<<

Endocrine-Related (2001) 8 161–173

Receptor signalling as a target for cancer intervention strategies

E Zwick, J Bange and A Ullrich

Max-Planck-Institut fu¨r Biochemie,Department of Molecular Biology,Am Klopferspitz 18A,82152 Martinsried, Germany (Requests for offprints should be addressed to A Ullrich; Email: [email protected])

Abstract In multicellular organisms,communication between individual cells is essential for the regulation and coordination of complex cellular processes such as growth,differentiation,migration and . The plethora of networks mediating these biological processes is regulated in part by polypeptide growth factors that can generate signals by activating cell surface receptors either in paracrine or autocrine manner. The primary mediators of such physiological cell responses are tyrosine (RTKs) that couple binding to downstream signalling cascades and transcription. Investigations over the past 18 years have revealed that RTKs are not only key regulators of normal cellular processes but are also critically involved in the development and progression of . Therefore,signalling pathways controlled by tyrosine kinases offer unique opportunities for pharmacological intervention. The aim of this review is to give a broad overview of RTK signalling involved in tumorigenesis and the possibility of target-selective intervention for anti-cancer therapy. Endocrine-Related Cancer (2001) 8 161–173

Introduction phosphorylated and activated, such as Src and phospholipase Cγ, or adaptor molecules that link RTK activation to On the basis of their structural characteristics, receptor downstream signalling pathways (Pawson 1995). One tyrosine kinases (RTKs) can be divided into 20 subfamilies important adaptor complex is the SHC–Grb2–Sos which share a homologous domain that specifies the catalytic complex that couples RTKs via Ras to the extracellular function (van der Geer et al. 1994). All RTKs regulated kinase (ERK)/-activated protein (MAP) consist of a single transmembrane domain that separates the kinase pathway which is crucial for RTK-induced cell intracellular tyrosine kinase region from the extracellular proliferation (Dhanasekaran & Reddy 1998) (Fig. 1). portion (Ullrich & Schlessinger 1990). The latter exhibit a variety of conserved elements including immunoglobulin (Ig)-like or epidermal (EGF)-like domains, Deregulation of RTKs fibronectin type III repeats or -rich regions that are characteristic for each subfamily of RTKs. Since the first connection between a viral , a The catalytic domain that displays the highest level of mutated RTK and human cancer was made in 1984 (Ullrich conservation includes the ATP- that catalyses et al. 1984), it is well known that aberrant signalling by receptor and tyrosine RTKs is critically involved in human cancer and other of RTK substrates (Yarden & Ullrich 1988). Ligand binding hyper-proliferative diseases. Constitutive activation of RTKs, to the extracellular domain leads to conformational changes which has been shown to be important for malignant that induce and stabilize receptor dimerization leading to transformation and tumour proliferation, can occur by several increased kinase activity and autophosphorylation of tyrosine mechanisms (Kolibaba & Druker 1997). In most cases gene residues (Greenfield et al. 1989, Ullrich & Schlessinger amplification, overexpression or are responsible 1990, Heldin 1995). Phosphorylation of distinct tyrosine for the acquired transforming potential of oncogenic RTKs. residues of the activated receptor creates binding sites for Somatic and germline mutations, which are associated with Src homology 2 (SH2)- and phosphotyrosine binding (PTB) distinct inherited and spontaneous human cancer syndromes, domain-containing . Molecules recruited via these have been observed in at least ten different RTK families binding motifs are either that are tyrosine (Robertson et al. 2000). These alterations include deletion or

Endocrine-Related Cancer (2001) 8 000–000 Online version via http://www.endocrinology.org 1351-0088/01/008–000  2001 Society for Endocrinology Printed in Great Britain Downloaded from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Zwick et al.: RTKs in anti-cancer strategies

Figure 1 Target-selective intervention of RTK signalling. Important signalling pathways induced by RTKs and a summary of the most successful strategies towards the prevention and interception of RTK signalling. Abbreviations: ERK, extracellular-regulated kinase; JNK,c-Jun terminal kinase; PI3 kinase,phosphatidylinositol-3 kinase; PLC γ, γ.

within the extracellular region or alterations of the expressed or overexpressed in the presence of its cognate catalytic domain, especially of the ATP-binding motif, which ligand, or when overexpression of the ligand occurs in the all result in a constitutive active RTK. In addition, mutations presence of its associated receptor. In many solid tumours it within the transmembrane domain have been shown to lead has been shown that elevated levels of both growth factor to ligand-independent kinase activation as reported for the receptor and its ligand are expressed concomitantly (Salomon RTK HER2/neu. et al. 1995). Activation of autocrine growth factor loops is another mechanism of aberrant RTK signalling and has been Intervention strategies frequently described for the epidermal (EGFR) and -like growth factor-I receptor (IGF-IR) Increasing knowledge of the structure and activation family (Derynck et al. 1987, Kaleko et al. 1990). This potent mechanism of RTKs and the signalling pathways controlled mechanism of activation occurs when a RTK is aberrantly by tyrosine kinases provided the possibility for the

162 Downloadedwww.endocrinology.org from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Endocrine-Related Cancer (2001) 8 161–173 development of target-specific drugs and new anti-cancer potency and selectivity, higher in vitro and in vivo efficacy therapies (Plowman et al. 1994). Approaches towards the and decreased toxicity have emerged (Klohs et al. 1997, prevention or interception of deregulated RTK signalling Al-Obeidi & Lam 2000). include the development of selective components that target Recombinant immunotoxins provide another possibility either the extracellular ligand-binding domain or the of target-selective . They are composed of a intracellular tyrosine kinase or binding region bacterial or plant either fused or chemically conjugated (Fig. 1). to a specific ligand such as the variable domains of the heavy The most successful strategy to selectively kill tumour and light chains of mAbs or to a growth factor (Kreitman cells is the use of monoclonal (mAbs) that are 1999). Immunotoxins either contain the bacterial directed against the extracellular domain of RTKs which Pseudomonas exotoxin A or diphtheria toxin or the plant are critically involved in cancer and are expressed at the toxins ricin A or clavin. These recombinant molecules can surface of tumour cells (Fan & Mendelsohn 1998). In the selectively kill their target cells when internalized after past years, recombinant technology has made an binding to specific cell surface receptors (Frankel et al. enormous progress in the design, selection and production 2000). of new engineered antibodies and it is possible to generate The use of antisense oligonucleotides represents another humanized antibodies, human–mouse chimeric or bispecific strategy to inhibit the activation of RTKs. Antisense antibodies for targeted cancer therapy (Farah et al. 1998, oligonucleotides are short pieces of synthetic DNA or RNA Hudson 1999). Mechanistically, anti-RTK mAbs might that are designed to interact with the mRNA to block the work by blocking the ligand–receptor interaction and transcription and thus the expression of specific-target therefore inhibiting ligand-induced RTK signalling and proteins (Marcusson et al. 1999). These compounds interact increasing RTK downregulation and internalization. In with the mRNA by Watson–Crick base-pairing and are addition, by binding of mAbs to certain epitopes on the therefore highly specific for the target protein. On the other cancer cells they induce immune-mediated responses such hand bioavailability may be poor since oligonucleotides can as opsonization and complement-mediated lysis and trigger be degraded upon internalization by cellular endo- and antibody-dependent cellular cytotoxicity by macrophages or . Nevertheless, several preclinical and clinical natural killer cells. In recent years, it became evident that studies suggest that antisense therapy might be mAbs control tumour growth by altering the intracellular therapeutically useful for the treatment of solid tumours signalling pattern inside the targeted tumour cell, leading (Pawlak et al. 2000). to growth inhibition and/or apoptosis (Cragg et al. 1999). In contrast, bispecific antibodies can bridge selected surface molecules on a target cell with receptors on an effector cell triggering cytotoxic responses against the target cell Epidermal growthfactor receptor (EGFR) that is thus killed (Segal et al. 1999). Despite the toxicity family that has been seen in clinical trials of bispecific antibodies, advances in antibody engineering, characterization of The receptor (EGFR) family of tumour antigens and immunology might help to produce RTKs comprises four members: the archetypal EGFR/ErbB1, rationally designed bispecific antibodies for anti-cancer which was the first RTK to be molecularly cloned (Ullrich therapy. et al. 1984), HER2/ErbB2, HER3/ErbB3 and HER4/ErbB4. Another promising approach to inhibit aberrant RTK The four receptors share two extracellular cysteine-rich signalling are small molecule drugs that selectively interfere domains and an intracellular portion with a long C-terminal with the intrinsic tyrosine kinase activity and thereby block tail carrying most of the autophosphorylation sites. Receptors receptor autophosphorylation and activation of downstream of the EGFR family are frequently co-expressed in various signal transducers (Levitzki 1999). The tyrphostins, which combinations and depending on the activating ligand they belong to the quinazolines, are one important group of such can form various homo- or heterodimers generating a inhibitors that compete with ATP for the ATP binding site complex signal transduction network (Alroy & Yarden 1997, at the receptor’s tyrosine kinase domain and some members Riese & Stern 1998). EGFR family members are activated have been shown to specifically inhibit the EGFR by a large group of EGF-related growth, which all contain a (Levitzki & Gazit 1995). But also potent and selective conserved EGF-like domain and are synthesized as inhibitors of receptors involved in neovascularization have transmembrane precursor proteins (Heldin 1996). These been developed and are now undergoing clinical evaluation. include EGF, transforming growth factor-α (TGFα), Using the advantages of structure-based drug design, , , heparin-binding EGF-like growth crystallographic structure information, combinatorial factor, and the large family of chemistry and high-throughput screening new structural alternatively-spliced (Burden & Yarden 1997). classes of tyrosine kinase inhibitors (TKIs) with increased These growth factors bind with different specificities and www.endocrinology.org Downloaded from Bioscientifica.com163 at 09/28/2021 02:58:10PM via free access Zwick et al.: RTKs in anti-cancer strategies affinities to EGFR, HER3 and HER4, while no ligand for autophosphorylation and constitutive signalling (Lonardo et HER2 has yet been identified. al. 1990, Brennan et al. 2000). It was shown that overexpression of HER2 led to a prolonged and enhanced cellular signalling through the MAP kinase pathway HER2 (Karunagaran et al. 1996, Harari & Yarden 2000). The RTK HER2 was originally identified as both the transforming gene in a chemically transformed rat The role ofHER2 in tumour growth cell line (Schechter et al. 1984) and as an Recently it became evident that the transforming ability of EGFR-related cDNA clone (Coussens et al. 1985), and has HER2 is linked to cell survival and the ability of HER2 to been most frequently implicated in human neoplasias and directly affect components of the machinery. cancer (King et al. 1985, Slamon et al. 1987, Hynes & Stern Progression through the cell cycle is critically dependent on 1994). active complex formation of cyclin-dependent kinases Despite the fact that no ligand is known to bind with (CDKs) with cyclin D1, which is a key regulator of G1/S high affinity to HER2, it is well known that HER2 acts as a phase transition of the cell cycle. Interestingly, cyclin D1 co-receptor for the other EGFR family members. In has been identified as a downstream target of HER2/neu in particular, either the EGFR ligands or the neuregulins that transgenic mice or HER2-overexpressing cell lines, which is bind HER3 or HER4 are able to induce heterodimer necessary for HER2/neu-induced transformation (Lee et al. formation between a high-affinity ligand binding receptor 2000). It was shown that cyclin D1 expression is upregulated and HER2. These heteromolecular interactions are of by activated HER2/neu and that blocking HER2 specifically pathophysiological relevance, because such receptor inhibits the growth of tumour cells by arresting the cells in combinations show strong mitogenic signalling and the G1 phase of the cell cycle. Concomitantly, accumulation tumorigenicity. Signalling pathways involving Ras, Src, of p27Kip1, a CDK inhibitor, and inactivation of – PI3-kinase and the MAP kinases ERK and JNK have been CDK2 complexes were seen (Lane et al. 2000). Moreover, shown to be activated by HER2-expressing cell lines and the level of c- and D-cylins, proteins that are involved seem to be important for tumour management (Zwick et al. in the sequestration of p27Kip1, decreased in the absence of 2000). functional HER2 (Neve et al. 2000). These results suggest HER2 gene mutation and high level overexpression are that HER2 induces the potentiation of cyclin E–CDK2 other potent mechanisms resulting in HER2 activation. A activity via regulation of p27Kip1 sequestration. As a single point mutation at position 664 within the consequence, HER2 overexpression deregulates the G1/S transmembrane region of HER2, encoding a change from the phase transition and results in aberrant proliferation hydrophobic valine to the negatively charged and tumour formation. Very recently it was demonstrated glutamic acid residue, renders the receptor constitutively that activation of the PI3-kinase/Akt pathway by active (Bargmann et al. 1986, Weiner et al. 1989). Although HER2-overexpressing cells promotes cell survival by the mutation is not observed in human tumours, a phosphorylating the CDK inhibitor p21Cip1 (Zhou et al. polymorphism at codon 655, which results in a valine to 2001). Phosphorylated p21Cip1 localizes to the isoleucine substitution, has been identified in the where it can form a complex with apoptosis-signal-regulating transmembrane coding region of the HER2 gene (Papewalis kinase 1 (ASK1) conferring its anti-apoptotic effect. At the et al. 1991). Performing a large-scale population-based, same time the HER2-induced cytoplasmic localization of case-control study in China, an association of the genetic p21Cip1 suppresses its growth-inhibiting activity in the HER2 polymorphism and an increased risk of , nucleus and consequently leads to HER2-mediated cell particularly among younger women, was recently identified growth. (Xie et al. 2000). Overexpression and/or gene amplification of HER2 was HER2 as a prognostic and predictive indicator found in various types of human cancer and especially in The relatively low expression level of HER2 in normal human breast where HER2 gene amplification epithelial cells is significantly enhanced in several types of has been identified with a frequency of 30% (Slamon et al. human cancers, including breast, ovarian, gastric, , 1987, 1989). Subsequently, it became evident that aberrantly bladder and carcinomas (Hynes & Stern 1994). elevated levels of HER2, either with or without gene Especially in breast cancer a significant correlation between amplification, correlated with a more aggressive progression HER2 overexpression and reduced survival of breast cancer of disease and a reduced patient survival time (Paik et al. patients exists (Slamon et al. 1987). Moreover, clinical as 1990). well as laboratory data revealed that overexpression of HER2 The mechanism underlying the oncogenic potential of an increases the metastatic potential of human breast and lung overexpressed HER2 may relate to the increased availability cancer cells and correlates with the number of lymph node for heterodimer formation and thus for high level metastases in node-positive breast cancer patients (Yu &

164 Downloadedwww.endocrinology.org from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Endocrine-Related Cancer (2001) 8 161–173

Hung 2000). Therefore, HER2 expression represents a combination with paclitaxel or doxorubicin with pivotal biological marker that can help to determine more cyclophosphamide significantly increased response duration, accurately the prognosis for individual patients (Cooke et al. time to progression and survival in first-line metastatic breast 2001). cancer patients (Burris 2000, Stebbing et al. 2000). In recent years, it became evident that HER2 is also a Moreover, preclinical experiments have shown that predictive marker for responses to various therapeutic agents Herceptin modulates repair of radiation-induced DNA used in cancer therapy. It has been reported that HER2 damage and enhances radiosensitivity of HER2- overexpression negatively influences the response rate of overexpressing breast cancer cells (Pietras et al. 1999). breast tumours to chemotherapeutic agents (Yu & Hung Therefore, it seems reasonable that Herceptin should be 2000). In addition, elevated levels of a processed HER2 considered as one of the routine treatment options in extracellular domain in patients metastatic breast cancer. seem to reduce the efficacy of certain Other strategies against HER2-positive breast and combinations (Colomer et al. 2000). Of most important that are currently under development are clinical relevance is the fact that HER2 overexpression recombinant immunotoxins directed against HER2 (Dean et correlates with a lack of response to anti-oestrogen hormonal al. 1998, Maurer-Gebhard et al. 1998, Rosenblum et al. therapy (Houston et al. 1999) and confers resistance to 1999). In addition, downregulation of HER2 expression by tamoxifen, an anti-oestrogen that is administered as the usage of HER2 antisense oligonucleotides enhances the endocrine therapy in breast cancer patients. growth inhibitory and pro-apoptotic activity of several distinct chemotherapeutic agents and thus may play a role in Herceptin – the first RTK-specific anti-oncogene future cancer therapy (Roh et al. 1999). drug Several approaches towards the selective prevention and EGFR interception of HER2-relevant signalling are possible (Bange et al. 2001). When it was demonstrated that MAb 4D5, a The EGFR is frequently overexpressed in non-small cell mAb directed against the extracellular domain of HER2, lung, bladder, cervical, ovarian, kidney, and pancreatic could specifically inhibit the proliferation of HER2- cancer and occurs with very high incidence in squamous cell overexpressing tumour cell lines and prevent tumour growth carcinomas of the head and neck (Salomon et al. 1995, in nude mice, an excellent weapon towards the suppression Grandis et al. 1996, Fontanini et al. 1998). The predominant of HER2-mediated signalling was established (Hudziak et al. mechanism leading to EGFR overexpression is EGFR gene 1989, Shepard et al. 1991). Herceptin, the recombinant amplification, with more than 15 copies per cell reported in ‘humanized’ version of murine MAb 4D5 developed by certain tumours (Velu 1990). In general, elevated levels of Genentech, has passed all clinical trials and has been EGFR expression are associated with late stage of disease approved by the US Food and Drug Administration (FDA) progression and often correlate with high metastatic rate and in 1998 for treatment of women with metastatic breast cancer increased rate of tumour proliferation (Pavelic et al. 1993). (Pegram et al. 1998) (Table 1). Using Herceptin as a In addition, tumorigenic changes in EGFR activity can also single-agent therapy for HER2-overexpressing breast cancer, occur via mutations that activate the receptor in the absence responses up to 15% have been reported (Baselga et al. 1996, of ligand binding. A number of EGFR deletions have been Cobleigh et al. 1999). Application of Herceptin in identified in human cancer and most of these mutations alter

Table 1 A summary of the most successful drugs against signalling which are currently evaluated in clinical phases or have already been approved by the FDA. A more detailed description is found in the text RTK Drug Company Description Status EGFR ZD18539 Iressa AstraZeneca TKI that inhibits EGFR signalling Phase III EGFR C225 ImClone Systems MAb directed against EGFR Phase III EGFR EGF fusion protein Seragen Recombinant diphtheria toxin–hEGF fusion Phase II protein HER2 Herceptin Genentech MAb directed against HER2 Approved by the FDA in 1998 IGF-IR INX-4437 INEX USA Antisense oligonucleotides targeting IGR-IR Phase I VEGFR SU5416 SUGEN TKI that inhibits VEGFR2 Phase II VEGFR/FGFR/ SU6668 SUGEN RTK inhibition of VEGFR,FGFR and PDGFR Phase I PDGFR FDA,Food and Drug Administration; MAb,monoclonal antibody; RTK,receptor tyrosine kinase; TKI,tyrosine kinase inhibitor. www.endocrinology.org Downloaded from Bioscientifica.com165 at 09/28/2021 02:58:10PM via free access Zwick et al.: RTKs in anti-cancer strategies the extracellular ligand binding domain of the receptor and shows a significant anti-tumour effect on human breast and result in a truncated EGFR with constitutively active kinase colon cancer cells that is synergistically enhanced in function (Tang et al. 2000). combination with various cytotoxic agents (Ciardiello et al. A very potent mechanism of constitutive EGFR 2000) and is currently being evaluated in phase II clinical activation in a variety of human cancers is autocrine trials. stimulation via growth factor loops. The most prominent ligand, which is involved in autocrine growth receptor Insulin growthfactor receptor (IGFR) α activation, is TGF (Seth et al. 1999, Hsieh et al. 2000). family Coexpression of TGFα and EGFR is frequently observed in glioblastomas and squamous cell carcinomas of the head and Structurally quite different from the EGFR family are the neck where it is correlated with poor prognosis (Grandis et (IR) and the insulin-like growth factor (IGF) al. 1998). receptor (IGF-IR). Both receptors consist of two extracellular Interestingly, it was recently shown that the G α subunits, which are responsible for ligand binding, and two protein-coupled receptor (GPCR)-induced cleavage of membrane-spanning β subunits bearing the tyrosine kinase EGF-like growth factors leads to EGFR transactivation and domain and the autophosphorylation sites (Yarden & Ullrich EGFR-related signalling in cancer cells (Gschwind et al. 1988). The ligands for the two receptors include insulin, 2001). Mechanistically, GPCR stimulation activates a IGF-I and IGF-II. While insulin is mostly a metabolic metalloprotease that cleaves a transmembrane EGF-like , IGF-I and IGF-II are crucial for normal ligand precursor and allows the released growth factor to development and . Interestingly, in the transactivate the EGFR (Prenzel et al. 2001). Moreover, circulation IGFs are found to be complexed to a number of Prenzel et al. demonstrated that the metalloprotease inhibitor different IGF-binding proteins (IGFBPs), which serve as batimastat blocks bombesin-induced transactivation of the transport vehicles for these ligands and modify the stability EGFR in PC3 human cells and significantly and the proliferative effect of the growth factors (Jones & reduces the high constitutive level of EGFR tyrosine Clemmons 1995). phosphorylation (Prenzel et al. 1999). This finding suggests In the last 10 years it has been demonstrated that IGF-IR that GPCR–EGFR cross-communication plays a prominent and its ligands are involved in the pathogenesis of a variety role in the development and progression of human cancer. of human tumours, particular in breast and prostate cancer (Perks & Holly 2000, Surmacz 2000). In primary breast EGFR as a target for anti-cancer therapies tumours, IGF-I and IGF-II are predominantly expressed in Since aberrant EGFR signalling is implicated in many stromal fibroblasts surrounding the normal and malignant cancers and seems to correlate with poor prognosis, it is an breast (Gebauer et al. 1998, Rasmussen & Cullen excellent target for anti-cancer therapy (Huang & Harari 1998), whereas the IGF-IR is overexpressed in breast cancer 1999, Hong & Ullrich 2000). Currently, the monoclonal cells and shows enhanced tyrosine kinase activity (Resnik et antibody-based treatments using cetuximab (C225) represent al. 1998). These findings indicate that IGFs are able to the most successful anti-EGFR therapies (Table 1). C225 is stimulate breast growth in a paracrine manner. directed against the extracellular domain of the EGFR and Moreover, it has been demonstrated that high plasma IGF-I inhibits receptor activation and downstream signalling by levels correlate with an elevated prostate cancer risk and blocking ligand binding to the EGFR and inducing receptor several prostate cancer cell lines were shown to be internalization. The growth inhibiting and anti-tumour effect responsive to IGF-I (Chan et al. 1998). On the basis of these of C225 was shown in a variety of human cancer cells results several approaches have been initiated to develop new including pancreatic, renal and breast carcinomas (Prewett et small molecule inhibitors (Blum et al. 2000) or other al. 1998, Overholser et al. 2000). Therefore, it is now strategies to interfere with the pathogenic IGF-IR signalling undergoing a number of clinical trials either alone or in (Brodt et al. 2000) (Table 1). combination with chemotherapeutic treatments such as cisplatin, paclitaxel or gemcitabine (Baselga et al. 2000, Vascular endothelial growth factor Bruns et al. 2000, Inoue et al. 2000). receptor (VEGFR) family Inhibition of the tyrosine kinase activity by TKIs represents another promising approach towards the The vascular endothelial growth factor (VEGF) is one of the intervention of EGFR signalling in cancer cells. The most main inducers of endothelial cell proliferation and advanced of these compounds are ATP analogues of the permeability of blood vessels. The two RTKs that bind quinazoline and pyridopyrimidine family, which interact with VEGFs, VEGFR-1 and VEGFR-2, are expressed on the conserved ATP-binding site in the kinase domain of the endothelial cells during embryonic development and are the receptor and thus block EGFR activation (Levitzki 1992, key regulators for , a process which leads to the Noonberg & Benz 2000). The quinazoline ZD-1839 (Iressa) formation of new blood vessels developing from pre-existing

166 Downloadedwww.endocrinology.org from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Endocrine-Related Cancer (2001) 8 161–173 ones. formation is usually quiescent in the adult only in normal development but also in tumour formation organism except for wound repair and the female menstrual and progression. Two classes of FGFRs were discovered in cycle. However, expansion of solid tumours beyond a the past. The first class comprise the four high affinity diameter of 1–2 mm requires de novo formation of a vascular FGFRs, whereas the second class is defined by low affinity network that provides the growing tumour with oxygen and FGF binding sites. Considerable evidence indicates that those essential nutrients. Many studies have provided evidence for low affinity binding sites represent heparan sulphate the role of the VEGF–VEGFR ligand–receptor system in proteoglycan molecules (HSPG) located on the cell surface. tumour vascularization and metastasis. Mechanistically, the These HSPG receptors may support the fine tuning of cell tumour secretes VEGF that in turn activates VEGFR-2 and responses to the FGFs present and also regulate their induces proliferation of stromal endothelial cells and availability and their transport within a tissue. sprouting of new blood vessels. The critical role of VEGFR-2 The first members of the large FGF family to be in the above-mentioned process was demonstrated by identified were the FGF-1 (aFGF) and FGF-2 (bFGF). They delivering of a retrovirus encoding a dominant negative were purified on the basis of their mitogenic activity towards VEGFR-2 into nude mice which prevented glioblastoma fibroblasts. Both ligands are potent for a variety xenograft growth (Millauer et al. 1994). These findings have of cells of mesodermal, ectodermal and endodermal origin opened a new avenue of cancer therapies and several (Basilico & Moscatelli 1992). In addition, they play a role as strategies have been developed for targeting the VEGFR positive regulators for endothelial and pathway (Table 1). angiogenesis (Folkman & Shing 1992). Inhibition of angiogenesis has several advantages The family of the high affinity FGFRs comprises four compared with other attempts in cancer therapy. This members: FGFR1 (flg), FGFR2 (bek), FGFR3 and FGFR4. includes the fact that endothelial cells are easily accessible Ligand binding to FGFRs induces dimerization and from the blood stream and are not likely to develop resistance phosphorylation of the cytoplasmic tyrosine residues, but full to cancer therapy, since endothelial cells are genetically activation is only achieved in the presence of heparin stable compared with tumour cells (Boehm et al. 1997). (Spivak-Kroizman et al. 1994). A common explanation is Therefore, over 20 anti-angiogenic drugs, which prevent that heparin is able to bind a number of monovalent FGFs endothelial cell proliferation directly by blocking activators allowing the formation of receptor oliogomeres, which bind of angiogenesis like the VEGF and fibroblast growth factor to the clustered FGFs. (FGF) receptors, are currently undergoing evaluation in phase I, II or III clinical trials (Table 1). For example the FGFs and FGFRs in cancer development small molecule inhibitor SU5416, which specifically inactivates VEGFR-2 (Fong et al. 1999), is the most The fact that a remarkable number of FGFs were identified advanced in clinical trials for the treatment of a variety of as isolated from tumours due to their ability to induce solid tumours (National Institutes of Health). The drug was proliferation or transformation of fibroblasts indicates their well tolerated in phase I studies and stable disease was functional role in tumorigenesis. Indeed, a number of studies demonstrated in a variety of tumours like Kaposi’s , point out that changes in the expression patterns of FGFs non-small cell , and breast may contribute to growth deregulation of human tumour cells cancer (Mendel et al. 2000, Fong et al. 1999). Another (Eguchi et al. 1992, Kornmann et al. 1997). Furthermore, it synthetic RTK inhibitor currently being evaluated is the has been demonstrated that expression of FGF-1, FGF-2 or tyrosine kinase inhibitor SU6668 that targets not only the FGF-8b in fibroblasts leads to transformation and tumour VEGFR but also the fibroblast growth factor receptor formation in nude mice. In addition, strategies to interfere (FGFR) and the -derived growth factor receptor with growth factor action, such as dominant negative FGFRs (PDGFR) (Laird et al. 2000). Both receptors are additionally or anti-sense oligonucleotides, result in impaired tumour involved in endothelial cell growth and angiogenesis. Other growth in nude mice (Becker et al. 1992, Li et al. 1994, active VEGFR-2 inhibitors that are now undergoing clinical Wang & Becker 1997, Auguste et al. 2001). These data trials are the TKIs PTK787, ZK22584 and a mAb that targets indicate that endogenous FGFs are autocrine mediators of the VEGFR-2 ligand. neoplastic cell growth. However, changes at the level of FGFRs, such as point growthfactors receptor (FGFR) mutations, elevated expression or different splicing, result in family dis-regulated FGFR signalling and have been identified in a variety of human tumours. For example, FGFR The fibroblast growth factors (FGFs) represent the largest overexpression was observed in tumours arising from family of growth factor ligands. To date more than 20 numerous tissues, including breast, prostate, , distinct members have been identified. The FGFs and their and salivary gland (Myoken et al. 1996, Shingu et designated receptors (FGFRs) appear to play critical roles not al. 1998, Giri et al. 1999). Somatic mutations in the FGFR-3, www.endocrinology.org Downloaded from Bioscientifica.com167 at 09/28/2021 02:58:10PM via free access Zwick et al.: RTKs in anti-cancer strategies similar to the activating mutations in skeletal dysplasias, (Jeffers et al. 1998). Somatic mutations in the HGFR have have been identified in and in multiple been demonstrated in childhood hepatocellular (Park et al. myeloma (Chesi et al. 1997, Capellen et al. 1999). 1999) as well as head and neck squamous cell carcinomas Since FGFR signalling shows a physiological profile of (Di Renzo et al. 2000). action that includes mitogenic and angiogenic activity and is Since HGF/SF is synthesized as a single chain precursor frequently altered in human tumours, this system might be a polypeptide devoid of biological activity and can be activated useful target for anti-cancer therapy. Suramin, a compound through a critical step involving proteolytic cleavage, this that complexes heparin-binding growth factors, such as initial step of HGF/SF activation provides a possible point FGF-2, has been shown to inhibit cell migration and of interference by potential inhibitors. In the past few years FGF-2-mediated induction of -plasminogen numerous inhibitors and antagonists have been identified activator (Takano et al. 1994). However, efficacy in clinical (Parr & Jiang 2001). These factors have demonstrated a trials is often limited by toxicity. Targeting the FGFRs was possible role in minimizing the action of HGF/SF on cancer first achieved by using a toxin chimera between FGF-2 and cells, and may be of therapeutic value in the future. the potent ribosome-inactivating protein saporin and shows anti-tumour activity towards melanoma cells expressing high RET receptor tyrosine kinase FGFR levels (Beitz et al. 1992). Another possibility to block FGFR signalling is Multiple endocrine neoplasia type 2 (MEN2) is a dominant represented by the use of small molecule inhibitors such as autosomal inherited cancer syndrome which exists in three SU6668, which interferes with FGFR tyrosine kinase different subtypes and is characterized by the development activity. SU6668 has been shown to induce striking of medullary thyroid carcinoma. The gene responsible for regression of large established human tumour xenografts and MEN2 was identified as the Rearranged during investigations are ongoing in phase I clinical trials (Laird et Transformation (RET) RTK. The RET proto-oncogene al. 2000) (Table 1). encodes a protein that is characterized by a cadherin-like and a cysteine-rich domain in the extracellular part of the Hepatocyte growthfactor receptor receptor. RET is expressed during embryogenesis in the peripheral nervous system and in the urogenital system and (HGFR) is involved in the development of the and the The receptor (HGFR), which is kidney (Edery et al. 1997). encoded by the proto-oncogene met (Naldini et al. 1991), MEN2 cancer syndromes are caused by dominant was identified as a regulator of a variety of morphogenic activating germline mutations in the RET proto-oncogene. processes like cell migration, cell scattering and invasion of For example, mutations at five different cysteine residues extracellular matrices (Birchmeier & Gherardi 1998). HGFR have been found in the extracellular domain, which result in is a disulphide-linked heterodimer with a glycosylated an unpaired cysteine and intermolecular disulphide bonding, extracellular α-chain and a β-chain, which consists of the leading to constitutively activated receptors (Chappuis- transmembrane and cytoplasmic tyrosine kinase domain Flament et al. 1998). (Giordano et al. 1989). Hepatocyte growth factor (HGF) or Interestingly, in 50% of patients with Hirschsprung scatter factor (SF), the corresponding ligand, is expressed in disease (HSCR), a disorder characterized by the absence of mesenchymal-derived cells where it is suggested to act in a enteric ganglia, gene deletions or point mutations that are paracrine manner on epithelial cells in close proximity and predicted to inactivate the RET receptor have been identified has been described as a growth modulator for hepatocytes, (Pasini et al. 1996, Eng & Mulligan 1997). Therefore, the melanocytes and in vitro (Kan et al. 1991, RET proto-oncogene is a unique example of either dominant Sonnenberg et al. 1993). Since HGF/SF is an important gain-of-function or dominant loss-of-function mutations in motility factor it might influence the migration of tumour the same gene associated with severe human disorders. cells and may facilitate invasive growth and metastasis. For example, HGF overexpression was demonstrated in a variety Platelet-derived growthfactor receptor of human tumours, such as thyroid and colorectal carcinomas (PDGFR) family and seems to have prognostic significance for non-small lung and breast cancer (Di Renzo et al. 1995, Siegfried et al. The two RTKs platelet-derived growth factor receptor 1997). HGFR mutations have been identified in patients with (PDGFR) and Kit are members of the PDGFR family. These inherited predisposition to develop multiple papillary renal proteins are characterized by an extracellular domain with cell carcinomas (HPRCC) (Schmidt et al. 1997). Most of five Ig-like domains and an intracellular tyrosine kinase these mutations lie adjacent to the kinase domain, leading to domain split by 100 amino acids. Two genes encoding enhanced enzymatic activity, transformation of fibroblasts PDGFR-α and PDGFR-β have been identified and both and invasive growth as shown by in vitro experiments receptors are activated by a ligand dimer consisting of

168 Downloadedwww.endocrinology.org from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Endocrine-Related Cancer (2001) 8 161–173

PDGF-A and/or PDGF-B. This in turn leads to receptor Auguste P, Gursel DB, Lemiere S, Reimers D, Cuevas P, Carceller dimerization with three possible configurations: αα, ββ, αβ F, Di Santo JP & Bikfalvi A 2001 Inhibition of fibroblast (Heldin et al. 1998). Coexpression of PDGFR and its ligands growth factor/fibroblast growth factor receptor activity in glioma cells impedes tumor growth by both angiogenesis-dependent and has been identified in glioblastoma and other human -independent mechanisms. Cancer Research 61 1717–1726. astrocytotic tumours, whereas normal brain tissue does Bange J, Zwick E & Ullrich A 2001 Molecular targets for breast not express these proteins (Hermanson et al. 1992). These cancer therapy and prevention. Nature Medicine 7 8–12. findings suggest an autocrine loop that stimulates the Bargmann CI, Hung MC & Weinberg RA 1986 Multiple uncontrolled growth of human brain tumours. However, the independent activations of the neu oncogene by a point mutation PDGF receptor–ligand system may exert its main function in altering the transmembrane domain of p185. Cell 45 649–657. Baselga J, Tripathy D, Mendelsohn J, Baughman S, Benz CC, tumour progression via the stimulation of neovascularization Dantis L, Sklarin NT, Seidman AD, Hudis CA, Moore J, Rosen of solid tumours (Plate et al. 1994, Dunn et al. 2000). PP, Twaddell T, Henderson IC & Norton L 1996 Phase II study The RTK Kit is predominantly expressed in mast cells, of weekly intravenous recombinant humanized anti-p185HER2 melanocytes and marrow (Wang et al. 1989), whereas monoclonal antibody in patients with HER2/neu-overexpressing its cognate ligand, (SCF), is found in stromal metastatic breast cancer. Journal of Clinical Oncology 14 737– cells, fibroblasts and endothelial cells (Heinrich et al. 1993). 744. The involvement of Kit in human malignancy is paradoxical. Baselga J, Pfister D, Cooper MR, Cohen R, Burtness B, Bos M, D’Andrea G, Seidman A, Norton L, Gunnett K, Falcey J, On the one hand functional expression of Kit has a positive Anderson V, Waksal H & Mendelsohn J 2000 Phase I studies of effect on the growth of small lung cancer cells (Krystal et al. anti-epidermal growth factor receptor chimeric antibody C225 1996) and mutations in Kit have been identified in patients alone and in combination with cisplatin. Journal of Clinical with familial and sporadic gastrointestinal tumours Oncology 18 904–914. (Moskaluk et al. 1999). On the other hand, loss of Kit is Basilico C & Moscatelli D 1992 The FGF family of growth factors associated with progression of melanoma and thyroid and . Advances in Cancer Research 59 115–165. carcinoma (Natali et al. 1992, 1995), suggesting a tumour Becker D, Lee PL, Rodeck U & Herlyn M 1992 Inhibition of the fibroblast growth factor receptor 1 (FGFR-1) gene in human suppressor function in this particular tumour type. melanocytes and malignant leads to inhibition of proliferation and signs indicative of differentiation. Oncogene 7 2303–2313. Conclusion Beitz JG, Davol P, Clark JW, Kato J, Medina M, Frackelton AR, Lappi DA, Baird A & Calabresi P 1992 Antitumor activity of Over the past decade tremendous progress has been made in basic fibroblast growth factor-saporin mitotoxin in vitro and in the elucidation of cellular signalling mechanisms and the vivo. Cancer Research 52 227–230. establishment of molecular pathways activated by RTKs and Birchmeier C & Gherardi E 1998 Developmental roles of hgf/sf their ligands in normal and malignant cells. Empirical and its receptor, the c-met tyrosine kinase. Trends in Cell observations and experimental studies led to an emerging Biology 8 404–410. consensus that several classes of RTKs and polypeptide Blum G, Gazit A & Levitzki A 2000 Substrate competitive growth factors play an important role in cancer management. inhibitors of IGF-I receptor kinase. Biochemistry 39 15705– 15712. Therefore, the potential of RTKs and their relevant signalling Boehm T, Folkman J, Browder T & O’Reilly MS 1997 as selective anti-cancer targets for therapeutic intervention has Antiangiogenic therapy of experimental cancer does not induce been recognized. As a consequence, a variety of successful acquired drug resistance. Nature 390 404–407. target-specific drugs such as mAbs and RTK inhibitors have Brennan PJ, Kumogai T, Berezov A, Murali R & Greene MI 2000 been developed and are currently evaluated in clinical trials. HER2/neu: mechanisms of dimerization/oligomerization. Moreover, the growing field of rational drug design and Oncogene 19 6093–6101. Brodt P, Samani A & Navab R 2000 Inhibition of the type I computational prediction, in combination with advanced insulin-like growth factor receptor expression and signaling: molecular understanding of cell signalling and genomic novel strategies for antimetastatic therapy. Biochemical profiling, may make possible the development of an entire new 60 1101–1107. generation of target-specific and molecularly based drugs and Bruns CJ, Solorzano CC, Harbison MT, Ozawa S, Tsan R, Fan D, this promises a new era of anti-cancer therapy. Abbruzzese J, Traxler P, Buchdunger E, Radinsky R & Fidler IJ 2000 Blockade of the epidermal growth factor receptor signaling by a novel tyrosine kinase inhibitor leads to apoptosis of References endothelial cells and therapy of human pancreatic carcinoma. Cancer Research 60 2926–2935. Al-Obeidi FA & Lam KS 2000 Development of inhibitors for Burden S & Yarden Y 1997 Neuregulins and their receptors: a protein tyrosine kinases. Oncogene 19 5690–5701. versatile signaling module in organogenesis and oncogenesis. Alroy I & Yarden Y 1997 The ErbB signaling network in Neuron 18 847–855. embryogenesis and oncogenesis: signal diversification through Burris HA 2000 Docetaxel (Taxotere) in HER-2-positive patients combinatorial ligand–receptor interactions. FEBS Letters 410 and in combination with trastuzumab (Herceptin). Seminars in 83–86. Oncology 27 19–23. www.endocrinology.org Downloaded from Bioscientifica.com169 at 09/28/2021 02:58:10PM via free access Zwick et al.: RTKs in anti-cancer strategies

Cappellen D, De Oliveira C, Ricol D, de Medina S, Bourdin J, Di Renzo MF, Olivero M, Martone T, Maffe A, Maggiora P, Sastre-Garau X, Chopin D, Thiery JP & Radvanyi F 1999 Stefani AD, Valente G, Giordano S, Cortesina G & Comoglio Frequent activating mutations of FGFR3 in human bladder and PM 2000 Somatic mutations of the met oncogene are selected cervix carcinomas. Nature Genetics 23 18–20. during metastatic spread of human hnsc carcinomas. Oncogene Chan JM, Stampfer MJ, Giovannucci E, Gann PH, Ma J, 19 1547–1555. Wilkinson P, Hennekens CH & Pollak M 1998 Plasma Dunn IF, Heese O & Black PM 2000 Growth factors in glioma insulin-like growth factor-I and prostate cancer risk: a angiogenesis: FGFs, PDGF, EGF, and TGFs. Journal of prospective study. Science 279 563–566. Neurooncology 50 121–137. Chappuis-Flament S, Pasini A, De Vita G, Segouffin-Cariou C, Edery P, Eng C, Munnich A & Lyonnet S 1997 Ret in human Fusco A, Attie T, Lenoir GM, Santoro M & Billaud M 1998 development and oncogenesis. Bioessays 19 389–395. Dual effect on the RET receptor of MEN 2 mutations affecting Eguchi J, Nomata K, Kanda S, Igawa T, Taide M, Koga S, specific extracytoplasmic . Oncogene 17 2851–2861. Matsuya F, Kanetake H & Saito Y 1992 and Chesi M, Nardini E, Brents LA, Schrock E, Ried T, Kuehl WM & immunohistochemical localization of basic fibroblast growth Bergsagel PL 1997 Frequent translocation t(4;14)(.3;q32.3) factor in renal cell carcinoma. Biochemical and Biophysical in multiple myeloma is associated with increased expression and Research Communications 183 937–944. activating mutations of fibroblast growth factor receptor 3. Eng C & Mulligan LM 1997 Mutations of the ret proto-oncogene Nature Genetics 16 260–264. in the multiple endocrine neoplasia type 2 syndromes, related Ciardiello F, Caputo R, Bianco R, Damiano V, Pomatico G, De sporadic tumours, and Hirschsprung disease. Human Mutation 9 Placido S, Bianco AR & Tortora G 2000 Antitumor effect and 97–109. potentiation of cytotoxic drugs activity in human cancer cells by Fan Z & Mendelsohn J 1998 Therapeutic application of ZD-1839 (Iressa), an epidermal growth factor receptor-selective anti-growth factor receptor antibodies. Current Opinion in tyrosine kinase inhibitor. Clinical Cancer Research 6 2053– Oncology 10 67–73. 2063. Farah RA, Clinchy B, Herrera L & Vitetta ES 1998 The Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, development of monoclonal antibodies for the therapy of cancer. Fehrenbacher L, Wolter JM, Paton V, Shak S, Lieberman G & Critical Reviews in Eukaryotic Gene Expression 8 321–345. Slamon DJ 1999 Multinational study of the efficacy and safety Folkman J & Shing Y 1992 Angiogenesis. Journal of Biological Chemistry 267 10931–10934. of humanized anti-HER2 monoclonal antibody in women who Fong TA, Shawver LK, Sun L, Tang C, App H, Powell TJ, Kim have HER2-overexpressing metastatic breast cancer that has YH, Hirth KP & McMahon G 1999 Su5416 is a potent and progressed after chemotherapy for metastatic disease. Journal of selective inhibitor of the vascular endothelial growth factor Clinical Oncology 17 2639–2648. receptor (flk-1/kdr) that inhibits tyrosine kinase catalysis, tumor Colomer R, Montero S, Lluch A, Ojeda B, Barnadas A, Casado vascularization, and growth of multiple tumor types. Cancer AB, Cortes-Funes H & Lloveras B 2000 Circulating HER2 Research 59 99–106. extracellular domain and resistance to chemotherapy in advanced Fontanini G, De Laurentiis M, Vignati S, Chine S, Lucchi M, breast cancer. Clinical Cancer Research 6 2356–2362. Silvestri V, Mussi A, De Placido S, Tortora G, Bianco AR, Cooke T, Reeves J, Lannigan A & Stanton P 2001 The value of Gullick W, Angeletti CA, Bevilacqua G & Ciardiello F 1998 the human epidermal growth factor receptor-2 (HER2) as a Evaluation of epidermal growth factor-related growth factors and prognostic marker. European Journal of Cancer 37 3–10. receptors and of neoangiogenesis in completely resected stage I– Coussens L, Yang-Feng TL, Liao YC, Chen E, Gray A, McGrath IIIA non-small-cell lung cancer: amphiregulin and microvessel J, Seeburg PH, Libermann TA, Schlessinger J, Francke U, count are independent prognostic indicators of survival. Clinical Levinson A & Ullrich A 1985 Tyrosine kinase receptor with Cancer Research 4 241–249. extensive homology to EGF receptor shares chromosomal Frankel AE, Kreitman RJ & Sausville EA 2000 Targeted toxins. location with neu oncogene. Science 230 1132–1139. Clinical Cancer Research 6 326–334. Cragg MS, French RR & Glennie MJ 1999 Signaling antibodies in Gebauer G, Jager W & Lang N 1998 Mrna expression of cancer therapy. Current Opinion in Immunology 11 541–547. components of the insulin-like growth factor system in breast Dean GS, Pusztai L, Xu FJ, O’Briant K, DeSombre K, Conaway cancer cell lines, tissues, and metastatic breast cancer cells. M, Boyer CM, Mendelsohn J & Bast RC 1998 Cell surface Anticancer Research 18 1191–1195. density of p185(c-erbB-2) determines susceptibility to Giordano S, Ponzetto C, Di Renzo MF, Cooper CS & Comoglio anti-p185(c-erbB-2)-ricin A chain (RTA) immunotoxin therapy PM 1989 Tyrosine kinase receptor indistinguishable from the alone and in combination with anti-p170(EGFR)-RTA in ovarian c-met protein. Nature 339 155–156. cancer cells. Clinical Cancer Research 4 2545–2550. Giri D, Ropiquet F & Ittmann M 1999 Alterations in expression of Derynck R, Goeddel DV, Ullrich A, Gutterman JU, Williams RD, basic fibroblast growth factor (FGF) 2 and its receptor FGFR-1 Bringman TS & Berger WH 1987 Synthesis of messenger RNAs in human prostate cancer. Clinical Cancer Research 5 1063– for transforming growth factors α and β and the epidermal 1071. growth factor by human tumors. Cancer Research 47 707–712. Grandis JR, Melhem MF, Barnes EL & Tweardy DJ 1996 Dhanasekaran N & Reddy PE 1998 Signaling by dual specificity Quantitative immunohistochemical analysis of transforming kinases. Oncogene 17 1447–1455. growth factor-alpha and epidermal growth factor receptor in Di Renzo MF, Olivero M, Giacomini A, Porte H, Chastre E, patients with squamous cell carcinoma of the head and neck. Mirossay L, Nordlinger B, Bretti S, Bottardi S & Giordano S Cancer 78 1284–1292. 1995 Overexpression and amplification of the met/hgf receptor Grandis JR, Chakraborty A, Zeng Q, Melhem MF & Tweardy DJ gene during the progression of colorectal cancer. Clinical 1998 Downmodulation of TGF-alpha protein expression with Cancer Research 1 147–154. antisense oligonucleotides inhibits proliferation of head and neck

170 Downloadedwww.endocrinology.org from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Endocrine-Related Cancer (2001) 8 161–173

squamous carcinoma but not normal mucosal epithelial cells. Vande Woude GF 1998 The mutationally activated met receptor Journal of Cellular Biochemistry 69 55–62. mediates motility and metastasis. PNAS 95 14417–14422. Greenfield C, Hiles I, Waterfield MD, Federwisch M, Wollmer A, Jones JI & Clemmons DR 1995 Insulin-like growth factors and Blundell TL & McDonald N 1989 Epidermal growth factor their binding proteins: biological actions. Endocrine Reviews 16 binding induces a in the external domain 3–34. of its receptor. EMBO Journal 8 4115–4123. Kaleko M, Rutter WJ & Miller AD 1990 Overexpression of the van der Geer P, Hunter T & Lindberg RA 1994 Receptor human insulin-like growth factor I receptor promotes protein-tyrosine kinases and their signal transduction pathway. ligand-dependent neoplastic transformation. Molecular and Annual Reviews in 10 251–337. Cellular Biology 10 464–473. Gschwind A, Zwick E, Prenzel N, Leserer M & Ullrich A 2001 Kan M, Zhang GH, Zarnegar R, Michalopoulos G, Myoken Y, Cell communication networks: epidermal growth factor receptor McKeehan WL & Stevens JI 1991 Hepatocyte growth factor/ transactivation as the paradigm for inter-receptor signal hepatopoietin a stimulates the growth of rat kidney proximal transmission. Oncogene 20 1594–1600. tubule epithelial cells (RPTE), rat nonparenchymal cells, Harari D & Yarden Y 2000 Molecular mechanisms underlying human melanoma cells, mouse keratinocytes and stimulates ErbB2/HER2 action in breast cancer. Oncogene 19 6102–6114. anchorage-independent growth of sv-40 transformed RPTE. Heinrich MC, Dooley DC, Freed AC, Band L, Hoatlin ME, Keeble Biochemical and Biophysical Research Communications 174 WW, Peters ST, Silvey KV, Ey FS, Kabat D et al. 1993 331–337. Constitutive expression of steel factor gene by human stromal Karunagaran D, Tzahar E, Beerli RR, Chen X, Graus-Porta D, cells. Blood 82 771–783. Ratzkin BJ, Seger R, Hynes NE & Yarden Y 1996 ErbB-2 is a Heldin CH 1995 Dimerization of cell surface receptors in signal common auxiliary subunit of NDF and EGF receptors: transduction. Cell 80 213–223. implications for breast cancer. EMBO Journal 15 254–264. Heldin CH 1996 Protein tyrosine kinase receptors. Cancer Surveys King CR, Kraus MH & Aaronson SA 1985 Amplification of a 27 7–24. novel v-erbB-related gene in a human mammary carcinoma. Heldin CH, Ostman A & Ronnstrand L 1998 Signal transduction Science 229 974–976. via platelet-derived growth factor receptors. Biochimica et Klohs WD, Fry DW & Kraker AJ 1997 Inhibitors of tyrosine Biophysica Acta 1378 F79–F113. kinases. Current Opinion in Oncology 9 562–568. Hermanson M, Funa K, Hartman M, Claesson-Welsh L, Heldin Kolibaba KS & Druker BJ 1997 Protein tyrosine kinases and CH, Westermark B & Nister M 1992 Platelet-derived growth cancer. Biochimica et Biophysica Acta 1333 F217–F248. factor and its receptor in human glioma tissue: expression of Kornmann M, Ishiwata T, Beger HG & Korc M 1997 Fibroblast messenger RNA and protein suggests the presence of autocrine growth factor-5 stimulates mitogenic signaling and is and paracrine loops. Cancer Research 52 3213–3219. overexpressed in human : evidence for Hong WK & Ullrich A 2000 The role of EGFR in solid tumors autocrine and paracrine actions. Oncogene 15 1417–1424. and implications for therapy. Oncology Biotherapeutics 1 Kreitman RJ 1999 Immunotoxins in cancer therapy. Current 1–29. Opinion in Immunology 11 570–578. Houston SJ, Plunkett TA, Barnes DM, Smith P, Rubens RD & Krystal GW, Hines SJ & Organ CP 1996 Autocrine growth of Miles DW 1999 Overexpression of c-erbB-2 is an independent small cell lung cancer mediated by coexpression of c- and marker of resistance to endocrine therapy in advanced breast stem cell factor. Cancer Research 56 370–376. cancer. British Journal of Cancer 79 1220–1226. Laird AD, Vajkoczy P, Shawver LK, Thurnher A, Liang C, Hsieh ET, Shepherd FA & Tsao MS 2000 Co-expression of Mohammadi M, Schlessinger J, Ullrich A, Hubbard SR, Blake epidermal growth factor receptor and transforming growth RA, Fong TA, Strawn LM, Sun L, Tang C, Hawtin R, Tang F, factor-alpha is independent of ras mutations in lung Shenoy N, Hirth KP, McMahon G & Cherrington 2000 SU6668 adenocarcinoma. Lung Cancer 29 151–157. is a potent antiangiogenic and antitumor agent that induces Huang SM & Harari PM 1999 Epidermal growth factor receptor regression of established tumors. Cancer Research 60 4152– inhibition in cancer therapy: biology, rationale and preliminary 4160. clinical results. Investigational New Drugs 17 259–269. Lane HA, Beuvink I, Motoyama AB, Daly JM, Neve RM & Hynes Hudson PJ 1999 Recombinant antibody constructs in cancer NE 2000 ErbB2 potentiates breast tumor proliferation through therapy. Current Opinion in Immunology 11 548–557. modulation of p27Kip1–Cdk2 complex formation: receptor Hudziak RM, Lewis GD, Winget M, Fendly BM, Shepard HM & overexpression does not determine growth dependency. Ullrich A 1989 p185HER2 monoclonal antibody has Molecular and Cellular Biology 20 3210–3223. antiproliferative effects in vitro and sensitizes human breast Lee RJ, Albanese C, Fu M, D’Amico M, Lin B, Watanabe G, tumor cells to tumor necrosis factor. Molecular and Cellular Haines GK, Siegel PM, Hung MC, Yarden Y, Horowitz JM, Biology 9 1165–1172. Muller WJ & Pestell RG 2000 Cyclin D1 is required for Hynes NE & Stern DF 1994 The biology of erbB-2/neu/HER-2 transformation by activated Neu and is induced through an and its role in cancer. Biochimica et Biophysica Acta 1198 E2F-dependent signaling pathway. Molecular and Cellular 165–184. Biology 20 672–683. Inoue K, Slaton JW, Perrotte P, Davis DW, Bruns CJ, Hicklin DJ, Levitzki A 1992 Tyrphostins: tyrosine kinase blockers as novel McConkey DJ, Sweeney P, Radinsky R & Dinney CP 2000 antiproliferative agents and dissectors of signal transduction. Paclitaxel enhances the effects of the anti-epidermal growth FASEB Journal 6 3275–3282. factor receptor monoclonal antibody ImClone C225 in mice with Levitzki A 1999 Protein tyrosine kinase inhibitors as novel metastatic human bladder transitional cell carcinoma. Clinical therapeutic agents. Pharmacology and Therapy 82 231–239. Cancer Research 6 4874–4884. Levitzki A & Gazit A 1995 Tyrosine kinase inhibition: an Jeffers M, Fiscella M, Webb CP, Anver M, Koochekpour S & approach to drug development. Science 267 1782–1788. www.endocrinology.org Downloaded from Bioscientifica.com171 at 09/28/2021 02:58:10PM via free access Zwick et al.: RTKs in anti-cancer strategies

Li Y, Basilico C & Mansukhani A 1994 Cell transformation by Papewalis J, Nikitin AY & Rajewsky MF 1991 G to A fibroblast growth factors can be suppressed by truncated polymorphism at amino acid codon 655 of the human erbB-2/ fibroblast growth factor receptors. Molecular and Cellular HER2 gene. Nucleic Acids Research 19 5452. Biology 14 7660–7669. Park WS, Dong SM, Kim SY, Na EY, Shin MS, Pi JH, Kim BJ, Lonardo F, Di Marco E, King CR, Pierce JH, Segatto O, Aaronson Bae JH, Hong YK, Lee KS, Lee SH, Yoo NJ, Jang JJ, Pack S, SA & Di Fiore PP 1990 The normal erbB-2 product is an Zhuang Z, Schmidt L, Zbar B & Lee JY 1999 Somatic atypical receptor-like tyrosine kinase with constitutive activity in mutations in the kinase domain of the met/hepatocyte growth the absence of ligand. New Biology 2 992–1003. factor receptor gene in childhood hepatocellular carcinomas. Marcusson EG, Yacyshyn BR, Shanahan WR Jr & Dean NM 1999 Cancer Research 59 307–310. Preclinical and clinical pharmacology of antisense Parr C & Jiang WG 2001 Hepatocyte growth factor activators, oligonucleotides. Molecular Biotechnology 12 1–11. inhibitors and antagonists and their implication in cancer Maurer-Gebhard M, Schmidt M, Azemar M, Altenschmidt U, intervention. Histology and Histolpathology 16 251–268. Stocklin E, Wels W & Groner B 1998 Systemic treatment with a Pasini B, Ceccherini I & Romeo G 1996 Ret mutations in human recombinant erbB-2 receptor-specific tumor toxin efficiently disease. Trends in Genetics 12 138–144. reduces pulmonary metastases in mice injected with genetically Pavelic K, Banjac Z, Pavelic J & Spaventi S 1993 Evidence for a modified carcinoma cells. Cancer Research 58 2661–2666. role of EGF receptor in the progression of human lung Mendel DB, Laird AD, Smolich BD, Blake RA, Liang C, Hannah carcinoma. Anticancer Research 13 1133–1138. AL, Shaheen RM, Ellis LM, Weitman S, Shawver LK & Pawlak W, Zolnierek J, Sarosiek T & Szczylik C 2000 Antisense Cherrington JM 2000 Development of SU5416, a selective small therapy in cancer. Cancer Treatment Reviews 26 333–350. molecule inhibitor of VEGF receptor tyrosine kinase activity, as Pawson T 1995 Protein modules and signalling networks. Nature an anti-angiogenesis agent. Anticancer Drug Design 15 29–41. 373 573–580. Millauer B, Shawver LK, Plate KH, Risau W & Ullrich A 1994 Pegram MD, Lipton A, Hayes DF, Weber BL, Baselga JM, Glioblastoma growth inhibited in vivo by a dominant-negative Tripathy D, Baly D, Baughman SA, Twaddell T, Glaspy JA & flk-1 mutant. Nature 367 576–579. Slamon DJ 1998 Phase II study of receptor-enhanced Moskaluk CA, Tian Q, Marshall CR, Rumpel CA, Franquemont chemosensitivity using recombinant humanized anti-p185HER2/ DW & Frierson HF 1999 Mutations of c-kit jm domain are neu monoclonal antibody plus cisplatin in patients with HER2/ found in a minority of human gastrointestinal stromal tumors. neu-overexpressing metastatic breast cancer refractory to Oncogene 18 1897–1902. chemotherapy treatment. Journal of Clinical Oncology 16 2659– Myoken Y, Okamoto T, Sato JD, Kan M, McKeehan WL, 2671. Nakahara M & Takada K 1996 Immunohistochemical study of Perks CM & Holly JM 2000 Insulin-like growth factor binding overexpression of fibroblast growth factor-1 (FGF-1), FGF-2, proteins (IGFBPs) in breast cancer. Journal of and FGF receptor-1 in human malignant salivary gland tumours. Biology and Neoplasia 5 75–84. Journal of Pathology 178 429–436. Pietras RJ, Poen JC, Gallardo D, Wongvipat PN, Lee HJ & Naldini L, Vigna E, Narsimhan RP, Gaudino G, Zarnegar R, Slamon DJ 1999 Monoclonal antibody to HER-2/neureceptor Michalopoulos GK & Comoglio PM 1991 Hepatocyte growth modulates repair of radiation-induced DNA damage and factor (hgf) stimulates the tyrosine kinase activity of the receptor enhances radiosensitivity of human breast cancer cells encoded by the proto-oncogene c-met. Oncogene 6 501–504. overexpressing this oncogene. Cancer Research 59 1347–1355. Natali PG, Nicotra MR, Winkler AB, Cavaliere R, Bigotti A & Plate KH, Breier G & Risau W 1994 Molecular mechanisms of Ullrich A 1992 Progression of human cutaneous melanoma is developmental and tumor angiogenesis. Brain Pathology 4 associated with loss of expression of c-kit proto-oncogene 207–218. receptor. International Journal of Cancer 52 197–201. Plowman GD, Ullrich A & Shawver LK 1994 Receptor tyrosine Natali PG, Berlingieri MT, Nicotra MR, Fusco A, Santoro E, kinases as targets for drug intervention. Drug News & Bigotti A & Vecchio G 1995 Transformation of thyroid Perspectives 7 334–339. epithelium is associated with loss of c-kit receptor. Cancer Prenzel N, Zwick E, Daub H, Leserer M, Abraham R, Wallasch Research 55 1787–1791. C & Ullrich A 1999 EGF receptor transactivation by National Institutes of Health www.nci.nih.gov G-protein-coupled receptors requires cleavage Neve RM, Sutterluty H, Pullen N, Lane HA, Daly JM, Krek W & of proHB-EGF. Nature 402 884–888. Hynes NE 2000 Effects of oncogenic ErbB2 on G1 cell cycle Prenzel N, Fischer OM, Streit S, Hart S & Ullrich A 2001 regulators in breast tumour cells. Oncogene 19 1647–1656. The epidermal growth factor receptor family as a central Noonberg SB & Benz CC 2000 Tyrosine kinase inhibitors targeted element for cellular signal transduction and diversification. to the epidermal growth factor receptor subfamily: role as Endocrine-Related Cancer 8 11–31. anticancer agents. Drugs 59 753–767. Prewett M, Rothman M, Waksal H, Feldman M, Bander NH & Overholser JP, Prewett MC, Hooper AT, Waksal HW & Hicklin Hicklin DJ 1998 Mouse–human chimeric anti-epidermal growth DJ 2000 Epidermal growth factor receptor blockade by antibody factor receptor antibody C225 inhibits the growth of human IMC-C225 inhibits growth of a human pancreatic carcinoma renal cell carcinoma xenografts in nude mice. Clinical Cancer xenograft in nude mice. Cancer 89 74–82. Research 4 2957–2966. Paik S, Hazan R, Fisher ER, Sass RE, Fisher B, Redmond C, Prewett M, Huber J, Li Y, Santiago A, O’Connor W, King K, Schlessinger J, Lippman ME & King CR 1990 Pathologic Overholser J, Hooper A, Pytowski B, Witte L, Bohlen P & findings from the national surgical adjuvant breast and bowel Hicklin DJ 1999 Antivascular endothelial growth factor receptor project: prognostic significance of erbB-2 protein overexpression (fetal liver kinase 1) monoclonal antibody inhibits tumor in primary breast cancer. Journal of Clinical Oncology 8 103– angiogenesis and growth of several mouse and human tumors. 112. Cancer Research 59 5209–5218.

172 Downloadedwww.endocrinology.org from Bioscientifica.com at 09/28/2021 02:58:10PM via free access Endocrine-Related Cancer (2001) 8 161–173

Rasmussen AA & Cullen KJ 1998 Paracrine/autocrine regulation and survival with amplification of the HER2/neu oncogene. of breast cancer by the insulin-like growth factors. Breast Science 235 77–82. Cancer Research Treatment 47 219–233. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith Resnik JL, Reichart DB, Huey K, Webster NJ & Seely BL 1998 DE, Levin WJ, Stuart SG, Udove J, Ullrich A & Press MF 1989 Elevated insulin-like growth factor I receptor autophosphory- Studies of the HER-2/neu proto-oncogene in human breast and lation and kinase activity in human breast cancer. Cancer ovarian cancer. Science 244 707–771. Research 58 1159–1164. Sonnenberg E, Meyer D, Weidner KM & Birchmeier C 1993 Riese DJ II & Stern DF 1998 Specificity within the EGF family/ Scatter factor/hepatocyte growth factor and its receptor, the ErbB receptor family signaling network. Bioessays 20 41–48. c-met tyrosine kinase, can mediate a signal exchange between Robertson SC, Tynan JA & Donoghue DJ 2000 RTK mutations and epithelia during mouse development. Journal and human syndromes when good receptors turn bad. Trends in of Cell Biology 123 223–235. Spivak-Kroizman T, Lemmon MA, Dikic I, Ladbury JE, Pinchasi Genetics 16 265–271. D, Huang J, Jaye M, Crumley G, Schlessinger J & Lax I 1994 Roh H, Hirose CB, Boswell CB, Pippin JA & Drebin JA 1999 Heparin-induced oligomerization of FGF molecules is Synergistic antitumor effects of HER2/neu antisense responsible for FGF receptor dimerization, activation, and cell oligodeoxynucleotides and conventional chemotherapeutic proliferation. Cell 79 1015–1024. agents. Surgery 126 413–421. Stebbing J, Copson E & O’Reilly S 2000 Herceptin (trastuzamab) in Rosen L 2000 Antiangiogenic strategies and agents in clinical advanced breast cancer. Cancer Treatment Reviews 26 287–290. trials. Oncologist 5 (Suppl 1) 20–27. Surmacz E 2000 Function of the IGF-I receptor in breast cancer. Rosenblum MG, Shawver LK, Marks JW, Brink J, Cheung L & Journal of Mammary Gland Biology and Neoplasia 5 95–105. Langton-Webster B 1999 Recombinant immunotoxins directed Takano S, Gately S, Neville ME, Herblin WF, Gross JL, Engelhard against the c-erb-2/HER2/neu oncogene product: in vitro H, Perricone M, Eidsvoog K & Brem S 1994 Suramin, an cytotoxicity, , and in vivo efficacy studies in anticancer and angiosuppressive agent, inhibits endothelial cell xenograft models. Clinical Cancer Research 5 865–874. binding of basic fibroblast growth factor, migration, Salomon DS, Brandt R, Ciardiello F & Normanno N 1995 proliferation, and induction of urokinase-type plasminogen Epidermal growth factor-related and their receptors in activator. Cancer Research 54 2654–2660. human malignancies. Critical Reviews in Oncology and Tang CK, Gong XQ, Moscatello DK, Wong AJ & Lippman ME Hematology 19 183–232. 2000 Epidermal growth factor receptor vIII enhances Schechter AL, Stern DF, Vaidyanathan L, Decker SJ, Drebin JA, tumorigenicity in breast cancer. Cancer Research 60 3081–3087. Greene MI & Weinberg RA 1984 The neu oncogene: an Ullrich A & Schlessinger J 1990 Signal transduction by receptors erb-B-related gene encoding a 185,00-Mr tumour antigen. with tyrosine kinase activity. Cell 61 203–212. Ullrich A, Coussens L, Hayflick JS, Dull TJ, Gray A, Tam AW, Nature 312 513–516. Lee J, Yarden Y, Libermann TA, Schlessinger J, Downward J, Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, Mages ELV, Whittle N, Warfield MD & Seeburg PH 1984 Scherer SW, Zhuang Z, Lubensky I, Dean M, Allikmets R, Human epidermal growth factor receptor cDNA sequence and Chidambaram A, Bergerheim UR, Feltis JT, Casadevall C, aberrant expression of the amplified gene in A431 epidermoid Zamarron A, Bernues M, Richard S, Lips CJ, Walther MM, Tsui carcinoma cells. Nature 309 418–425. LC, Geil L, Orcutt ML, Stackhouse T, Zbar B et al. 1997 Velu TJ 1990 Structure, function and transforming potential of the Germline and somatic mutations in the tyrosine kinase domain epidermal growth factor receptor. Molecular and Cellular of the met proto-oncogene in papillary renal carcinomas. Nature Endocrinology 70 205–216. Genetics 16 68–73. Wang C, Curtis JE, Geissler EN, McCulloch EA & Minden MD Segal DM, Weiner GJ & Weiner LM 1999 Bispecific antibodies in 1989 The expression of the proto-oncogene c-kit in the blast cancer therapy. Current Opinion in Immunology 11 558–562. cells of acute myeloblastic leukemia. Leukemia 3 699–702. Seth D, Shaw K, Jazayeri J & Leedman PJ 1999 Complex Wang Y & Becker D 1997 Antisense targeting of basic fibroblast post-transcriptional regulation of EGF-receptor expression by growth factor and fibroblast growth factor receptor-1 in human EGF and TGF-alpha in human prostate cancer cells. British melanomas blocks intratumoral angiogenesis and tumor growth. Journal of Cancer 80 657–669. Nature Medicine 3 887–893. Shepard HM, Lewis GD, Sarup JC, Fendly BM, Maneval D, Weiner DB, Liu J, Cohen JA, Williams WV & Greene MI 1989 A Mordenti J, Figari I, Kotts CE, Palladino MA & Ullrich A 1991 point mutation in the neu oncogene mimics ligand induction of Monoclonal antibody therapy of human cancer: taking the HER2 receptor aggregation. Nature 339 230–231. protooncogene to the clinic. Journal of Clinical Immunology 11 Xie D, Shu XO, Deng Z, Wen WQ, Creek KE, Dai Q, Gao YT, Jin F & Zheng W 2000 Population-based, case-control study of 117–127. HER2 genetic polymorphism and breast cancer risk. Journal of Shingu K, Fujimori M, Ito K, Hama Y, Kasuga Y, Kobayashi S, National Cancer Institute 92 412–417. Itoh N & Amano J 1998 Expression of fibroblast growth factor-2 Yarden Y & Ullrich A 1988 Growth factor receptor tyrosine and fibroblast growth factor receptor-1 in thyroid diseases: kinases. Annual Reviews in Biochemistry 57 443–478. difference between and hyperplastic lesions. Yu D & Hung MC 2000 Overexpression of ErbB2 in cancer and Endocrinology Journal 45 35–43. ErbB2–targeting strategies. Oncogene 19 6115–6121. Siegfried JM, Weissfeld LA, Singh-Kaw P, Weyant RJ, Testa Zwick E, Wallasch C & Ullrich A 2000 HER2/neu: a target for JR & Landreneau RJ 1997 Association of immunoreactive breast cancer therapy. Breast Disease 11, 7–18. hepatocyte growth factor with poor survival in resectable Zhou BP, Liao Y, Xia W, Spohn B, Lee M-H & Hung M-C 2001 non-small cell lung cancer. Cancer Research 57 433–439. Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A & phosphorylation in HER2/nwu-overexpressing cells. Nature Cell McGuire WL 1987 Human breast cancer: correlation of relapse Biology 3 245–252. www.endocrinology.org Downloaded from Bioscientifica.com173 at 09/28/2021 02:58:10PM via free access Downloaded from Bioscientifica.com at 09/28/2021 02:58:10PM via free access