Sie sind auf Seite 1von 16

Critical Reviews in Oncology/Hematology 50 (2004) 23–38

Tyrosine kinase receptors as attractive targets of cancer therapy


Amar Bennasroune, Anne Gardin1 , Dominique Aunis, Gérard Crémel, Pierre Hubert∗
INSERM Unit 575, 5 rue Blaise Pascal, 67084 Strasbourg Cedex, France

Accepted 11 August 2003

Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2. RTK signaling: ligand-binding, dimerization, kinase activity, substrate phosphorylation, protein docking 25
2.1. RTK structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
2.2. Aberrations affecting growth factor receptors in tumor cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
2.3. Signaling pathways of tyrosine kinase receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
2.4. Regulation of RTK activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
3. RTKs and cancer: experimental tools . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
4. RTK as targets in clinical research: what is going on? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
4.1. Antagonists of ligand-binding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
4.2. Antibodies to RTKs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
4.3. Tyrosine kinase inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.4. Protein–protein interaction inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
4.4.1. Inhibitors of signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
4.4.2. Inhibitors of dimerization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
5. Conclusion and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
Reviewers (MA 496) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
Biography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38

Abstract

Receptor tyrosine kinases (RTKs) are the main mediators of the signaling network that transmit extracellular signals into the cell, and
control cellular differentiation and proliferation. Recent and rapid advances in our understanding of cellular signaling by receptor tyrosine
kinases, in normal and malignant cells, have brought to light the potential of RTKs as selective anti-cancer targets. Their activity is normally
tightly controlled and regulated. Overexpression of RTK proteins or functional alterations caused by mutations in the corresponding genes
or abnormal stimulation by autocrine growth factor loops contribute to constitutive RTK signaling, resulting in dysregulated cell growth
and cancer. The mechanisms of uncontrolled RTK signaling that leads to cancer has provided the rationale for anti-RTK drug development.
Herceptin, Gleevec, and Iressa are the first examples of drugs which have successfully translated basic research on oncogenes into cancer
therapeutics. RTKs can be viewed as multifunctional targets, and strategies towards the prevention and inhibition of RTK signaling include
antibodies, antagonist ligands, small molecule inhibitors of protein kinase activity, and inhibitors of protein–protein interactions. Progresses
in the field of rational drug design and computational chemistry will vastly benefit from the availability of increasing structural knowledge of
both the kinase domains and the ligand-binding sites of these receptors.
© 2003 Elsevier Ireland Ltd. All rights reserved.

Keywords: Tyrosine protein kinase; Receptors; Oncology; Signaling; Therapy

∗ Corresponding author. Tel.: +33-3-88-45-67-03; fax: +33-3-88-60-08-06.


E-mail address: hubert@neurochem.u-strasbg.fr (P. Hubert).
1 Present address: Clinical Pharmacology Department, Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland.

1040-8428/$ – see front matter © 2003 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.critrevonc.2003.08.004
24 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

1. Introduction drug targets for the 21st century [11], in cancer and other
diseases.
To this day (early 2003), Herceptin (trastuzumab) for the The breakthrough discovery of Varmus and Bishop that
treatment of advanced breast cancer, Gleevec (Imatinib Me- cancer-inducing genes of animal retroviruses such as v-src
sylate, STI-571) for the therapy of patients with Philadel- represent mutated host genes that were recombined into the
phia chromosome-positive chronic myelogenous leukemia viral genome raised the question of whether the oncogene
and GIST (gastrointestinal stromal tumors), and Iressa (Gefi- concept was also relevant to human cancer [12]. This first
tinib, ZD1839, in Japan) for the treatment of lung cancer are connection between a human gene product that regulates
the first examples of drugs which have successfully trans- normal cell proliferation and a viral oncogene strongly sug-
lated basic research on oncogenes into cancer therapeutics. gested that human cancer development may also involve ab-
They have been heralded as the most significant develop- normalities in the expression and structure of endogenous
ment toward a new era of target-directed therapies [1]. These genes that have regulatory roles in cell proliferation. The
agents provide at last clinical validation of the emerging first cloning and sequence analysis of a cDNA encoding
field of molecular oncology, specifically therapies targeting a cell surface protein, the human epidermal growth factor
deregulated cellular pathways that play a critical role in tu- (EGF) receptor, provided a partial answer to this question
morigenesis, which were anticipated at the very beginning by revealing a close relationship with the v-erbB oncogene
of our understanding of molecular oncogenesis in the 1980s [13,14]. Interestingly, this cloning was performed from a
[2–4]. Very interestingly, these three drugs are inhibitors cell line which had been established from a human cancer,
of protein tyrosine kinases, a class of enzymes which have an epidermoı̈d sarcoma termed A431. A search for simi-
been at the early onset of the discovery of oncogenes [5–7]. lar genetic aberrations in tumor tissues using cDNA probes
Furthermore, they inhibit a subclass of these oncogenes, the of EGFR and an accidentally cloned EGFR-related gene,
receptor tyrosine kinases (RTKs), which present distinctive termed human EGFR-related gene (HER2), resulted in the
advantages as potential drug targets. This article reviews the discovery that the gene encoding the HER2/neu receptor ty-
links between the still growing field of our basic understand- rosine kinase is amplified up to 100-fold in the tumor cells
ing of the functions of these RTKs and their deregulation of about 30% of patients with invasive breast cancer. A sig-
in cancer, and the ongoing developments of new drugs tar- nificant clinical correlation was shown between HER2/neu
geting RTKs which will hopefully be as successful as Her- gene amplification and overexpression and parameters of
ceptin and Gleevec [8,9]. The development of the pertain- malignancy, including reduced survival and reduced time to
ing literature is so fast that we have had to operate a very relapse, relative to patients with normal receptor levels [15].
subjective selection. We have chosen to present the RTKs This explains the current emphasis on the ErbB/HER family
as a whole family, stressing the common features that make as promising targets for new drugs.
them attractive targets, rather than discussing details about The importance of discovering new targets and path-
each receptor or subgroup of receptors. ways for therapeutic intervention in cancer is clear, and
The mutation and deregulation of cancer genes lead to criteria for validation and selection of new drug targets
a wide range of changes in cellular structure and func- can be summarized as follows: (1) frequency of genetic or
tion, all of which contribute to the malignant phenotype epigenetic deregulation of the target or pathway in human
and pathological behavior of human cancer. Hanahan and cancer; (2) demonstration in a model system that the target
Weinberg [10] have usefully characterized cancer in terms contributes to the malignant phenotype; (3) evidence of
of six hallmark traits: (1) self-sufficiency in growth signals; the reversal of the malignant phenotype by expression of
(2) insensitivity to growth inhibitory signals; (3) evasion of dominant negative protein, antisense strategies, antibodies,
apoptosis; (4) limitless replicative potential; (5) induction inhibitory peptides or drug leads; (4) practical feasibility,
of sustained angiogenesis; and (6) invasion and metastasis. tractability or drugability of the target; (5) availability of a
At least half of these traits involve RTKs and their signaling robust, efficient biological test cascade to support the drug
pathways: clearly, activating mutations and/or overexpres- discovery program; (6) ability to run a robust cost-effective
sion of RTKs and their ligands can drive deregulated cell high-throughput screen; (7) availability of a structure-based
growth; RTKs such as the insulin-like growth factor 1 drug design approach [9]. As a family, oncogenic RTKs
(IGF1) receptor are key components of the cell death control do fulfill these criteria. Proof-of-concept for the drugability
machinery; and, last but not least, angiogenesis-initiating of these molecular targets is, of course, already established
signaling involves RTKs such as the vascular endothe- with the regulatory approval of the anti-ErbB2 antibody
lium growth factor (VEGF), platelet-derived growth factor Herceptin and the tyrosine kinase inhibitors Gleevec and
(PDGF) and fibroblast growth factor (FGF) receptors. As Iressa. Although many efforts in designing new anti-cancer
all of these traits can be targeted by the new generation of drugs have so far been mostly directed at the ErbB
molecular therapeutic agents, this should place RTKs at a sub-family, one may reasonably believe that the established
very prominent place among targets to attack malignant dis- proof-of-concept will hold true for most if not all oncogenic
ease. Indeed, Sir Philip Cohen has very recently proposed RTKs. Most noteworthy is the fact that structural knowledge
that protein kinases in general may prove to be the major of the crystal structure of many tyrosine kinase domains
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 25

Fig. 1. Schematic structure of receptor tyrosine kinases, and their mechanism of activation: (a) extracellular ligand-binding domain; (b) unique
transmembrane domain; (c) juxtamembrane region; (d) kinase domain; (e) C-terminus. (1) The first step in receptor activation consists in ligand-induced
dimerization/conformational change; (2) this brings the catalytic domains in close proximity for transphosphorylation, yielding full receptor activation.

has already been instrumental in the very rapid molecular ceptor families, have been identified [19]. This catalog has
understanding of clinical resistance to STI-571 [16]. recently been confirmed by analysis of the human genome
Furthermore, RTKs are multifunctional transmembrane [20]. All RTKs contain a large, glycosylated, extracellu-
proteins, expressed at the cell surface. These characteristics lar ligand-binding domain, a single transmembrane region,
make them potential multisite targets, which can be inhibited and a cytoplasmic portion with a conserved protein tyro-
by water soluble molecules such as antibodies or binding sine kinase domain (Fig. 1). In addition to the catalytic do-
antagonists, as well as by cell permeant substances such as main, a juxtamembrane region and a carboxyl-terminal tail
kinase inhibitors. can be identified in the cytoplasmic portion. They further
contain regulatory sub-domains which negatively or posi-
tively influence substrate-binding and phosphorylation, as
2. RTK signaling: ligand-binding, dimerization, kinase well as sub-domains involved in the obligatory dimeriza-
activity, substrate phosphorylation, protein docking tion and/or structural changes during kinase activation af-
ter ligand-binding. Each of these structural domains, and
2.1. RTK structure their functions, are potential targets for drug developments
(Fig. 2), and have indeed been addressed in a number of ex-
Growth factors mediate their diverse biologic responses perimental studies (see below). Because of their structure,
(control of cellular proliferation, differentiation, migration RTKs can be visualized as membrane-associated allosteric
and metabolism) by binding to and activating cell-surface enzymes with the ligand-binding and protein tyrosine ki-
receptors with intrinsic protein kinase activity [17,18]. To nase domains separated by the plasma membrane [5,17,21].
date, about 60 RTKs, which belong to ∼16 different re- Their role is to catalyze the transfer of the gamma-phosphate

Fig. 2. Receptor tyrosine kinases as multisite targets. The different steps involved in RTK activation, regulation and synthesis are schematically depicted.
Each step is associated with actual (bold face) or potential (italicized) means of inhibition for cancer therapy.
26 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

of adenosine triphosphate (ATP) to tyrosine residues within neu/ErbB2, enhances its transforming properties [40]. The
their own polypeptide chain and to that of exogenous sub- transmembrane mutation in neu may have a stabilizing ef-
strates. fect on its conformation, which results in dimerization and
On the basis of sequence similarity, it is possible to constitutive activation of receptor signaling [41]. Genetic al-
classify these receptors into related groups [17,18,21]. terations in this domain have, in fact, demonstrated an active
Characteristic structural features of the extracellular do- role of this region in RTK dimerization and demonstrated
mains of these groups include, among others, cysteine-rich that dimerization is not sufficient, but proper alignment must
motifs, immunoglobulin-like repeats, fibronectin type III also occur for activation and signaling [42,43].
repeats, and EGF motifs that can be present singly or in The activation of intrinsic protein kinase activity results in
different combinations. These different domains determine autophosphorylation of specific tyrosine residues in the cyto-
specificity for ligand-binding. It is beyond the scope of this plasmic portion of the RTK. Moreover, tyrosine phosphory-
review to provide detailed information on specific RTKs or lation in the kinase domain stimulates the intrinsic catalytic
RTK families. A number of excellent reviews is published activity of the receptor. Recently, biochemical and structural
each year on these topics (e.g. EGFR-ErbB family [22–24], studies have revealed some of the molecular mechanisms
platelet-derived growth factor receptors (PDGFRs) [25], in- that mediate such activation [44,45]. There is substantial ev-
sulin receptor (IR) family [26,27], FGFRs [28], c-Kit [29], idence that autophosphorylation occurs in trans by a second
Met [30], VEGFRs [28,31], Trks [32], Ret [33,34], ephrin receptor tyrosine kinase after dimerization and/or conforma-
receptors [35], etc.). tional changes induced by ligand-binding. In the unphospho-
There is substantial evidence that ligand-induced activa- rylated state, the receptor possesses a low catalytic activity
tion of the kinase domain and its signaling potential are me- due to the particular conformation of a specific domain in
diated by receptor dimerization [36–38]. This event stabi- the kinase region (the activation loop), which interferes with
lizes interactions between adjacent cytoplasmic domains and the phosphotransfer event. Phosphorylation of the kinase
controls the activation of kinase activity. Receptor oligomer- domain removes this inhibition, and the catalytic activity is
ization appears to be a common phenomenon among growth enhanced and persists for some time independently of the
factor receptors. Dimerization can take place between two presence of the ligand. Although kinase activity is at a low
identical receptors (homodimerization), or between differ- basal level in the monomeric state, this activity is sufficient
ent members of the same receptor family (heterodimeriza- to induce trans-autophosphorylation once the dimer forms.
tion) [17]. Heterodimerization of RTKs has been shown to Autophosphorylation also occurs outside the kinase domain
increase the repertoire of ligands that can be recognized by and serves the important function of creating docking sites
each receptor alone and, on the other hand, to expand the for downstream signal transduction molecules.
diversity of signaling pathways that can be recruited by a The juxtamembrane sequence that separates the trans-
given receptor [17,22,23]. membrane and cytoplasmic domains is not well conserved
How ligands bind to the receptors and induce oligomer- between different families of receptors. However, jux-
ization is specific for each class of RTKs [5,36–38]. PDGF, tamembrane sequences are highly similar among members
for example, induces receptor dimerization by virtue of its of the same family, and some studies indicate that this
dimeric nature [25]. Interestingly, the insulin receptor family domain plays a role in modulation of receptor functions
exists as preformed disulfide-bonded homo- or heterodimers by heterologous stimuli [46]. This region may also play a
of receptor subunits. Thus, in this case, ligand-binding does role in signaling, as has been suggested by the capacity to
not induce receptor dimerization but presumably causes a bind specific substrates in a ligand-dependent manner. For
conformational change in the preformed dimeric receptor, example, it has been shown that eps8 directly binds to the
which leads to receptor activation. The existence of pre- juxtamembrane domain of EGFR in a phosphotyrosine- and
formed, but inactive, receptor dimers has also been evoked SH2-independent manner.
for other RTKs, indicating that ligand-induced conforma- The tyrosine kinase domain is the best conserved among
tional alterations may be the universal mechanism of activa- tyrosine kinase receptors, and an intact protein tyrosine ki-
tion. In any case, it is now well established that dimerization nase domain is absolutely required for receptor signaling.
is necessary for RTK activation. Nevertheless, recent data For example, mutation of a single lysine in the ATP-binding
have shown that receptor dimerization may not be sufficient site, which blocks the ability of the receptor to phosphory-
for activation. This notion stems mostly from experimental late tyrosine residues, completely inactivates receptor bio-
modifications of the transmembrane domain of some RTKs logic function. The kinase domain of some receptor tyrosine
and other related receptors [39]. The main function of the kinases (e.g. PDGF and FGF receptors) is split by inser-
transmembrane domain is to anchor the receptor in the plane tions of up to 100 amino acid residues. Kinase insertion se-
of the plasma membrane, thereby connecting the extracel- quences are highly conserved between species, and contain
lular environment with internal compartments of the cell. It autophosphorylation sites that have the function of coupling
was initially thought that this domain represented a passive with signal-transducing molecules.
anchor of the receptor to the membrane. However, a point The carboxyl-terminal tail sequences are the most diver-
mutation in the transmembrane domain of one receptor, the gent between known RTKs. The carboxy-terminal domain is
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 27

thought to play an important role in regulating kinase activ- from two neighboring monomers. A rather similar configu-
ity. This region typically contains several tyrosine residues, ration has also been found for the ligand-binding region of
which are phosphorylated by the activated kinase. In fact, ErbB3/HER3 [50]. Other examples of resolved extracellular
the receptor itself is often the major tyrosine-phosphorylated domain structures include the neurotrophin-binding domain
species observed following ligand stimulation. The pres- of TrkA and TrkB [51,52], the complex between VEGF and
ence of specific amino acid sequences in this domain plays the second domain of Flt-1 [53], or the complex formed
an important role in determining activation of specific between the receptor EphB2 and ephrin-B2 [54]. As for the
signal-transducing molecules by RTKs. intracellular kinase domain, several structures have been
Although membrane proteins in general are major drug reported, demonstrating a common overall architecture sim-
targets and represent an important class of proteins en- ilar to that of other protein kinases, as had been inferred
coded by about 30% of all genes, little progress has been from the strong sequence homology. Data obtained with
achieved in elucidating their detailed structures at the the inactive and active form of the insulin receptor kinase
atomic level [47]. No complete structure of a RTK has have provided a molecular explanation for the repression of
yet been established. Nevertheless, the modular nature of the catalytic activity in the absence of ligand [55,56]. They
RTKs has allowed the determination of a few extracellular have confirmed the importance of dimerization, and shown
domain and a larger number of kinase domain structure that an activation loop in the kinase domain is blocking
through X-ray crystallography [44] (Fig. 3). Among others, the ATP-binding site in the inactive form. Phosphorylation
the detailed structures of the EGF receptor extracellular of this loop allows for full kinase activity. Role of such
domain complexed with its ligands (TGFalpha or EGF) an activation loop has been confirmed for other RTKs (see
has very recently been published [48,49]. These structures Hubbard and Till [45]). These are extremely important
show that ligand-induced dimerization is achieved by direct progresses, since they will allow for rational design of bind-
receptor–receptor interactions between a loop protruding ing, dimerization, and kinase inhibitors. Strikingly, such

Fig. 3. An overview of selected structures of RTK domains and related inhibitors and signaling proteins. Structures were extracted from the Protein Data
Bank (PDB, http://www.pdb.org/, [153]), and drawn with Rasmol [154]. EGF receptor: (A) extracellular domains I–IV (green ribbon diagram) in complex
with egf (orange spacefill display) (PDB access code: 1IVO [48]); (B) tyrosine kinase domain (blue ribbon) with inhibitor Erlotinib (Tarceva, red spacefill
representation) (PDB 1M17 [155]). ErbB2: (C) extracellular domain of human ErbB2/HER2 (green ribbon) in complex with Herceptin Fab fragment (red
spacefill display); (D) helical membrane spanning fragment (PDB 1IIJ [156]). FGFR: (E) complex of the extracellular domain of FGFR2 (green) with
FGF1 (orange) and heparin (blue spacefill display) (PDB 1E0O [157]); (F) kinase domain of FGFR1 (blue) in complex with SU4984 inhibitor (red) (PDB
1AGW [158]). Adaptor protein and signaling domains (purple ribbons): (G) Grb2 (PDB 1GRI [159]; (H) Pleckstrin homology–phosphotyrosine-binding
(PH–PTB) domain of IRS-1 (PDB 1QQG [160]); (I) SH2 domain of the regulatory subunit of PI3 kinase (PDB 1BFI [161]). Dotted lines denote the
fragments of unknown structure (usually, transmembrane and juxtamembrane domains, as well as distal N- an C-terminus).
28 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

structural data have already been instrumental in the molec- tivated by autocrine stimulation by one of its ligands, most
ular understanding of clinical resistance to Gleevec [16,57]. commonly TGFalpha.
Also, the binding domain of Herceptin on the extracellular
region of HER2 has been solved very recently [58]. 2.3. Signaling pathways of tyrosine kinase receptors

2.2. Aberrations affecting growth factor receptors in tumor Knowledge of the cascade of biochemical events trig-
cells gered by ligand stimulation of tyrosine kinase receptors has
increased rapidly in recent years and provides further evi-
Recent reviews discuss the mechanisms by which RTKs dence of the importance of these signaling pathways in can-
participate in malignant transformation and tumor prolif- cer. In order to effectively coordinate signaling cascades,
eration [18,59,60]. Growth factor receptors can be consti- nature has created a variety of molecules known as adap-
tutively activated by autocrine loops, a number of other tor and scaffolding proteins [61]. These proteins play a role
mechanisms have been identified by which growth factor in intracellular signaling by both recruiting various pro-
receptors can become transforming, among which muta- teins to specific locations and by assembling networks of
tions and overexpression represent prominent anomalies. proteins particular to a cascade. Adaptor proteins, through
Retroviral transduction of proto-oncogenes can result in protein–protein interactions via specific motifs, provide a
mutation of the normal version of the gene. link between molecules of a signaling cascade and proteins
The paradigm for such alterations is v-erbB, the onco- such as RTKs. One such adaptor, Grb2, is important in the
genic counterpart of the EGFR receptor, transduced as the activation of downstream signaling pathways such as the
viral oncogene of avian erythroblastosis virus [61]. The ras/raf/MAPK pathway. These adaptor proteins often con-
mechanism of v-ErbB activation involves deletion of its tain a variety of motifs that mediate protein–protein interac-
ligand-binding domain, resulting in a truncated EGFR. An- tions. Src homology 2 (SH2) and phosphotyrosine-binding
other example of extracellular deletion is the avian v-ros (PTB) domains are protein motifs that bind to specific phos-
oncogene which differs from its cellular counterpart, c-ros, phorylated tyrosine-containing sequences, dictating partic-
in that this domain is deleted with the exception of six ular binding partners. SH3 domains recognize and bind to
amino acids and contains a three amino acid insertion in the proline-rich sequences in target proteins. Thus, as in the case
middle of the transmembrane domain. This results in con- of Grb2 which contains both SH2 and SH3 sequences, an
stitutive activation of the molecule [62]. Neu (the murine adaptor protein can bring a cytoplasmic protein via its SH3
homologue of ErbB2/HER2) was initially identified as an domain to an activated RTK via an SH2 domain-binding to
oncogene by NIH-3T3 transfection analysis of cDNA from phosphorylated tyrosine residues of the receptor.
ethylnitrosourea-induced rat neuroblastomas. The trans- Recently, new signaling pathways have been described
forming gene was identified as having a specific mutation for the RTKs, in addition to the classical plasma membrane
in its transmembrane domain responsible for oncogenic ac- effectors which activate the ras/MAPK and PI3kinase/akt
tivation, again responsible for constitutive activation of the pathways [65] (schematized in Fig. 4). Notably, EGFR has
kinase [40]. been shown to stimulate directly phosphorylation and nu-
In human malignancies, overexpression of a normal re- clear translocation of signal transducer and activator of tran-
ceptor, with or without the concomitant presence of the scription (STATs) proteins. RTKs or their proteolytic frag-
ligand, contributes to neoplastic transformation. Examples ments may also be active inside cells, and surprisingly in
include the EGFR, ErbB2, the insulin-like growth factor 1 the nucleus where they could act directly as transcription
receptor, etc. The best characterized mechanisms involve factors.
EGFR family members [38,63,64]. ErbB2/HER2 was ini- Again, it is beyond the scope of this review to discuss in
tially identified as an amplified gene in a primary human detail the diverse signaling cascades of RTKs, the reader is
breast carcinoma and a salivary gland tumor. ErbB2 overex- referred to recent reviews [17,60,66–70].
pression beyond some critical threshold level in NIH/3T3 fi-
broblasts was shown to be sufficient to induce the malignant 2.4. Regulation of RTK activity
phenotype. Clinical studies have indicated that the normal
erbB2 gene is frequently amplified and/or overexpressed in Because of the critical roles played by RTKs in cellu-
human breast carcinomas and in ovarian carcinomas, and lar signaling processes, their catalytic activity has to be
detection in breast carcinomas of high ErbB2 levels is a normally under tight control. This is achieved by a vari-
prognostic indicator of poor survival. Thus, ErbB2 appears ety of means, which can be briefly summarized as follows:
to be most commonly altered in human malignancies by a first level of regulation is constituted by the kinase do-
mechanisms leading to its overexpression. Whereas ErbB2 main itself where the state of phosphorylation controls di-
overexpression has been observed primarily in adenocarci- rectly kinase activity [71]. Phosphorylation of tyrosines on
nomas, overexpression of an apparently normal EGFR has the receptor will also dictate the binding of SH2 and PTB
been reported frequently in squamous cell carcinomas and domains-containing substrates and adapters. Obviously, the
glioblastomas. In many cases, the EGFR appears to be ac- phosphorylation state of a RTK will also be controlled in
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 29

Fig. 4. A very schematic view of RTK signaling. The main pathways are indicated, with kinase names italicized. Elements of these pathways for
which inhibitors are under development are also underlined. Abbreviations (pathways from left to right): PI3K, phosphatidylinositol 3-kinase; Pdk1,
phosphoinositide-dependent protein kinase-1; Akt, oncogenic kinase initially isolated from a transforming mouse retrovirus (also named PKB, or related to
A and C protein kinase: RAC-PK); p70S6K , ribosomal S6 kinase; Shc, (src homology collagen); Sos, (son of sevenless); Grb2, (growth factor receptor-bound
protein 2) are adaptor proteins; Ras, oncogene first isolated in rat sarcomas; Raf, oncogenic kinase initially isolated from a transforming mouse virus;
Mek, Map/Erk kinase (or Mkk: map kinase kinase); Erk/MapK, extracellular signal-regulated kinase/mitogen-activated protein kinase; Jak, janus kinase;
Stat, signal transducer and activator of transcription; PLC, phospholipase C; PKC, protein kinase C; Src, oncogene of the chicken Rous sarcoma
virus.

a specific manner by the activity of protein tyrosine phos- cancer, especially because they do not allow for the study
phatases, a number of which have recently been identified of interactions with the tumor host, their use is certainly
[72,73]. still justified [78]. More sophisticated models can also be
Another important means of regulation in cells is developed from genetically engineered cells. For instance,
“down-regulation” whereby ligand-binding leads to disap- Brandt et al. [79] have described an elegant model of breast
pearance of receptors from cell surface. This phenomenon cancer in the mouse, by injecting mammary fat pads with
is due to accelerated endocytosis of ligand–receptor com- cells expressing an oncogenic ErbB2 receptor.
plexes, and has been extensively studied for the ErbB/HER Laboratory mice have also been a central player in cancer
receptors [74]. Endocytosis occurs through clathrin- research. The techniques of gene targeting and transgenesis
dependent and independent mechanisms, although the pre- have allowed the creation and study of many transgenic
cise nature of clathrin-independent pathways remains un- and knock-out strains. More recently, genetic systems al-
clear. Quite recently a role for ubiquitinylation of receptors lowing for precise conditional control of the expression
has been evidenced [75–77]. of specific genes have been introduced [80]. The benefits
All these findings may yield new developments in drugs and disadvantages of these animal models have recently
aiming at attenuating RTK signaling, for instance by acti- been discussed [81–84]. As far as RTKs are concerned,
vating specific phosphatases, or accelerating receptor down- numerous transgenic mice have been created (see Table 1
regulation, or preventing receptor nuclear translocation. for some selected examples). The first such transgenic mice
carried an activated neu/erbB2 oncogene driven by a mouse
mammary tumor virus (MMTV) promoter, and developed
3. RTKs and cancer: experimental tools mammary adenocarcinomas [85]. Besides providing direct
evidence supporting the role of RTKs and their ligands in
Historically, discovery and preclinical development of tumorigenesis, these models have allowed and will allow for
anti-cancer drugs have mostly relied on empiric cell-based increasing our understanding of the development of tumors
screening for inhibition of proliferation. Together with the in a whole organism, and for in vivo preclinical drug testing
use of xenografts of human cancers in immunologically in a more sophisticated setting than cultured cells [86].
deficient mice, this approach has yielded much information High-throughput discovery tools are always desirable in
and allowed the discovery of many useful therapeutics. Re- the quest for effective new drugs. Peptide chips represent
cently, targeting specific molecules such as oncogenes has a very recent development in the search for performant as-
provided a more mechanistically-based approach. Many says to discover kinase inhibitors. Houseman et al. [103]
cell lines are available that express mutated RTKs, or over- have chemically immobilized a short peptide substrate of
express normal receptors, or such cell lines can be easily the tyrosine kinase src on gold surfaces. Phosphorylation of
constructed by transfection. Although these cell lines can- the peptide in the presence of the enzyme can be quanti-
not replicate the full spectrum of characteristics of any tatively analyzed by plasmon resonance, fluorescence and
30 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

Table 1
Selected examples of transgenic mouse overexpressing one RTK as cancer models
Type of cancer Oncogenic RTK or ligand Method Phenotype Reference

Breast Transforming c-neu MMTV promoter Synchronous early mammary [85]


adeno-carcinoma + glandular
dysplasias
Breast Transforming c-neu MMTV promoter Stochastic late mammary [87]
adenocarcinoma + glandular
dysplasias
Breast ErbB2/neu + TGFalpha MMTV promoter Progressive mammary tumors [88,89]
Breast ErbB2 Neu promoter [90]
Breast ErbB2/neu MMTV promoter Late focal mammary tumors [91]
+ lung metastasis
Breast Transforming c-neu MMTV promoter Synchronous mammary [92]
adenocarcinoma in each gland
+ glandular dysplasias
Breast ErbB2/neu MMTV promoter Late unifocal mammary tumors [93]
Breast Mutated met Metallothionein promoter Metastatic mammary carcinoma [94]
Thyroid RET/PTC chimera Thryroglobulin promoter Papillary carcinomas [95]
Thyroid RET with MEN2 mutation Calcitonin promoter Medullary carinoma [96]
Angiogenesis VEGF Keratin promoter Skin papilloma [97]
Diverse RET-Men2B Dopamine hydroxylase Ganglioneuromas + renal [98]
promoter malformations
Skin and other epithelia Activated erbB2 Keratin promoter Epithelial hyperplasia, [99]
+ tetracyclin-induction up-regulation of TGFalpha
Skin ErbB2 Keratin promoter [100]
Brain tumor Transforming c-neu MBP promoter Low incidence of brain tumors [101]
Pituitary adenoma Truncated FGFR4 Prolactin promoter Lactotroph pituitary tumors [102]

phosphorimaging. Such a system can easily be used for Suramin was introduced in 1920 for the treatment of try-
high-throughput assays monitoring the activity of protein ki- panosomiasis, and was later found to be a potent inhibitor
nases, and should be instrumental in the discovery of new of the reverse transcriptase of retroviruses, including the
kinase inhibitors. HTLV-III. It was also found to possess anti-neoplasic prop-
erties, notably by inhibiting angiogenesis [104]. Suramin
does so by inhibiting the action of aFGF and bFGF at the re-
4. RTK as targets in clinical research: what is ceptor level. Recent clinical studies have shown little bene-
going on? fits in prostate cancer, with neurotoxicity being a significant
complication [105].
In this section, we aim at presenting an necessarily Another way to disrupt the interaction of key growth
non-exhaustive overview of current development of drugs factors with their RTK targets in the cell membrane has
targeting RTKs. As schematized in Fig. 2, RTKs can be been developed by Sebti and Hamilton [106]. They used
viewed as multisite targets, and we chose to follow the same an entirely novel strategy based on the design of synthetic
logical order of presentation, from the extracellular-binding agents which contain a large and functionalized surface
site to the intracellular events. Gene therapies will not be area that selectively and strongly bind to the complemen-
discussed here, as they are still in their prime infancy for tary surface of the growth factor and so block its oncogenic
RTKs. Table 2 presents a short overview of some drugs cur- signaling function. Sebti and Hamilton [106] have placed
rently in clinical trials, belonging to the two largest groups, particular focus on platelet-derived growth factor and its
namely antibodies and small kinase inhibitors. complementary RTK, platelet-derived growth factor recep-
tor because its overexpression is seen in many carcinomas
4.1. Antagonists of ligand-binding and some cancer patients have high serum levels of PDGF.
This approach to growth factor binding agents involves the
As ligand-binding is the first step in RTK activation, tar- attachment of several peptide loops onto a core scaffold in
geting the ligands themselves, or the extracellular-binding direct analogy to the six hypervariable loops that are found
site on the receptor, represent a first straightforward ap- in the antigen recognition region of an antibody Fab frag-
proach. The relatively large size of peptidic growth factors ment [106], The synthesis of a library of binding agents has
represents a major obstacle in the development of small an- permitted to identify a molecule, GFB-111 that targets the
tagonists. Nevertheless, quite a few RTK antagonists have regions of PDGF (loops I and III) that are involved in bind-
been developed. ing to its receptor. By preventing PDGF from binding to its
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 31

Table 2
Some examples of drugs targeting RTKs (monoclonal antibody (MAb))
RTK Drug Company Description Status Indication

HER2 Trastuzumab/Herceptin Genentech MAb directed against Approved Approved for breast
HER2 Phases I cancer; other solid
and II tumours
EGFR/HER2 PKI 166 Novartis Inhibitor of EGFR and Phase I Prostate cancer,
HER2 renal cell carcinoma
EGFR OSI-774 (Tarceva) Roche/Genentech/OSI Inhibitor of EGFR Phase III NSCLC, head and
neck, ovarian cancer
EGFR C225/cetuximab ImClone Systems MAb directed against Phase III Colorectal, head
EGFR and neck, pancreas
cancers
EGFR ABX-EGF Abgenix MAb against EGFR Phase II NSCLC
EGFR ZD1839 (Iressa) AstraZeneca Inhibitor of EGFR Phase II Approved in Japan
signalling for NSCLC many
cancers
Abl/PDGFR/c-Kit STI-571 (Gleevec) Novartis Inhibitor of PDGFR, Approved Approved by the
c-Kit (and abl) Phase IV FDA for GIST (and
CML) glioma,
sarcoma
VEGFR SU5416 SUGEN Inhibitor of VEGFR-2 Phase III Angiogenesis
VEGFR/FGFR/PDGFR SU6668 SUGEN Inhibitor Phase I Angiogenesis
Trk CEP2563 Cephalon Inhibitor of Trk Phase II Ovarian, prostate,
pancreas cancers

RTK, GFB-111 blocks PDGF-induced receptor autophos- phosphorylation of ErbB2 [110]. Recently, it has been pro-
phorylation, activation of Erk1 and Erk2 kinases, and DNA posed that the anti-tumor effects of trastuzumab can be ex-
synthesis. This molecule is highly potent (IC50 = 250 nM) plained by inhibition of HER2 cleavage and prevention of
and selective for PDGF over EGF, IGF-1, aFGF, and bFGF the production of an active truncated HER2 fragment [111].
(IC50 values > 100 ␮M) but inhibits also VEGF-induced As for ErbB2/HER2, there is a relationship between over-
Flk-1 tyrosine phosphorylation and Erk1/Erk2 activation expression of epidermal growth factor receptor and aggres-
with an IC50 of 10 ␮M. Furthermore, GFB-111 treatment sive behavior of tumor cells. The anti-EGFR monoclonal
of nude mice bearing human tumors resulted in significant antibody C225 (cetuximab) was generated and has been
inhibition of tumor growth and angiogenesis. Thus, these shown to have anti-tumor activity in vitro and in xenograft
results demonstrate that selective PDGF-binding molecules models [112]. This antibody has been chimerized with
such as GFB-111 that are capable of blocking signaling, human IgG1 in its constant region to increase its clinical
tumor growth, and angiogenesis may lead to the develop- utility by decreasing the potential for generation of human
ment of a new class of anti-cancer drugs capable of treating anti-mouse in recipients [113]. In fact, the anti-tumor ac-
a wide spectrum of human cancers [106,107]. tivity of cetuximab has been attributed to several distinct
mechanisms: a therapeutic strategy combining C225 and
4.2. Antibodies to RTKs chemotherapeutic agents such as doxorubicin or cisplatin
showed that anti-EGFR MAbs substantially enhance the ef-
Today, more than 70 monoclonal antibodies (MAb) are fects of these agents against well-established xenografts of
currently in commercial trials beyond Phases I and II. These tumor cells expressing high levels of EGF receptors. Com-
include reagents against cancer, transplant rejection, a vari- bination of chemotherapeutic agents and C225 may block
ety of immunological conditions such as Crohn’s disease, activation of receptor tyrosine kinase and induce EGFR
antiviral prophylaxis, and thrombosis [108]. down-regulation [114]. Another mechanism of action is that
Herceptin (trastuzumab, Genentech) is the first commer- both cetuximab and M225, its murine progenitor, inhibit
cially available antibody targeting a RTK. Herceptin is a hu- cell-cycle progression in many cell lines, causing cells to
manized monoclonal antibody, so called because all the orig- arrest in the G1 gap phase that occurs prior to DNA syn-
inal mouse-derived components except the antigen-binding thesis. A series of experiments has shown that anti-EGFR
region have been replaced by human counterparts. Several antibody treatment causes an increase in the expression of
mechanisms have been defined to explain its anti-tumor ef- the cell-cycle inhibitor p27kip1 . This in turn results in an
fects: trastuzumab down-regulates HER2 expression by ac- increase in the formation of inhibitory p27kip1 –Cdk2 com-
celerating receptor endocytosis and degradation [109], act- plexes which prevent cells from exiting the G1 phase of the
ing as a partial agonist of the ligand of this receptor. It can cell cycle [115]. Many data show that cetuximab has also
also inhibit cell-cycle progression preceded by a rapid de- anti-angiogenic properties, which were first reported by Petit
32 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

et al. [116]. This group found that established A431 tumor [124]. Although small molecules and enzymes are most pop-
xenografts treated with cetuximab displayed a significant ular among drug designers, the idea of developing specific
decrease in the production of angiogenic factors, and these inhibitors of oncogenic RTKs was initially met with skep-
data have since been confirmed with additional tumor cell ticism, mostly for fear of poor specificity. This was due to
lines. Cetuximab has also been shown to influence apop- the nature of most inhibitors, which are competitive for the
tosis. Indeed, in DiFi human colon carcinoma cells which well conserved ATP-binding site in the kinase domain. Nev-
express high level of EGFR, cetuximab treatment-induced ertheless, as exemplified by Gleevec and Iressa, this class
programmed cell death by activation of multiple apoptotic of molecules may prove to be much more useful than was
caspases [117]. A Phase Ib study of C225 in combination earlier expected (see [11]).
with cisplatin in patients with recurrent squamous cell car- STI-571 (GleevecTM ) is a protein-tyrosine kinase in-
cinoma of the head and neck has been conducted in order hibitor that inhibits the Bcr-Abl tyrosine kinase, the constitu-
to determine the optimal biological dose of C225 and to tive abnormal tyrosine kinase created by Philadelphia chro-
establish a safety profile of C225 in a different range of mosome abnormality in chronic myeloid leukemia (CML)
dose levels in combination with cisplatin [118]. [125]. STI-571 inhibits proliferation and induces apoptosis
ABX-EGF (Abgenix), a fully human IgG2 monoclonal in Bcr-Abl positive cell lines as well as in fresh leukemic
antibody specific for human EGFR has been generated. cells from Philadelphia chromosome-positive CML. In fact,
ABX-EGF binds EGFR with high affinity (5 × 10−11 M), STI-571, first known as CGP 57148, was initially described
blocks the binding of both EGF and TGFalpha to var- as an inhibitor of both Abl and platelet-derived growth fac-
ious EGFR-expressing human carcinoma cell lines, and tor receptor [126]. It inhibits at nanomolar concentrations
inhibits tumor cell activation and cell proliferation. In vivo, the kinase activity of v-abl, bcr-abl, c-Kit and PDGFR, and
ABX-EGF prevents completely the formation of human this was found to translate into both in vivo anti-tumor ac-
epidermoı̈d carcinoma A431 xenografts in athymic mice, tivity [127]. Small cell lung cancer (SCLC) is an aggressive
and administration of ABX-EGF without concomitant cancer characterized by deregulation of several autocrine
chemotherapy results in complete eradication of established growth mechanisms including stem cell factor and its re-
tumors. Human pancreatic, renal, breast and prostate tu- ceptor c-Kit. It has been shown that STI-571 also inhibits
mor xenografts which express more than 17,000 EGFR growth of SCLC cells through a mechanism that involves
molecules per cell showed significant growth inhibition inactivation of the tyrosine kinase c-Kit. The effectiveness
when treated with ABX-EGF. Thus, Yang et al. [119] have of STI-571 in this study suggests this drug may be use-
demonstrated a potent anti-tumor activity of ABX-EGF and ful for patients with SCLC [128]. STI-571 has also been
its therapeutic potential for the treatment of multiple human approved for treatment of gastro-intestinal stromal cancer
solid tumors that overexpress EGFR. [129,130].
A very original approach, exocyclic mimetics of antibod- Anilinoquinazolines are potent and selective inhibitors
ies, has been recently proposed [120]. A short peptide (called of the ErbB family of receptor tyrosine kinases, and three
AHNP, 12 aminoacids) was rationally designed to mimic the analogues (two reversible and one irreversible inhibitor)
structure of one loop of Herceptin, and was found to mimic have been evaluated clinically as anti-cancer drugs [131].
many effects of the native antibody. Analogues of this pep- Among these quinazoline-derived agents that have been
tide with better activity have been developed [121]. Such tested as anti-cancer drugs in vitro and in preclinical mod-
short and stable antibody analogs may offer potentially sig- els, ZD1839 (Gefitinib, Iressa, AstraZeneca) is a highly
nificant therapeutic advantages. specific EGF receptor tyrosine kinase inhibitor. Inhibition
In general, antibodies have started to fulfill their promise of EGFR transphosphorylation by ZD 1839 blocks signal
as anti-cancer therapeutics with four antibodies now mar- transduction at its first step, thus providing anti-proliferative
keted in the United States: Rituxan (IDEC Pharmaceuticals, effects on several human cancer cell lines overexpressing
Genentech) used against non-Hodgkin’s lymphoma, Her- EGFR, including ovarian, breast and colon cancer [132].
ceptin, Mylotarg (Wyeth Laboratories) used against acute Preclinical studies demonstrated efficacy and good bioavail-
myelogenous leukemia, and Campath (Alemtuzumab, Mil- ability. In Phase I studies, toxicity was manageable. More
lenium and ILEX Partners) used against B-cell chronic lym- Phases II and III studies are ongoing, and Iressa has already
phocytic leukemia, and other MAbs are now in advanced been approved for treatment of NSCLC in Japan.
oncology trials [122]. Vascular endothelial growth factor, fibroblast growth fac-
It should also be stressed that RTKs being membrane tor and platelet-derived growth factor and their cognate re-
proteins, they represent also a target for combination therapy ceptor tyrosine kinases are strongly implicated in angiogen-
coupling toxic drugs to ligands or antibodies [123]. esis associated with solid tumors; the sustained growth of
solid tumors is dependent on angiogenesis, the growth of
4.3. Tyrosine kinase inhibitors new blood vessels from existing host vasculature [133]. Us-
ing rational drug design coupled with traditional screening
The design of small molecule inhibitors of kinases in gen- technologies, a novel inhibitor of these receptors has been
eral, and RTKs in particular, seems now to be very promising discovered: SU6668 (Sugen/Pharmacia). SU6668 is a small
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 33

molecule synthetic kinase inhibitor of the tyrosine activity of from Sos linked by a lysine spacer was designed using struc-
Flk-1/KDR or VEGF receptor, PDGFR, an FGFR. Indeed, tural data from Grb2 and a proline-rich peptide complexed
in several cellular systems, it inhibits tyrosine phosphoryla- with its SH3 domains. This proline-rich “peptidimer” which
tion and mitogenesis after stimulation of cells with appro- has two VPPPVPPRRR sequences, which specifically rec-
priate ligands. Administration of SU6668 in athymic mice ognize Grb2, blocks selectively Grb2–Sos complexation in
resulted in significant growth inhibition of a diverse panel of ER22 (CCL39 fibroblasts overexpressing EGFR) cellular
human tumor xenografts of glioma, melanoma, lung, colon, extracts. Moreover, this “peptidimer” was modified to en-
ovarian and epidermoid origin. The attractive and validated ter by coupling it to penetratin, a peptide consisting of 16
targets of SU6668, coupled with its broad activity in tumor aminoacids issued from the Antennapedia homeodomain.
xenograft models have motivated its entry into clinical de- The results showed that at 10 ␮M, the conjugate inhibits the
velopment. Accordingly, SU6668 has recently entered Phase Grb2–Sos interaction (100%) and MAPK phosphorylation
I clinical trials, and its safety and efficacy profile in humans (ERK1 and ERK2) was largely decreased (60%) in ER22
will emerge in the near future [134]. without modifying cellular growth. When tested for its
Other small molecule inhibitors targeted against tyrosine anti-proliferative effect, the conjugate was an efficient in-
kinase are currently in clinical trials [1]. hibitor of colony formation of transformed NIH 3T3/HER2
cells grown in soft agar, with an IC50 of 1 ␮M. Thus, as
4.4. Protein–protein interaction inhibitors these “peptidimers” do not seem to develop toxicity in sys-
tem which do not overexpress the Ras signaling pathway,
In living cells, many regulated processes are initiated their design appear to be interesting leads to investigate
or inhibited through specific protein–protein complex for- signaling and intracellular processes and for designing se-
mation. That is why peptides and small molecules that lective inhibitors of tumorigenic Ras-dependent processes
modify the overall quaternary structure of a selection of [137,139].
receptor–ligand interactions and oligomeric viral enzymes Concerning the possibility of inhibiting Grb2 at the
have been developed recently [135]. This part presents sev- level of its SH2 domain, this approach was expected to
eral approaches by which RTK can be inhibited by peptide be easier because small phosphopeptides were reported to
and peptidominetics which interfere with protein–protein have affinities for SH2 domains in the 10−8 to 10−7 M
interactions. range. However, these peptides are negatively charged and
do not enter cells easily; moreover, cellular phosphatases
4.4.1. Inhibitors of signaling degrade the phosphotyrosine group which is necessary
The small adaptor protein Grb2 is essential in the Ras for phosphopeptide activity. To design peptidomimetic
signaling pathway. It is made up of one SH2 domain of inhibitors, the three-dimensional structure of the Grb2
approximately 100 aminoacids surrounded by two SH3 do- SH2 domain complexed with a phosphopeptide has been
mains, each containing about 60 aminoacids. The SH2 do- used. Then, a series of small peptides with the sequence
main bind to target proteins at the level of phosphotyrosine mAZ-pTyr-Xaa-Asn-NH2, where Xaa denotes methylphos-
motifs, whereas the SH3 domains bind to proline-rich mo- photyrosine or its carboxylic mimetics, were synthesized as
tifs of these target proteins [136]. The binding of growth inhibitors of the Grb2 SH2 domains. Peptide with (Me)pTyr
factor on its receptor triggers dimerization and phosphoryla- as Xaa has the highest affinity for Grb2 (Kd = 3 ± 1 nM)
tion on its carboxyl-terminal tyrosine residues. This allows and exhibits to date the best inhibitory activity (IC50 =
recruitment under the membrane near the Ras protein of 11 ± 1 nM) to displace PSpYVNVQN–Grb2 interaction in
the Grb2–Sos complex. Sos, the exchange factor of Ras, is an ELISA test [137,140]. These compound which showed
thus able to activate Ras under its GTP form. Then, Ras can the highest affinity for Grb2 was chosen in order to prepare
recruit Raf and activate the MAPK cascade which induce a prodrug in which the phosphate groups were protected
cell division and/or differentiation [137]. As overexpression with S-acetyl thioethyl ester (SATE). The prodrugs obtained
of ErbB receptors induce a downstream signaling deregula- are very hydrophobic and can easily enter cells, where they
tion, and particularly of Ras signaling pathway [138], Grb2 are degraded by esterases, and the chemically unstable in-
constitutes a target for the design of therapeutic agents as termediates liberate the active groups which can inhibit the
anti-tumor agents. Two possible ways of inhibiting the Ras growth of NIH3T3 cells transfected by the oncogene HER2
signaling pathway at the level of Grb2 have been described in colonies on soft agar [137].
by Garbay et al. [137]: they first describe a strategy to in-
hibit Grb2–Sos interaction at the level of its SH3 domains 4.4.2. Inhibitors of dimerization
and then report the results obtained in blocking Grb2 SH2 Reports have shown that short peptides (10–20 amino
domain interaction with EGFR or Shc. acids) can compete with target proteins [141]. In the context
Concerning the interruption of the growth factor- of cancer, a few works have shown that small peptidomimet-
stimulated Ras signaling pathway at the level of the ics can be used to inhibit dimerization of RTKs which are
Grb2–Sos interaction by inhibition of Grb2 SH3 domains, implicated by a mutation or overexpression in this disease.
a “peptidimer” made of two identical proline-rich sequence In fact, these peptides act as antagonists of RTKs.
34 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

Appropriate ligand-induced dimerization of RTKs implies respect, relative lack of specificity of some effective drugs
the participation of different interaction loops on the recep- may turn out to be one more advantage for this class of
tor surface. One such dimerization interface has recently new molecules. Indeed, although initially developed as an
been characterized for the EGF receptor (see above, and inhibitor of the abl tyrosine kinase in CML, STI-571 has al-
[142]). Berezov et al. [143] have designed a series of 12–30 ready proved as an effective inhibitor of the mutated c-kit
amino acids long peptides derived from potential such sur- RTK in GIST [148], and has been approved for this indica-
faces in ErbB/HER receptors [143]. These peptides are ef- tion. Of course, such a poor specificity is much less desir-
fective binders of ErbB receptor ectodomains, inhibit ErbB2 able in purely experimental settings, where kinase inhibitors
dimerization and growth of cells overexpressing these recep- are very often used in cell-based assays for the dissection of
tors. Such small peptides may be developed as therapeutic signaling pathways [149].
agents. One possible drawback for strategies aiming at RTKs in-
Another example is given by the activation of the rat hibition is the necessity for molecular characterization of
neu/erbB2 oncogene by a point mutation within its trans- individual tumors in order to ascertain the target status. This
membrane domain that results in the substitution of glu- holds true for any drug aiming at specific targets deregu-
tamic acid for valine at position 664, and is associated with lated in cancer, whether be it a RTK, or any component
constitutive activation of the tyrosine kinase [40]. In order of the ras/MAP kinase pathway, or an anti-oncogene. Nev-
to investigate the role of the alpha-helical transmembrane ertheless, this is not a trivial issue, as the experience with
sequence in the function of neu, Lofts et al. [144] have con- Herceptin has already proved that only those patients who
structed an expression vector to produce a variety of short show overexpression of HER2/ErbB2 would benefit from
transmembrane neu proteins, lacking ligand-binding or in- treatment. However, it is not yet clear whether responsive
tracellular kinase domain. Such sequences should interact patients can be identified by target expression levels, as
with full-length receptors and prevent receptor dimeriza- is the case with Herceptin, which is approved for use in
tion. They showed that the short transmembrane molecules ErbB2-positive cancer and requires the use of an assay to
are expressed at the cell surface and can retard the growth measure expression of this receptor for patient selection.
of neu-transformed cells in monolayers, as colonies in soft Standardization of reproducible procedures to evaluate the
agar and as tumors in animals. We have recently extended precise status of the tumor of individual patients will most
this work and shown that short hydrophobic peptides are probably benefit from the ongoing progresses in gene anal-
able to inhibit both EGFR and ErbB2/HER2 kinases in cul- ysis techniques such as microarrays, as well as more classi-
tured human cancer cells (Bennasroune et al., manuscript cal immuno-cytological methods. On the other hand, there
submitted). In an another work, Qian et al. [145] have is a possibility that inhibition of an oncogenic pathway at
shown that the transforming activity of oncogenic p185neu a level not obviously altered may be beneficial. For in-
(also termed Tneu) can be inhibited in vitro and in vivo stance, inhibition of the met RTK is effective in a cellular
by co-expression of a truncated neu protein with a large system with a ras mutation [150]. In reverse, the use of a
cytoplasmic deletion which includes the tyrosine kinase farnesyl transferase inhibitor, targeting oncogenic ras, does
domain. Indeed, in cell lines co-expressing full-length and inhibit proliferation of an ErbB2 overexpressing cell line
truncated neu proteins, they observed co-dimerization be- [151].
tween full-length p185neu and truncated peptides resulting Another serious problem has very recently emerged when
in the formation of a kinase-inactive heteromeric complex the use of Gleevec and Herceptin has been extended to more
[145]. These peptides may be important for the design patients. Resistance to these molecules has been observed,
of future therapies directed against ErbB family oncopro- as with any anti-cancer drug. The extent of the problem has
teins. yet to be assessed, and it is most probably a witness of the
enormous genetic instability of cancerous cells. Indeed, a
point mutation in the kinase domain of bcr-abl has been
5. Conclusion and future directions demonstrated in leukemic patients who became resistant to
Gleevec [16,152].
One should not overlook the possibility for RTK-targeting Nevertheless, the various implications of RTKs through
drugs to potentially target more than one RTK (as exempli- mutations and overexpression in cancer, together with the
fied by Gleevec, and other molecules in development), and ever increasing knowledge about their structure, mechanism
thus more than one type of cancer, and even more than one of activation and regulation, allows for reasonable hopes in
disease [146]. The position of RTKs at the very heart of future developments of efficient treatments targeting these
many of the cellular processes that are deregulated in can- oncogenes.
cer makes them even more attractive since inhibition of one
RTK pathway may yield effects on more than simply cell
Reviewers (MA 496)
proliferation. For instance, Herceptin has very recently been
shown in a mouse model of breast cancer to exert also its Prof. Jacques Robert, Institute Bergonié, 229, Cours de
beneficial effects by inhibiting angiogenesis [147]. In this l’Argonne, F-33076 Bordeaux Cedex, France.
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 35

Prof. Christiane Garbay, Laboratoire de Pharmacochimie [25] Heldin CH, Ostman A, Ronnstrand L. Signal transduction via
Moléculaire et Structurale, UFR des Sciences Pharmaceu- platelet-derived growth factor receptors. Biochim Biophys Acta
1998;1378:F79–F113.
tiques & Biologiques, 4 av. de l’Observatoire, F-75270 Paris [26] Marino-Buslje C, Martin-Martinez M, Mizuguchi K, Siddle K,
Cedex 06, France. Blundell TL. The insulin receptor: from protein sequence to struc-
ture. Biochem Soc Trans 1999;27:715–26.
[27] De Meyts P, Whittaker J. Structural biology of insulin and IGF1
receptors: implications for drug design. Nat Rev Drug Discov
References 2002;1:769–83.
[28] Gerwins P, Skoldenberg E, Claesson-Welsh L. Function of fibroblast
growth factors and vascular endothelial growth factors and their
[1] Sawyers CL. Rational therapeutic intervention in cancer: kinases as receptors in angiogenesis. Crit Rev Oncol Hematol 2000;34:185–94.
drug targets. Curr Opin Genet Dev 2002;12:111–5. [29] Taylor ML, Metcalfe DD. Kit signal transduction. Hematol Oncol
[2] Busch H. Onc genes and other new targets for cancer chemotherapy. Clin North Am 2000;14:517–35.
J Cancer Res Clin Oncol 1984;107:1–14. [30] Longati P, Comoglio PM, Bardelli A. Receptor tyrosine kinases as
[3] Levitzki A. Signal-transduction therapy. A novel approach to disease therapeutic targets: the model of the MET oncogene. Curr Drug
management. Eur J Biochem 1994;226:1–13. Targets 2001;2:41–55.
[4] Huber BE. Therapeutic opportunities involving cellular oncogenes: [31] Matsumoto T, Claesson-Welsh L. VEGF receptor signal transduc-
novel approaches fostered by biotechnology. FASEB J 1989;3:5–13. tion. Sci STKE 2001;2001:RE-21.
[5] Ullrich A, Schlessinger J. Signal transduction by receptors with [32] Nakagawara A. Trk receptor tyrosine kinases: a bridge between
tyrosine kinase activity. Cell 1990;61:203–12. cancer and neural development. Cancer Lett 2001;169:107–14.
[6] Yarden Y, Ullrich A. Growth factor receptor tyrosine kinases. Annu [33] Santoro M, Melillo RM, Carlomagno F, Fusco A, Vecchio G.
Rev Biochem 1988;57:443–78. Molecular mechanisms of RET activation in human cancer. Ann N
[7] Cantley LC, Auger KR, Carpenter C, et al. Oncogenes and signal Y Acad Sci 2002;963:116–21.
transduction. Cell 1991;64:281–302. [34] Manie S, Santoro M, Fusco A, Billaud M. The RET receptor: func-
[8] Walsh TP, Grant GH. Computer modelling of the receptor-binding tion in development and dysfunction in congenital malformation.
domains of VEGF and PIGF. Protein Eng 1997;10:389–98. Trends Genet 2001;17:580–9.
[9] Workman P, Kaye SB. Translating basic cancer research into new [35] Nakamoto M, Bergemann AD. Diverse roles for the Eph family
cancer therapeutics. Trends Mol Med 2002;8(4):S1–9. of receptor tyrosine kinases in carcinogenesis. Microsc Res Tech
[10] Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2002;59:58–67.
2000;100:57–70. [36] Heldin CH. Dimerization of cell surface receptors in signal trans-
[11] Cohen P. Protein kinases—the major drug targets of the twenty-first duction. Cell 1995;80:213–23.
century. Nat Rev Drug Discov 2002;1:309–15. [37] Weiss A, Schlessinger J. Switching signals on or off by receptor
[12] Varmus H, Bishop JM. Biochemical mechanisms of oncogene ac- dimerization. Cell 1998;94:277–80.
tivity: proteins encoded by oncogenes. Introduction. Cancer Surv [38] Brennan PJ, Kumagai T, Berezov A, Murali R, Greene MI, Ku-
1986;5:153–8. mogai T. HER2/neu: mechanisms of dimerization/oligomerization.
[13] Downward J, Yarden Y, Mayes E, et al. Close similarity of epidermal Oncogene 2000;19:6093–101.
growth factor receptor and v-erb-B oncogene protein sequences. [39] Jiang G, Hunter T. Receptor signaling: when dimerization is not
Nature 1984;307:521–7. enough. Curr Biol 1999;9:R568–71.
[14] Ullrich A, Coussens L, Hayflick JS, et al. Human epidermal [40] Bargmann CI, Weinberg RA. Oncogenic activation of the
growth factor receptor cDNA sequence and aberrant expression of neu-encoded receptor protein by point mutation and deletion. EMBO
the amplified gene in A431 epidermoid carcinoma cells. Nature J 1988;7:2043–52.
1984;309:418–25. [41] Weiner DB, Liu J, Cohen JA, Williams WV, Greene MI. A point
[15] Slamon DJ, Godolphin W, Jones LA. Studies of the HER-2/neu mutation in the neu oncogene mimics ligand induction of receptor
proto-oncogene in human breast and ovarian cancer. Science aggregation. Nature 1989;339:230–1.
1989;244:707–12. [42] Burke CL, Lemmon MA, Coren BA, Engelman DM, Stern DF.
[16] Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance Dimerization of the p185(neu) transmembrane domain is necessary
to STI-571 cancer therapy caused by BCR-ABL gene mutation or but not sufficient for transformation. Oncogene 1997;14:687–96.
amplification. Science 2001;293:876–80. [43] Bell CA, Tynan JA, Hart KC, Meyer AN, Robertson SC, Donoghue
[17] Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell DJ. Rotational coupling of the transmembrane and kinase domains
2000;103:211–25. of the Neu receptor tyrosine kinase. Mol Biol Cell 2000;11:3589–
[18] Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature 99.
2001;411:355–65. [44] Hubbard SR. Structural analysis of receptor tyrosine kinases. Prog
[19] Robinson DR, Wu YM, Lin SF. The protein tyrosine kinase family Biophys Mol Biol 1999;71:343–58.
of the human genome. Oncogene 2000;19:5548–57. [45] Hubbard SR, Till JH. Protein tyrosine kinase structure and function.
[20] Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. Annu Rev Biochem 2000;69:373–98.
The protein kinase complement of the human genome. Science [46] Hubbard SR. Theme and variations: juxtamembrane regulation of
2002;298:1912–34. receptor protein kinases. Mol Cell 2001;8:481–2.
[21] Fantl WJ, Johnson DE, Williams LT. Signalling by receptor tyrosine [47] Werten PJ, Remigy HW, de Groot BL, et al. Progress in the
kinases. Annu Rev Biochem 1993;62:453–81. analysis of membrane protein structure and function. FEBS Lett
[22] Olayioye MA, Neve RM, Lane HA, Hynes NE. The ErbB signaling 2002;529:65–72.
network: receptor heterodimerization in development and cancer. [48] Ogiso H, Ishitani R, Nureki O, et al. Crystal structure of the com-
EMBO J 2000;19:3159–67. plex of human epidermal growth factor and receptor extracellular
[23] Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. domains. Cell 2002;110:775–87.
Nat Rev Mol Cell Biol 2001;2:127–37. [49] Garrett TP, McKern NM, Lou M, et al. Crystal structure of a
[24] de Bono JS, Rowinsky EK. The ErbB receptor family: a therapeutic truncated epidermal growth factor receptor extracellular domain
target for cancer. Trends Mol Med 2002;8:S19–26. bound to transforming growth factor alpha. Cell 2002;110:763–73.
36 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

[50] Cho HS, Leahy DJ. Structure of the extracellular region of HER3 [74] Waterman H, Yarden Y. Molecular mechanisms underlying endo-
reveals an interdomain tether. Science 2002;297:1330–3. cytosis and sorting of ErbB receptor tyrosine kinases. FEBS Lett
[51] Wiesmann C, Ultsch MH, Bass SH, de Vos AM. Crystal structure 2001;490:142–52.
of nerve growth factor in complex with the ligand-binding domain [75] Shtiegman K, Yarden Y. The role of ubiquitylation in signal-
of the TrkA receptor. Nature 1999;401:184–8. ing by growth factors: implications to cancer. Semin Cancer Biol
[52] Banfield MJ, Naylor RL, Robertson AG, Allen SJ, Dawbarn D, 2003;13:29–40.
Brady RL. Specificity in Trk receptor:neurotrophin interactions: [76] Katz M, Shtiegman K, Tal-Or P, et al. Ligand-independent degra-
the crystal structure of TrkB-d5 in complex with neurotrophin-4/5. dation of epidermal growth factor receptor involves receptor ubiq-
Structure (Camb) 2001;9:1191–9. uitylation and Hgs, an adaptor whose ubiquitin-interacting motif
[53] Wiesmann C, Fuh G, Christinger HW, Eigenbrot C, Wells JA, de targets ubiquitylation by Nedd4. Traffic 2002;3:740–51.
Vos AM. Crystal structure at 1.7 A resolution of VEGF in complex [77] Citri A, Alroy I, Lavi S, et al. Drug-induced ubiquitylation and
with domain 2 of the Flt-1 receptor. Cell 1997;91:695–704. degradation of ErbB receptor tyrosine kinases: implications for
[54] Himanen JP, Rajashankar KR, Lackmann M, Cowan CA, Henke- cancer therapy. EMBO J 2002;21:2407–17.
meyer M, Nikolov DB. Crystal structure of an Eph receptor-ephrin [78] Masters JR. Human cancer cell lines: fact and fantasy. Nat Rev
complex. Nature 2001;414:933–8. Mol Cell Biol 2000;1:233–6.
[55] Hubbard SR, Wei L, Ellis L, Hendrickson WA. Crystal structure of [79] Brandt R, Wong AM, Hynes NE. Mammary glands reconsti-
the tyrosine kinase domain of the human insulin receptor. Nature tuted with Neu/ErbB2 transformed HC11 cells provide a novel
1994;372:746–54. orthotopic tumor model for testing anti-cancer agents. Oncogene
[56] Hubbard SR. Crystal structure of the activated insulin receptor 2001;20:5459–65.
tyrosine kinase in complex with peptide substrate and ATP analog. [80] Lewandoski M. Conditional control of gene expression in the mouse.
EMBO J 1997;16:5572–81. Nat Rev Genet 2001;2:743–55.
[57] Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B, [81] Klausner RD. Studying cancer in the mouse. Oncogene
Kuriyan J. Structural mechanism for STI-571 inhibition of abelson 1999;18:5249–52.
tyrosine kinase. Science 2000;289:1938–42. [82] DePinho RA, Jacks T. The laboratory mouse in cancer research.
[58] Cho H, Mason K, Ramyar K, et al. Structure of the extracellular Semin Cancer Biol 2001;11:175–6.
region of HER2 alone and in complex with the Herceptin Fab. [83] Hann B, Balmain A. Building ‘validated’ mouse models of human
Nature 2003;421:756–60. cancer. Curr Opin Cell Biol 2001;13:778–84.
[59] Kolibaba KS, Druker BJ. Protein tyrosine kinases and cancer. [84] Herzig M, Christofori G. Recent advances in cancer research: mouse
Biochim Biophys Acta 1997;1333:F217–48. models of tumorigenesis. Biochim Biophys Acta 2002;1602:97–113.
[60] Porter AC, Vaillancourt RR. Tyrosine kinase receptor-activated sig- [85] Muller WJ, Sinn E, Pattengale PK, Wallace R, Leder P. Single-step
nal transduction pathways which lead to oncogenesis. Oncogene induction of mammary adenocarcinoma in transgenic mice bearing
1998;17:1343–52. the activated c-neu oncogene. Cell 1988;54:105–15.
[61] Pawson T, Scott JD. Signaling through scaffold, anchoring, and [86] Rosenberg MP, Bortner D. Why transgenic and knockout animal
adaptor proteins. Science 1997;278:2075–80. models should be used (for drug efficacy studies in cancer). Cancer
[62] Zong CS, Poon B, Chen J, Wang LH. Molecular and biochem- Metastasis Rev 1998;17:295–9.
ical bases for activation of the transforming potential of the [87] Bouchard L, Lamarre L, Tremblay PJ, Jolicoeur P. Stochastic ap-
proto-oncogene c-ros. J Virol 1993;67:6453–62. pearance of mammary tumors in transgenic mice carrying the
[63] Yarden Y. The EGFR family and its ligands in human cancer. MMTV/c-neu oncogene. Cell 1989;57:931–6.
Signalling mechanisms and therapeutic opportunities. Eur J Cancer [88] Muller WJ, Arteaga CL, Muthuswamy SK. Synergistic interaction
2001;37(4):S3–8. of the Neu proto-oncogene product and transforming growth factor
[64] Klapper LN, Kirschbaum MH, Sela M, Yarden Y. Biochemical and alpha in the mammary epithelium of transgenic mice. Mol Cell
clinical implications of the ErbB/HER signaling network of growth Biol 1996;16:5726–36.
factor receptors. Adv Cancer Res 2000;77:25–79. [89] Lenferink AE, Simpson JF, Shawver LK, Coffey RJ, Forbes
[65] Wells A, Marti U. Signalling shortcuts: cell-surface receptors in the JT, Arteaga CL. Blockade of the epidermal growth factor re-
nucleus. Nat Rev Mol Cell Biol 2002;3:697–702. ceptor tyrosine kinase suppresses tumorigenesis in MMTV/Neu
[66] Khosravi-Far R, Der CJ. The Ras signal transduction pathway. + MMTV/TGF-alpha bigenic mice. Proc Natl Acad Sci USA
Cancer Metastasis Rev 1994;13:67–89. 2000;97:9609–14.
[67] Palmer BD, Rewcastle GW, Thompson AM, et al. Tyrosine kinase [90] Weinstein EJ, Kitsberg DI, Leder P. A mouse model for breast
inhibitors. Part 4. Structure-activity relationships among N- and cancer induced by amplification and overexpression of the neu
3-substituted 2,2 -dithiobis(1H-indoles) for in vitro inhibition of promoter and transgene. Mol Med 2000;6:4–16.
receptor and nonreceptor protein tyrosine kinases. J Med Chem [91] Guy CT, Webster MA, Schaller M, Parsons TJ, Cardiff RD,
1995;38:58–67. Muller WJ. Expression of the neu protooncogene in the mammary
[68] Cantley LC. The phosphoinositide 3-kinase pathway. Science epithelium of transgenic mice induces metastatic disease. Proc Natl
2002;296:1655–7. Acad Sci USA 1992;89:10578–82.
[69] Pawson T. Regulation and targets of receptor tyrosine kinases. Eur [92] Guy CT, Cardiff RD, Muller WJ. Activated neu induces rapid tumor
J Cancer 2002;38:S3–S10. progression. J Biol Chem 1996;271:7673–8.
[70] Vivanco I, Sawyers CL. The phosphatidylinositol 3-kinase AKT [93] DiGiovanna MP, Lerman MA, Coffey RJ, Muller WJ, Cardiff RD,
pathway in human cancer. Nat Rev Cancer 2002;2:489–501. Stern DF. Active signaling by Neu in transgenic mice. Oncogene
[71] Hubbard SR, Mohammadi M, Schlessinger J. Autoregulatory mech- 1998;17:1877–84.
anisms in protein-tyrosine kinases. J Biol Chem 1998;273:11987– [94] Jeffers M, Fiscella M, Webb CP, Anver M, Koochekpour S, Vande
90. Woude GF. The mutationally activated Met receptor mediates motil-
[72] Hunter T. Protein kinases and phosphatases: the yin and yang of ity and metastasis. Proc Natl Acad Sci USA 1998;95:14417–22.
protein phosphorylation and signaling. Cell 1995;80:225–36. [95] Santoro M, Chiappetta G, Cerrato A, et al. Development of thyroid
[73] Ostman A, Bohmer FD. Regulation of receptor tyrosine ki- papillary carcinomas secondary to tissue-specific expression of the
nase signaling by protein tyrosine phosphatases. Trends Cell Biol RET/PTC1 oncogene in transgenic mice. Oncogene 1996;12:1821–
2001;11:258–66. 6.
A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38 37

[96] Michiels FM, Chappuis S, Caillou B, et al. Development of [116] Petit AM, Rak J, Hung MC, et al. Neutralizing antibodies against
medullary thyroid carcinoma in transgenic mice expressing the RET epidermal growth factor and ErbB-2/neu receptor tyrosine kinases
protooncogene altered by a multiple endocrine neoplasia type 2A down-regulate vascular endothelial growth factor production by
mutation. Proc Natl Acad Sci USA 1997;94:3330–5. tumor cells in vitro and in vivo: angiogenic implications for signal
[97] Larcher F, Murillas R, Bolontrade M, Conti CJ, Jorcano JL. transduction therapy of solid tumors. Am J Pathol 1997;151:1523–
VEGF/VPF overexpression in skin of transgenic mice induces an- 30.
giogenesis. Oncogene 1998;17:303–11. [117] Liu B, Fang M, Schmidt M, Lu Y, Mendelsohn J, Fan Z. Induction
[98] Sweetser DA, Froelick GJ, Matsumoto AM, et al. Ganglioneuromas of apoptosis and activation of the caspase cascade by anti-EGF
and renal anomalies are induced by activated RET(MEN2B) in receptor monoclonal antibodies in DiFi human colon cancer cells
transgenic mice. Oncogene 1999;18:877–86. do not involve the c-jun N-terminal kinase activity. Br J Cancer
[99] Xie W, Chow LT, Paterson AJ, Chin E, Kudlow JE. Conditional 2000;82:1991–9.
expression of the ErbB2 oncogene elicits reversible hyperplasia in [118] Shin DM, Donato NJ, Perez-Soler R, et al. Epidermal growth factor
stratified epithelia and up-regulation of TGFalpha expression in receptor-targeted therapy with C225 and cisplatin in patients with
transgenic mice. Oncogene 1999;18:3593–607. head and neck cancer. Clin Cancer Res 2001;7:1204–13.
[100] Kiguchi K, Bol D, Carbajal S, et al. Constitutive expression of [119] Yang XD, Jia XC, Corvalan JR, Wang P, Davis CG. Development of
erbB2 in epidermis of transgenic mice results in epidermal hyper- ABX-EGF, a fully human anti-EGF receptor monoclonal antibody,
proliferation and spontaneous skin tumor development. Oncogene for cancer therapy. Crit Rev Oncol Hematol 2001;38:17–23.
2000;19:4243–54. [120] Park BW, Zhang HT, Wu C, et al. Rationally designed
[101] Hayes C, Kelly D, Murayama S, Komiyama A, Suzuki K, Popko anti-HER2/neu peptide mimetic disables P185HER2/neu tyrosine
B. Expression of the neu oncogene under the transcriptional control kinases in vitro and in vivo. Nat Biotechnol 2000;18:194–8.
of the myelin basic protein gene in transgenic mice: generation of [121] Berezov A, Zhang HT, Greene MI, Murali R. Disabling erbB
transformed glial cells. J Neurosci Res 1992;31:175–87. receptors with rationally designed exocyclic mimetics of antibodies:
[102] Ezzat S, Zheng L, Zhu XF, Wu GE, Asa SL. Targeted expression structure-function analysis. J Med Chem 2001;44:2565–74.
of a human pituitary tumor-derived isoform of FGF receptor-4 [122] Carter P. Improving the efficacy of antibody-based cancer therapies.
recapitulates pituitary tumorigenesis. J Clin Invest 2002;109:69–78. Nat Rev Cancer 2001;1:118–29.
[103] Houseman BT, Huh JH, Kron SJ, Mrksich M. Peptide chips for the [123] Allen TM. Ligand-targeted therapeutics in anticancer therapy. Nat
quantitative evaluation of protein kinase activity. Nat Biotechnol Rev Cancer 2002;2:750–63.
2002;20:270–4. [124] Garrett MD, Workman P. Discovering novel chemotherapeutic drugs
[104] Zaniboni A. Suramin: the discovery of an old anticancer drug. Med for the third millennium. Eur J Cancer 1999;35:2010–30.
Oncol Tumor Pharmacother 1990;7:287–90. [125] Druker BJ, Lydon NB. Lessons learned from the development of
[105] Kaur M, Reed E, Sartor O, Dahut W, Figg WD. Suramin’s devel- an abl tyrosine kinase inhibitor for chronic myelogenous leukemia.
opment: what did we learn? Invest New Drugs 2002;20:209–19. J Clin Invest 2000;105:3–7.
[106] Sebti SM, Hamilton AD. Design of growth factor antagonists with [126] Buchdunger E, Zimmermann J, Mett H, et al. Inhibition of
antiangiogenic and antitumor properties. Oncogene 2000;19:6566– the Abl protein-tyrosine kinase in vitro and in vivo by a
73. 2-phenylaminopyrimidine derivative. Cancer Res 1996;56:100–4.
[107] Blaskovich MA, Lin Q, Delarue FL, et al. Design of GFB-111, [127] Morin MJ. From oncogene to drug: development of small molecule
a platelet-derived growth factor binding molecule with antiangio- tyrosine kinase inhibitors as anti-tumor and anti-angiogenic agents.
genic and anticancer activity against human tumors in mice. Nat Oncogene 2000;19:6574–83.
Biotechnol 2000;18:1065–70. [128] Wang WL, Healy ME, Sattler M, et al. Growth inhibition and mod-
[108] Glennie MJ, Johnson PW. Clinical trials of antibody therapy. Im- ulation of kinase pathways of small cell lung cancer cell lines by the
munol Today 2000;21:403–10. novel tyrosine kinase inhibitor STI-571. Oncogene 2000;19:3521–
[109] Sarup JC, Johnson RM, King KL. Characterization of an 8.
anti-p185HER2 monoclonal antibody that stimulates receptor func- [129] Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety
tion and inhibits tumor cell growth. Growth Regul 1991;1:72–82. of imatinib mesylate in advanced gastrointestinal stromal tumors.
[110] Lane HA, Beuvink I, Motoyama AB, Daly JM, Neve RM, Hynes N Engl J Med 2002;347:472–80.
NE. ErbB2 potentiates breast tumor proliferation through mod- [130] Hernandez-Boluda JC, Cervantes F. Imatinib mesylate (Gleevec,
ulation of p27(Kip1)-Cdk2 complex formation: receptor overex- Glivec): a new therapy for chronic myeloid leukemia and other
pression does not determine growth dependency. Mol Cell Biol malignancies. Drugs Today (Barc) 2002;38:601–13.
2000;20:3210–23. [131] Denny WA. The 4-anilinoquinazoline class of inhibitors of the erbB
[111] Molina MA, Codony-Servat J, Albanell J, Rojo F, Arribas J, Baselga family of receptor tyrosine kinases. Farmacol 2001;56:51–6.
J. Trastuzumab (Herceptin), a humanized anti-Her2 receptor mono- [132] Ciardiello F, Caputo R, Bianco R, et al. Antitumor effect and
clonal antibody, inhibits basal and activated Her2 ectodomain cleav- potentiation of cytotoxic drugs activity in human cancer cells by
age in breast cancer cells. Cancer Res 2001;61:4744–9. ZD-1839 (Iressa), an epidermal growth factor receptor-selective
[112] Masui H, Kawamoto T, Sato JD, Wolf B, Sato G, Mendelsohn tyrosine kinase inhibitor. Clin Cancer Res 2000;6:2053–63.
J. Growth inhibition of human tumor cells in athymic mice by [133] Folkman J. Role of angiogenesis in tumor growth and metastasis.
anti-epidermal growth factor receptor monoclonal antibodies. Can- Semin Oncol 2002;29:15–8.
cer Res 1984;44:1002–7. [134] Laird AD, Vajkoczy P, Shawver LK, et al. SU6668 is a potent
[113] Goldstein NI, Prewett M, Zuklys K, Rockwell P, Mendelsohn J. antiangiogenic and antitumor agent that induces regression of es-
Biological efficacy of a chimeric antibody to the epidermal growth tablished tumors. Cancer Res 2000;60:4152–60.
factor receptor in a human tumor xenograft model. Clin Cancer [135] Zutshi R, Brickner M, Chmielewski J. Inhibiting the assembly of
Res 1995;1:1311–8. protein–protein interfaces. Curr Opin Chem Biol 1998;2:62–6.
[114] Baselga J, Norton L, Masui H, et al. Antitumor effects of dox- [136] Chardin P, Cussac D, Maignan S, Ducruix A. The Grb2 adaptor.
orubicin in combination with anti-epidermal growth factor receptor FEBS Lett 1995;369:47–51.
monoclonal antibodies. J Natl Cancer Inst 1993;85:1327–33. [137] Garbay C, Liu WQ, Vidal M, Roques BP. Inhibitors of Ras
[115] Baselga J. The EGFR as a target for anticancer therapy—focus on signal transduction as antitumor agents. Biochem Pharmacol
cetuximab. Eur J Cancer 2001;37(Suppl 4):S16–22. 2000;60:1165–9.
38 A. Bennasroune et al. / Critical Reviews in Oncology/Hematology 50 (2004) 23–38

[138] Kim H, Muller WJ. The role of the epidermal growth factor receptor of Tyr-253 in the Abl kinase domain P-loop. Proc Natl Acad Sci
family in mammary tumorigenesis and metastasis. Exp Cell Res USA 2002;99:10700–5.
1999;253:78–87. [153] Berman HM, Westbrook J, Feng Z, et al. The protein data bank.
[139] Cussac D, Vidal M, Leprince C. A Sos-derived peptidimer blocks Nucleic Acids Res 2000;28:235–42.
the Ras signaling pathway by binding both Grb2 SH3 domains and [154] Sayle RA, Milner-White EJ. RASMOL: biomolecular graphics for
displays antiproliferative activity. FASEB J 1999;13:31–8. all. Trends Biochem Sci 1995;20:374.
[140] Liu WQ, Vidal M, Gresh N, Roques BP, Garbay C. Small peptides [155] Stamos J, Sliwkowski MX, Eigenbrot C. Structure of the epider-
containing phosphotyrosine and adjacent alphaMe-phosphotyrosine mal growth factor receptor kinase domain alone, in complex with
or its mimetics as highly potent inhibitors of Grb2 SH2 domain. J a 4-anilinoquinazoline inhibitor. J Biol Chem 2002;277:46265–
Med Chem 1999;42:3737–41. 72.
[141] Cochran AG. Antagonists of protein–protein interactions. Chem [156] Goetz M, Carlotti C, Bontems F, Dufourc EJ. Evidence for an
Biol 2000;7:R85–94. alpha-helix → pi-bulge helicity modulation for the neu/erbB-2
[142] Schlessinger J. Ligand-induced, receptor-mediated dimerization and membrane-spanning segment. A 1H NMR and circular dichroism
activation of EGF receptor. Cell 2002;110:669–72. study. Biochemistry 2001;40:6534–40.
[143] Berezov A, Chen J, Liu Q, Zhang HT, Greene MI, Murali R. [157] Pellegrini L, Burke DF, von Delft F, Mulloy B, Blundell TL. Crystal
Disabling receptor ensembles with rationally designed interface structure of fibroblast growth factor receptor ectodomain bound to
peptidomimetics. J Biol Chem 2002;277:28330–9. ligand and heparin. Nature 2000;407:1029–34.
[144] Lofts FJ, Hurst HC, Sternberg MJ, Gullick WJ. Specific short trans- [158] Mohammadi M, McMahon G, Sun L. Structures of the tyrosine
membrane sequences can inhibit transformation by the mutant neu kinase domain of fibroblast growth factor receptor in complex with
growth factor receptor in vitro and in vivo. Oncogene 1993;8:2813– inhibitors. Science 1997;276:955–60.
20. [159] Maignan S, Guilloteau JP, Fromage N, Arnoux B, Becquart J,
[145] Qian X, O’Rourke DM, Zhao H, Greene MI. Inhibition of p185neu Ducruix A. Crystal structure of the mammalian Grb2 adaptor. Sci-
kinase activity and cellular transformation by co-expression of a ence 1995;268:291–3.
truncated neu protein. Oncogene 1996;13:2149–57. [160] Dhe-Paganon S, Ottinger EA, Nolte RT, Eck MJ, Shoelson SE.
[146] Cohen P. The development and therapeutic potential of protein Crystal structure of the pleckstrin homology-phosphotyrosine bind-
kinase inhibitors. Curr Opin Chem Biol 1999;3:459–65. ing (PH-PTB) targeting region of insulin receptor substrate 1. Proc
[147] Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK. Tumor biology: Natl Acad Sci USA 1999;96:8378–83.
Herceptin acts as an anti-angiogenic cocktail. Nature 2002;416:279– [161] Siegal G, Davis B, Kristensen SM, et al. Solution structure of the
80. C-terminal SH2 domain of the p85 alpha regulatory subunit of
[148] Joensuu H, Roberts PJ, Sarlomo-Rikala M. Effect of the tyrosine phosphoinositide 3-kinase. J Mol Biol 1998;276:461–78.
kinase inhibitor STI-571 in a patient with a metastatic gastrointesti-
nal stromal tumor. N Engl J Med 2001;344:1052–6.
[149] Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mech-
Biography
anism of action of some commonly used protein kinase inhibitors.
Biochem J 2000;351:95–105.
[150] Furge KA, Kiewlich D, Le P, et al. Suppression of Ras-mediated Pierre Hubert obtained his M.D. in 1979 and a Ph.D.
tumorigenicity and metastasis through inhibition of the Met receptor in cell biology in 1992 at the University Louis Pasteur,
tyrosine kinase. Proc Natl Acad Sci USA 2001;98:10722–7. Strasbourg, France. He is currently Chargé de Recherche
[151] Norgaard P, Law B, Joseph H, et al. Treatment with farnesyl-protein
at the National Institute for Health and Medical Research
transferase inhibitor induces regression of mammary tumors in trans-
forming growth factor (TGF) alpha and TGF alpha/neu transgenic (INSERM, Unit 575). He heads a small research group on
mice by inhibition of mitogenic activity and induction of apoptosis. the role of membrane domains in receptor tyrosine kinase
Clin Cancer Res 1999;5:35–42. activation.
[152] Roumiantsev S, Shah NP, Gorre ME, et al. Clinical resistance to the
kinase inhibitor STI-571 in chronic myeloid leukemia by mutation

Das könnte Ihnen auch gefallen