 reverse signaling in endothelial cells

    Arterioscler. Thromb. Vasc. Biol.

    (2003)
  • J.P. Himanen et al.

    Crystal structure of an Eph receptor-ephrin complex

    Nature

    (2001)
  • U.H. Wang et al.

    Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4

    Cell

    (1998)
  • A. Kania et al.

    Mechanisms of ephrin-Eph signalling in development, physiology and disease

    Nat. Rev. Mol. Cell Biol.

    (2016)
  • A. Barquilla et al.

    Eph receptors and ephrins: therapeutic opportunities

    Annu. Rev. Pharmacol. Toxicol.

    (2015)
  • Y. Chen et al.

    Targeting receptor tyrosine kinase EphB4 in cancer therapy

    Semin. Cancer Biol.

    (2017)
  • N.K. Noren et al.

    Paradoxes of the EphB4 receptor in cancer

    Cancer Res.

    (2007)
  • D. Vaught et al.

    Eph receptors in breast cancer: roles in tumor promotion and tumor suppression

    Breast Cancer Res.

    (2008)
  • E.M. Lisabeth et al.

    Eph receptor signaling and ephrins

    Cold Spring Harb. Perspect. Biol.

    (2013)
  • A.H. Zisch et al.

    Replacing two conserved tyrosines of the EphB2 receptor with glutamic acid prevents binding of SH2 domains without abrogating kinase activity and biological responses

    Oncogene

    (2000)
  • X. Huang et al.

    Induction of cell retraction by the combined actions of Abl-CrkII and Rho-ROCK1 signaling

    J. Cell Biol.

    (2008)
  • Cited by (20)

    • Homoharringtonine suppresses LoVo cell growth by inhibiting EphB4 and the PI3K/AKT and MAPK/EKR1/2 signaling pathways

      2020, Food and Chemical Toxicology
      Citation Excerpt :

      MTT and apoptosis assays using EphB4-overexpression LoVo cells further indicated that EphB4 suppression by HHT could inhibit proliferation and induce LoVo cell apoptosis. EphB4 receptor stimulation by Ephrins drives the subsequent recruitment of downstream signaling molecules, including the PI3K/AKT/mTOR and MAPK/ERK1/2 signaling (Chen et al., 2017; Salgia et al., 2018). Phosphatidylinositol-3-kinases (PI3Ks) catalyze phosphoinositide phosphorylation at the 30-hydroxyl group, and produce a vital second messenger phosphatidylinositol-3, 4, 5-trisphosphate (PIP3).

    • Discovery of novel anti-angiogenesis agents. Part 9: Multiplex inhibitors suppressing compensatory activations of RTKs

      2019, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Angiopoietin (Ang) and its receptor (Tie-2) could inhibit endothelial cell apoptosis, and promote vascular bud, branch, reshape and mature [3]. Erythropoietin producing hepatocyte receptor B4 (EphB4) and its membrane adherent ligand (EphrinB2) are essential for angiogenesis, vascular maturation, and pericytes recruitment [4]. All of them mentioned above belong to the receptor tyrosine kinase family (RTK), which plays an important role in signal transduction and is an indispensable regulator of angiogenesis [5].

    • Ephrin type-B receptor 4 affinity chromatography: An effective and rapid method studying the active compounds targeting Ephrin type-B receptor 4

      2019, Journal of Chromatography A
      Citation Excerpt :

      The erythropoietin-producing hepatoma (Eph) family is the largest class of receptor tyrosine kinases (RTKs). Eph receptors are type I transmembrane proteins, which transduce bidirectional intracellular signals via interacting with their membrane-bound ligands (ephrins) [1]. Ephrin stimulation induces the dimerization and autophosphorylation of the Eph receptor driving the activation of down-stream signaling molecules which include Src family kinases, PI3K, MAP kinases, and so on [2,3].

    • Therapeutic potential of targeting the Eph/ephrin signaling complex

      2018, International Journal of Biochemistry and Cell Biology
      Citation Excerpt :

      We will summarize here the latest studies on the field of Eph-related pharmacology, concentrating on Eph/ephrin/ADAM-based antibodies and small molecules, as well as Henipavirus targeting, and will discuss the future of genomics/proteomics-based medicine. Many different approaches have been employed in efforts to target the Eph/ephrin signaling system; antibodies, peptides and soluble fragments of the receptors/ligands, small molecule inhibitors of protein-protein interactions (PPIs), kinase inhibitors, and siRNAs (Lodola et al., 2017; Charmsaz et al., 2017; Chen et al., 2017; Barquilla and Pasquale, 2015; Charmsaz and Boyd, 2017; Salgia et al., 2018). Although few have entered clinical trials, the relative infancy of the field and the enormous potential of various ‘druggable’ domains, should guarantee the continuation of these efforts.

    • Biological Significance of EphB4 Expression in Cancer

      2024, Current Protein and Peptide Science
    View all citing articles on Scopus
    View full text