Progress in Growlh Facror Research, Vol. 4, pp. l-24. Printed in Great Britain. All rights reserved.

0955-2235/92 SC 1992 Pergamon

1992

THE TYPE 1 (EGFR-RELATED) GROWTH FACTOR RECEPTORS LIGANDS

$15.00 Press Ltd

FAMILY OF AND THEIR

Sally A. Prigent and Nicholas R. Lemoine ICRF Oncology Group Royal Postgraduate Medical School, Hammersmith Hospital Du Cane Road, London WI2 ONN, U.K.

This review considers the biology of the type 1 growth factor receptor family which is increasingly recognised as important in the control of normal cellproltferation and in the pathogenesis of human cancer. The family currently comprises three closely related members: the epidermal growth factor (EGF) receptor, c-erbB-2 and c-erbB-3, all of which show abnormalities of expression in various human tumours. The family offactors related to EGF has also expanded recently and now includes transforming growth factor alpha, heparin-binding EGF, amphiregulin, cripto and heregulin, as well as several other potential ligands for the c-erbB2-2 receptor. The involvement of these receptors and growth factors in human cancer has implications for the design of novelforms of therap?, ,for cancer, and we review recent advances andfuture avenues for investigation. Keywords: EGF receptor, c-erbB-2 (HERyneu), c-erbB-3 (HER3). amphiregulin, heregulin, oncogene, growth factor.

EGF, TGF alpha,

INTRODUCTION Epidermal growth factor was first extracted from mouse submaxillary glands [ 1] and was subsequently shown to have growth promoting effects and to stimulate uptake of glucose and amino acids by cells. Isolation of this ligand made possible the purification of the epidermal growth factor receptor (EGFR) from the squamous carcinoma cell line A43 1 in which it is overexpressed [2], and some years later its cDNA sequence [3,4, 5, 61 and genomic structure [7] were determined. The techniques of molecular biology facilitated the identification of two related putative receptors, c-erbB-2 (HER2) and more recently c-erbB-3 (HER3), by low stringency probing of human cDNA libraries with sequences from the EGFR (c-erbB-1) gene or its avian viral homologue v-erbB. While the signalling mechanism of the EGF receptor has been carefully examined, the Acknowledgements-We arc grateful to Bill Gullick for his critical reading of the manuscript and his helpful comments, and to Zoe Redley for her excellent typing.

2

S.A. Prigent and N.R. Lemoine

biology of c-erbB-2 and c-erbB-3 has been poorly defined, mainly because their natural ligands have not been identified. It is now clear that EGF is just one member of an expanding family of related peptides, of which some are capable of binding to the EGF receptor and others are candidate ligands for the c-erbB-2 and c-erbB-3 receptors. This review will describe our current knowledge of the members of the c-erbB family of receptors, their ligands and their involvement in human neoplasia. THE EPIDERMAL

GROWTH

FACTOR

RECEPTOR

The EGF receptor is a 170 kDa glycoprotein consisting of three domains: a 621 amino acid extracellular domain responsible for ligand recognition which contains a high proportion of cysteine residues clustered in two distinct regions; a 23 amino acid hydrophobic transmembrane region; and a 542 amino acid intracellular region containing a highly conserved tyrosine kinase domain (Fig. 1). Binding of ligand leads to receptor dimerisation which increases the catalytic activity of its tyrosine kinase [8]. Receptor autophosphorylation on tyrosine residues occurs at three major sites towards the C-terminus: Y-1068, Y-l 148, Y-l 173 [3], although two other potential sites have been reported 19, lo]. Mutational analysis suggests that phosphorylation of the three major sites is important for maximum biological activity [l 1, 121, and may be a necessary prerequisite to permit access of substrates to the catalytic site of the EGFR. The C-terminal tail may also contain a more proximal region involved in negative regulation and internalisation [13, 141. Following activation, the EGFR kinase phosphorylates a number of cellular substrates including phospholipase C gamma, MAP kinase and the ras GTPase-activating protein GAP, which, at least in the case of phospholipase C gamma, leads to an increase in catalytic activity [ 15, 16, 17, 18, 191. It has recently been shown that a small soluble actin-binding protein called profilin binds to the substrate phosphatidylinositol 4, 5-biphosphate and inhibits its hydrolysis by phospholipase C-gamma unless this enzyme has been phosphorylated by the EGF receptor tyrosine kinase [20]. The involvement of profilin may be the link that could explain the effects induced by EGF or TGF alpha on cellular morphology and motility. The EGF receptor can itself be a substrate for phosphorylation on threonine residues. Phosphorylation of threonine-654 has been shown to inhibit EGFR tyrosine kinase activity [21]. Recently two additional growth factor-stimulated kinases have been isolated which phosphorylate the EGF receptor at threonine669 [22]. This site has been reported to regulate internalisation and substrate phosphorylation on tyrosine residues 1231. Phosphorylation of EGFR by serine/ threonine kinases may be the mechanism by which platelet-derived growth factor, phorbol esters and other factors can influence the activity of the EGF receptor ~241. The activated receptor/ligand complex is endocytosed and degraded within the lysosomes, except in some cell types such as hepatocytes, where the receptor is recycled to the cell membrane. This process of internalisation may be essential for control of normal mitogenic signalling since a truncated EGFR mutant with normal kinase activity is not internalised and possesses increased transforming activity u41.

rvpe I Growth Factor Receptors

% amino acid

EGFR c-erbB2 c-erbB3

EGFFU

sequence EGFFU

Identity c-tiB21

42

I

46

46

45

45

46

44

41

50

41

82

60

62

I

TK TK

FIGURE 1. Diagrammatic representation of the sequence similarities of ditfere-nt domains of the EGF receptor, c-erbBt and c-erbB3. The percentage figares indicate the proportion of identical residoes shared by the indicated pairs of receptors. (Reproduced by permission from Cancer Topics, 1992 Vol. 8.)

Abnormalities of EGFR Signal Transduction

In pathological conditions the signal transduction pathway involving the EGF receptor can be subverted in various ways. Overexpression of the stimulating ligands TGF alpha and EGF, and/or the EGF receptor itself, occurs in a wide variety of human tumour types. EGF receptor overexpression may be associated with amplification of the gene or may occur in the presence of the normal gene copy number, presumably due to increased transcription or increased stability of the transcribed messenger RNA. Amplification of the EGF receptor locus may involve large amplicons of DNA spanning up to 1000 kb, and the contribution of flanking sequences and co-amplified genes to cellular transformation in these cases is not yet clear. In addition, structural alterations have been reported affecting both extracellular and intracellular domains of the receptor in the A431 vulva1 carcinoma cell line and in malignant gliomas, one type of human brain tumour [25, 26, 27, 28, 291. Overexpression of either ligand or receptor in the absence of the other does not usually result in full neoplastic transformation in vitro [30], but high expression of both components together can lead to transformation of a variety of cell types [31, 32, 33, 34, 351. However, transgenic animal experiments suggest that overexpression of the EGF receptor does not transform cells in vivo even when ligand is available [36].

4

S.A. Prigent and N.R. Lemoine

It is beyond the scope of this review to discuss the aberrant expression of EGFR in tumours and this topic has recently been reviewed in detail elsewhere [37]. In summary, the EGF receptor is overexpressed at high frequency in a wide range of tumours, commonly in squamous carcinomas of various sites and less commonly in adenocarcinomas (particularly in pancreatic and gastric cancers) [38, 391. Overexpression, sometimes associated with gene amplification, may be associated with poor prognosis in breast cancer, lung cancer and bladder cancer. THE c-e&El-2 PROTO-ONCOGENE The human c-erbB-2 proto-oncogene was identified independently by two groups [40,41] who probed human genomic libraries at low stringency with probes from the v-erbB oncogene (a truncated version of the EGF receptor from the avian erythroblastosis retrovirus). Amplification of the gene was found in a human breast cancer (MAC1 17) [42] and the MKN-7 gastric cancer cell line [43]. Subsequent work has shown that c-erbB-2 is amplified and overexpressed in a significant proportion of adenocarcinomas of various sites, but rarely in other types of malignant tumour. Analysis of the c-erbB-2 coding sequence shows that it has structural features of a growth factor receptor with close similarity to the EGF receptor protein [44] and the gene is sometimes referred to as HER-2, based on its homology to the human EGF Ieceptor. To determine whether c-erbB-2 can convey a mitogenic signal an elegant strategy has been employed [45, 461 to create a chimaeric protein consisting of the extracellular ligand binding domain of the EGF receptor fused to the cytoplasmic sequence of the human c-erbB-2 protein or the equivalent rat protein, neu. The chimaeric protein bound EGF, which stimulated autophosphorylation and mitogenesis, and was shown to induce increased transcription of fos, jun, the glucose transporter and ornithine decarboxylase genes [47]. A number of groups have shown that activation of the EGF receptor can result in cross-phosphorylation of the c-erbB-2 protein in cells that express both receptors [48, 49, 501. This might be achieved, at least in part, by heterodimerisation of the EGF receptor and the c-erbB-2 protein, which is induced by EGF binding and dramatically increases the capacity for self-phosphorylation of the dimerised receptor [51, 521. It seems likely that while there may be cellular substrates that are common to both receptor tyrosine kinases, there may be others that are specific for either the EGF receptor or c-erbB-2. In experiments with chimaeric receptors regions within the cytoplasmic domain have been identified that may determine this specificity [53, 541. The EGF receptor and c-erbB-2 protein can act in a synergistic fashion to transform rodent cells [55], but the mechanisms involved in this cooperation are still unclear although the capacity for formation of heterodimers is obviously a possibility [5 1,521. Mechanisms

of Activation

of c-erbB-2 to a Transforming

Oncogene

As with most proto-oncogenes c-erbB-2 can be activated to a transforming oncogene by several, quite different, mechanisms. The first indication of the role of the gene in cancer came from a rat experimental carcinogenesis model. Treatment of pregnant BDIX rats with the chemical carcinogen ethylnitrosourea leads to the development of

Type 1 Growth Factor Receptors

5

tumours of the CNS at high frequency. Bargmann and Weinberg [56] subsequently demonstrated that this was due to a single point mutation in the c-erbB-2 gene (in the rat referred to as the neu gene) causing a valine residue in the transmembrane region of the molecule to be converted to glutamic acid. This mutation stabilises receptors in a dimeric form in which they are catalytically active [57]. When the same mutation is introduced into the equivalent position (codon 659) of the human gene it will transform cells in culture [58] and the same mutation in the Drosophila EGF receptor (DER) greatly increases its tyrosine kinase activity [59]. It is intriguing that similar mutations in the transmembrane domain of the human EGF receptor do not apparently produce oncogenic activation [60,61]. We have used sensitive polymerase chain reaction (PCR) assays to detect such activating point mutations of c-e&B-2 and found no evidence of their occurrence in a large series of human tumours of various types [38,62,63,64,65]. Other groups have reached the same conclusion by sequence analysis in smaller numbers of human cancer DNAs [66, 671. Mutations at other codons in the transmembrane encoding region do not activate the transforming function of the neu or c-erhB-2 genes. There is some polymorphism of the surrounding sequence in normal individuals [68]. Expression of the mutant rat neu gene under the control of mouse mammary tumour virus long terminal repeat (MMTV LTR) causes the frequent development of breast cancers in transgenic mice [69,70] and introduction of the gene into mammary epithelial cells in the breast tissue of living rats produces tumours [71]. Expression of the wild-type or a mutant c-erbB-2 gene under the control of the MMTV promoter produced adenocarcinomas of several sites as well as B cell lymphomas [72]. The c-erbB-2 protein can be partially activated by truncation of the gene so that the extracellular portion of the molecule is deleted [58, 731. Such changes are observed in human tumours as rearrangements of the c-erbB-2 gene, but relatively few instances of this have been reported [see for example 74, 75, 76,771. The chief mechanism of activation of the c-erbB-2 gene in human cancers is by aberrantly high expression of the receptor protein. Gene amplification appears to be the predominant mechanism leading to elevated protein expression, at least in breast cancer. but overexpression by increased mRNA transcription alone does occur in a minority of cases [78, 791. In several breast cancer cell lines there is evidence of increased transcription of individual copies of c-erbB-2 contained in the amplicon, which suggests that increased transcription may precede amplification and be sustained after the increase in gene copy number [78]. The elements involved in the control of c-erbB-2 expression are under intensive study and it has recently been shown that oestrogen may have an inhibitory effect on c-erbB-2 transcription in breast cancer cells [80]. The role of abnormal c-erbB-2 expression in human tumours is currently uncertain, but it does not appear to be directly associated with cell proliferation. Attempts have been made to correlate c-erbB-2 overexpression with high rates of cell proliferation in invasive and in situ breast cancer [81,82,83] but experimental studies suggest that there may not be a simple relationship between expression of c-erbB-2 and human breast cancer cell proliferation in vivo and in vitro [80,84,85]. There is evidence that c-erbB-2 may contribute to the invasion and spread of tumours. Clinical studies have demonstrated that c-erbB-2 amplification and overexpression are correlated with aggressive tumour growth and poor prognosis in breast and ovarian cancer [66] and

6

S.A. Prigent and N.R. Lemoine

experimental studies show that the rat neu oncogene can induce the metastatic phenotype in transfected cells [86]. Expression

of Human c-erbB-2 in Normal Tissues and Tumours

The c-erbB-2 protein is expressed widely on normal human and animal tissues, but characteristically on secretory epithelia [87, 88, 89,901. The expression of c-erbB-2 in human tumours is reviewed in detail elsewhere [91] but we will briefly outline the major points here. Overexpression of the c-erbB-2 growth factor receptor occurs, usually as a consequence of gene amplification, in a fifth of breast, stomach, pancreatic, bladder and ovarian cancers. Overexpression occurs less frequently in some other adenocarcinomas and in squamous tumours of the lung. Overexpression is associated with poor prognosis in breast and ovarian cancers and possibly non-small cell lung cancers. THE c-e&B-3 PROTO-ONCOGENE Two groups have recently cloned a third member of the erbB receptor tyrosine kinase family. Aaronson and colleagues [92] detected the new member of the erbB protooncogene family by reduced stringency hybridisation of a v-erbB probe to normal human genomic DNA. Todaro and colleagues cloned a gene of almost identical sequence from the MDA-MB-361 and A431 cell lines using hybridisation with degenerate oligonucleotides encoding a conserved 7 amino acid sequence in the tyrosine kinase domain of the EGF receptor [93]. The predicted structure of the c-erbB-3 protein is closely similar to that of both the EGF receptor and the c-erbB-2 growth factor receptor (Fig. 1). The highest degree of sequence identity is in the tyrosine kinase domain where 60% of the residues are identical in EGFR and c-erbB-3. The kinase domains of EGFR and c-erbB-2 are more similar to each other (82% homology) than to that of c-erbB-3. The mature protein is extensively glycosylated and has a molecular weight of 160 kDa. Expression of c-erbB-3 transcripts was demonstrated in cultured keratinocytes and melanocytes but not in stromal fibroblasts, and was detected in 36 of 38 carcinomas but in only 2 of 12 sarcomas and 0 of 7 haematopoietic malignant cell lines. Overexpression of c-erbB-3 mRNA (in the absence of gene amplification or gross rearrangement) was found in 6 of 17 breast cancer cell lines. The possibility that c-erbB-3 expression might be relatively restricted to cells of epithelial or neuroectodermal origin is confirmed by our own studies using a panel of specific antibodies to this protein [94]. We find that while it is widely expressed in normal epithelial and some mesenchymal tissues, it is not generally found in haematopoietic or lymphoid cells. THE EGF FAMILY

OF GROWTH

FACTORS

Purification and sequence analysis of EGF revealed the presence of six conserved cysteine residues which cross-bond to create three peptide loops [95]. More recently several other peptides which interact with the EGF receptor have been purified and cloned and contain the same generalised motif X,CX,CX,,,CX,,CXCX,GX,CX,, where X represents any non-cysteine amino acid, and n is a variable number. These include TGF-alpha, amphiregulin, heparin-binding EGF and some virally-encoded

/

Type I Growth Fuctor Receptors

peptides. An interesting feature of these peptides is that they are all synthesised as much larger membrane-bound, glycosylated precursors which (at least in the case of EGF and TGF-alpha) have been shown to possess biological activity. Very recently it has been reported that a ligand for c-erbB-2 called Heregulin also possesses the same general structure and is synthesised as a larger precursor. The predicted secondary structure of five members of the EGF ligand family is shown schematically in Fig. 2. EGF-like sequences are not confined to growth factors but have been observed in a variety of cell-surface and extracellular proteins which have interesting properties in cell adhesion, protein-protein interaction and development [96]. These proteins include blood coagulation factors (factors VI, IX, X, XII, protein C, protein S, protein Z, tissue plasminogen activator, urokinase); extracellular matrix components (laminin, cytotactin, entactin); cell surface receptors (LDL receptor, thrombomodulin receptor) and immunity-related proteins (complement Clr, uromodulin). Interestingly, the general structural pattern of EGF-like precursors is preserved through lower organisms as well as in mammalian cells. A number of genes with developmental significance have been identified in invertebrates with EGF-like repeats. Perhaps the best characterised of these is the Notch gene of Drosophila which encodes 36 tandemly arranged 40 amino acid repeats which show homology to EGF [97]. Hydropathy plots indicate a putative membrane spanning domain, with the EGFrelated sequences being located on the extracellular side of membrane. Other homeotic genes with EGF-like repeats include Delta, 95F and 5ZD which were identified using probes based on Notch, and the nematode gene U-I.2 which encodes a putative receptor for a developmental signal transmitted between two specified cells. The conservation of EGF-like motifs between invertebrates and mammals is intriguing. It is interesting to speculate that EGF-like growth factors and the many other molecules containing EGF-like sequences which participate in a wide variety of functional systems, may have evolved from an ancestral cell-to-cell communication system. This review will consider only those proteins with the characteristics of growth factors. Epidermal Growth Factor

The history of the discovery of EGF and its structural and functional properties have recently been reviewed by Laurence and Gusterson [96]. During studies of nerve growth factor, a peptide was detected in extracts of submaxillary glands which induced precocious eyelid opening and incisor eruption by direct stimulation of epidermal growth and keratinisation [98]. A 53 amino acid peptide was purified from mouse submaxillary glands and sequenced [l]. The human equivalent, urogastrone, was subsequently isolated from urine [99]. The mRNA for EGF is approximately 4750 base pairs and encodes an EGF-precursor of 1217 amino acids [loo, 1011. Contained within this long coding region are the EGF coding region itself and nine other sequences with a high degree of homology to EGF. The mature EGF unit is that closest to the carboxyl terminus in the precursor protein. The protein begins with a 29 amino acid signal sequence and the precursor structure resembles a transmembrane receptor with a hydrophobic alpha helical membrane spanning domain and a region of high homology to the low density lipoprotein receptor [ 1021. Several mouse tissues, particularly distal renal tubule cells, do not process the 1217 amino acid precursor [ 1031, suggesting that EGF and the nine additional EGF-like sequences in the precursor may exist as a cell

8

S.A. Prigent and N.R. Lemoine

surface protein and have some recognition function. Studies using an EGF mini-gene expression system have recently shown that the membrane-anchored form of EGF is capable of stimulating EGF receptors on adjacent cells, similar to the situation for TGF-alpha [ 1041. The 3D structure of EGF has been determined by high resolution ‘H NMR and computational analysis, and functionally important residues have been located by a combination of site-directed mutagenesis and ‘H NMR [105]. EGF binds exclusively to the EGF receptor and not to c-erbB-2 or c-erbB-3. It is a potent stimulator of cell multiplication and a modulator of the differentiation and function of cells of various types [99, 106, 1071. EGF inhibits gastric acid secretion and protects the gastric mucosa from damage by the acid [IO& 1091 and has recently been associated with a novel cell lineage responsible for mucosal repair of the gastrointestinal tract [I 10, 111, 1121. It has been localised immunohistochemically to the submandibular salivary gland, serous glands of the nasal cavity, stomach, Brunner’s glands of the duodenum, anterior pituitary, bone marrow, sweat glands, mammary gland, ovary, uterus and placenta [ 113, 114, 115, 116, 1171. Synthesis of EGF in mouse salivary glands is strongly induced by both androgens and Padrenergic agonists [98]. It is detectable in saliva, milk and urine but, in man, circulates primarily in alpha granules in platelets [I 181, suggesting a role in wound healing analogous to PDGF and TGF alpha, which are also found in platelet granules [119]. EGF accelerates eyelid opening and tooth eruption in new born mice [ 1051, skin development in foetal lambs [120] and lung maturation in foetal rabbits [121] and lambs [122]. It also stimulates growth of gastric mucosa in suckling rats [ 1231 and of hypoproliferative intestinal epithelium in adult rats maintained on total parenteral nutrition 11241. Recently it has been shown that EGF antisense oligonucleotides can block the initiation of odontogenesis in murine mandibular explants [ 1251. Little is known of the involvement of this growth factor in neoplasia, but there are reports of elevated expression in 12% of pancreatic cancers [ 112],86% of pleomorphic adenomas of salivary glands [I 26],68% of prostatic carcinomas [ 1271, and around 20% of gastric carcinomas [39, 1281. Elevated levels of “EGF-like” factor have been described in about 30% of ovarian, endometrial and cervical carcinomas and 16% of breast carcinomas [ 1291. Transforming

Growth

Factor-Alpha

Transforming growth factor-alpha (TGF-alpha) is a potent mitogen [130]. TGFalpha structurally and functionally resembles epidermal growth factor [EGF] and induces a mitogenic response by binding to and activating the tyrosine kinase activity of the EGF receptor [131, 132, 1331. TGF-alpha does not bind c-erbB-2 and its interaction with c-erbB3 has not been examined. The mature form of TGF-alpha is synthesised as part of a 160 amino acid cell surface precursor from a 4.8kb mRNA [134, 1351. The mature 50 amino acid TGF-alpha is released from the extracellular domain by proteolytic cleavage between alanine-valine residues at both termini. Larger soluble forms of TGF-alpha representing incompletely-processed intermediates have also been identified [136, 1371. Such precursor molecules may accumulate on the surface of certain cells [138] and may be biologically active through interaction with EGF receptors on the surface of adjacent cells [138, 1391. TGF-alpha may therefore be biologically active as either a secreted peptide or a membrane-bound ligand.

9

rvpe I Growth Factor Receptors

There is now a great deal of evidence to implicate TGF-alpha in processes of normal development. Exogenously administered EGF or TGF-alpha can influence the development of several tissues. Exogenous TGF-alpha accelerates tooth eruption and eyelid opening in new born mice [140, 1411. TGF-alpha mRNA has been identified in preimplantation mouse embryos [142] and at low levels in several tissues of foetal day 9 and 10 mice [143]. In adults, TGF-alpha mRNA and/or protein have been detected in the epithelial cells of the anterior pituitary [144], maternal decidua [145], skin keratinocytes [ 1461, bronchus, intestine, kidney tubules, female genital tract [ 1471, brain [148] and activated macrophages [149]. TGF alpha has also been implicated in inflammation and wound healing [ 112, 1501, cell migration [ 1511, angiogenesis [ 1521. and bone resorption [ 153, 1541. Elevated expression of TGF-alpha has been frequently associated with neoplastic transformation. TGF-alpha activity was first described in the culture medium of retrovirally transformed cells by virtue of its ability to collaborate in the reversible transformation of cultured NRK cells [155, 156, 1571. Elevated production of TGFalpha is frequently found in human cancers [158] and malignantly transformed cultured cells [ 1591 including malignancies of the liver [ 160, 16 1] breast [ 1621, stomach [163], lung [164, 1651, colon [164], kidney [166], pancreas [112, 167, 1681, ovary [147] and tumours of the thyroid [169]. Overexpression of TGF-alpha under certain conditions can lead to hyperproliferation of cells in the absence of neoplasia as in the case of psoriasis [170]. Direct evidence to suggest that TGF-alpha can contribute to neoplastic transformation has been provided by reconstruction experiments in which recombinant TGF-alpha is expressed in cultured cells in vitro. Transfection of TGF-alpha plasmids into rat I [31] and NRK fibroblasts [32] and mouse mammary epithelial cells [33, 341 can produce full transformation as assayed by anchorage independence in soft agar and tumorigenicity in nude mice. However, other workers have failed to observe transformation of NRK fibroblasts [33] or NIH 3T3 fibroblasts [I711 expressing high levels of TGF-alpha. It has been shown that expression of membrane-anchored proTGF-alpha can be sufficient to produce this transformation, and that this form can interact with EGF receptors on adjacent cells without further processing [172]. Transformation of NIH 3T3 fibroblasts by expression of TGF-alpha may be dependent upon the simultaneous overexpression of EGF receptors [35, 171, 1731. Expression of TGF-alpha under the control of the metallothionein gene promoter in transgenic mice leads to disordered growth and differentiation in breast, liver and pancreas, and ultimately to neoplasia in liver and breast [174, 1751. Amphiregulin

Todaro and colleagues have identified a new growth factor which was isolated from the conditioned medium of MCF-7 cells treated with phorbol ester [176, 1771. This growth factor, termed amphiregulin, was found to be both structurally and functionally related to EGF and TGF-alpha. The mature protein of either 78 or 84 amino acids is embedded within a 252 amino acid transmembrane precursor, much like that found in TGF-alpha. In contrast to EGF and TGF alpha amphiregulin also has a hydrophilic 43 amino acid extension rich in lysine and arginine residues at its Nterminus (Fig. 2). This domain also includes motifs usually associated with nuclear localisation and DNA binding [I781 and specific antibodies have been used to

S.A. Prigent and N.R. Lemoinr

10

demonstrate nuclear immunoreactivity in both cultured cells and human biopsy material [179, and our own unpublished data]. High level expression of amphiregulin occurs in normal placentae, testis and ovaries and significant amounts in pancreas, colon, breast and some other tissues [178]. As its name suggests amphiregulin has both stimulatory and inhibitory effects on cell growth depending on the cell type [ 1761 and concentration of ligand [ 1791. Amphiregulin stimulates proliferation of normal human fibroblasts and keratinocytes but dramatically inhibits the growth of two human breast cancer cell lines that overexpress both TGF alpha and EGF receptor (MDA-MB-468 and MDA-MB-231). The expression of amphiregulin is inversely correlated with the expression of TGF-alpha in individual cell lines [l SO]. The mechanism by which amphiregulin produces its bifunctional effects on cell growth is not yet clear. Ciardiello et al. [ 18 l] report overexpression of amphiregulin (together with cripto) in 60-70% of primary or metastatic colorectal carcinomas compared with normal colonic mucosa. Recently heparin-affinity chromatography has been used to purify from the conditioned medium of human keratinocytes a peptide called keratinocyte autocrine factor (KAF) that is closely related or possibly identical to amphiregulin [182]. The function of this growth factor can be inhibited by heparin. Schwannoma-Derived

Growth Factor (SDGF)

This heparin-binding factor which has mitogenic activity on glial cells was isolated from the conditioned medium of the JSl cell line established from a rat Schwann ccl! tumour [183]. Screening of a cDNA library from JSl cells with degenerate oligonucleotides based on the amino acid sequence allowed cloning of nucleic acid sequence for a 243 residue prepro-protein and a 3’-untranslated sequence. It seems likely that SDGF is the rat homologue of amphiregulin since the amino acid sequence of SDGF is 76% identical to human amphiregulin, but there are distinct differences between the two molecules such as the greater molecular weight of the most abundant form of SDGF compared with amphiregulin. There are also differences in their biological activities, with amphiregulin being a less potent mitogen than SDGF for 3T3 cells and SDGF lacking any inhibitory effect on the growth of A43 1 cells. Heparin-Binding

EGF-Like

Growth

Factor (HB-EGF)

An apparently novel growth factor of the EGF family has been purified from the conditioned medium of the human U-937 histiocytic lymphoma cell line [184] and cDNA subsequently cloned from cells stimulated with phorbol ester. The primary translation product comprises 208 amino acids with a central domain, including an EGF-like motif rich in cysteine residues. HB-EGF most closely resembles amphiregulin, since these two polypeptides consist of a comparable number of amino acids and both have a notably hydrophilic domain upstream of the EGF-like motif [184]. These authors have shown that HB-EGF can bind to the EGF receptor on A43 1 cells and smooth muscle cells with high affinity, and is mitogenic for keratinocytes and postulate that it may be involved in wound healing.

Type I Growth Factor Receptors TGF-alpha

EGF

Cripto

FIGURE 2. Schematic representation of tive members of the EGF family. The secondary stroctore of hmnao EGF is redrawn from Hommel et aL, 1991 [lw aod contairrs two antiparallel /&sheets. Cysthe bridges are indicated by bars. Tke representethos of the other ECF-like ligaods are not based on NMR data, bat have been drawn to iaclade tke EGF core. Completely conserved residues are shown in black and positiveiy-ckarf@ residues in the N-termioal extensions of HB-EGF nod ampkiregolin are represented by shaded circles. Where indicated (?), N- and C-termini have not been determioed.

Viral Growth

Factors

DNA sequences encoding EGF-like peptides were first demonstrated in the genomes of several pox-viruses including Vaccinia virus [185], Shope fibroma virus [186], Molluscum contagiosum [187] and Myxoma virus [188] by searching genetic

12

S.A. Prigent and N.R. Lemoine

databanks. The processed gene product of one of these sequences, Vaccinia virus growth factor (WGF) was subsequently purified from the medium of Vaccinia virus infected cells, and was found to be a 77 amino acid glycosylated peptide [189]. VVGF is structurally similar to EGF (37% amino acid homology) and TGF-alpha (30% homology) with an identical pattern of cysteine residues to that in TGF-alpha. It is derived from a large precursor protein and the glycosylated processed form has a molecular weight of 23 kDa. It stimulates proliferation of human diploid fibroblasts by binding to the EGF receptor. The putative growth factors encoded in the genomes of other pox-viruses have not been purified, however a synthetic 55 amino acid peptide encoding the ( trboxyl portion of the Shope fibroma virus growth factor is capable of inducing EGF like bioeffects [190]. The role of these growth factors in viral replication and cytopathology is unclear. Cripto

The cripto gene was cloned accidentally [ 19 l] from a human embryonal carcinoma cell line as a composite cDNA with an unrelated gene. The open reading frame of the gene encodes a protein of 188 amino acids with a central domain that includes a structural cysteine-rich motif common to other members of the EGF family including EGF, TGF-alpha and human amphiregulin. Distinct from other family members cripto does not possess the A loop which is created when cysteines Cl and C3 (numbered from the N-terminus) form a disulphide bond, and it does not appear to exist in a transmembrane form. The interaction of cripto with the EGF receptor, c-erbB-2 or c-erbB-3 receptors has not been investigated. It has recently been shown that expression of cripto under the control of the RSV long terminal repeat in immortalised mouse NOG-8 mammary epithelial cells produces partial transformation, characterised by anchorage independence but not tumorigenicity [192]. Expression of cripto, and also amphiregulin, is elevated in colorectal and gastric neoplasms compared to the normal surrounding mucosa [ 18 I, 1931, but other tumours have not yet been examined. POTENTIAL

LIGANDS

FOR THE c-erbB-2 RECEPTOR

Lupu et al. [194] purified a growth factor secreted by the MDA-MB-231 human breast cancer cell line and identified it as a 30kDa glycoprotcin. This growth factor, which they termed gp30, was able to stimulate phosphorylation of both c-erbB-2 p 185 and the EGF receptor directly and independently, and has been shown to specifically inhibit the growth and suppress the soft agar colony formation of SK-BR-3 cells overexpressing c-erbB-2. Treatment of cells with anti-EGFR antibody had no effect on the inhibition of SK-BR-3 cell colony formation produced by gp30, and direct binding of gp30 to c-erbB-2 growth factor receptor was confirmed by binding competition experiments with anti-c-erbB-2 antibody. It thus appears that gp30 is a ligand for c-erbB-2, and also for the EGF receptor. The gene encoding this protein has not yet been cloned. Recently M. Shepard’s group at Genentech (also working with the MDA-MB-231 cell line) have reported the cloning of a gene for a novel protein which they have christened Heregulin (H. M. Shepard, Cold Spring Harbor Symposium, 1991). This

13

Type I Growth Factor Receptors

protein appears to have the appropriate credentials to be a specific ligand for the cerbB-2 receptor. The mature glycosylated 45 kDa protein, structurally related to EGF. is derived from a larger 645 amino acid precursor. Heregulin binds to and activates c-erbB-2, but not the EGF receptor, and has properties that appear to be distinct from those of gp30. Yarden and Weinberg [195] found that conditioned medium from cells transformed with an activated ras oncogene could induce phosphorylation of the neu ~185 protein and stimulate proliferation in haemopoietic cells transfected with the rat neu oncogene. The active factor in this conditioned medium has been partially purified and identified as a 35kDa glycoprotein which is heat-stable but sensitive to reduction [196]. It is capable of stimulating the proto-oncogenic receptor but is ineffective on the oncogenic rat neu protein which is constitutively active. The factor is also active on the EGF receptor at a similar concentration to that required to activate c-erbB-2. The relationship of this factor and gp30 is not yet clear. Another activating factor for the neu protein has been termed NAF (for neuactivating factor) by Dobashi et al. [197]. NAF was partially purified from medium conditioned by the human T cell line ATL-2. The heat-stable peptide has a molecular weight of less than 30kDa and specifically binds to neu resulting in tyrosine kinase activation and receptor internalisation. The growth-promoting effects of NAF are reported to be specific to cells expressing the neu protein. Tarakhovsky et al. [198] have used a ligand-trapping assay to show that mouse peritoneal macrophages activated by muramyl dipeptide secrete a 25 kDa polypeptide that interacts with the rat neu receptor. The identity of the gene encoding this factor is not yet known. It is not presently clear to what extent these peptides represent distinct ligands, since in most cases they have not been purified to homogeneity and no sequence information is available. It is possible that several ligands exist for c-erbB-2, as is the case for the EGF receptor. IMPLICATIONS

FOR CANCER THERAPY

There is increasing evidence that genetic alterations in growth factor signalling pathways are closely linked to developmental abnormalities and to chronic diseases including cancer [ 1991. Members of the EGF receptor family are frequently implicated in human cancer. As already discussed amplification or overexpression of the genes for EGF receptor and c-erbB-2 are commonly observed in certain malignancies. Overexpression of either gene under appropriate conditions in mammalian cells confers the transformed phenotype [30,73]. Additionally there is one report to suggest that c-erbB-3 is overexpressed in certain breast carcinomas, although at present there is no evidence that this gene product is able to transform mammalian cells or that it possesses tyrosine kinase activity [92]. At least in the case of EGF receptor, transformation appears to be dependent on the presence of an activating ligand. TGFalpha is frequently detected in carcinomas expressing large amounts of EGF receptors thus establishing autocrine loops where the EGF receptor is chronically stimulated [35]. It is possible that such autocrine loops are more prevalent than current knowledge suggests, since any one of an increasing number of EGF-like peptides could act as the stimulating ligand. Similarly for c-erbB-2, autocrine stimulation may be achieved by as

14

S.A. Prigent and N.R. Lemoine

yet unidentified peptides, although there is some evidence to suggest that the c-erbB-2 tyrosine kinase may possess a relatively high basal activity [200]. Potentially activating mutations in the genes for EGF-like growth factor receptors are not commonly observed in human malignancies, one notable exception being human gliomas where rearrangement of the EGF receptor gene frequently occurs resulting in the production of a truncated protein. Our present knowledge of the role of EGF receptors and ligands in cancer offers possibilities for improvements in diagnosis and prognosis, and opportunities for therapeutic intervention. Since gene amplification of EGF receptor and c-erbB-2 is confined to tumour tissue, identification of multiple copies of these genes by quantitative PCR could provide a diagnostic indicator. There is now strong evidence that the presence of large amounts of EGF receptor or c-erbB-2 protein is an indicator of poor prognosis in breast cancer, although for other cancers this relationship is less well defined. Immunocytochemical screening of tumour biopsies would therefore identify those patients for which more aggressive therapy would be appropriate. Perhaps the most challenging question is whether these growth factor signalling pathways could be manipulated therapeutically. Such intervention could theoretically be achieved by inhibiting ligand binding, receptor dimerisation, tyrosine kinase activation or protein expression of ligand or receptor. Monoclonal antibodies have been raised against both ligands and receptors for the EGF receptor system and against the extracellular domain of c-erbB-2. Neutralising antibodies against EGF and TGF alpha are effective in vitro, but have not been used successfully in vivo [201]. Recent reports of the existence of other stimulating ligands for EGF receptor may explain the lack of success of this approach. Antibodies against the extracellular domain of both receptors have however produced more promising results both in vitro and in vivo. Such antibodies could potentially be useful per se to down-regulate surface receptors, to inhibit ligand binding or as vehicles for localising toxic molecules to the site of the tumour. Perhaps the most success has been achieved with an antibody known as 4D5 which reacts with an extracellular epitope on c-erbB-2 and reversibly inhibits in vitro proliferation of human breast cancer cell lines expressing this oncogene product [202]. This antibody has been shown to increase the sensitivity of cancer cells to tumour necrosis factor alpha [202] and to enhance the cytotoxic effect of diammedichloroplatinum on breast cancer cell lines expressing high levels of c-erbB-2 both in vitro and in vivo [203]. It is currently being used in clinical trials for tumour localisation and possible efficacy in combination with CDDP [204]. The 4D5 antibody has recently been humanised, and bispecific antibodies containing Fv fragments of both 4D5 and anti-CD3 antibodies have been engineered which should promote T-cell recruitment to the tumour site [205]. In the case of human gliomas where gene rearrangement of the EGF receptor results in unique protein sequences at the splice junctions, antibodies recognising these sequences could provide very specific tools for tumour targeting. Another promising approach has been to synthesise fusion proteins of TGF-alpha and toxin sequences (such as Pseudomonas endotoxin) which bind to EGF receptors and have cytotoxic activity on tumour cells but not on normal tissues [206]. Other experimental approaches to interfere with receptor signalling have included the design of inhibitors of receptor dimerisation using either molecules which interpose within the cell membrane [201] or isolated domains of the receptor [207]. Inhibitors of tyrosine kinases have been developed which, though interesting, are probably insufficiently specific or of high enough affinity for clinical use [201].

IS

i’J+pe I Growth Fuctor Receptors

Clearly the EGF receptor family is potentially a very useful target for cancer therapy. However progress in this direction is limited by our fundamental understanding of the biochemical processes involved in normal receptor function. Transcription factors responsible for overexpression of these receptors in the absence of gene amplification might provide a site for intervention in malignancy. A knowledge of the threedimensional structure of these receptors will assist the design of peptides or other molecules capable of inhibiting dimerisation. An improved understanding of the mechanism of action of recently-described EGF-like ligands, particularly that of amphiregulin which is able to inhibit growth of some cells, might reveal new strategies for compromising receptor activity. The recent cloning of Heregulin, the ligand for cerbB-2 should greatly improve our understanding of the function of the c-erbB-2 protein. The c-erbB-3 protein meanwhile remains completely uncharacterised. It is hoped that our improved understanding of the role of these new molecules might provide rational new approaches for cancer therapy.

REFERENCES I, Savage CR. Jr, lnagami T. Cohen S. The primary structure of epidermal growth factor. J Biol Chew!. 1972;247: 7612-7621. 2. Wrann MM, Fox CF. Identification of epidermal growth factor receptors in a hyperproducing epldermal carcinoma cell line. J Biol Chem. 1979; 254: 8083-8086. 3. Downward J, Yarden Y, Mayes E, Scrace G, Totty N, Stockwell P, Ullrich A, Schlessinger J, Waterfield MD. Close similarity of epidermal growth factor receptor and v-erbB oncogene protein sequences. Nature 1984; 307: 521-527. 4. Lin CR, Chen WS, Kruiger W, Stolarsky LS, Weber W. Evans RM, Verma IM, Gill GN , Rosenfeld MC. Expression cloning of human EGF receptor complementary DNA: gene amplification and three related messenger RNA products in A43 1 cells. Science 1984; 224: 843-848. 5. Ullrich A, Coussens L, Hayflick JS, Dull TJ, Gray A, Tam AW, Lee J, Yarden Y, Libermann TA. Schlessinger J, Downward J, Mayes ELV, Whittle N. Waterfield MD, Seeburg PH. Human epidermal growth factor receptor cDNA sequence and aberrant expression of the amplified gene in A431 epidermoid carcinoma cells. Nature 1984; 309: 418425. 6. Xu YH, lshii S, Clark AJ, Sullivan M. Wilson RK. Ma DP, Roe BA, Merlin0 GT, Pastan 1. Human epidermal growth factor receptor cDNA is homologous to a variety of RNAs overproduced in A431 carcinoma cells. Nature 1984; 309: 806-810. 7. Haley J, Whittle N, Bennett P, Kinchington D, Ullrich A, Waterfield M. The human EGF receptor gene: structure of the 1 IOkb locus and identification of sequences regulating its transcription. Oncogene Rcs. 1987; I: 375-396. 8. Y arden Y. Schlessinger J. Epidermal growth factor induces rapid, reversible aggregation of the purified epidermal growth factor receptor. Biochemislry 1987; 26: 1443-1451. 9. Hsuan JJ, Totty N. Waterfield MD. Identification of a novel autophosphorylation site (P4) on the epidermal growth factor receptor. Biochem J. 1989; 262: 659-663. 10. Walton GM, Chen WS, Rosenfeld MG, Gill GN. Analysis of deletions of the carboxyl terminus of the epidermal growth factor receptor reveals self-phosphorylation at tyrosine 992 and enhanced in viva tyrosine phosphorylation of cell substrates. J Biol Chem. 1990; 265: 175@1754. I I. Velu TJ. Vass WC, Lowy DR. Beguinot L. Functional heterogeneity of proto-oncogene tyrosine kinases: the C terminus of the human epidermal growth factor receptor facilitates cell proliferation. Mol CeMBiol. 1989;9: 1772-1778. 12. Helin K. Velu T, Martin P, Vass WC, Allevato G, Lowy DR. Beguinot L. The biological activity of the human epidermal growth factor receptor is positively regulated by its C-terminal tyrosines. Oncogene 1991; 6: 825-832. 13. Chen WS, Lazar CS, Lund KA. Welsh JB. Chang C-P, Walton GM, Der CJ. Wiley HS. Gill GN. Rosenfeld MG. Functional independence of the epidermal growth factor receptor from a domain required for ligand-induced internalization and calcium regulation. Cell 1989: 59: 3343.

16

S.A. Prigent and N.R. Lemoine

14. Wells A, Welsh JB, Lazar CS, Wiley HS, Gill GN, Rosenfeld MG. Ligand-induced transformation by a noninternalizing epidermal growth factor receptor. Science 1990; 247: 962-964. 15. Margolis B, Rhee SG, Felder S, Mervic M, Lyall R, Levitzki A, Ullrich A, Zilberstein A, Schlessinger J. EGF induces tyrosine phosphorylation of phospholipase C-II: A potential mechanism for receptor signalling. Cell 1989; 57: 1101-I 107. 16. Meisenhelder J, Suh PG, Rhee SG, Hunter T. Phospholipase C-gamma is a substrate for the PDGF and EGF receptor protein-tyrosine kinases in vivo and in v&o. Cell 1989; 57: 1109-l 122. 17. Rossomando AJ, Payne DM, Weber MJ, Sturgill TW. Evidence that pp42, a major tyrosine kinase target protein, is a mitogen-activated serine/threonine protein kinase. Proc NatI Acad Sci USA. 1989; 86: 6940-6943. 18. Wahl M, Nishibe S, Suh PG, Rhee SG, Carpenter G. Epidermal growth factor stimulates tyrosine phosphorylation of phospholipase C-II independently of receptor internalization and extracellular calcium. Proc NatfArad Sci USA. 1989; 86: 1568-1572. 19. Ellis C, Moran M, McCormick F, Pawson T. Phosphorylation of GAP and GAP-associated proteins by transforming and mitogenic tyrosine kinases. Nature 1990; 343: 377-38 1. 20. Goldschmidt-Clermont PJ, Kim JW, Machesky LM, Rhee SG, Pollard TD. Regulation of phospholipase C-gamma 1 by profilin and tyrosine phosphorylation. Science 1991: 251: 1231-1233. 21. Countaway JL, McQuilkin P, Girones N, Davis RJ. Multisite phosphorylation of the epidermal growth factor receptor. J Biol Chem. 1990; 265: 3407-3416. 22. Northwood IC, Gonzalez FA, Wartmann M, Raden DL, Davis RJ. Isolation and characterization of two growth factor-stimulated protein kinases that phosphorylate the epidermal growth factor receptor at threonine 669. J Biol Chem. 1991; 266: 1526615276. 23. Heisermann GJ, Wiley HS, Walsh BJ, Ingraham HA, Fiol CJ, Gill GN. Mutational removal of the thr669 and ser671 phosphorylation sites alters substrate specificity and ligand-induced internalization of the epidermal growth factor receptor. J Biol Chem. 1990; 265: 12820-12827. 24. Lin CR, Chen WS, Lazar CS, Carpenter CD, Gill GN, Evans RM, Rosenfeld MG. Protein kinase C phosphorylation at Thr654 of the unoccupied EGF receptor and EGF binding regulate functional receptor loss by independent mechanisms. Cell 1986; 44: 8399848. 25. Libermann TA, Razon N, Bartal AD, Yarden Y, Schlessinger J, Soreq H. Expression of epidermal growth factor receptors in human brain tumours. Cancer Res. 1984; 44: 753-760. 26. Libennann TA, Nusbaum HR, Razon N, Kris R, Lax 1. Soreq H. Whittle N, Waterfield MD, Ullrich A, Schlessinger J. Amplification, enhanced expression and possible rearangement of EGF receptor gene in primary human brain tumours of glial origin. Nafure 1985: 313: 144147. 27. Merlin0 GT, Ishii S, Whang-Peng J, Knutsen T, Xu YH. Clark AJ. Stratton RH, Wilson RK, Ma DP, Roe BA, Hunts JH, Shimizu N, Pastan I. Structure and localisation of genes encoding aberrant and normal epidermal growth factor receptor RNAs from A431 human carcinoma cells. Mel Cell Biol. 1985; 5: 172221734. 28. Humphrey PA. Wong AJ, Vogelstein B, Friedman HS, Werner MH, Bigner DD, Bigner SH. Amplification and expression of the epidermal growth factor receptor gene in human glioma xenografts. Cancer Rex 1988; 48: 223 l-2238. 29. Humphrey PA, Wong AJ, Vogelstein B, Zalutsky MR, Fuller GN, Archer GE, Friedman HS, Kwatra MM. Bigner SH, Bigner DD. Anti-synthetic peptide antibody reacting at the fusion junction of deletion-mutant epidermal growth factor receptors in human glioblastomas. Proc Nail Acad Sci USA. 1990; 87: 4207-42 I I. 30. Di Fiore PP, Pierce JH, Fleming TP, Hazan R. Ullrich A, King CR, Schlessinger J, Aaronson SA. Overexpression of the human EGF receptor confers an EGF-dependent transformed phenotype to NIH 3T3 cells. Cell 1987; 51: 1063-1070. 3 I. Rosenthal A, Lundquist PB, Bringman TS, Goeddel DV, Derynck R. Expression in rat fibroblasts of a human transforming growth factor-alpha cDNA results in transformation, Cell 1986; 46: 301-309. 32. Watanabe S, Lazar E, Sporn MB. Transformation of normal rat kidney (NRK) cells by an infectious retrovirus carrying a synthetic rat type alpha transforming growth factor gene. Proc Nat1 Acad Sci USA. 1987; 84: 125881262. 33. McGeady ML, Kerby S, Shankar V, Ciardiello F, Salomon D, Seidman M. Infection with a TGFalpha retroviral vector transforms normal mouse mammary epithelial cells but not normal rat fibroblasts. Oncogene 1989; 4: 1375-1382. 34. Shankar V, Ciardiello F, Kim N, Derynck R, Liscia DS, Merlo G, Langton BC, Sheer D, Callahan R, Bassin RH, Lippman ME, Hynes N, Salomon DS. Transformation of an established mouse mammary

Type I Growth Factor Receptors

17

epithelial

cell line following transfection with a human transforming growth factor alpha cDNA. MO/ 1989; 2: I-l 1. 35. DiMarco E, Pierce JH, Fleming TP, Kraus MH, Molloy CJ, Aaronson SA, Di Fiore PP. Autocrine interaction between TGF-alpha and the EGF receptor: quantitative requirements for induction of the malignant phenotype. Oncogene 1989; 4: 831-838. 36. Merlin0 CT. Epidermal growth factor receptor regulation and function. Semin Cuncer Biol. 1990; I: Curcinogenesis

217-284. 37.

38.

39. 40.

41.

42. 43.

44.

45. 46. 47.

48.

49.

Gullick WJ. Prevalence of aberrant expression of the epidermal growth factor receptor in human cancers. Br Med Bulletin 1991; 47: 87-98. Lemoine NR, Jain S, Silvestre F, Lopes C, Hughes CM, McLelland E. Gullick WJ. Filipe MI. Amplification and overexpression of the EGF receptor and c-erhB-2 proto-oncogenes in human stomach cancer. Br J Cuncer 1991; 64: 79983. Lemoine NR, Hughes CM, Gullick WJ, Brown CL, Wynford-Thomas D. Abnormalities of the EGF receptor system in human thyroid neoplasia. In( J Carzcer 1991; 49: l-4. Coussens L, Yang-Feng TL. Liao Y-C, Chen E, Gray A. McGrath J. Seeburg PH. Libermann TA. Schlessinger J. Francke U. Levinson A, Ullrich A. Tyrosine kinase receptor with extensive homology to EGF receptor shares chromosomal location with neu oncogene. Science 1985; 230: 1132-I 139. Yamamoto T. Ikawa S. Akiyama T, Semba K, Nomura N, Miyajima N. Saito T. Toyoshima K. Similarity of protein encoded by the human c-e&B-2 gene to epidermal growth factor receptor. Nuturc 1986; 3 19: 230-234. Kmg CR, Draus MH, Aaronson SA. Amplification of a novel v-r&B-related gene in a human mammary carcinoma. Science 1985; 229: 974976. Fukushige S-I, Matsubara K-I, Yoshida M, Sasaki M. Suzuki T. Semba K. Toyoshima K, Yamamoto T. Localisation of a novel v-erbB-related gene, c-erbB-2. on human chromosome I7 and its amplification in a gastric cancer cell line. Mol Cell Biol. 1986; 6: 955-958. Gullick WJ. A comparison of the structures of single polypeptide chain growth factor receptors that possess protein tyrosine kinase activity. In: Cooke BA, King RJB. van de Molen HJ eds. Hormones and their uctions. Amsterdam, The Netherlands: Elsevier; 1988: 349-360. Lehvaslaiho H, Lehtola L, Sistonen L, Alitalo K. A chimeric EGF-R-neu proto-oncogene allows EGF to regulate neu tyrosine kinase and cell transformation EMBO J. 1989; 8: 159-166. Lee J. Dull TJ, Lax 1. Schlessinger J, Ulrich A. HER2 cytoplasmic domain generates normal mitogemc and transforming signals in a chimeric receptor. EMBO J. 1989; 8: 167-173. Sistonen L. Holtta E. Lehvaslaiho H, Lehtola L, Alitalo K. Activation of the neu tyrosine kinase induces the FosiJun transcription factor complex, the glucose transporter and ornithine decarboxylase. J Cell Biol. 1989; 109: 191 l-1919. King CR. Borrello I. Bellot F. Comoglio P, Schlessinger J. EGF binding to its receptor triggers a rapid tyrosine phosphorylation of the erbB-2 protein in the mammary tumour cell line SK-BR-3. EMBO J. 1988; 7: 1647-1651. Kokai Y. Dobashi K. Weiner DB, Myers JN, Nowell PC, Green MI. Phosphorylation process induced by epidermal growth factor alters the oncogenic and cellular neu (NGL) gene products. Proc Noi/ Ad Sc,i USA.

1988:

85: 5389-5393.

50. Stern DF, Kamps

MP, Cao H. Oncogenic activation of pl85neu stimulates tyrosine phosphorylation 1988: 8: 3969-3973. Wada T, Qian X. Greene MI. Intermolecular association of the p185neu protein and EGF receptor modulator EGF receptor function. Cell 1990; 61: 133991347. Goldman R, Levy RB, Peler E, Yarden Y. Heterodimerization of the c-erbB-1 and c-erbB-2 receptors in human breast carcinoma cells: a mechanism for receptor transregulation. Biochemistq, 1990: 29: 1102411028. Di Fiore PP, Segatto 0, Taylor WG, Aaronson SA, Pierce JH. EGF receptor and erbB-2 tyrosine kinase domains confer cell specificity for mitogenic signalling. Science 1990:248: 79-83.. Segatto 0. Lonardo F, Wexler D, Fazioli F, Pierce JH. Bottaro DP, White MF. Di Fiore PP. The juxtamembrane regions of the epidermal growth factor receptor and gpl85erbB-2 determine the specificity of signal transduction, Mel Cell Biol. 1991; II: 3191-3202. Kokai Y. Myers JN, Wada T. Brown VI, LeVea CM, Davis JG. Dobashi K. Greene MI. Synergistic interaction of pl85c-neu and the EGF receptor leads to transformation of rodent fibroblasts. Cell in viva.

51. 52.

53. 54.

55.

1989; 56.

Mol

Cell

Biol.

58: 281-292.

Bargmann mutation

CL Weinberg RA. Oncogenic activation and deletion. EMBO J. 1988: 7: 2043-2052.

of the neu-encoded

receptor

protein

by point

18

S.A. Prigent and N.R. Lemoine

57. Weiner DB, Liu J, Cohen JA, Williams WV, Greene MI. A point mutation in the neu oncogene mimics ligand induction of receptor aggregation. Nature 1989; 339: 230-231. 58. Segatto 0, King CR, Pierce JH, Di Fiore PP, Aaronson SA. Different structural alterations upregulate in vitro tyrosine kinase activity and transforming potency of the e&B-2 gene. Mel Cell Biol. 1988; 8: 5570-5574. 59. Wides RJ, Zak NB, Shilo B-Z. Enhancement of tyrosine kinase activity of the Drosophila epidermal growth factor receptor homolog by alterations in the transmembrane domain. Eur J Biochem. 1990: 189: 637645. 60. Kashles 0, Szabary D, Bellot F, Ullrich A, Schlessinger J, Schmidt A. Ligand-induced stimulation of epidermal growth factor receptor mutants with altered transmembrane regions. Proc Nat1 Acud Sci USA. 1988; 85: 9567-9571. 61. Carpenter CD, lngraham HA, Cachet C, Walton GM, Lazar CS, Sowadski JM, Rosenfeld MG, Gill GN. Structural analysis of the transmembrane domain of the epidermal growth factor receptor. J Biol Chem. 1991; 266: 5750-5755. 62. Hall PA, Hughes CM, Staddon SL, Richman PA, Gullick WJ, Lemoine NR. The c-erbB-2 protooncogene in human pancreatic cancer. J Pathol. 1990; 16 1: 195-200. 63. Lemoine NR, Staddon SL, Dickson C, Barnes DM, Gullick WJ. Absence ofactivating transmembrane mutations in the c-e&B-2 proto-oncogene in human breast cancer. Oncogene 1990; 5: 237-239. 64. Lemoine NR, Wyllie FS, Lillehaug JR, Staddon SL, Hughes CM, Aasland R, Shaw J. Varhaug JE. Brown CL, Gullick WJ, Wynford-Thomas D. Absence of abnormalities of the c-erbB and c-erbB-2 oncogenes in human thyroid neoplasia. Eur J Cancer 1990; 26: 7777779. 65. Tuzi NL, Venter DJ, Kumar S, Staddon SL, Lemoine NR, Gullick WJ. Expression of growth factor receptors in human brain tumours. Br J Cancer 1991; 63: 227-233. 66. Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A. Press MF. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 1989; 244: 707-712. 67. Saya H, Ara S, Lee PSY, Hung MC. Direct sequencing analysis of transmembrane region of human neu gene by polymerase chain reaction. MO/ Curcinogenesis 1990; 3: 198-201. 68. Papewalis J, Nitikin AY, Rajewsky MF. G to A polymorphism at amino acid codon 655 of the human erbB-Z/HER2 gene. Nucleic Acid Res. 1991; 19: 5452. 69. Muller WJ, Sinn E. Pattengale PK, Wallace R. Leder P. Single step induction of mammary adeno carcinoma in transgenic mice bearing the activated c-neu oncogene. Cell 1988; 54: 105-l 15. 70. Bouchard L, Lamarre L, Tremblay PJ. Jolicoeur P. Stochastic appearance of mammary tumours in transgenic mice carrying the MMTV/c-neu oncogene. Cell 1989; 57: 93 l-936. 71. Wang B, Kennan WS, Yasukawa-Barnes J, Lindstrom MJ, Gould MN. Frequent induction of mammary carcinomas following neu oncogene transfer into in situ mammary epithelial cells of susceptible and resistant rat strains. Cancer Res. 1991; 51: 5649-5654. 72. Suda Y, Aizawa S, Furuta Y, Yagi T, Ikawa Y, Saitoh K, Yamada Y, Toyoshima K, Yamamoto T. Induction of a variety of tumours by c-erhB-2 and clonal nature of lymphomas even with the mutated gene (Va16599Glu659). EMBO J. 1990; 9: 181-190. 73. Di Fiore PP. Pierce JH. Kraus MH, Segatto 0. King CR, Aaronson SA. erbB-2 is a potent oncogene when overexpressed in NIH/3T3 cells. Science 1987; 237: 178-182. 74. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987; 235: 177-182. 75. Tal M, Wetzler M, Josefberg Z, Deutch A. Gutman M, Assaf D. Kris R, Shiloh Y, Givol D. Schlessinger J. Sporadic amplification of the HERZ/neu protooncogene in adenocarcinomas ofvarious tissues. Cancer RPS. 1988; 48: 15 17-l 520. 76. Yokota J, Yamamoto T, Miyajima N, Toyoshima K, Nomura N, Sakamoto H, Yoshida T, Terada M, Sugimura T. Genetic alterations of the c-e&B-2 oncogene occur frequently in tubular adenocarcinoma of the stomach and are often accompanied by amplification of the v-erbA homologue. Oncogene 1988; 2: 283-287. 77. Park J-B. Rhim JS, Park S-C. Kimm S-W, Kraus MH. Amplification, overexpression. and rearrangement of the erbB-2 protooncogene in primary human stomach carcinomas. Cancer Res. 1989; 49: 6605-6609. 78. Kraus MH, Popescu NC, Amsbaugh SC, King CR. Overexpression of the EGF receptor-related proto-oncogene erbB-2 in human mammary tumour cell lines by different molecular mechanisms. EMBO J. 1987: 6: 6055610.

Type

1 Growth

Factor

Receptors

19

79. Parkes HC. Lillicrop K, Howell A, Craig RK. c-erbB-2 mRNA expression in human breast tumours : comparison with c-erbB-2 DNA amplification and correlation with prognosis. Br J Cancer 1990; 61: 3945. X0. Wlrri AM, Laine AM, Majasuo KE, Alitalo KK, Harkiinen PL. Estrogen suppression of erbB-2 expression is associated with increased growth rate of ZR-75-1 human breast cancer cells in vitro and in nude mice. ht J Cancer 1991; 49: 616-623. Xl. Borg A, Linell F, Idvall I, Johansson S, Sigurdsson H, Ferno M, Killander D. HER-Zineu amplification and comedo type breast carcinoma. Lancer 1989; i: 12681269. X2. Bacus SS, Ruby SG. Weinberg DS, Chin D, Ortiz R. Bacus JW. HER-?/neu oncogene expression and proliferation in breast cancers. Am J Pathol. 1990; 137: 103-l Il. 83. Barnes DM, Meyer JS, Gonzalez JG, Gullick WJ. Millis RR. Relationship between c-erbB-2 immunorcactivity and thymidine labelling index in breast cancer in situ. Breast Can Res Treat. 1991; 18: II -17. X4. Dati C, Antoniotti S, Taverna D, Perroteau I, DeBortoli M. lnhibition of c-erbB-2 oncogene expression by estrogens in human breast cancer cells. Oncogene 1990: 5: 1001-1006. X5. Read LD. Keith D. Slamon DJ, Katzanellenbogen BS. Hormonal modulation of HER-Z/neu protooncogene messenger ribonucleic acid and ~185 protein expression in human breast cancer cell lines. Ctrncer Res. 1990: 50: 3947-395 1. 86. Yu D. Hung MC. Expression of activated rat neu oncogene is sufficient to induce experimental metastasis in 3T3 cells. Oncogene 1991; 6: 1991-1996. X7. Gullick WJ. Berger MS, Bennett PL, Rothbard JB, Waterfield MD. Expression of the c-erbB-2 protein in normal and transformed cells. Inf J Cancer 1987; 40: 246254. X8. Quirke P. Pickles A, Tuzi NL, Mohamdee 0, Gullick WJ. Pattern of expression of c-erbB-2 oncoprotein in human fetuses. Br J Cancer 1989; 60: 6469. X9. Natali PG. Nicotra MR, Bigotti A, Venturo I, Slamon DJ, Fendly BM, Ullrich A. Expression of the pl85 encoded by HER-2 oncogene in normal and transformed human tissues. Int J Cancer 1990; 45: 457461, 90. Press MF. Cordon-Card0 C, Slamon DJ. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene 1990; 5: 953-962. 91. Lofts FJ. Gullick, WJ. c-erbB-2 amplification and overexpression in human tumours. In: Dickson RB. Lippman ME, eds. Breasr cancer: cellulur und nroleculur biology, Vol III. Norwell, USA: Kluwer Academic Publishers; 1992: in press. 92. Kraus MH, lssing W. Miki T. Popescu NC, Aaronson SA. Isolation and characterisation of ERBB3, a third member of the ERBB/epidermal growth factor receptor family: evidence for overexpression in a subset of human mammary tumours. Proc Nat1 Acad Sci USA. 1989; 86: 9193-9197. 93. Plowman GD. Whitney GS, Neubauer MG, Green JM, McDonald VL. Todaro GJ. Shoyab M. Molecular cloning and expression of an additional epidermal growth factor receptor-related gene. Proc Natl Acud Sci USA. 1990; 87: 49054909. 94. Prigent SA, Lemoine NR. Hughes CM, Plowman GD, Selden C. Gullick WJ. Expression ofthe c-erbB3 protein in normal human adult and fetal tissues. Oncogene: in press. 95. Savage CR, Jr, Hash JH. Cohen S. Epidermal growth factor. J Biol Chem. 1973: 248: 7669-7672. 96. Laurence DJR, Gusterson BA. The epidermal growth factor: a review of structural and functional relationships in the normal organism and in cancer cells. Tumor Biol. 1990; I I: 229-261. 97. Wharton W, Johansen KM, Xu T, Artavanis-Tsakonas S. Nucleotide sequence from the neurogenic locus notch implies a gene product that shares homology with proteins containing EGF-like repeats. C’ell 1985; 43: 557-581. 98. (arpenter G. Cohen S. Epidermal growth factor. Annu Rev Biochem. 1979; 48: 193-216. 99. Gregory H. Isolation and structure of urogastrone and its relationship to epidermal growth factor. Nature 1975: 251: 325-321. 100. Gray A. Dull TJ. Ullrich A. Nucleotide sequence of epidermal growth factor cDNA predicts a t 28.000molecular weight protein precursor. Nature 1983; 303: 7222725. 101. Scott J, Urdea M, Quiroga M, Sanchez-Pescador R, Fong N, Selby M, Rutter WJ. Bell GI. Structure of a mouse submaxillary messenger RNA encoding epidermal growth factor and seven related proteins. Science 1983: 221: 236-240. 102. Russell DW. Schneider WJ. Yamamoto T. Luskey KL. Brown MS, Goldstein JL. Domain ofthe LDL receptor: sequence homology with the epidermal growth factor precursor. Cell 1984; 37: 577-585.

20

S.A.

Prigent

and N.R.

Lemoine

103. Rail LB, Scott J, Bell GI, Crawford RJ, Penschow JD, Niall HD, Coghlan JP. Mouse preproepidermal growth factor synthesis by the kidney and other tissues, Nurure 1985; 3 13: 228-23 1. 104. Dobashi Y, Stern DF. Membrane-anchored forms of EGF stimulate focus formation and intercellular communication. Oncogene 1991; 6: 1151-I 159. 105. Cohen S. Isolation of a mouse submaxillary gland protein accelerating incisor eruption and eyelid opening in the new-born animal. J Biol Chem. 1962; 237: 155551562. 106. Gregory H. Willshire IR. The isolation of urogastrones -inhibitors of gastric acid secretion - from human urine. Hoppe-Seylers Z Physiol Chem. 1975; 356: 1765-1774. 107. Hommel U, Dudgeon TJ, Fallon A, Edwards RM, Campbell IA. Structure-function relationships in human epidermal growth factor studied by site-directed mutagenesis and ‘H NMR. Biochemi.stry 1991; 30: 8891-8898. 108. Kirkegaard P, Olsen PS, Poulsen SS, Nexo E. Epidermal growth factor inhibits cysteamine-induced duodenal ulcers. Gusrroenferology 1983; 85: 127771283. 109. Olsen PS, Poulsen SS, Kirkegaard P, Nexo P. The role of submandibular saliva and epidermal growth factor in gastric cytoprotection. Gustroenterology 1984; 87: 103-108. 110. Wright NA. Pike C, Elia G. Induction of a novel growth factor-secreting cell lineage by mucosal ulceration in human gastrointestinal stem cells. Nature 1990; 343: 82-85. I I I. Wright NA, Poulsom R. Stamp GWH, Hall PA. Jeffery RE. Longcroft JM, Rio M-C, Tomasetto C. Chambon P. Epidermal growth factor (EGF/URO) induces expression of regulatory peptides in damaged human gastrointestinal tissues. J Pathol. 1990; 162: 279-284. 112. Barton CM, Hall PA, Hughes CM, Gullick WJ, Lemoine NR. Transforming growth factor alpha and epidermal growth factor in human pancreatic cancer. J Purhol. 1991; 163: Ill-1 16. 113. Elder JB, Williams G, Lacey E, Gregory H. Cellular localisation of human urogastrone/epidermal growth factor. Nature 1978; 271: 466467. 114. Heitz U, Kasper M, van Noorden S, Polak J. Gregory H, Pearse GE. Immunohistochemical localisation of urogastrone to human duodenal and submandibular glands. Gur 1978: 19: 408413. 115. Kasselberg AG, Orth DN, Gray ME, Stahlmann MT. Immunocytochemical localisation of human epidermal growth factor/urogastrone in several human tissues. J Histochem Cyrochem. 1985: 33: 3 15322. 116. Paulsen SS. Skov-Olsen P, Hess J, Kirkegaard P. Immunohistochemical localisation of epidermal growth factor in rat and man. Histochemistry 1986; 85: 389-394. 117. Kajikawa K, Yasui W. Sumiyoshi H, Yoshida K. Nakayama H. Ayhan A, Yokozaki H. Ito H. Tahara E. Expression of epidermal growth factor in human tissues: immunohistochemical and biochemical analysis. Virchows Arch A: Pathol Anat. 1991; 418: 27-32. 118. Oka Y, Orth DN. Human plasma epidermal growth factor/p urogastrone is associated with blood platelets. J Clin Invest. 1983: 12: 249-259. 119. Ross R, Raines EW. Bowen-Pope DF. The biology of platelet-derived growth factor. Cell 1986; 46: 155-169. 120. Dolling N. Thorburn GE, Young IR. Effect of epidermal growth factor in the skin of the fetal lamb. J Anat. 1983; 136: 656. 121, Catterton WZ. Escobedo BN, Sexson WR, Gray ME, Sundell HW, Stahlman MT. Effect ofepidermal growth factor on lung maturation in fetal rabbits. Pediufr Res. 1979; 13: 104108. 122. Sundell HW, Gray ME, Serenius F’S, Escobedo MB, Stallman MT. Effects of epidermal growth factor on lung maturation in fetal lambs. Am J Pathol. 1980; 100: 707-726. 123. Dembinski AB, Johnson LR. Effect of epidermal growth factor on the development of rat gastric mucosa. Endocrinology 1985: 116: 90-94. 124. Goodlad RA, Wilson TGJ. Lenton W, Gregory H, McCullagh KG, Wright NA. Proliferative effects of urogastrone-EGF on the intestinal epithelium. Gur 1987; 28 SI: 3743. 125. Kronmiller JE, Upholt WB, Kollar EJ. EGF antisense oligonucleotides block murine odontogenesis in vitro. Dev Biol. 1991; 147: 4855488. 126. Yamahara M, Fujito T, Ishikawa T, Shimosato T, Yokozaki H, Yasui W, Tahara E. Phenotypic expression of human epidermal growth factor in foetal submandibular gland and pleomorphic adenoma of salivary gland. Virchows Arch A: Pathol Anat. 1988; 412: 301-306. 127. Fowler JE. Lau JLT, Ghash L, Mills SE. Mounzer A. Epidermal growth factor and prostatic carcinoma: an immunohistochemical study. J Urology 1988: 139: 857-861. 128. Sugiyama K, Yonemura Y, Miyazaki I. Immunohistochemical study of epidermal growth factor and epidermal growth factor receptor in gastric carcinoma. Cancer 1989: 63: 1557-l 561.

Type

1 Growth

Factor

Receptors

171

129. Bauknecht T, Kohler M. Jam I, P8eiderer A. The occurrence of epidermal growth factor receptors and the characterisation of EGF-like factors in human ovarian, endometrial, cervical and breast cancer. J Cuncer Res C/in Oncol. 1989; 115: 193-199. 130. Derynck R. Transforming growth factor-alpha. Cell 1988; 54: 593-595. 13 I Marquardt H. Hunkapiller MW, Hood LE, Twardzik DR, DeLarco JE, Stephenson JR, Todaro GJ. Transforming growth factors produced by retrovirus transformed rodent fibroblasts and human melanoma cells: amino acid sequence homology with epidermal growth factor. Proc Nut/ Acod Scr USA. 1983; 80: 46844688. 132. Marquardt H. Hunkapiller MW, Hood LE. Todaro GJ. Rat transforming growth factor type I: Structure and relation to epidermal growth factor. Science 1984; 223: 1079-1082. 133. Massague J. Epidermal growth factor-like transforming growth factor II. Interaction with epidermal growth factor receptors on human placenta membrane and A431 cells. J Biol Chem. 1983; 258: 13614. 13620. 134. Derynck R. Roberts AB, Winkler ME, Chen EY, Goeddel DV. Human transforming growth factoralpha: precursor structure and expression in E. co/i. Cell 1984; 38: 287-297. 135. Lee DC. Rose TM, Webb NR, Todaro GJ. Cloning and sequence analysis of a cDNA for rat transforming growth factor-alpha. Nature 1985; 313: 489491. 136. Bringman TS. Lindquist PB. Derynck R. Different transforming growth factor alpha species are derived from a glycosylated and palmitoylated transmembrane precursor. CeN 1987; 48: 429440. 137. Teixido J. Massague JM. Structural properties of a soluble bioactive precursor for transforming growth factor-alpha. J Eiol Chem. 1988; 263: 3924-3929. 138. Brachmann R, Lindquist PB, Nagashima N, Kohr W, Lipari T. Napier M, Derynck R. Transmembrane TGF-alpha precursors activate EGF/TGF-alpha receptors. Cell 1989; 56: 691-700. 139. Wong ST, Winchell LF. McCune BK, Earp HS, Teixido J, Massague J, Herman B, Lee DC. The TGFalpha precursor expressed on the cell surface binds to the EGF receptor on adjacent cells. leading to signal transduction. Ceil 1989; 56: 495-506. 140. Smith JM, Sporn MB, Roberts AB, Derynck R, Winkler ME. Gregory H. Human transforming growth factor-alpha causes precocious eyelid opening in newborn mice. Nuture 1985: 315: 5155516. 141. Tam JP. Physiological effects of transforming growth factor in the new born mouse. Science 1985: 229: 613-675. 142. Rappolee DA, Brenner CA, Shuhz R, Mark D, Werb Z. Developmental expression of PDGF. TGFalpha and TGF-beta genes in preimplantation mouse embryos. Science 1988; 241: 182331825. 143. Wilcox JN. Derynck R. Developmental expression of transforming growth factors alpha and beta in mouse fetus. MO/ Cell Biol. 1988; 8: 3415-3422. 144. Samsoondar J. Kobrin MS, Kudlow JE. Alpha transforming growth factor secreted by untransformed bovine anterior pituitary cells in culture. Int J Biol Chem. 1986; 261: 14408814413. 145. Han VKM. Hunter ES. Pratt RM, Zendegui JG, Lee DC. Expression of rat transforming growth factor alpha mRNA during development occurs predominantly in the maternal decidua. Mel Cell Biol. 1987; 7: 2335-2343. 146. (‘offey RJ, Derynck R, Wilcox JN, Bringman TS. Goustin AS. Moses HL, Pittelkow MR. Production and autoinduction of transforming growth factor-alpha in human keratinocytes. Nature 1987: 328: X 17820. 147. Kommoss F. Wintzer HO, Von Kleist S. Kohler M, Walker R. Langton B, Van Tran K, Pfleiderer A. Bauknecht T. In siru distribution of transforming growth factor alpha in normal human tissues and in malignant tumours of the ovary. J Puthol. 1990; 162: 223-230. 148. Wilcox JN. Derynck R. Localisation of cells synthesising transforming growth factor-alpha mRNA in the mouse brain JNeurosci. 1988; 8: 1901-1904. 149. Madtes DK, Raines EW, Sakariassen KS, Assoian RK, Sporn MB, Bell GI, Ross R. Induction of transforming growth factor-alpha in activated human alveolar macrophages. Cell 1988; 53: 285-293. 150. Schultz GS, White M. Mitchell R, Brown G, Lynch J, Twardzik DR, Todaro GJ. Epithelial wound healing enhanced by transforming growth factor-alpha and vaccinia growth factor, Science 1987; 235: 350-352. 15 I. Barrandon Y. Green H. Cell migration is essential for sustained growth of keratinocyte colonies: the roles of transforming growth factor alpha and epidermal growth factor. Ceil 1987; 50: 1131-1137. 152. Schreiber AB, Winkler ME, Derynck R. Transforming growth factor alpha: a more potent angiogenic mediator than epidermal growth factor. Science 1986: 232: 1250-1253.

22

S.A. Prigent and N.R. Lemoine

153. Stern PH, Krieger MS, Nissenson RA, Williams RD. Winkler ME, Derynck R, Strewler GJ. Human transforming growth factor-alpha stimulates bone resorption in vitro. J C/in Invesf. 1985; 76: 2016-2019. 154. Ibbotson KJ, Harrod J, Gowen M, D’Souza S, Smith DD. Winkler ME, Derynck R, Mundy GR. Human recombinant transforming growth factor-alpha stimulates bone resorption and inhibits formation in vitro. Proc Narl Acad Sci USA. 155. DeLarco JE, Todaro GJ. Growth factors from murine sarcoma virus-transformed cells. Proc Nafl Acad Sci USA. 1978: 75: 400-4005. 156. Ozanne B, Fulton RJ, Caplan PL. Kirsten Murine sarcoma virus transformed cell lines and a spontaneously transformed rat cell line produce transforming factors. J Cell Phvsiol. 1980; 105: 1633 180. 157. Todaro GJ, Fryling C, DeLarco JE. Transforming growth factors produced by certain human tumor cells: polypeptides that interact with epidermal growth factor receptors. Pm Nat1 Acad Sci USA. 1980; 77; 5258-5262. 158. Nickel1 KA, Halper J, Moses HL. Transforming growth factors in solid human malignant neoplasms. Cancer Res. 1983; 43: 1966-1971. 159. Derynck R, Goeddel DV, Ullrich A. Gutterman JU, Williams RD, Bringman TS, Berger WH. Synthesis of mRNAs for transforming growth factors alpha and beta and the epidermal growth factor receptor by human tumours. Cancer Res. 1987; 47: 7077712. 160. Liu C, Tsao MS, Grisham JW. Transforming growth factors produced by normal and neoplastically transformed rat liver epithelial cells in culture. Cancer Res. 1988; 48: 850-855. 161. Raymond VW, Lee DC, Grisham JW, Earp HS. Regulation of transforming growth factor-alpha messenger RNA expression in a chemically transformed rat hepatic epithelial cell line by phorbol ester and hormones. Cancer Rex 1989; 49: 3608-36 12. 162. Salomon DS, Perroteau I, Kidwell WR, Turn J, Derynck R. Loss of growth responsiveness to epidermal growth factor and enhanced production of alpha-transforming growth factors in cells. J Cell Phwiol. 1987; 130: 397.. ras transformed mouse mammary epithelial 409. 163. Bennett C, Paterson IM, Corbishley CM, Luqmani YA. Expression of growth factor and epidermal growth factor receptor encoded transcripts in human gastric tissues. Cuncer Res. 1989; 49: 2104-21 Il. 164. Liu C. Woo A, Tsao MS. Expression of transforming growth factor alpha in primary colon and lung carcinomas. Br J Cancer 1990; 62: 425429. 165. Tateishi M, Ishida T, Mitsudomi T, Sugimachi K. Prognostic implication of transforming growth factor alpha in adenocarcinoma of the lung - an immunohistochemical study. Br J Cancer 1991; 63: 130-133. 166. Petrides PE, Bock S, Bovens J, Hofmann R. Jakse G. Modulation ofpro-epidermal growth factor. protransforming growth factor alpha and epidermal growth factor receptor gene expression in human renal carcinomas. Cancer Res. 1990; 50: 39343939. 167. Smith JJ, Derynck R, Korc M. Production of transforming growth factor-alpha in human pancreatic cancer cells: evidence for a superagonist autocrine cycle. Pror Nat1 Acad Sci USA. 1987; 84: 756777570. 168. Ohmura E. Okada M. Onoda N, Kamiya Y, Murakami H, Tsushima T, Shizume K. Insulin-like growth factor I and transforming growth factor alpha as autocrine growth factors in human pancreatic cancer cell growth. Cancer Res. 1990: 50: 1033107. 169. Aasland R, Akslen LA, Varhaug JE, Lillehaug JR. Co-expression of the genes encoding transforming growth factor-alpha and its receptor in papillary carcinomas of the thyroid. Int J Cancer 1990; 46: 382387. 170. Elder JT, Fisher GJ. Lindquist PB, Bennett GL. Pittelkow MR, Coffey RJ, Jr, Ellingsworth L. Derynck R, Voorhees JJ. Overexpression of transforming growth factor alpha in psoriatic epidermis. Science 1989: 243: 81 l-813. 171, Finzi E. Fleming T, Segatto 0, Pennington CY. Bringman TS. Derynck R, Aaronson SA. The human transforming growth factor type alpha coding sequence is not a directly acting oncogene when overexpressed in NlH/3T3 cells. Proc Nat/ Acad Sci USA. 1987: 84: 3733-3737. 172. Blasband AJ, Gilligan DM, Winchell LF, Wong ST, Luetteke NC. Rogers KT, Lee DC. Expression of the TGF-alpha integral membrane precursor induces transformation of NRK cells. Onrogene 1990: 5: 1213-1221. 173. Finzi E, Fleming T, Pierce JH. Retroviral expression of transforming growth factor alpha does not transform fibroblasts or keratinocytes. J Invest Dernmtol. 1990; 95: 382-387.

Tape

I Growth

Factor

Receptors

23

174. Sandgren EP, Luetteke NC, Palmiter RD, Brinster RI, Lee DC. Overexpression of TGF-alpha in transgenic mice: induction of epithelial hyperplasia, pancreatic metaplasia, and carcinoma of the breast. CeN 1990; 61: 1121-1135. 175. Jhappan C. Stable C, Harkins RN, Faust0 N, Smith GH, Merlin0 GT. TGF-alpha overexpression in transgenic mice indices liver neoplasia and abnormal development of the mammary gland and pancreas. Cell 1990; 61: 1137-l 146. 176. Shoyab M. McDonald VL, Bradley JF, Todaro GJ. Amphiregulin: a bifunctional growth-modulating glycoprotein produced by the phorbol 12-myristate 13-acetate-treated human breast adenocarcinoma cell line MCF-7. Proc Nut1 Acad Sci USA. 1988; 85: 6528-6532. 177. Shoyab M, Plowman CD, McDonald VL, Bradley JG. Todaro GJ. Structure and function of human amphiregulin: a member of the epidermal growth factor family. Science 1989; 243: 10741076. 178. Plowman GD, Green JM. McDonald VL, Neubauer MG. Disteche CM, Todaro GJ. Shoyab M. The amphiregulin gene encodes a novel epidermal growth factor-related protein with tumor-inhibitory activity. Mel Cell Biol. 1990: IO: 1969-1981. 179. Johnson GR. Saeki T, Auersberg N, Gordon AW, Shoyab M, Salomon DS, Stromberg K. Response to and expression of amphiregulin by ovarian carcinoma and normal ovarian surface epithehal cells: nuclear localisation of endogenous amphiregulin. Biochem Biophys Res Commun. 1991; 180: 481488. 180. Todaro GJ. Rose TM. Spooner CE, Shoyab M, Plowman GD. Cellular and viral ligands that interact with the EGF receptor. Semin Cancer Biol. 1990; I: 257-263. I8 I. Ciardiello F. Kim N, Saeki T, Dono R, Persico MG, Plowman GD, Garrigues J, Radke S, Todaro GJ, Salomon DS. Differential expression of epidermal growth factor-related proteins in human colorectal tumors. Proc Nat1 Acad Sci USA. 1991; 88: 7792-7796. IX?. Cook PW, Mattox PA, Keeble WW, Pittelkow MR. Plowman GD, Shoyab M, Adelman JP. Shipley GD. A heparin sulfate-regulated human keratinocyte autocrine factor is similar or identical to amphiregulin. Mel Cell Bio/. 1991; 1 I: 2547-2557. 183. Kimura H, Fischer WH, Schubert D. Structure, expression and function of schwannoma-derived growth factor. Nature 1990; 348: 257-260. 184. Higashiyama S, Abraham JA. Miller J. Fiddes JC, Klagsbrun M. A heparin-binding growth factor secreted by macrophage-like ceils that is related to EGF. Science 1991: 251: 936-939. 185. Reisner AH. Similarity between the vaccinia virus 19K early protein and epidermal growth factor. Nu/ure 1985; 313: 801-803. 186. Chang W. Upton C. Hu S-L, Purchio AF. McFadden G. The genome of shope fibroma virus, a tumorigenic poxvirus, contains a growth factor gene with sequence similarity to those encoding epidermal growth factor and transforming growth factor alpha. Mel Cell Biol. 1987; 7: 535-540. I X7. Porter CD. Archard LC. Characterization and physical mapping of molluscum contagiosum virus DNA and location of a sequence capable of encoding a conserved domain of epidermal growth factor. J Gen Viral. 1987; 68: 673-682. 188. Upton C. Macen JL. McFadden G. Mapping and sequencing of a gene from myxoma virus that is related to those encoding epidermal growth factor and transforming growth factor alpha. J Viral. 1987: 61: 1271-1275. 189. Stroobant P, Rice AP, Gullick WJ, Cheng DJ, Kerr IM. Waterfield MD. Purification and characterisation of Vaccinia virus growth factor Cell 1985; 42: 383-393. 190. Lin Y-Z, Caporaso G, Chang P-Y, Ke X-H, Tam JP. Synthesis of a biological active tumor growth factor from the predicted DNA sequence of Shope fibroma virus. Biochemistry 1988; 27: 5640-5645. 191. Ciccodicola A. Dono R, Obici S. Simeone A, Zollo M, Persico MG. Molecular characterization of a gene of the ‘EGF family’ expressed in undifferentiated human NTERA2 teratocarcinoma cells. EMBCI J. 1989; 8: 1987-1991. 192. Ciardiello F. Dono R, Kim N, Persico MG. Salomon DS. Expression of cripto, a novel gene of the epidermal growth factor gene family, leads to in viva transformation of a normal mouse mammary epithelial cell line. Cancer Res. 1991; 51: 1051-1054. 193. Kuniyasu H. Yoshida K. Yokozaki H, Yasui W, Ito H, Toge T, Ciardiello F, Persico MG, Saeki T, Salomon DS. Tahara E. Expression of cripto, a novel gene of the epidermal growth factor family. in human gastrointestinal carcinomas, Jpn J Cancer Res. 1991; 82: 969-973. 194. Lupu R, Colomer R, Zugmaier G, Sarup J, Shepard M, Slamon D. Lippman ME. Direct interaction of a ligand for the erbB2 oncogene product with the EGF receptor and p185erbB2. Science 1990: 249: 155221555.

24

S.A. Prigent and N.R. Lemoine

195. Yarden Y, Weinberg RA. Experimental approaches to hypothetical hormones: detection of a candidate ligand of the neu proto-oncogene. Proc Nat1 Acad Sci USA. 1989; 86: 3179-3183. 196. Yarden Y, Peles E. Biochemical analysis of the ligand for the neu oncogenic receptor. Biochemistry 1991; 30: 3543-3550. 197. Dobashi K, Davis JG, Mikani Y, Freeman JK, Hamuro J, Greene MI. Characterisation of a neu/ c-erbB-2 protein-specific activating factor. Proc Nat1 Acad Sci USA. 1991; 88: 8582-8586. 198. Tarakhovsky A, Zaichuk T, Prassolov V, Butenko ZA. A 25kDa polypeptide is the ligand for p185neu and is secreted by activated macrophages. Oncogene 1991; 6: 2187-2196. 199. Aaronson SA. Growth factors and cancer. Science 1991; 254: 1146-I 153. 200. Lonardo F, DiMarco E, King CR, Pierce JH, Segatto 0, Aaronson SA, Di Fiore PP. The normal erbB-2 product is an atypical receptor-like tyrosine kinase with constitutive activity in the absence of ligand. New Biol. 1990; 2: 992-1003. 201. Gullick WJ. Inhibitors of growth factor receptors. In: Carney D, Sikora K eds. Genes and cancer. Chichester, U.K. Wiley; 1990: 263-273. 202. Hudziak RM, Lewis GD, Winget M, Fendly BM, Shepard HM, Ullrich A. Monoclonal antibody has antiproliferative effects in vitro and sensitizes human breast tumor cells to tumor necrosis factor. MO/ Cell Biol. 1989; 9: 1165-l 172. 203. Hancock MC, Langton BC, Chan T, Toy P, Monahan JJ, Mischak RP. Shawver LK. A monoclonal antibody against the c-erbB-2 protein enhances the cytotoxicity of cis-diammedichloroplatinum against human breast and ovarian tumor cell lines. Cancer Res. 1991; 5 I: 45754580. 204. Shepard HM, Lewis GD, Sarup JC, Fendly BM, Maneval D, Mordenti J, Figari I, Kotts CE, Palladino MA, Jr, Ullrich A, Slamon D. Monoclonal antibody therapy of human cancer: taking the HER2 protooncogene to the clinic. J Clin Immunol. 1991; 11: 117-127. 205. Shalaby MR, Shepard HM, Presta L, Rodrigues ML, Beverley PCL, Feldmann M, Carter P. Development of humanized bispecitic antibodies reactive with cytotoxic lymphocytes and tumor cells overexpressing the HER2 protooncogene. J Exp Med. 1992: 175: 217-225. 206. Heimbrook DC, Stirdivant SM, Ahern JD, Balishin NL, Patrick DR, Edwards GM, Defeo-Jones D, FitzGerald DJ, Pastan I, Oliff A. Transforming growth factor alpha-Pseudomonas exotoxin fusion protein prolongs survival of nude mice bearing tumor xenografts. Proc Nat1 Acad Sci USA. 1990; 87: 4697470 1. 207. Basu A, Raghunath M, Bishayee S, Das M. Inhibition of tyrosine kinase activity of the epidermal growth factor (EGF) receptor by a truncated receptor form that binds to EGF: role for interreceptor interaction in kinase regulation. Mel Cell Biol. 1989; 9: 671-677.

The type 1 (EGFR-related) family of growth factor receptors and their ligands.

This review considers the biology of the type 1 growth factor receptor family which is increasingly recognised as important in the control of normal c...
2MB Sizes 0 Downloads 0 Views