You are on page 1of 10

Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

Targeting receptor tyrosine kinase EphB4 in cancer therapy


⁎ ⁎
Yinnan Chenb, Hongmei Zhangc, , Yanmin Zhanga,
a
School of Pharmacy, Health Science Center, Xi’an Jiaotong University, No. 76, Yanta West Street, #54, Xi’an, Shaanxi Province 710061, PR China
b
School of Molecular Sciences, Arizona State University, Tempe, AZ 85287, USA
c
Department of Endocrinology, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: Eph receptors and their Eph receptor-interacting (ephrin) ligands together form an important cell commu-
Receptor tyrosine kinases nication system with diverse roles. Experimental evidence demonstrated Eph receptor bidirectional signaling
EphB4 receptor with both tumor-suppressing and tumor-promoting activities in cancer cells. The tyrosine kinase EphB4, a
Ephrin signaling member of the Eph receptor family, has been associated with tumor angiogenesis, growth and metastasis, thus
Cancer therapy
making it a valuable and attractive target for drug design for therapeutic applications. In the past decade, many
Drug target
studies have focused on elucidating the structure and function of EphB4 in complex with its ligand ephrinB2 for
their role in carcinogenesis. Meanwhile, an array of compounds targeting EphB4 have been studied and several
selective inhibitors have been tested in clinical studies. This review discusses the structure and function of the
EphB4 receptor, analyzes its potential as a target for anticancer therapy, and summarizes the information about
inhibitors of EphB4 kinase activity. Conclusively, EphB4 is a challenging but promising therapeutic target in
cancer.

1. Introduction on their ligand-binding characteristics, Ephs have been subdivided into


Eph A and Eph B receptors, although there is significant redundancy
Kinases present one of the largest families of enzymes. Protein ki- and cross talk between subclasses [4].
nases regulate key biological processes including cell differentiation, In general, EphA receptors (EphA1-A10) are promiscuously acti-
proliferation, and motility [1]. Receptor tyrosine kinases (RTKs) are vated by ephrinA ligands (ephrinA1-ephrinA6), whereas EphB receptors
transmembrane proteins that undergo a conformational change upon (EphB1-EphB6) are activated by ephrinB ligands (ephrinB1-ephrinB3).
the binding of signaling molecules to the extracellular (N-terminal) Cross interactions have been observed between EphB2 and ephrinA5 as
domain. The conformational change triggers autophosphorylation of well as between EphA4 and ephrinB2/B3. Unlike other receptors in the
tyrosine residues in the intracellular domain of the RTK. Subsequently, family, EphB4 display a distinct specificity for its ligand ephrinB2,
the activated RTKs can phosphorylate a variety of cytoplasmic signaling while exhibiting a very weak binding affinity for either ephrinB1 or
proteins, which transduce a cellular response. Among other processed, ephrinB3 [5]. The EphB4-ephrinB2 interaction plays a prominent role
this signaling may induce the transcription of specific genes [1]. The in cardiovascular development and regulates vascularization in malig-
Eph receptors family in erythropoietin-producing hepatocellular carci- nant tumors [6].
noma belongs to the largest class of RTKs. Like other RTKs, they The Eph receptor/ephrin upregulation in cancer cells, the angio-
transduce signals from the cell exterior to the cell interior via the li- genic vasculature, and injured or diseased tissues offer opportunities for
gand-induced activation of their kinase domains. Eph receptor tyrosine Eph/ephrin-targeted drug delivery [7]. In fact, EphB4 overexpression
kinases constitute a large family of transmembrane proteins, containing has been linked to several tumor types such as breast, prostate, colon,
a single cytoplasmic kinase domain that is activated in response to the uterus, melanoma, and ovarian cancer among others [8–13]. Tyrosine
binding of ephrin ligands to the extracellular globular domain of the kinase EphB4 has been associated with angiogenesis, tumor growth and
receptor [2]. Eph receptors, however, have distinctive features: instead metastasis, making it a valuable and attractive target for drug design for
of binding soluble ligands, they generally mediate contact-dependent therapeutic applications. However, the role of EphB4 in cancer remains
cell-cell communication by interacting with surface-associated ligands a matter for debate, since Eph-ephrin interactions have been shown to
(ephrins on neighboring cells). Ephrins can also attenuate signaling either promote or inhibit tumor growth [14]. The primary focus of this
through Eph receptors that are co-expressed on the same cell. [3]. Based review is to analyze the structure and function of EphB4, briefly discuss


Corresponding authors.
E-mail addresses: zhanghongmei02@xinhuamed.com.cn (H. Zhang), zhang2008@mail.xjtu.edu.cn (Y. Zhang).

http://dx.doi.org/10.1016/j.semcancer.2017.10.002
Received 15 June 2017; Received in revised form 13 September 2017; Accepted 4 October 2017
1044-579X/ © 2017 Elsevier Ltd. All rights reserved.

Please cite this article as: Chen, Y., Seminars in Cancer Biology (2017), http://dx.doi.org/10.1016/j.semcancer.2017.10.002
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

its potential as a target for anticancer therapy and summarize updated 2.2. Structure of EphB4 binding domain
research about inhibitors of EphB4 kinase activity.
EphB and ephrinB are both transmembrane proteins. The B ephrins
are composed of an extracellular Eph receptor- binding domain, a
2. The structure of the EphB4 protein single-pass transmembrane region, and an intracellular domain con-
taing several phosphorylation sites and a PDZ binding motif at the C-
2.1. The molecular structure of the EphB receptor family terminus (Fig. 1). Chrencik et al. identified that the ligand- binding
domain of EphB4 forms a complex with the extracellular domain of
Himanen and colleagues. have reported the crystal structure of the ephrinB2 [20]. The overall structure of the EphB4-ephrinB2 complex is
EphB2 and ephrinB2 complex [15]. Thus. a tetramer is formed by two similar to that of the EphB2-ephrinB2 complex, and a second portion of
Eph-ephrin receptor/ligand dimers bound to each other. The extra- the high- affinity heterodimerization interface forms immediately ad-
cellular domains of EphB family members consistently include a glob- jacent to the ligand- binding cavity. The high-affinity EphB4- ephrinB2
ular N-terminal ligand-binding domain, a cysteine-rich region, com- heterodimer is formed through the insertion of the solvent-exposed li-
posed of an epidermal growth factor (EGF)-like motif, and two gand G-H loop into the upper convex, hydrophobic surface of the EphB4
fibronectin type III repeats [15]. The ephrin-binding domains of EphBs receptor. The G-H loop of ephrin-B2 is buttressed by the G-H and J-K
have a high- affinity binding site that mediates the biochemical com- loops of EphB4, and should form similar main chain hydrogen bonds
munication between Ephs and the ephrins in adjacent cells [16], as well and numerous van der Waals interactions with EphB4 (Fig. 2A). This
as low affinity ephrin- binding sites that mediate the clustering of Eph- high-affinity binding interface is highly hydrophobic, and includes re-
ephrin complexes [17]. The extracellular ligand-binding domain of the sidues Phe-120, Pro-122, Leu-124, Trp-125, and Leu-127 of ephrin-B2.
Eph receptor is connected to its cytoplasmic portion, which contains a Phe-120 forms hydrophobic interactions with Leu-95, Leu-100, and
tyrosine kinase domain and protein-protein interacting modules, in- Pro-101, whereas Leu-124 interacts with Thr-147, in the receptor J-K
cluding a sterile-α-motif (SAM) and a PSD95, DLG, ZO1-binding motif loop. Meanwhile, Trp-125 extends to the surface of the receptor, in-
(PDZ)-binding motif [18]. The SAM domain is a protein-interacting between the J-K and G-H loops, participating in hydrophobic interac-
domain that promotes homodimerization and the oligomerization of tions with residues Leu-48, Glu-50, Val-159, Leu-188, and Ala-186 of
receptors. The PDZ-binding motif mediates the organization of protein EphB4. The conserved Cys-61-Cys-184 disulfide bridge of EphB4 is
complexes at the plasma membrane [19] (Fig. 1). stabilized by Pro-122, in the conserved FSPN segment of the ephrin-B2

Fig. 1. Schematic domain structure of EphB4 and its


ligand, ephrinB2. Eph receptors have an extracellular
region including an ephrin-binding domain, a cy-
steine-rich region and two fibronectin type III re-
peats, with an intracellular region containing a tyr-
osine kinase domain, a sterile alpha motif domain
and a PDZ-binding motif. EphrinB ligands are
transmembrane proteins with a cytoplasmic tail and
a PDZ-binding domain. Bi-directional signaling re-
sults in forward signaling through Eph receptors and
reverse signaling through ephrin ligands.

2
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Fig. 2. The binding domain of the EphB4 receptor in complex with the ephrinB2 extracellular domain (yellow) (PDB ID: 2HLE and 2BBA) [20,21]. (A) The EphB4 receptor consists of a
jellyroll folding topology with 13 anti-parallel β-sheets (cyan) connected by loops of varying lengths, whereas the ephrin ligand (yellow) resembles the Greek key folding topology. The
interface is formed through the insertion of the ligand G-H loop into the hydrophobic binding cleft of EphB4. Key residues directly interacting with ephrinB2 are colored in purple. (B)
Surface representation of the three-dimensional structure of the EphB4 binding surface with EphrinB2. Key residues interacting with Ephrin B2 colored in purple.

Fig. 3. Crystal structure of the kinase domain of EphB4 (PDB ID: 3zew) [29]. Left: Backbone ribbon representation of the EphB4 kinase domain (alpha-helixes and beta sheets are shown
in red and cyan respectively). Right: ATP- binding site of EphB4, with highlighted residues used as targets for kinase inhibitors. The key side chains that can potentially interact with
kinase inhibitors are shown as colored sticks.

G-H loop [20] (Fig. 2B). preferentially interacts with only one ephrin ligand, ephrinB2. Several
The structure of the EphB4 receptor in complex with the high-affi- amino acid residues that make important contacts with the ephrin G-H
nity (40 nM) antagonistic TNYL-RAW (TNYLFSPNGPIARAW) peptide loop in the high-affinity dimerization interface of EphB2 are not con-
has been elucidated. The peptide is inserted into the same cleft occu- served in EphB4. For example, Ser-194 of EphB2 is conserved in other
pied by ephrinB2, along the hydrophobic upper convex portion of the EphB receptors, but not in EphB4, where an alanine is present at the
EphB4 receptor, which is situated on top of a β-sheet ‘floor’, formed by corresponding position. Moreover, EphB receptors have an aromatic
the D and E β strands. In addition, loops D-E, E-F, G-H, and J-K effec- residue at the position corresponding to Tyr-57 of EphB2. In EphB4, this
tively buttress the peptide in the cavity, forming numerous van der position is occupied by Leu 48, which cannot form a hydrogen bond
Waals interactions and main chain hydrogen bonds that stabilize with the main chain oxygen of Pro-150 of ephrinB2 or an aromatic-
binding. These studies reveal structural features for drug discovery of aromatic interaction with Phe-113 of ephrinB2, as observed with Tyr-
EphB4 antagonists [21]. 57 of EphB2. Rather, Leu-48 can only form weak hydrophobic inter-
EphB4 is the sole member of the Eph receptor family that actions with ephrinB2. A leucine that replaces the arginine at position

3
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

95 of the EphB4 receptor appears to be sufficient to cause a substantial 3.1. EphB4 as a tumor repressor
structural rearrangement of the receptor J-K loop, resulting in the ab-
sence of another salt bridge that is present in the dimerization interface 3.1.1. Forward signaling in cancer cells
of EphB2 with both ephrinB2 and ephrinA5 [15,20]. Further studies Eph receptor signaling induced by ephrin binding is initiated though
from Dai et al. demonstrated that two pairs of dynamic salt bridges are autophosphorylation and Src family kinase- mediated phosphorylation
unique to EphB4, resulting in ephrinB2 recognition and conformational of intracellular tyrosine residues, resulting in the activation of the tyr-
selection [22]. These specificities for the interaction between EphB4 osine kinase catalytic domain [32].
and ephrinB2 could be applied to target therapy with decreased side In many cancer cell lines, EphB4 receptors appear to be highly ex-
effects [23], which attribute to decrease the non-specific interaction of pressed but poorly activated by ephrins, as evaluated by the low level of
inhibitor and its potential targets. The ability to modulate the EphB4- tyrosine phosphorylation. For example, EphB4 tyrosine phosphoryla-
ephrinB2 binding will be critical to dissect the roles of these molecules tion is much lower in breast cancer cell lines, when compared with non-
in tumorigenesis and angiogenesis. The high-resolution structure of the transformed MCF10A epithelial cells [14,33,34]. This is one of the clues
ephrin binding domain of EphB4 in complex with a high-affinity se- suggesting that ephrin- dependent Eph forward signaling might be
lective antagonist will allow for the design of novel compounds that detrimental to tumor progression. Moreover, forcing EphB4 receptor
recapitulate the critical contacts of the peptide with EphB4, while activation with soluble Fc fusion proteins of ephrin ligands can inhibit
presenting good pharmacokinetic properties. proliferation, survival, and migration of many types of cancer cells in
culture as well as tumor growth in mouse models [34,35]. EphB4 re-
2.3. Design of EphB4 inhibitors for targeting the kinase domain ceptors that are activated by ephrins acquire the remarkable ability to
inhibit oncogenic signaling pathways, including Abl-Crk, PI3K-Akt and
Previously Tokarski et al. reported a co-structure dasatinib, which is HRAS-Erk [36]. Xiao et al. reported that the functional coupling of
a multitargeted tyrosine kinase inhibitor, in complex with kinase do- EphB4 to different downstream effectors determines the roles of EPhB4
main [24]. Tyrosine kinase domains are well conserved among Eph in inhibition or promotion of signaling. For instance, activation of
receptors. A series of studies have reported the design of EphB4 in- EphB4 resulted in inhibition of Ras/ERK in endothelial cells (HUVEC)
hibitors by modifying the leading compound based on the structure of because of the effectors like p120 RasGAP. However, MCF7 cells are
the kinase domain (Fig. 3), and those of the available co- structures stimulated by activation of EphB4 in due to PP2A is the effector [37]. In
[25–28]. Several groups of compounds have been designed to bind the esophageal squamous cell carcinoma (ESCC) compared with the paired
catalytic domain of EphB4. For instance, some inhibitors can form a key normal tissues, the expression of EphB4 was up regulated while
hydrogen bond or present a donor-acceptor pair to Met-696. Moreover, ephrinB2 was down regulated. Phosphorylation of EphB4 induced by its
some can be buried in the selectivity pocket, the hydrophobic region ligand, ephrinB2-Fc, inhibited growth, migration, and colony formation
beyond the Thr-693 gatekeeper residue, to disrupt ATP binding [28]. in ESCC cells. Moreover, overexpression of EphB4 or that of an inactive
Furthermore, the hinge and Glu-697, or the glycine-rich loop and Ile- mutant EphB4 kinase in ESCC cells promoted cell growth and migra-
621, can form sulfonamides that promote hydrogen bonding with some tion, suggesting EphB4 promoted cell growth and migration in-
inhibitors in the active site of the kinase [27]. Some compounds provide dependently of its kinase activity, and that repressed tumorigenesis
hydrogen donor and acceptor bonds to the carbonyl side chain of [38]. Consistent with this result, Rutkowski et al. found that EphB4
Glu664 and the backbone amide of Asp-758. This helps to explain why overexpressing cells demonstrated an enhanced anchorage-independent
these compounds retain affinity for the kinase despite the presence of a growth, migration and invasion, all characteristics associated with an
polar group in the relatively lipophilic environment of the back pocket aggressive phenotype. This supports the hypothesis that the over-
of the ATP-binding site [26]. Overman et al. presented a crystallization expression of EphB4 facilitates tumor progression. However, these ef-
analysis of human EphB family kinases, including the structure of fects were reversed in the presence of ephrinB2, which led to a reduc-
staurosporine-bound EphB4 [29]. Staurosporine-bound EphB4 adopts tion in EphB4 protein levels, demonstrating that ligand-dependent
the most closed conformation observed, with the Gly-rich loop folding signaling is tumor suppressive in prostate cancer [39].
tightly over the ligand. In contrast, the apoprotein structures of EphB1 EphB4 mutations may also contribute to the disruption of forward
and EphB2 are more open, with the Gly-rich loops partly disordered. signaling by impairing ephrin binding or kinase activity. The over-
The rational design and optimized synthetic gene constructs of the expression of EphB4 in breast cancer produced less malignancy in ad-
EphB4 intracellular kinase domain utilized by Overman et al. provided vanced stages [40]. Furthermore, EphB4 has been shown to reprogram
a useful platform for researchers to study or improve the derivatives of transcription, leading to anti-proliferative and anti-invasive outcomes.
EphB4 kinase inhibitors [30]. A dominant negative form of EphB4 has been shown to promote col-
orectal cancer proliferation and invasion [41]. Mutations that impair
3. The rgulatiory function of EphB4 in cancer Eph receptor signaling or promote the up regulation of tyrosine phos-
phatases that dephosphorylate Eph receptors may also promote tu-
The functions of the Eph/ephrin system in cancer are complex be- morigenesis [42]. Using dose and time dependent studies in breast
cause many tumor cells express varying degrees of Eph receptors and cancer cells, Barneh et al. showed that EphB4 can differentially inhibit
ligands. Eph signaling controls cell morphology, adhesion, migration, cell growth in a post-confluent state and that the presence of ligands
invasion, and the epithelial phenotype by modifying the organization of promotes the growth-inhibitory properties of the EphB4 receptor. In
the actin cytoskeleton and influencing the activities of integrins and this case, the inhibition of growth may have occurred due to the ex-
intercellular adhesion molecules [18,31]. Eph-ephrin complexes trans- tended treatment with ligand, a process that leads to the down reg-
duce emanate bidirectional signals: forward signals depend on Eph ki- ulation of the receptor [43].
nase activity for propagation in receptor-expressing cells, whereas re-
verse signals depend on Src family kinases for propagation in the 3.1.2. EphrinB2 reverse signaling in cancer cells
ephrin-expressing cells [18]. Eph receptors and ephrins are widely ex- Unlike Eph receptors, B ephrins do not possess intrinsic catalytic
pressed in cancer cells and tumor stroma, but may be down regulated in activity, and thus rely on the recruitment of signaling molecules. Ephrin
advanced stages of cancer [14]. Moreover, dysregulating mutations B signaling is initiated by the recruitment and activation of Src family
affecting Eph function also play a role in cancer pathogenesis [14]. kinases, which phosphorylate specific tyrosine residues in the in-
However, although bidirectional signaling promotes tumor angiogen- tracytoplasmic domain of B ephrins, resulting in receptor engagement
esis, the mechanism underlying its role in cancer progression is poorly and clustering [32]. Ephrin reverse signaling may also contribute to
understood. tumor suppression in some cancers. Eph receptors may further decrease

4
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

tumor invasiveness by promoting the formation of tight junctions in overexpressed and may play an unknown role in cancer progression
neighboring epithelial cells through the stimulation of ephrinB. Similar [63]. Rutkowski et al. showed the evidence that overexpression of
to its role in neurons, ephrinB reverse signaling may also inhibit the EphB4 leads to a transformed phenotype in MCF-10A cells and an in-
migratory and invasive effects of the CXCR4G protein-coupled chemo- creased metastatic phenotype in 22Rv1 cancer cells. It is restrained by
kine receptor in cancer cells [44,45]. Loss of ephrinB2 has also been ephrin-B2 stimulation due to reduce EphB4 levels [39]. Heroult, et al.
reported leading to carcinogenesis in a mouse mammary tumor model. reported expression of EphB4 by tumor cells confers a preferential site-
The Wnt/β- catenin/Tcf4 pathway in colorectal cancer cells are down specific metastasis phenotype to ephrinB2-expressing vascular beds
regulated with over-expression of ephrinB2 [46,47]. [64].

3.2. EphB4 as a tumor promotor 3.2.4. Regulation of angiogenesis by EphB4


Blood vessels are essential for tumor growth and represent an im-
3.2.1. Forward signaling in cancer cells portant venue for metastatic dissemination. Eph receptors and ephrins
It has been reported that EphB4 is over expressed in various types of promote angiogenesis by mediating communications among vascular
cancer, including colon, prostate, breast, esophageal, pancreatic, lung, cells and with tumor cells. Moreover, the EphB4 extracellular domain
mesothelioma and colorectal cancers [33,38,48–52]. The forward and can induce angiogenic responses by stimulating ephrinB2 reverse sig-
reverse Eph-ephrin signaling has been shown to enhance malignant naling in cultured endothelial cells [57]. During development, EphB4
transformation in some cases. Mounting evidence supports the notion receptor expression was detected in the endothelial cells of arteries and
that Eph receptors are also capable of unconventional signaling activ- veins. Suggestively, EphB4 regulates arterial-venous vessel segregation
ities independent of ephrin ligand activation. Meanwhile, it is well es- and remodeling in vasculature [65,66]. Furthermore, the reverse sig-
tablished that the Eph-ephrin system promotes cancer −associated naling medicated by ephrinB2 in endothelial tumor cells, pericytes, and
angiogenesis. Yang et al. reported ephrinB2 dependent EphB4 signaling smooth muscle cells is important for blood vessel assembly and en-
that enhances the migratory and invasive ability of melanoma cells, largement, and for decreasing blood vessel permeability, as well as for
which also requires the activation of RhoA GTPase [53]. EphB4 sti- promoting the interaction between endothelial cells and pericytes or
mulated by ephrinB2 can promote proliferation of breast cancer in vascular smooth muscle cells [57,67,68]. Additionally, ephrinB may
MCF7 by the activation of Ras/ERK signaling [37]. Moreover, Becerikli have other roles in the tumor-located endothelium, including enhan-
et al. found that EphB4 was overexpressed in soft tissue sarcomas. cing recruitment of bone marrow derived endothelial progenitor cells
Therefore, either decreasing the expression of EphB4 or inhibiting its that participate in tumor vascularization, which involves the EphB4
kinase activity can reduce the proliferation of sarcoma cells [54]. dependent up regulation [69]. Pfaff et al. showed that EphB-ephrinB
interactions during a cascade of events can lead to monocyte adhesion
3.2.2. Reverse signaling and transmigration through the vascular endothelium [70].
EphrinB reverse signaling involves Src family kinases, that phos-
phorylate the ephrinB cytoplasmic domain and regulate its interaction 4. EphB4 as a therapeutic target in cancer
with signaling molecules [45]. EphrinB2 is up regulated in the invading
cells of glioma and melanoma biopsy samples. The overexpression of Eph receptors and ephrins are promising new therapeutic targets in
EphrinB2 in cultured cancer cells enhances integrin mediated attach- cancer. Various strategies have been employed to evaluate the inter-
ment, migration and invasion [55,56]. Tumor cells expressing domi- ference of tumor-promoting effects or the enhancement of tumor sup-
nant negative EphB4 are incapable of forward signaling, but remain pressive effects. The inhibition of the Eph-ephrin system may be par-
able to stimulate ephrinB2 reverse signaling, that in turn promotes cell ticularly useful for anti-angiogenic therapies. The EphB4/ephrinB2
invasion and proliferation. The overexpression of EphB4 correlates with bidirectional signaling has an established role in the formation of the
tumors with larger blood vessels and a higher blood content [57]. Sa- vascular system, as evidenced by embryonic lethality in knockout
wamiphak, et al. have shown that ephrinB2 can up-regulate vascular mouse studies, which arise due to malformed vascular architecture
endothelial growth factor receptor type 2 (VEGFR2) signaling through [71].
PDZ interactions. Thus, ephrinB2 reverse signaling positively controls
sprouting and branching in pathological angiogenesis [58]. In a recent 4.1. Regulation of expression
study, Alam et al. demonstrated that ephrinB2 could induce ABCG2
expression in mutant p53 colorectal cancer cells, which promotes tu- The EphB4-ephrinB2 interaction plays a distinct role in diverse
morigenesis and is responsible for the acquired chemo-resistance in the biological processes such as neuronal development, bone homeostasis,
cells [59]. The growth promoting effects of ephrinB2 are also found in angiogenesis, migration and tumor invasiveness [57,72]. Several stu-
leukemia cell lines, indicating a role in the tumorigenesis [60]. dies point to significant levels of EphB4 expression in different malig-
nant tumors, whereas the inhibition of EphB4 expression inihibits
3.2.3. Unconventional EphB4 receptor activities tumor progression [9,33,54]. The critical importance of EphB4 in tumor
Besides the kinase activity of EphB4, many studies have shown progression is demonstrated by studies employing over expression and
ephrinB2 independent cancer promoting activity. Down regulation of knockdown strategies. The forced over-expression of EphB4 in non-tu-
EphB4 by small interfering RNAs or antisense oligonucleotides has been morigenic MCF10A breast cells and in 22Rv1 prostate cancer cells led to
shown to decrease cancer cell malignancy in culture and mouse cancer transformation in the MCF10A cell line and to an increase in the me-
models [33,61]. In these studies, the EphB4 tyrosine was poorly phos- tastatic phenotype of 22Rv1 cells [39]. Consequently, the knockdown
phorylated indicating this receptor is independent of the ligand medi- of EphB4 in several cancer cell lines consistently resulted in a 70–80%
ated kinase activation [8,44]. Noren et al. demonstrated that down reduction in cancer cell viability, an 8–16-fold increase in apoptosis,
regulation of EphB4 inhibits integrin-mediated cell substrate adhesion, and in an up to 80% reduction in cell migration and invasion [8,73].
spreading and migration, and reduces beta1-integrin protein levels in The downregulation of EphB4 expression using siRNAs or antisense
breast cancer cells, while low expression of the EphB4 ligand, ephrin- oligonucleotides has been shown to inhibit malignant cell behavior in
B2, does not show these above effects [62]. different types of culture and tumor growth in vivo [8,9,61,74]. Kras-
Knockdown of EphB4 in prostate cancer causes a significant re- noperov et al. reported that monoclonal antibodies inhibit tumor an-
duction in cell motility in vitro and tumor growth in vivo [9]. One of the giogenesis and growth by binding to fibronectin like domain 1. This
explanation from Mertens-Walker et al. is that lower expression of subsequently leads to the degration of EphB4 and inhibits the organi-
EphB4 can lead to down regulation of integrin beta 8, which is zation of human endothelial cell into tube-like structures in vitro [75].

5
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

However, the down regulation of EphB4 is not effective in preventing reported to inhibit VEGF driven angiogenesis in vivo [28,87].
ephrinB2 reverse signaling. Tumor cells expressing dominant negative Potent inhibitors of the kinase domain of EphB4 receptors have been
EphB4 were incapable of forward signaling, but could stimulate reported and the binding modes have been disclosed for these mole-
ephrinB2, attract endothelial cells, and stimulate cell invasion, survival cules [88]. Type I inhibitors are ATP competitors that recognize the
and proliferation. This characteristic correlated with tumors with larger activated state of the kinase [89–91]. Type II inhibitors interact with an
blood vessels and higher blood content [57]. Therefore, anti-ephrinB2 inactive conformation of the kinase and generally offer better se-
antibodies have been proposed to reduce tumor growth by inhibiting lectivity profiles. Like type I, type I1/2 inhibitors bind to the kinase in
the formation of tumor blood and lymphatic vessels independent of the active DFG-in (DFG “in” is based on the position of the conserved
EphB4 [76]. aspartate-phenylalanine-glycine triad at the beginning of the activation
loop) conformation, but extend their interactions to the hydrophobic
4.2. Disrupting Eph-ephrin interactions in anti-angiogenic therapy back pocket, establishing the characteristic hydrogen bonds present in
type II inhibitors [92]. Based on structural studies of EphB4 in complex
EphB4/ephrinB2 signaling is important for angiogenesis. Thus, po- with its inhibitors, it is feasible to optimize the leading compound as
tential medical applications targeting the angiogenesis-related tumor well as design better inhibitors. However, there are a few potential
progress, such as tumor growth and metastasis, could be devised by problems regarding the use of these Eph kinase inhibitors, including
hindering this protein-protein interaction. EphB4-ephrinB2 binding their anti-angiogenic activity that might also block possible Eph tumor
occurs at contact sites between cells. EphrinB2 induces EphB4 clus- suppressive activities, since these compounds target the ATP- binding
tering and increase kinase activity, thus generating downstream signals site and therefore their selectivity tends to be low. Additionally, EphB4
that affect cellular behavior. Monoclonal antibodies and peptides that kinase inhibitors will not be effective in preventing ephrinB2 reverse
block the EphB4-ephrinB2 interaction have been described, and some signaling.
have been utilized with high efficiency. Therefore, the disruption of Thus, as Xiao et al. reported, EphB4 can promote or suppress ele-
EphB4-ephrinB2 binding represents a potential avenue for anti- ments of the signaling pathway, such as Ras/MEK/ERK, due to its
angiogenic cancer therapies. Therefore, ephrinB2-Fc has also been used functional coupling to various downstream effectors in different cells
as an agonist that slows breast cancer xenografts by activating anti- [37]. It represents a potential challenge in targeting EphB4 for cancer
oncogenic signaling [34,57]. Various peptides and competitive small therapy, due to its conflicting effects on cancer cells and endothelial cell
compound inhibitors have been designed to mimic the binding of compartments.
the15-amino acid long G-H loop of ephrins, which is the main region
mediating high affinity binding of the ephrins to the C-terminal pocket 5. Tumor inhibitors targeting EphB4
of Eph receptors [21,77–79]. These EphB4 antagonistic compounds
may prove useful as a starting point for the generation more potent EphB4 inhibitors have been studied over the past several years, with
small molecule derivatives that inhibit the binding of ephrinB2 to this many different approaches used to test for inhibition of activity or
receptor. competitive binding affinity as a novel therapeutic modality to treat
The monomeric EphB4 ectodomain can inhibit tumorigenesis pre- malignancy. Several potent Eph kinase inhibitors were reported either
sumably by blocking the EphB4-ephrinB2 forward and reverse sig- based on high molecular weight compounds like peptides [23,93–95],
naling, resulting in the inhibition of angiogenesis. In fact, several which block the extracellular domain of the appropriate receptor, or on
groups have demonstrated that the soluble monomeric extracellular small organic molecules, which bind to the intramolecular ATP- binding
domain of EphB4 is able to block the interaction between ephrinB2and pocket and inhibit EphB4 kinase activity [95].
its receptor, leading to the inhibition of angiogenesis and tumor growth
[80,81]. Soluble fusion proteins, containing the extracellular domain of 5.1. Laboratorial or preclinical inhibitors of EphB4
EphB4 that antagonize EphB4-ephrinB2 interaction could inhibit tumor
growth and angiogenesis in tumor xenograft models, thus confirming 5.1.1. Synthetic EphB4 kinase inhibitors or high-affinity ligands of the
the importance of EphB4 in oncology. Furthermore, the EphB4-human EphB4 receptor
serum albumin fusion protein has shown therapeutic promise by in- Small molecule inhibitors are a rapidly emerging area for dis-
hibiting EphB4-ephrinB2 signaling in tumors [82]. The extracellular covering potential therapeutic candidates [96]. The targeted synthesis
cysteine- rich region and fibronectin-like domains of EphB4 can be used of specific EphB4 inhibitors has shown great promise. However, no
as epitope targets by antibodies, which may potentially affect the in- specific small-molecule EphB4 inhibitors exist, except for antibodies
teraction between EphB4 and ephrinB2 [75,83]. Thus, EphB4 agonists and peptides, and most EphB4 inhibitors show multi-target effects.
that also antagonize ephrin binding may prove particularly beneficial, A series of reports on EphB4 tyrosine kinase inhibitors emerged in
by enhancing EphB4-dependent tumor suppression in cancer cells and 2008, 2010, and 2011 [97–100]. Among these, Bardelle, et al. reported
by inhibiting ephrinB2-dependent angiogenesis [34,57]. a series of synthetic inhibitors based on 2,4-bis-anilinopyrimidines and
3,5-bis substituted anilinopyrimidines, in two papers in 2008 [97,98].
4.3. Kinase inhibitors for EphB4 Investigators such as Bardelle have used structural information from
two distinct species bound in the active site of EphB4 to generate a
The ephrin- binding pocket in the extracellular N-terminal domain novel series of 2,4-bis-anilinopyrimidine inhibitors. Structure–activity
of Eph receptors and the ATP- binding pocket in the intracellular kinase relationship (SAR) studies around the C-2 hinge-binding aniline in-
domain represent potential binding sites for peptides and small mole- dicated a strong preference for meta-substitution, particularly with re-
cules [84]. Efforts to identify small molecules that target the Eph kinase gard to electron-withdrawing groups. One example from this work,
domain have begun to yield some high-affinity inhibitors. Moreover, compound 28, has shown promise in its selectivity and pharmacokinetic
some inhibitors designed to target other kinases are effective against profile for kinase inhibition and served as the basis for development.
Eph receptors, due to the conserved structure of the kinase domain Further optimization, based on non-benzodioxole, led to compound 20,
[85,86]. Therefore, multi-targeted tyrosine kinase inhibitors could also a potent inhibitor of EphB4 and Src kinases, with good pharmacoki-
inhibit the kinase activity of EphB4; some of these, such as EXEL-7647 netics in various preclinical species and high fraction unbound in
and dasatinib, are already being tested in clinical trials. Moreover, some plasma [99]. The best IC50 obtained for EphB4 was 1 nM. Mamat et al.
new inhibitors have been sourced from kinase inhibitor libraries pos- presented the development of indazolylpyrimidine derivatives for
11
sessing specificity for EphB4, for which the efficacy has already been C-/18F-radiolabeling as high-affinity EphB4 receptor ligands [95].
determined. The selective NVP-BHG712 EphB4 inhibitor has been Recently, a series of commercially available xanthine derivatives were

6
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

identified as micromolar inhibitors of EphB4 by high-throughput frag- The various synthesized tumor inhibitors targeting EphB4 are listed
ment-based docking into the ATP-binding site of the kinase domain. in Table 1.
Karine et al. exploited the binding mode obtained by automatic docking
for the optimization of these EphB4 inhibitors by chemical synthesis. 5.1.2. Natural compounds derived from plants
Compound 66 employed threonine as the gatekeeper residue (Abl, Lck, Natural products, with their extensive structural diversity, are a
and Src) of all the inhibitors shows very high affinity for a few other major source for the currently available anticancer drugs. Several plants
tyrosine kinases. On the other hand, it is selective against kinases with a derived natural products have been described as potential inhibitors.
larger gatekeeper [86]. However, we did not find any recent publications investigating EphB4
NVP-BHG712, a potent and selective inhibitor of the EphB4 receptor inhibitors from plants. Therefore, many potential screening studies
tyrosine kinase, has an ED50 of 25 nM for the inhibition of EphB4 au- could be performed on plant products targeting EphB4. For instance,
tophosphorylation in cell-based assays, and is thereby estimated to be the Astragalus polysaccharide (APS), obtained from Astragalus mem-
roughly 200-fold more potent against EphB4 than against VEGFR2. branaceus, displays a range of activities in many systems. Yang et al.
However, it cannot discriminate among different Eph receptor kinases investigated the effects of APS on erythroid differentiation and its
[87]. Moreover, NVP-BHG712 also displays a synergistic effect with mechanism of action in K562 cells using microarrays. Interesting, the
others chemotherapy drugs for solid tumors. Kathawala, et al. in- mechanism underlying the action of APS involved the down-regulation
vestigated the combination of NVP-BHG712 and paclitaxel, and showed of EphB4 mRNA [115].
that the administration of NVP-BHG712 significantly increased the le-
vels of paclitaxel in tumors but not in plasma compared to paclitaxel 5.1.3. Peptides
alone. The combination of NVP-BHG712 and paclitaxel could serve as a Many peptides that selectively bind to EphB4 have been identified
novel and useful therapeutic strategy to attenuate paclitaxel resistance by phage display [78]. The inhibition of EphB4 signaling by using so-
mediated by the expression of the ABCC10 transporter [101]. luble extracellular domains of EphB4 has been shown to inhibit tumor

Table 1
Summary of laboratorial or preclinical studies of synthetic tumor inhibitors targeting EphB4.

Compounds Mechanism of action Pharmacodynamic effects on cancer Ref.

Inhibitors of kinase activity


Pyrazolo[3,4-d]pyrimidine (NVP- High selectivity for targeting the EphB4 kinase, inhibits Inhibits RTK autophosphorylation in stable transfected A375 [86,87]
BHG712) EphB4 and other kinases including c-Raf, c-Src and c-Abl melanoma cells and A375 xenografts
Golvatinib Inhibits kinase activities of c-Met, Tie2, and EphB4 Strongly inhibits tumor growth and tumor angiogenesis in [102,103]
xenograft models, and potently represses the growth of both c-
Met amplified tumor cells and that of endothelial cells
stimulated with either HGF or VEGF
Dasatinib A multi-targeted kinase inhibitor including EphB4 Used in numerous cancers treatments, including advanced [104,105]
prostate cancer
Nilotinib A selective Bcr-Abl kinase inhibitor; also inhibits EphB4 Treatment of imatinib-resistant chronic myelogenous [106,107]
leukemia
(18)F-labeled benzodioxolylpyrimidines Inhibits both the EphB4 receptor tyrosine kinase and the Inhibits the growth of human melanoma cells [108]
Src kinase, also inhibits the EphB4
Imidazopyridines Inhibits multiple receptor tyrosine kinases involved in More potent for the inhibition of endothelial tube formation [109]
angiogenic and tumorigenic signaling in an in vitro angiogenesis co-culture model; inhibits the
VEGF-driven autophosphorylation of lung KDR in mice and
rats
Imidazo[1,2-a]pyrazine diaryl ureas Multikinase inhibitor targets EphB4, EphA2, VEGF —— [84,110]
receptor 2 and the Tie2 receptor
Quinazoline (EXEL-7647) A potent inhibitor of EGFR, ErbB2, KDR, and EphB4 Inhibits cellular proliferation and EGFR pathway activation in [91]
the erlotinib-resistant H1975 cell line in vitro and in vivo,
substantially inhibits the growth of H1975 and MDA-MB-
231xenograft tumors and reduces tumor vessel density
Pyrido[2–3]pyrimidine (PD173955) A potent inhibitor of several ephrin receptor tyrosine Antiproliferative effects on cell viability in KU812 leukemia [111]
kinases cells
Anilinopyrimidines derivatives EphB4 kinase inhibitors Inhibition of cancer cell viability [97–100]
Xanthine derivatives Inhibits EphB4 by docking into the ATP-binding site of Inhibits growth of the central nervous system (SNB-75), [1,86]
the kinase domain leukemia (K-562) and breast (HS578T) cancer cell lines;
inhibits tube formation of HUVECs
Quinoxalines derivatives Inhibitor of the EphB4 tyrosine kinase High antiproliferative activities against leukemia (K-562), [1]
lung (HOP-92), colon (HT-29), renal (A498) and breast cancer
cells (MDA-MB-231, HS 578T) in the low nM range; inhibits
the VEGF-A induced sproutin in HUVECs g; inhibits tumor
growth in MDA-MB-231 nude mice xenografts
Trifluoromethylphenyl derivatives Inhibitors of kinase activity —— [85]
Thieno[3,2-c]pyridine derivatives ATP competitive kinase inhibitors against angiogenic —— [89]
targets, VEGFR2, Tie-2, and EphB4
Phenylthiopene derivatives Inhibitor of the EphB4 tyrosine kinase —— [84,112]

Inhibitors of the Eph receptor-ephrin interaction


Indazolylpyrimidine derivatives for High-affinity EphB4 receptor ligands Inhibit cancer cell viability [95]
11
C-/18F-radiolabeling
Disalicylic acid-furanylderivative Binds to the ephrin-binding pocket of EphA4; targets Inhibits capillary-like tube formation on Matrigel as well as [113]
several other Eph receptors cell viability
Lithocholic acid A competitive and reversible Eph-receptor ligand, Lithocholic acid dose-dependently inhibits PC3 adhesion to [84,114]
inhibits ephrin binding to both EphA and EphB ephrinA1-Fc and antagonizes the EphA2 dependent PC3 cell
receptors; also inhibits the ephrin-induced EphA2 and rounding
EphB4 tyrosine phosphorylation

7
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

growth and angiogenesis with xenograft studies in vivo [80,81]. The 6. Conclusions and perspective
soluble monomeric derivative of the extracellular domain of EphB4
(sEphB4) was designed as an antagonist of EphB4- ephrinB2 signaling. EphB4 and its ligand ephrinB2 are increasingly being recognized as
sEphB4 blocks the activation of EphB4 by ephrinB2, suppresses en- therapeutic targets in cancer. However, we are still far from being able
dothelial cell migration, adhesion, and tube formation in vitro, and in- to reliably decipher their mechanism of action and to precisely predict
hibits the angiogenic effects of various growth factors (VEGF and bFGF) the outcomes of their modulation. The uncertainty is due to the com-
in vivo. sEphB4 also inhibits tumor growth in murine tumor xenograft plexity of the interacting cells that receive interdependent signals from
models [80]. sEphB4 is thus a therapeutic candidate for vascular pro- the same complex, EphB4 and ephrinB2, in association with other re-
liferative diseases and cancer. ceptor tyrosine kinases that may promote or repress tumorigenesis. The
TNYL (TNYLFSPNGPIA) was the most potent among several syn- functional coupling of EphB4 with downstream effectors can be dra-
thetic peptides examined, with an IC50 value of 50- 150 μM for the matically differentially activated in a cell or tissue context-dependent
inhibition of EphB4–ephrin-B2 interaction in ELISA assays. However, a manner. Meanwhile, responses after initiation could be due to kinase-
modified version that contains the RAW motif found in other EphB4- dependent or kinase- independent signaling pathways that are cell or
binding peptides (TNYL-RAW) at the C terminus has dramatically im- tissue specific- manner. Thus, inhibiting kinase activity could be ben-
proved potency [84]. Jill et al. reported the crystal structure of the eficial in some settings, whereas in others, promoting kinase activity
EphB4 receptor in complex with a highly specific antagonistic peptide might be useful. The lack of specificity is always a challenge for ther-
(TNYL-RAW) at a resolution of 1.65 Å. The structure provides a mole- apeutic targeting, which requires the targeting of pathological EphB4
cular explanation for the remarkably high binding affinity of the pep- functions, without inadvertently affecting unrelated tissues or organs.
tide. TNYL-RAW is localized to a hydrophobic cleft in EphB4, corre- An important step forward will involve understanding the EphB4-
sponding to the cleft in EphB2 occupied by the ephrin-B2 G-H loop, ephrinB2 activities beyond bidirectional signaling and precisely eluci-
consistent with its antagonistic properties. The TNYL-RAW peptide is a dating the crosstalk with oncogenic pathways. Systematic studies will
particularly effective antagonist because it interferes with ephrinB2 also be critical in providing a comprehensive overview of the effects of
binding not only in the high- affinity binding cleft of EphB4, but also in the EphB4 bidirectional and unconventional signaling mechanisms in
the regions adjacent to the dimerization interface. These studies reveal cancer. Apart from their relatively well- understood role in angiogen-
structural features that will aid drug discovery initiatives, for devel- esis, it will be interesting to elucidate how the EphB4-ephrinB2 system
oping EphB4 antagonists for therapeutic applications [21]. Liu et al. influences metastasis, the epithelial-to-mesenchymal transition, tissue
produced an EphB4 inhibitor, sEphB4-HSA, which was prepared by invasion and other hallmarks of cancer.
fusing monomeric soluble EphB4 to human serum albumin [52]. For each type of target, there are some challenges: (1) the targets of
sEphB4-HSA was highly active as a single agent in inhibiting tumor protein- protein interactions at the surface of the EphB4- ephrinB2
growth, that was accompanied by apoptosis in tumor cells and the in- complex are typically hard to perceive and difficult to inhibit using
hibition of PI3K and Src signaling. A combination of sEphB4-HSA and small molecules; (2) small molecule compounds targeting the ATP-
the anti-VEGF antibody (Bevacizumab) was superior to each agent binding site could block the activity of other kinases, and therefore,
alone, and led to complete tumor regression [52]. their selectivity tends to be low; (3) some Eph kinase inhibitors with
anti-angiogenic activity might also block potential EphB4 tumor sup-
pressive activities. Thus, despite the challenges presented by the com-
5.2. Clinical studies on EphB4 inhibitors plex biology of the Eph receptor/ephrin system, exciting possibilities
exist for therapies exploiting these molecules. Multiple factors should
None of the currently approved angiogenesis inhibitors have been be taken into consideration such as tumor type, stage, and tumor mi-
reported to inhibit EphB4. JI-101 is an oral multi-kinase inhibitor that croenvironment. The optimizing strategies will require more details
targets VEGFR2, platelet derived growth factor receptor β (PDGFR-β), understanding of the signaling mechanisms via EphB4 in different
and ephrin type-B receptor 4 (EphB4) [116]. By targeting multiple tumor cells. Thus, a more in depth understanding of the structure,
angiogenesis signaling pathways in tumor vessel beds, JI-101 has the biogenesis, and mechanisms of EphB4 will provide invaluable in-
potential to inhibit multiple stages of tumor angiogenesis and thus formation for functional studies and an increase in the efficiency of
enhance its anti-tumor efficacy. JI-101 was previously studied in a rational drug design.
phase I trial in patients with advanced solid tumors [116]. Everolimus
is an FDA-approved drug for the treatment of advanced renal cell car- Conflict of interest
cinoma and advanced neuroendocrine tumors. The combination of
these two agents may provide synergistic benefits to patients with The authors declare that there are no conflicts of interest.
certain solid tumors.
XL647 is an oral small-molecule inhibitor of multiple receptor tyr- Acknowledgements
osine kinases, including endothelial growth factor receptor (EGFR),
VEGFR2, HER2 and EphB4. XL647 inhibits cellular proliferation and This work was supported by the National Natural Science
the activation of the EGFR-pathway in the erlotinib-resistant H1975 cell Foundation of China (Grant no. 81773772, 81370088, 81227802 and
line that harbors a double mutation (L858R and T790 M) in EGFR. In 81230079), the Fundamental Research Funds for the Central
vivo efficacy studies showed that XL647 substantially inhibited the Universities of Zhuizong.
growth of H1975 xenograft tumors, and reduced both tumor EGFR
signaling and tumor vessel density. Further, XL647 demonstrated an References
inhibitory effect on tumor cell growth and tumor vasculature in both
EGFR mutant xenografts and MDA-MB-231 xenografts that highly ex- [1] A. Unzue, K. Lafleur, H. Zhao, T. Zhou, J. Dong, P. Kolb, et al., Three stories on Eph
press VEGF [91]. Moreover, Pietanza et al. reported an open-label, kinase inhibitors: from in silico discovery to in vivo validation, Eur. J. Med. Chem.
112 (2016) 347–366.
multi-institutional Phase II study to investigate the efficacy and safety [2] A. Das, U. Shergill, L. Thakur, S. Sinha, R. Urrutia, D. Mukhopadhyay, et al.,
of XL647 for the treatment-naive non-small-cell lung cancer patients Ephrin B2/EphB4 pathway in hepatic stellate cells stimulates Erk-dependent VEGF
production and sinusoidal endothelial cell recruitment, Am. J. Physiol.
that show an enrichment in EGFR mutations. Patients with EGFR mu- Gastrointest. Liver Physiol. 298 (2010) G908–15.
tations had a 57% objective response rate when administered with [3] E.M. Lisabeth, G. Falivelli, E.B. Pasquale, Eph receptor signaling and ephrins, Cold
XL647 as a first-line treatment [117]. Spring Harb. Perspect. Biol. 5 (2013).
[4] E.B. Pasquale, The Eph family of receptors, Curr. Opin. Cell Biol. 9 (1997)

8
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

608–615. (2006) 815–U53.


[5] A. Kania, R. Klein, Mechanisms of ephrin-Eph signalling in development, phy- [35] N.K. Noren, E.B. Pasquale, Paradoxes of the EphB4 receptor in cancer, Cancer Res.
siology and disease, Nat. Rev. Mol. Cell Biol. 17 (2016) 240–256. 67 (2007) 3994–3997.
[6] A. Kania, R. Klein, Mechanisms of ephrin-Eph signalling in development, phy- [36] X.D. Huang, D. Wu, H. Jin, D. Stupack, J.Y.J. Wang, Induction of cell retraction by
siology and disease, Nat. Rev. Mol. Cell Biol. 17 (2016) 240–256. the combined actions of Abl-CrkII and Rho-ROCK1 signaling, J. Cell Biol. 183
[7] A. Barquilla, E.B. Pasquale, Eph receptors and ephrins: therapeutic opportunities, (2008) 711–723.
Annu. Rev. Pharmacol. Toxicol. 55 (2015) 465–487. [37] Z. Xiao, R.A. Carrasco, K. Kinneer, D. Sabol, B. Jallal, S. Coats, et al., EphB4
[8] S.R. Kumar, J. Singh, G.B. Xia, V. Krasnoperov, L. Hassanieh, E.J. Ley, et al., promotes or suppresses Ras/MEK/ERK pathway in a context-dependent manner
Receptor tyrosine kinase EphB4 is a survival factor in breast cancer, Am. J. Pathol. implications for EphB4 as a cancer target, Cancer Biol. Ther. 13 (2012) 630–637.
169 (2006) 279–293. [38] F.Q. Hu, Z. Tao, Z.Y. Shen, X.L. Wang, F. Hua, Down-regulation of EphB4 phos-
[9] G.B. Xia, S.R. Kumar, R. Masood, S.T. Zhu, R. Reddy, V. Krasnoperov, et al., EphB4 phorylation is necessary for esophageal squamous cell carcinoma tumorigenecity,
expression and biological significance in prostate cancer, Cancer Res. 65 (2005) Tumor Biol. 35 (2014) 7225–7232.
4623–4632. [39] R. Rutkowski, I. Mertens-Walker, J.E. Lisle, A.C. Herington, S.A. Stephenson,
[10] S.A. Stephenson, S. Slomka, E.L. Douglas, P.J. Hewett, J.E. Hardingham, Receptor Evidence for a dual function of EphB4 as tumor promoter and suppressor regulated
protein tyrosine kinase EphB4 is up-regulated in colon cancer, BMC Mol. Biol. 2 by the absence or presence of the ephrin-B2 ligand, Int. J. Cancer 131 (2012)
(2001) 15. E614–E624.
[11] G. Berclaz, E. Karamitopoulou, L. Mazzucchelli, V. Rohrbach, E. Dreher, [40] G. Berclaz, B. Flutsch, H.J. Altermatt, V. Rohrbach, V. Djonov, A. Ziemiecki, et al.,
A. Ziemiecki, et al., Activation of the receptor protein tyrosine kinase EphB4 in Loss of EphB4 receptor tyrosine kinase protein expression during carcinogenesis of
endometrial hyperplasia and endometrial carcinoma, Ann. Oncol. 14 (2003) the human breast, Oncol. Rep. 9 (2002) 985–989.
220–226. [41] H. Dopeso, S. Mateo-Lozano, R. Mazzolini, P. Rodrigues, L. Lagares-Tena, J. Ceron,
[12] X. Huang, Y. Yamada, H. Kidoya, H. Naito, Y. Nagahama, L. Kong, et al., EphB4 et al., The receptor tyrosine kinase EPHB4 has tumor suppressor activities in in-
overexpression in B16 melanoma cells affects arterial-venous patterning in tumor testinal tumorigenesis, Cancer Res. 69 (2009) 7430–7438.
angiogenesis, Cancer Res. 67 (2007) 9800–9808. [42] P. Huusko, D. Ponciano-Jackson, M. Wolf, J.A. Kiefer, D.O. Azorsa, S. Tuzmen,
[13] G. Castellano, J.F. Reid, P. Alberti, M.L. Carcangiu, A. Tomassetti, S. Canevari, et al., Nonsense-mediated decay microarray analysis identifies mutations of
New potential ligand-receptor signaling loops in ovarian cancer identified in EPHB2 in human prostate cancer, Nat. Genet. 36 (2004) 979–983.
multiple gene expression studies, Cancer Res. 66 (2006) 10709–10719. [43] F. Barneh, M. Moshayedi, H. Mirmohammadsadeghi, S. Haghjooy-Javanmard,
[14] E.B. Pasquale, Eph receptors and ephrins in cancer: bidirectional signalling and A.M. Sabzghabaee, S. Badri, EphB4 tyrosine kinase stimulation inhibits growth of
beyond, Nat. Rev. Cancer 10 (2010) 165–180. MDA-MB-231 breast cancer cells in a dose and time dependent manner, Dis.
[15] J.P. Himanen, K.R. Rajashankar, M. Lackmann, C.A. Cowan, M. Henkemeyer, Markers 93 (2013) 3–8.
D.B. Nikolov, Crystal structure of an Eph receptor-ephrin complex, Nature 414 [44] E.B. Pasquale, EPH receptor signalling casts a wide net on cell behaviour, Nat. Rev.
(2001) 933–938. Mol. Cell Biol. 6 (2005) 462–475.
[16] J.P. Himanen, N. Saha, D.B. Nikolov, Cell-cell signaling via Eph receptors and [45] D. Arvanitis, A. Davy, Eph/ephrin signaling: networks, Gene Dev. 22 (2008)
ephrins, Curr. Opin. Cell Biol. 19 (2007) 534–542. 416–429.
[17] J.P. Himanen, L. Yermekbayeva, P.W. Janes, J.R. Walker, K. Xu, L. Atapattu, et al., [46] A.C. Andres, A. Ziemiecki, Eph and ephrin signaling in mammary gland mor-
Architecture of Eph receptor clusters, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) phogenesis and cancer, J. Mammary Gland Biol. 8 (2003) 475–485.
10860–10865. [47] H. Clevers, E. Batlle, EphB/EphrinB receptors and Wnt signaling in colorectal
[18] E.B. Pasquale, Eph-ephrin bidirectional signaling in physiology and disease, Cell cancer, Cancer Res. 66 (2006) 2–5.
133 (2008) 38–52. [48] J.H. Lv, Q.Y. Xia, J.J. Wang, Q. Shen, J. Zhang, X.J. Zhou, EphB4 promotes the
[19] A.H. Zisch, C. Pazzagli, A.L. Freeman, M. Schneller, M. Hadman, J.W. Smith, et al., proliferation, invasion, and angiogenesis of human colorectal cancer, Exp. Mol.
Replacing two conserved tyrosines of the EphB2 receptor with glutamic acid Pathol. 100 (2016) 402–408.
prevents binding of SH2 domains without abrogating kinase activity and biolo- [49] J. Bai, Y.J. Wang, L. Liu, Y.L. Zhao, Ephrin B2 and EphB4 selectively mark arterial
gical responses, Oncogene 19 (2000) 177–187. and venous vessels in cerebral arteriovenous malformation, J. Int. Med. Res. 42
[20] J.E. Chrencik, A. Brooun, M.L. Kraus, M.I. Recht, A.R. Kolatkar, G.W. Han, et al., (2014) 405–415.
Structural and biophysical characterization of the EphB4(center dot)ephrinB2 [50] D.M. Brantley-Sieders, A.X. Jiang, K. Sarma, A. Badu-Nkansah, D.L. Walter,
protein-protein interaction and receptor specificity, J. Biol. Chem. 281 (2006) Y. Shyr, et al., Eph/ephrin profiling in human Breast cancer reveals significant
28185–28192. associations between expression level and clinical outcome, PLoS One 6 (2011).
[21] J.E. Chrencik, A. Brooun, M.I. Recht, M.L. Kraus, M. Koolpe, A.R. Kolatkar, et al., [51] B.D. Ferguson, R. Liu, C.E. Rolle, Y.H.C. Tan, V. Krasnoperov, R. Kanteti, et al., The
Structure and thermodynamic characterization of the EphB4/ephrin-B2 antagonist EphB4 receptor tyrosine kinase promotes lung cancer growth: a potential novel
peptide complex reveals the determinants for receptor specificity, Structure 14 therapeutic target, PLoS One 8 (2013).
(2006) 321–330. [52] R. Liu, B.D. Ferguson, Y. Zhou, K. Naga, R. Salgia, P.S. Gill, et al., EphB4 as a
[22] D.D. Dai, Q. Huang, R. Nussinov, B.Y. Ma, Promiscuous and specific recognition therapeutic target in mesothelioma, BMC Cancer (2013) 2013.
among ephrins and Eph receptors, Bba-Proteins Proteom. 1844 (2014) [53] N.Y. Yang, E.B. Pasquale, L.B. Owen, I.M. Ethell, The EphB4 receptor-tyrosine
1729–1740. kinase promotes the migration of melanoma cells through Rho-mediated actin
[23] J.E. Chrencik, A. Brooun, M.I. Recht, G. Nicola, L.K. Davis, R. Abagyan, et al., cytoskeleton reorganization, J. Biol. Chem. 281 (2006) 32574–32586.
Three-dimensional structure of the EphB2 receptor in complex with an antag- [54] M. Becerikli, B. Merwart, M.C. Lam, P. Suppelna, A. Rittig,
onistic peptide reveals a novel mode of inhibition, J. Biol. Chem. 282 (2007) A. Mirmohammedsadegh, et al., EPHB4 tyrosine-kinase receptor expression and
36505–36513. biological significance in soft tissue sarcoma, Int. J. Cancer 136 (2015)
[24] J.S. Tokarski, J.A. Newitt, C.Y.J. Chang, J.D. Cheng, M. Wittekind, S.E. Kiefer, 1781–1791.
et al., The structure of dasatinib (BMS-354825) bound to activated ABL kinase [55] S. Meyer, C. Hafner, M. Guba, S. Flegel, E.K. Geissler, B. Becker, et al., Ephrin-B2
domain elucidates its inhibitory activity against imatinib-resistant ABL mutants, overexpression enhances integrin-mediated ECM-attachment and migration of B16
Cancer Res. 66 (2006) 5790–5797. melanoma cells, Int. J. Oncol. 27 (2005) 1197–1206.
[25] B. Barlaam, R. Ducray, C. Lambert-van der Brempt, P. Ple, C. Bardelle, N. Brooks, [56] M. Nakada, E.M. Anderson, T. Demuth, S. Nakada, L.B. Reavie, K.L. Drake, et al.,
et al., Inhibitors of the tyrosine kinase EphB4. Part 4: discovery and optimization The phosphorylation of ephrin-B2 ligand promotes glioma cell migration and in-
of a benzylic alcohol series, Bioorg. Med. Chem. Lett. 21 (2011) 2207–2211. vasion, Int. J. Cancer 126 (2010) 1155–1165.
[26] C. Bardelle, B. Barlaam, N. Brooks, T. Coleman, D. Cross, R. Ducray, et al., [57] N.K. Noren, M. Lu, A.L. Freeman, M. Koolpe, E.B. Pasquale, Interplay between
Inhibitors of the tyrosine kinase EphB4. Part 3: identification of non-benzodioxole- EphB4 on tumor cells and vascular ephrin-B2 regulates tumor growth, Proc. Natl.
based kinase inhibitors, Bioorg. Med. Chem. Lett. 20 (2010) 6242–6245. Acad. Sci. U. S. A. 101 (2004) 5583–5588.
[27] C. Bardelle, T. Coleman, D. Cross, S. Davenport, J.G. Kettle, E.J. Ko, et al., [58] S. Sawamiphak, S. Seidel, C.L. Essmann, G.A. Wilkinson, M.E. Pitulescu, T. Acker,
Inhibitors of the tyrosine kinase EphB4 Part 2: structure-based discovery and et al., Ephrin-B2 regulates VEGFR2 function in developmental and tumour an-
optimisation of 3, 5-bis substituted anilinopyrimidines, Bioorg. Med. Chem. Lett. giogenesis, Nature 465 (2010) 487–U115.
18 (2008) 5717–5721. [59] S.K. Alam, V.K. Yadav, S. Bajaj, A. Datta, S.K. Dutta, M. Bhattacharyya, et al., DNA
[28] C. Bardelle, D. Cross, S. Davenport, J.G. Kettle, E.J. Ko, A.G. Leach, et al., damage-induced ephrin-B2 reverse signaling promotes chemoresistance and drives
Inhibitors of the tyrosine kinase EphB4 Part 1: structure-based design and opti- EMT in colorectal carcinoma harboring mutant p53, Cell Death Differ. 23 (2016)
mization of a series of 2, 4-bis-anilinopyrimidines, Bioorg. Med. Chem. Lett. 18 707–722.
(2008) 2776–2780. [60] Y. Takahashi, M. Itoh, N. Nara, S. Tohda, Effect of EPH-Ephrin signaling on the
[29] R.C. Overman, J.E. Debreczeni, C.M. Truman, M.S. McAlister, T.K. Attwood, growth of human leukemia cells, Anticancer Res. 34 (2014) 2913–2918.
Completing the structural family portrait of the human EphB tyrosine kinase do- [61] S.R. Kumar, R. Masood, W.A. Spannuth, J. Singh, J. Scehnet, G. Kleiber, et al., The
mains, Protein Sci. 23 (2014) 627–638. receptor tyrosine kinase EphB4 is overexpressed in ovarian cancer, provides sur-
[30] R.C. Overman, I. Green, C.M. Truman, J.A. Read, K.J. Embrey, M.S.B. McAlister, vival signals and predicts poor outcome, Br. J. Cancer 96 (2007) 1083–1091.
et al., Stability and solubility engineering of the EphB4 tyrosine kinase catalytic [62] N.K. Noren, N.Y. Yang, M. Silldorff, R. Mutyala, E.B. Pasquale, Ephrin-in-
domain using a rationally designed synthetic library, Protein Eng. Des. Sel. 26 dependent regulation of cell substrate adhesion by the EphB4 receptor, Biochem.
(2013) 695–704. J. 422 (2009) 433–442.
[31] E.B. Pasquale, Eph receptor signalling casts a wide net on cell behaviour, Nat. Rev. [63] I. Mertens-Walker, B.C. Fernandini, M.S.N. Maharaj, A. Rockstroh, C.C. Nelson,
Mol. Cell Biol. 6 (2005) 462–475. A.C. Herington, et al., The tumour-promoting receptor tyrosine kinase, EphB4,
[32] O. Salvucci, G. Tosato, Essential roles of EphB receptors and EphrinB ligands in regulates expression of Integrin-beta 8 in prostate cancer cells, BMC Cancer 15
endothelial cell function and angiogenesis, Adv. Cancer Res. 114 (2012) 21–57. (2015).
[33] S.R. Kumar, J.S. Scehnet, E.J. Ley, J. Singh, V. Krasnoperov, R. Liu, et al., [64] M. Heroult, F. Schaffner, D. Pfaff, C. Prahst, R. Kirmse, S. Kutschera, et al., EphB4
Preferential induction of EphB4 over EphB2 and its implication in colorectal promotes site-specific metastatic tumor cell dissemination by interacting with
cancer progression, Cancer Res. 69 (2009) 3736–3745. endothelial cell-expressed EphrinB2, Mol. Cancer Res. 8 (2010) 1297–1309.
[34] N.K. Noren, G. Foos, C.A. Hauser, E.B. Pasquale, The EphB4 receptor suppresses [65] S.P. Herbert, J. Huisken, T.N. Kim, M.E. Feldman, B.T. Houseman, R.A. Wang,
breast cancer cell tumorigenicity through an Abl-Crk pathway, Nat. Cell Biol. 8 et al., Arterial-venous segregation by selective cell sprouting: an alternative mode

9
Y. Chen et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

of blood vessel formation, Science 326 (2009) 294–298. Chem. 112 (2016) 347–366.
[66] S. Kuijper, C.J. Turner, R.H. Adams, Regulation of angiogenesis by Eph-Ephrin [93] M. Koolpe, M. Dail, E.B. Pasquale, An ephrin mimetic peptide that selectively
interactions, Trends Cardiovasc. Med. 17 (2007) 145–151. targets the EphA2 receptor, J. Biol. Chem. 277 (2002) 46974–46979.
[67] R. Erber, U. Eichelsbacher, V. Powajbo, T. Korn, V. Djonov, J.H. Lin, et al., EphB4 [94] B.Y. Ma, S. Kolb, M. Diprima, M. Karna, G. Tosato, Q.Q. Yang, et al., Investigation
controls blood vascular morphogenesis during postnatal angiogenesis, EMBO J. 25 of the interactions between the EphB2 receptor and SNEW peptide variants,
(2006) 628–641. Growth Factors 32 (2014) 236–246.
[68] O. Salvucci, D. Maric, M. Economopoulou, S. Sakakibara, S. Merlin, A. Follenzi, [95] K. Ebert, J. Wiemer, J. Caballero, M. Kockerling, J. Steinbach, J. Pietzsch, et al.,
et al., EphrinB reverse signaling contributes to endothelial and mural cell assembly Development of indazolylpyrimidine derivatives as high-affine EphB4 receptor
into vascular structures, Blood 114 (2009) 1707–1716. ligands and potential PET radiotracers, Bioorg. Med. Chem. 23 (2015) 6025–6035.
[69] P. Foubert, J.S. Silvestre, B. Souttou, V. Barateau, C. Martin, T.G. Ebrahimian, [96] Y. Chen, Y. Zhang, Functional and mechanistic analysis of telomerase: an anti-
et al., PSGL-1-mediated activation of EphB4 increases the proangiogenic potential tumor drug target, Pharmacol. Ther. 163 (2016) 24–47.
of endothelial progenitor cells, J. Clin. Invest. 117 (2007) 1527–1537. [97] C. Bardelle, D. Cross, S. Davenport, J.G. Kettle, E.J. Ko, A.G. Leach, et al.,
[70] D. Pfaff, M. Heroult, M. Riedel, Y. Reiss, R. Kirmse, T. Ludwig, et al., Involvement Inhibitors of the tyrosine kinase EphB4 Part 1: structure-based design and opti-
of endothelial ephrin-B2 in adhesion and transmigration of EphB-receptor-ex- mization of a series of 2, 4-bis-anilinopyrimidines, Bioorg. Med. Chem. Lett. 18
pressing monocytes, J. Cell Sci. 121 (2008) 3842–3850. (2008) 2776–2780.
[71] S.S. Gerety, H.U. Wang, Z.F. Chen, D.J. Anderson, Symmetrical mutant phenotypes [98] C. Bardelle, T. Coleman, D. Cross, S. Davenport, J.G. Kettle, E.J. Ko, et al.,
of the receptor EphB4 and its specific transmembrane ligand ephrin-B2 in cardi- Inhibitors of the tyrosine kinase EphB4 Part 2: structure-based discovery and
ovascular development, Mol. Cell 4 (1999) 403–414. optimisation of 3, 5-bis substituted anilinopyrimidines, Bioorg. Med. Chem. Lett.
[72] C. Zhao, N. Irie, Y. Takada, K. Shimoda, T. Miyamoto, T. Nishiwaki, et al., 18 (2008) 5717–5721.
Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis, Cell Metab. 4 [99] C. Bardelle, B. Barlaam, N. Brooks, T. Coleman, D. Cross, R. Ducray, et al.,
(2006) 111–121. Inhibitors of the tyrosine kinase EphB4. Part 3: identification of non-benzodioxole-
[73] G. Xia, S.R. Kumar, R. Masood, S. Zhu, R. Reddy, V. Krasnoperov, et al., EphB4 based kinase inhibitors, Bioorg. Med. Chem. Lett. 20 (2010) 6242–6245.
expression and biological significance in prostate cancer, Cancer Res. 65 (2005) [100] B. Barlaam, R. Ducray, C. Lambert-van der Brempt, P. Ple, C. Bardelle, N. Brooks,
4623–4632. et al., Inhibitors of the tyrosine kinase EphB4. Part 4: discovery and optimization
[74] G. Xia, S.P. Kumar, J.P. Stein, J. Singh, V. Krasnoperov, S. Zhu, et al., EphB4 of a benzylic alcohol series, Bioorg. Med. Chem. Lett. 21 (2011) 2207–2211.
receptor tyrosine kinase is expressed in bladder cancer and provides signals for cell [101] R.J. Kathawala, L.Y. Wei, N. Anreddy, K. Chen, A. Patel, S. Alqahtani, et al., The
survival, Oncogene 25 (2006) 769–780. small molecule tyrosine kinase inhibitor NVP-BHG712 antagonizes ABCC10-
[75] V. Krasnoperov, S.R. Kumar, E. Ley, X.Q. Li, J. Scehnet, R. Liu, et al., Novel EphB4 mediated paclitaxel resistance: a preclinical and pharmacokinetic study,
monoclonal antibodies modulate angiogenesis and inhibit tumor growth, Am. J. Oncotarget 6 (2015) 510–521.
Pathol. 176 (2010) 2029–2038. [102] Y. Nakazawa, S. Kawano, J. Matsui, Y. Funahashi, O. Tohyama, H. Muto, et al.,
[76] M.A. Abengozar, S. de Frutos, S. Ferreiro, J. Soriano, M. Perez-Martinez, Multitargeting strategy using lenvatinib and golvatinib: maximizing anti-angio-
D. Olmeda, et al., Blocking ephrinB2 with highly specific antibodies inhibits an- genesis activity in a preclinical cancer model, Cancer Sci. 106 (2015) 201–207.
giogenesis, lymphangiogenesis, and tumor growth, Blood 119 (2012) 4565–4576. [103] T. Nakagawa, O. Tohyama, A. Yamaguchi, T. Matsushima, K. Takahashi,
[77] S. Duggineni, S. Mitra, R. Noberini, X.F. Han, N. Lin, Y. Xu, et al., Design, synthesis S. Funasaka, et al., E7050: a dual c-Met and VEGFR-2 tyrosine kinase inhibitor
and characterization of novel small molecular inhibitors of ephrin-B2 binding to promotes tumor regression and prolongs survival in mouse xenograft models,
EphB4, Biochem. Pharmacol. 85 (2013) 507–513. Cancer Sci. 101 (2010) 210–215.
[78] M. Koolpe, R. Burgess, M. Dail, E.B. Pasquale, EphB receptor-binding peptides [104] M.W. Karaman, S. Herrgard, D.K. Treiber, P. Gallant, C.E. Atteridge,
identified by phage display enable design of an antagonist with ephrin-like affi- B.T. Campbell, et al., A quantitative analysis of kinase inhibitor selectivity, Nat.
nity, J. Biol. Chem. 280 (2005) 17301–17311. Biotechnol. 26 (2008) 127–132.
[79] R. Noberini, S. Mitra, O. Salvucci, F. Valencia, S. Duggineni, N. Prigozhina, et al., [105] Q. Chang, C. Jorgensen, T. Pawson, D.W. Hedley, Effects of dasatinib on EphA2
PEGylation potentiates the effectiveness of an antagonistic peptide that targets the receptor tyrosine kinase activity and downstream signalling in pancreatic cancer,
EphB4 receptor with nanomolar affinity, PLoS One (2011) 2011. Br. J. Cancer 99 (2008) 1074–1082.
[80] N. Kertesz, V. Krasnoperov, R. Reddy, L. Leshanski, S.R. Kumar, S. Zozulya, et al., [106] J.S. Melnick, J. Janes, S. Kim, J.Y. Chang, D.G. Sipes, D. Gunderson, et al., An
The soluble extracellular domain of EphB4 (sEphB4) antagonizes EphB4-EphrinB2 efficient rapid system for profiling the cellular activities of molecular libraries,
interaction, modulates angiogenesis, and inhibits tumor growth, Blood 107 (2006) Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 3153–3158.
2330–2338. [107] M. Breccia, Alimena G. Nilotinib, A second-generation tyrosine kinase inhibitor for
[81] G. Martiny-Baron, T. Korff, F. Schaffner, N. Esser, S. Eggstein, D. Marme, et al., chronic myeloid leukemia, Leukemia Res. 34 (2010) 129–134.
Inhibition of tumor growth and angiogenesis by soluble EphB4, Neoplasia 6 [108] C. Mamat, B. Mosch, C. Neuber, M. Kockerling, R. Bergmann, J. Pietzsch, Fluorine-
(2004) 248–257. 18 radiolabeling and radiopharmacological characterization of a benzodiox-
[82] D. Djokovic, A. Trindade, J. Gigante, M. Badenes, L. Silva, R. Liu, et al., olylpyrimidine-based radiotracer targeting the receptor tyrosine kinase EphB4,
Combination of Dll4/Notch and Ephrin-B2/EphB4 targeted therapy is highly ef- ChemMedChem 7 (2012) 1991–2003.
fective in disrupting tumor angiogenesis, BMC Cancer 10 (2010). [109] T.P. Burkholder, J.R. Clayton, M.E. Rempala, J.R. Henry, J.M. Knobeloch,
[83] S.A. Stephenson, E.L. Douglas, I. Mertens-Walker, J.E. Lisle, M.S.N. Maharaj, D. Mendel, et al., Discovery of LY2457546: a multi-targeted anti-angiogenic kinase
A.C. Herington, Anti-tumour effects of antibodies targeting the extracellular cy- inhibitor with a novel spectrum of activity and exquisite potency in the acute
steine-rich region of the receptor tyrosine kinase EphB4, Oncotarget 6 (2015) myelogenous leukemia-Flt-3-internal tandem duplication mutant human tumor
7554–7569. xenograft model, Invest. New Drug 30 (2012) 936–949.
[84] R. Noberini, I. Lamberto, E.B. Pasquale, Targeting Eph receptors with peptides and [110] S.A. Mitchell, M.D. Danca, P.A. Blomgren, J.W. Darrow, K.S. Currie, J.E. Kropf,
small molecules: progress and challenges, Sem. Cell Dev. Biol. 23 (2012) 51–57. et al., Imidazo[1,2-a]pyrazine diaryl ureas: inhibitors of the receptor tyrosine ki-
[85] Y. Choi, F. Syeda, J.R. Walker, P.J. Finerty, D. Cuerrier, A. Wojciechowski, et al., nase EphB4, Bioorg. Med. Chem. Lett. 19 (2009) 6991–6995.
Discovery and structural analysis of Eph receptor tyrosine kinase inhibitors, [111] M. Caligiuri, L. Molz, Q. Liu, F. Kaplan, J.P. Xu, J.Z. Majeti, et al., MASPIT: three-
Bioorg. Med. Chem. Lett. 19 (2009) 4467–4470. hybrid trap for quantitative proteome fingerprinting of small molecule-protein
[86] K. Lafleur, D. Huang, T. Zhou, A. Caflisch, C. Nevado, Structure-based optimiza- interactions in mammalian cells, Chem. Biol. 13 (2006) 711–722.
tion of potent and selective inhibitors of the tyrosine kinase erythropoietin pro- [112] T. Zhou, A. Caflisch, High-throughput virtual screening using quantum mechanical
ducing human hepatocellular carcinoma receptor B4 (EphB4), J. Med. Chem. 52 probes: discovery of selective kinase inhibitors, ChemMedChem 5 (2010)
(2009) 6433–6446. 1007–1014.
[87] G. Martiny-Baron, P. Holzer, E. Billy, C. Schnell, J. Brueggen, M. Ferretti, et al., [113] R. Noberini, S.K. De, Z. Zhang, B. Wu, D. Raveendra-Panickar, V. Chen, et al., A
The small molecule specific EphB4 kinase inhibitor NVP-BHG712 inhibits VEGF disalicylic acid-furanyl derivative inhibits ephrin binding to a subset of Eph re-
driven angiogenesis, Angiogenesis 13 (2010) 259–267. ceptors, Chem. Biol. Drug Des. 78 (2011) 667–678.
[88] F. Zuccotto, E. Ardini, E. Casale, M. Angiolini, Through the gatekeeper door: ex- [114] C. Giorgio, I.H. Mohamed, L. Flammini, E. Barocelli, M. Incerti, A. Lodola, et al.,
ploiting the active kinase conformation, J. Med. Chem. 53 (2010) 2681–2694. Lithocholic acid is an eph-ephrin ligand interfering with Eph-kinase activation,
[89] Y. Miyazaki, M. Nakano, H. Sato, A.T. Truesdale, J.D. Stuart, E.N. Nartey, et al., PLoS One 6 (2011).
Design and effective synthesis of novel templates, 3,7-diphenyl-4amino-thieno and [115] M. Yang, X.H. Qian, D.H. Zhao, S.Z. Fu, Effects of Astragalus polysaccharide on the
furo-[3,2-c]pyridines as protein kinase inhibitors and in vitro evaluation targeting erythroid lineage and microarray analysis in K562 cells, J. Ethnopharmacol. 127
angiogenetic kinases, Bioorg. Med. Chem. Lett. 17 (2007) 250–254. (2010) 242–250.
[90] K. Lafleur, J. Dong, D. Huang, A. Caflisch, C. Nevado, Optimization of inhibitors of [116] T.L. Werner, M.L. Wade, N. Agarwal, K. Boucher, J. Patel, A. Luebke, et al., A pilot
the tyrosine kinase EphB4: 2. Cellular potency improvement and binding mode study of JI-101, an inhibitor of VEGFR-2, PDGFR-beta, and EphB4 receptors, in
validation by X-ray crystallography, J. Med. Chem. 56 (2013) 84–96. combination with everolimus and as a single agent in an ovarian cancer expansion
[91] S.B. Gendreau, R. Ventura, P. Keast, A.D. Laird, F.M. Yakes, W. Zhang, et al., cohort, Invest. New Drug 33 (2015) 1217–1224.
Inhibition of the T790M gatekeeper mutant of the epidermal growth factor re- [117] M.C. Pietanza, S.M. Gadgeel, A. Dowlati, T.J. Lynch, R. Salgia, K.M. Rowland,
ceptor by EXEL-7647, Clin. Cancer Res. 13 (2007) 3713–3723. et al., Phase II study of the multitargeted tyrosine kinase inhibitor XL647 in pa-
[92] A. Unzue, K. Lafleur, H.T. Zhao, T. Zhou, J. Dong, P. Kolb, et al., Three stories on tients with non-small-cell lung cancer, J. Thorac. Oncol. 7 (2012) 856–865.
Eph kinase inhibitors: from in silico discovery to in vivo validation, Eur. J. Med.

10

You might also like