throbber
x i1&,}
`
`SIR
`
`MINI REVIEW
`
`Cyrokine & Growth Factor Reviews Vol. 7. No. 2, pp.
`
`l33——l4l, 1996
`
`PII: S1359-6101(96)00016-0
`
`Copyright if; 1996 Elsevier Science Ltd. All rights reserved
`Printed in Great Britain.
`
`l359—6l0l/96 $32.00-l—0.00
`
`The Epidermal Growth Factor Receptor and its Ligands as Therapeutic
`Targets in Human Tumors
`
`Valerie Rusch,*T¢ John Mendelsohn§1l and Ethan Dmitrovskyill
`
`The epidermal growth factor receptor (EGFR) is detected on many non—haematopoietic tissues and
`is frequently overexpressed in human tumors. With its ligand, TGF-at, it forms a well—defined
`autocrine growth loop. Several clinical approaches, using EGFR as a therapeutic target, are being
`investigated, particularly monoclonal antibodies combined with chemotherapy, and pharmacological
`inhibition of downstream components of the EGFR signaling pathway. Copyright «rt 1996 Elsevier Science rm.
`
`Key words: EGFR ° Ligands ° Therapeutic targets - Human tumors.
`
`Growth factors and their receptors are known to play an
`important role in normal cell proliferation and in neo-
`plastic growth, but their signaling mechanisms are not
`yet fully elucidated. The epidermal growth factor receptor
`(EGFR) and its ligand, transforming growth factor alpha
`(TGF—oc), form one of the best defined autocrine growth
`loops in human tumors. Methods of interrupting this
`stimulatory loop by specific antagonists exist. The use of
`EGFR or its ligands as targets for the treatment of human
`malignancies is now being explored. Thus understanding
`the structure, function and potential clinical implications
`of regulating the EGFR loop is a paradigm for the study
`of other growth factors and their receptors in cancer
`diagnosis and treatment. The role of the EGFR and its
`ligands as therapeutic targets in human tumors is the
`focus of this review.
`
`STRUCTURE AND FUNCTION OF THE EGFR
`
`The EGFR is a 53-amino acid, 170-kDa trans-
`membrane peptide present on many non-haematopoietic
`human tissues. It is composed of three major domains:
`
`§Laboratories of Receptor Biology, and iMolecular Medicine,
`1lDepartment of Medicine, Memorial Sloan-Kettering Cancer
`Center, New York, 10021, USA.
`* To whom correspondence should be addressed at: Memorial
`Sloan-Kettering Cancer Center, 1275 York Avenue, New York,
`NY 10021, U.S.A. Tel: 212-639-5873; Fax:2l2—639-2807;
`e-mail: ruschv@,mskcc.org
`
`an extracellular domain connected via a transmembrane
`
`lipophilic segment to an intracellular protein tyrosine
`kinase domain. The extracellular domain binds receptor
`specific ligands and activates the cytoplasmic domain,
`which then initiates a cascade of biological signals from
`the cytoplasm to the nucleus, ultimately resulting in mito-
`genesis [1, 2].
`Five ligands for the EGFR have been identified: the
`epidermal growth factor (EGF), TGF-oz,
`the vaccinia
`virus growth factor (VGF), amphiregulin and cripto.
`Although there is little overall sequence conservation
`(approximately 22%), these EGF—like molecules bind the
`EGFR with similar afiinities. The most widely expressed
`ligand for the EGFR in human tissues is TGF~oc. The
`active form of TGF—:x, a 50 amino acid peptide, is pro-
`duced by proteolytic cleavage of the extracellular com-
`ponent of its precursor form, pro—TGF-ac, which includes
`a transmembrane domain.
`
`Following ligand binding, the EGFR undergoes dimer-
`ization [3]. Since a single ligand molecule binds to a
`single EGFR molecule, this is thought to occur through
`a conformational change in the receptor [4-6]. Dimer-
`ization activates the intrinsic protein tyrosine kinase via
`intermolecular autophosphorylation (Figure 1). The
`tyrosine autophosphorylated regions function as high
`affinity binding sites for cytoplasmic target proteins
`involved in the transmission of biological signals to the
`nucleus [7, 8]. Protein binding is mediated by conserved
`regions of approximately 100 amino acids termed SRC
`homology 2 (SH2) domains. SH3 domains, another con-
`served region of 50 amino acids whose function is poorly
`understood, are often associated with SH2 domains.
`
`133
`
`APOTEX EX. 1017-001
`
`

`
`134
`
`V. Rusch et al.
`
`INACTIVE
`MONOMERS
`
`ACTIVE
`
`rnaus AUTO-
`PHOSPHORVLATION
`
`“
`
`DIMERS
`
`T Y R 05 I N E DE -
`PHOSPMORYLATION
`
`
`
`Although further investigation is required to define the
`mechanisms through which EGF R activation leads to
`mitogenesis, its effects on cell proliferation [15] are estab-
`lished. Introduction of either the TGF-at or EGF genes
`into cells expressing EGFRS can lead to cell
`trans-
`formation and enhanced proliferation [l6~l9]. TGF-oz
`over—expression is only weakly transforming in cultured
`cells that have a modest number of EGFRS on the cell
`
`surface. Conversely, EGFR overexpression at high levels
`only leads to transformation when cells are also exposed
`to high levels of EGF and TFG-oz. Thus,
`the estab-
`lishment of an autocrine loop responsible for trans-
`formation seems
`to
`require
`two distinct
`events,
`overexpression of EGFR and one of its ligands [20].
`Disruption of this growth stimulatory loop leads to Gl
`cell cycle arrest, and under some unusual circumstances,
`apoptosis [21]. This has been shown in several in vitro
`studies which also provide information about the poten-
`tial mediators of this G1 arrest. Apoptosis may result
`from interactions between the EGFR pathway and other
`growth regulatory loops. For instance, in a human col-
`orectal carcinoma cell line DiFi, which expresses high
`levels of EGFR, monoclonal antibodies to EGF R induce
`G1 cell cycle arrest and apoptosis. The addition of high
`concentrations of insulin or of insulin-like growth factor
`(IGF-1) delays apoptosis, but does not reverse G1 cell
`cycle arrest [22]. In these cells, blockade of the EGF
`receptor by monoclonal antibody results in elevation of
`the p27K"" inhibitor, accompanied by decreased cyclin-
`dependent kinase activity and decreased Rb phos-
`phorylation [22, 23]. Thus, the EGFR pathway plays an
`important role in cell transformation and proliferation
`and may also regulate aspects of apoptosis. For these
`reasons, it is of interest to examine expression of EGFR
`and its ligands in normal and neoplastic cells. A431
`human squamous carcinoma cells, which express high
`numbers of EGFR, are paradoxically growth-inhibited
`by the addition of nanomolar concentrations of EGF,
`which induces p2lC“"/WAR‘ expression, and reduces
`CDK2 activity. The induction of p2lC“°"“""F”‘ expression
`in these cells is inhibited by specific EGFR tyrosine kinase
`inhibitors, including anti-EGF receptor monoclonal anti-
`body 225 and tyrphostin AGl478, suggesting that its
`induction is a direct result of tyrosine kinase activation
`[24].
`
`EXPRESSION OF EGFR AND ITS LIGANDS IN
`HUMAN TISSUES AND TUMORS
`
`The EGFR is expressed in most normal human epi-
`thelial tissues and overexpressed in many human epi-
`thelial malignancies, including lung, breast, oesophageal,
`head and neck, and bladder cancers. Operexpression of
`EGFR ligands, particularly TGF-oz,
`is also frequently
`seen. EGFR mutations are rare, DNA amplification of
`the EGFR loci
`is more common, and EGFR over-
`expression often results from overexpression of its
`mRNA. In some tumors, EGFR overexpression is viewed
`
`APOTEX EX. 1017-002
`
`
`
`SUBST RATE
`PHOSPHOIIYLATION
`
`Figure 1. Ligand binding induces receptor dimerization, which
`leads to activation of the intrinsic protein tyrosine kinase
`activity and receptor autophosphorylation. Tyrosine auto-
`phosphorylation on multiple sites creates specific binding sites
`for target proteins, which bind to the activated receptor with
`their SH2 domains. (Schlessinger and Ullrich, Neuron 1992, 9,
`384, with permission.)
`
`Although not yet completely identified, target proteins
`are divided into two main groups: type-I proteins, which
`contain distinct enzymatic activities, and type-II proteins,
`which serve as regulatory subunits of downstream sig-
`naling proteins (Figure 2). Type-I proteins include phos-
`pholipase C-y (PCL-32) and the GTP-ase activating
`protein (GAP) of ras. Type-ll proteins include GRB-2
`which may also control ras signaling by stimulating a
`GDP-~GTP exchange factor or by inhibiting ras GTP-ase
`activity [1, 2]. The proteins further downstream in the
`cytoplasm which transmit signals to the nucleus are being
`defined. An important component of signaling appears
`to be the STAT proteins, so-called because they are recog-
`nized for their dual function in signal transduction in the
`cytoplasm and activation of transcription in the nucleus.
`The STAT proteins are known to be activated through
`the Jak kinases, a group of receptor-linked protein tyro-
`sine kinases. EGF causes activation of the STAT—l and -
`
`3 proteins in cultured hepatoma cells in the mouse liver,
`but whether the same mechanism is present in human
`tumors and whether other EGFR ligands function in a
`similar manner are not yet known [9—14].
`
`

`
`EGFR and its Ligands in Human Tumors
`
`135
`
`
`
`Ptd|ns—3K
`PLCV
`0
`O
`l
`fig
`DAG
`‘P3 Ptdlns(3)P
`
`l
`l Ca2+
`
`Figure 2. Direct phosphorylation (black dots on symbols) of substrates phospholipase C—y (PLCyi), phosphatidylinositol 3-kinase
`(ptdlns-3K), and ras GTPase—activating protein (GAP) to secondary events. including enzymatic activation and metabolite
`formation, activation of enzymatic functions by association, and serine/threonine phosphorylation (white dot on symbol) of
`substrates. Phosphorylation of raf is an indirect event, thought to be mediated via phosphokinase C. DAG = diaclyglycerol;
`1P3 = inositol l.4,5—triphosphate; Ptdlns(3)P = phosphatidylinositol 3-phosphate. (Modified from Ullrich and Schlessinger. Cell
`1990, 61, 208, with permission.)
`
`as an indicator of a poor clinical outcome, but this is not
`a universal
`finding [25—28]. Although EGFR over-
`expression is often present in epithelial carcinomas, its
`role in tumor initiation, growth and progression needs to
`be better defined. Non-small cell lung cancer (NSCLC)
`is one malignancy in which this has been extensively
`studied. and it serves as a paradigm for studies in other
`cancers.
`
`EGF R overexpression is found in lung cancer cell lines
`and primary tumors. Protein and binding assays show
`that NSCLC cell lines express higher levels of EGFR
`than do small cell lung cancer (SCLC) cell lines, and that
`this occurs in all histological subtypes of NSCLC [29,
`30]. Derynck at al. examined a large number of human
`tumors and tumor cell lines, including lung cancers, for
`the presence of TGF—oc and EGF R mRNA. By Northern
`analysis, TG F—a mRNA was strongly expressed in a squa-
`mous cell and a large cell carcinoma, but not in two
`adenocarcinomas of the lung or in normal lung tissue.
`Expression of EGFR mRNA was found in lung tumors
`of all histologies and in a normal lung specimen [31]. A
`larger study of lung cancers, derived from 68 patients,
`revealed ”5I—EGF binding in all NSCLC histologies, but
`especially in the squamous cell carcinomas. By Southern
`analysis, gene amplification was noted in two out of six
`samples of squamous cell carcinomas. In contrast to pre
`vious reports on breast and bladder cancers, there was
`no correlation between EGFR level and histological fea-
`tures of dilferentiation of tumor stage [32].
`Additional
`studies confirmed the frequent over-
`expression of EGFR in NSCLC, especially in squamous
`cell carcinomas, as assessed by binding assay and im-
`munohistochemistry [3342] Some of these studies cor-
`related EGFR overexpression with clinical outcome.
`Reported results vary widely with poorer survival being
`found in some reports [35, 37———39] and improved survival
`
`in others [36, 43]. These discrepant findings are probably
`explained by the small numbers of patients studied. the
`different techniques used to assess EGFR expression (e. g.
`paralfin embedded vs. frozen tissue, different monoclonal
`antibodies or binding assay) and the frequent lack of
`control specimens from normal lung tissue. One study
`examined EGFR overexpression by Northern analysis
`and immunohistochemistry on paired samples of primary
`tumor and benign lung obtained from 57 carefully staged
`patients and found no significant correlation with his-
`tology tumor stage or overall survival. This study also
`examined mRNA overexpression for
`three EGFR
`ligands, EGF, TGF-or and amphiregulin. EGF was not
`expressed in either primary tumors or benign lung tissue.
`whereas TGF-ac expression was increased in 61% of
`tumor vs. normal tissues, and amphiregulin expression
`was decreased in 63% of tumors [44]. Taken as a whole,
`these studies suggest that overexpression of EGFR and
`its ligand TGF-or is a frequent event, occurring in at least
`half of all NSCLC but does not clearly correlate with
`clinical or pathological indicators of aggressive tumor
`behavior.
`
`The presence of EGFR and TGF—:x overexpression in
`premalignant lesions of the lung, e. g. bronchial dysplasia,
`was only examined in a single study. in 62 preneoplastic
`bronchial lesions retrospectively identified in 34 patients
`who had resection of a NSCLC, overexpression of
`EGFR, TGF—oc and p53 was assessed by immuno-
`histochemistry [45]. Thirty (48%) of the bronchial lesions
`showed overexpression of EGF R, and this occurred as
`frequently in areas of metaplasia and atypia as it did in
`dysplasia and carcinoma in sizu. This study confirmed
`findings of the previous report
`[44]
`in which immu-
`nohistochemical staining for EGFR was consistently seen
`in the basal layer of the bronchial epithelium, but not in
`the pulmonary parenchyma. ln bronchial neoplasia. all
`
`APOTEX EX. 1017-003
`
`

`
`136
`
`V. Rusch et al.
`
`‘layers of the epithelium stain for EGFR (Figure 3). In
`contrast, staining for TGF-oz is consistently seen in nerves
`and submucosal salivary glands, but is variably present
`in both normal and premalignant bronchial epithelium.
`Although TGF-ac was strongly expressed in 16 out of
`34 primary tumors, overexpression of TGF-oz did not
`consistently accompany malignant transformation of the
`bronchial epithelium.
`These findings suggest that EGFR overexpression may
`be a more important step in tumor initiation than pro-
`gression of NSCLC. Overexpression of TGF-ac, although
`common in established NSCLC, is a much less frequent
`event in early bronchial neoplasia. On the other hand,
`autocrine expression of EGF or of cripto does not appear
`to play an important growth-regulating role in overt
`NSCLC ([44] and V. Rusch, Memorial Sloan-Kettering
`Cancer Center). The potential clinical implications of
`decreased amphiregulin expression in NSCLC vs. normal
`lung tissues are not yet known.
`
`EGFR: A POTENTIAL TARGET FOR CANCER
`THERAPY
`
`The frequent overexpression of EGF R in NSCLC and
`other human tumors and its apparent role in tumori-
`genesis make it a target for cancer treatment. Four poten-
`tial strategies using EGFR as a therapeutic target are
`now under study: (1) monoclonal antibodies alone, (2)
`immunotoxins, (3) monoclonal antibodies in conjunction
`with standard chemotherapy and (4) pharmacological
`agents inhibiting downstream components of the EGFR
`pathway, e. g. tyrosine kinase inhibitors.
`Several rodent monoclonal antibodies to the EGFR
`
`have been tested in vitro and in xenograft tumor models
`[46]. Dean et al. tested a panel of rat monoclonal anti-
`bodies to EGFR against three head and neck carcinoma
`cell lines, and A549, a squamous cell carcinoma of the
`lung, and found partial growth inhibition. These mono-
`clonal antibodies also caused regression of EGFR over-
`expressing tumors in athymic nude mice [47]. Aboud-
`Pirak et al. tested iodine—l25-labeled monoclonal anti-
`
`body 108.4 against subcutaneous xenografts in nude mice
`of a human oral epidermoid carcinoma cell with high
`levels of EGF R expression, and found that it inhibited
`tumor growth, and prolonged the survival of animals.
`Experimental lung metastases generated by intravenous
`injection of these cells also were growth-inhibited by this
`antibody. The simultaneous administration of cisplatin
`enhanced the growth suppressive effects of the antibody
`in the subcutaneous xenograft tumor model [48].
`The most extensively studied murine monoclonal anti-
`bodies against EGFR are 528 IgG2a and 225 IgGl [49,
`50]. These differ in their isotypes but bind to EGFR with
`similar affinities, down-regulate tyrosine protein kinase
`activity to a comparable degree, and block activation of
`the receptor. In vitro cytoxicity studies against A431 cells
`reveal that 528 IgG2a, but not 225 IgGl, induces partial
`complement-mediated cytoxocity.
`In
`thymic mice,
`
`tumors generated by subcutaneous injection of A431 cells
`with activated macrophages, showed enhancement of
`antitumour effects with suboptimal doses of 528 IgG2a,
`suggesting that immune mechanisms may contribute to
`the anti-tumor effect of this antibody. In contrast, the
`225 IgGl is thought to act primarily by altering EGFR
`function [51].
`j
`,
`Anti—EGFR murine monoclonal antibodies have been
`
`safely administered in high doses in human clinical trials.
`In a Phase I trial, Perez-Soler et al. treated 15 patients
`(13 with NSCLC, two with head and neck cancer) with
`escalating doses of an anti-EGFR monoclonal antibody,
`RG 83852, which activates the receptor kinase and down-
`regulates the receptor. No clinical toxicities were seen
`at doses that resulted in high tumor EGFR saturation, ac-
`companied by an antibody-medicated 3-4-fold upregu-
`lation of tyrosine kinase activity 24 h post therapy [52].
`In another Phase I trial, Divgi et al. studied the effect of
`indium-labeled (‘”In)225 IgGl antibody in 20 patients.
`At doses ranging from 1
`to 300 mg, no toxicity was
`observed, and tumors were imaged by single photon-
`emission-computed tomography (SPECT) in all patients
`who received doses of 20 mg or more [53]. All patients
`produced anti-murine antibodies (known as a HAMA
`response), underscoring a limitation to this therapeutic
`strategy in human clinical trials. A chimerized form of
`this anti-EGFR monoclonal antibody currently being
`tested in Phase I—Il trials, could allow repeated antibody
`administration since it has not been found to elicit a
`
`HAMA response (J. Mendelsohn, Memorial Sloan-Kett-
`ering Cancer Center). Another important limitation of
`clinical treatment only using anti-EGFR monoclonal
`antibodies, is that, in most cases, these appear to suppress
`the growth of cultured tumor cells without inducing cyto-
`toxicity. Therefore, combined treatment with other can-
`cer therapies is likely required to achieve a clinically
`meaningful response.
`Anti—EGFR monoclonal antibodies have been linked
`
`to plant or bacterial toxins to breast cell-specific immuno-
`toxins. Vollmar et al.
`tested two chimeric molecules,
`
`Ab 225 and EGF, both conjugated to ricin. Both caused
`growth inhibition of HeLa cells which have a high EGFR
`number, but not of 3T3-NR6 cells which are EGFR
`deficient [54]. Using gelonin, a 605-ribosome-inactivating
`hemotoxin, conjugated to the murine monoclonal anti-
`body B4G7, Ozawa et al. showed cytoxicity of four
`EGFR-rich squamous cell carcinoma cell lines but not of
`two EGFR-deficient cell lines [55]. Kirk et al. developed
`a novel chimeric recombinant cytotoxin composed of two
`independent domains, TGF-oz and a 40 kDa segment
`of the Pseudomonas exotoxin protein, designated PE-
`40. When tested against several human breast and lung
`cancer cell lines, toxicity was found to be directly related
`to the EGFR expression levels of each cell line tested [56].
`Although anti-EGFR immunotoxins are effective in in
`vitro models,
`there is no clinical experience with this
`therapeutic approach. The potential toxicities of immuno-
`toxins against normal human tissues could limit the
`therapeutic efficacy in the clinical setting.
`
`APOTEX EX. 1017-004
`
`

`
`EGFR and its Ligands in Human Tumors
`
`l37
`
`Figure 3. Positive immunohistochemical staining for EGFR in an invasive carcinoma (a). The normal respiratory epithelium
`generally shows positivity in the basal layer (b), while the superficial cell layers are negative. In areas of dysplasia, positivity is
`often seen in more superficial layers as well (0), while areas of CIS show full thickness staining (d).
`
`APOTEX EX. 1017-005
`
`

`
`MAb 523
`HHHHH
`DOXQ
`
`MN? 523
`llllllllll
`DOXO
`
`control
`
`V. Rusch et al.
`
`(B)
`
`A
`1")
`
`E
`3
`G.)
`3;:
`(I)
`
`E
`2
`
`MAb 528
`
`138
`
`(A)
`
`14
`
`12
`
`r‘\1O
`('3
`
`E
`3
`CD
`3;‘
`U)
`
`8
`
`E 6
`3
`
`4
`
`2
`
`o
`
`30
`
`.
`40
`
`50
`
`o
`
`10
`
`20
`
`4o
`
`50
`
`0
`
`10
`
`20
`30
`d3YS
`days
`Figure 4. Antitumour activity of MAb 528 in combination with doxorubicin (DOXO) on well-established A431 squamous cell
`carcinoma xenografts in athymic mice. Treatment was started when tumors reached a mean size of 0.4 cm‘ on day ll in Fig. 4A,
`or day 9 in Fig. 4B. Each treatment group consisted of at least five animals. A total of 10 mice were treated in the combination
`group in the experiment plotted in Fig. 4A. and eight animals in Fig. 4B. Results are given in mean tumor sizeiSE. Error bars
`are not present when fewer than three animals remained alive in a certain treatment group. Doxorubicin (100 pg/20 g body weight)
`was given intraperitoneally on days 1 and 2 of the treatment (day 11 and 12 or day 9 and 10 after tumor cell inoculation). MAb
`528 (1 mg) was given intraperitoneally on day l of the treatment and twice a week thereafter for a total of 10 doses. (A) Treatment
`with either doxorubicin alone or MAb alone partially inhibited tumor growth. Doxorubicin in combination with MAb 528
`completely eradicated all tumors in the animals surviving on day 30 (N = 8). (B) Doxorubicin in combination with non-specific
`mouse IgG did not result in a greater antitumor effect than doxorubicin alone, while doxorubicin in combination with MAb 528
`resulted in the disappearance of all tumors in the animals surviving on day 30 (N = 6). Arrows show the days on which the
`treatment was administered. (Baselga et al., J Nat Cancer Inst 1993, 85, 1330, with permission.)
`
`Recently, the strategy of eliciting tumor cytoxicity by
`combined administration of anti-EGFR antibodies and
`
`chemotherapy has been explored. Baselga et al. used
`monoclonal antibodies 528 IgG2a and 225 IgGl indi-
`vidually in combination with doxorubicin to effectively
`treat xenograft tumors from human A431 squamous cell
`carcinoma and human MDA-468 breast adenocarcinoma
`cell
`lines. These monoclonal antibodies enhanced the
`
`antitumor effects of doxorubicin producing at least addi-
`tive growth suppression. These results were further con-
`firmed by the treatment of wel1—established xenografts in
`athymic mice [57]. Combination treatment of mice bear-
`ing A431 xenografts resulted in tumor eradication in at
`least 40% of the mice, compared to only temporary
`growth inhibition when either antibody or doxorubicin
`was administered alone (Figures 4 and 5). Similar
`enhancement of antitumor activity against xenografts
`was observed when 225 IgAl therapy was combined with
`cisplatinum treatment of human tumor xenografts [58].
`As a result of these promising pre-clinical findings, clini-
`cal trials testing the use of standard chemotherapeutic
`
`agents (e.g. doxorubicin, cisplatin) in conjunction with
`humanized anti-EGFR antibodies, are now in progress
`in patients with metastatic cancer.
`Another therapeutic strategy currently under study is
`to block downstream steps involved in EGFR pathway
`signaling [59—62]. Tyrosine knase inhibitors antogonize
`the critical step in this pathway. Osherov tested two
`groups of tyrosine kinase inhibitors (tyrphostins), one
`specific for the EGFR,
`the other for the neu/erb-B2
`kinases, and found that these could block EGF-induced
`mitogenic signaling in NIH3T3 cells [63]. More recently,
`Fry er al. tested a small molecule called PD 143035 which
`preferentially inhibits the EGF R tyrosine kinase. In a
`human squamous cell carcinoma cell line and in Swiss
`3T3 fibroblasts, PD 153035 selectively blocked EGF-
`mediated events, including early gene expression (c-jun),
`cell transformation and mitogenesis [64]. Conceivably,
`identification of other
`important downstream com-
`ponents of the EGFR pathway could allow the devel-
`opment of additional inhibitors. Clinical application of
`such compounds may not be realistic until issues of effec-
`
`APOTEX EX. 1017-006
`
`

`
`MAW25
`HHH
`HDOXO
`
`control
`
`W 3.5
`
`3.0
`
`fig 35
`7: 2-0
`N
`‘a
`E 1.5
`3
`
`1'0
`
`0.5
`
`0
`
`0
`
`‘O
`
`MAb 225
`
`ooxo + MAb 225
`
`30
`
`40
`
`20
`days
`
`EGFR and its Ligands in Human Tumors
`
`139
`
`(B) 1'2
`
`1.0
`
`m,\_8
`5’
`(D
`.g.6
`5
`g 4
`- -
`
`-2
`
`00
`
`MAb528
`HHHHH
`uooxo
`
`ooxo
`control
`
`MAb528
`
`DOXO+MAb528
`
`1o
`
`20
`
`so
`
`40
`
`so
`
`days
`Figure 5. (A) Antitumour activity of MAb 225 in combination with doxorubicin (DOXO) on well-established A431 squamous
`cell carcinoma xenografts. Treatment was started when tumors reached a mean size of 0.3 cm‘. A total of 10 mice were treated in
`the combination group. Results are given in mean tumors size:SE. Doxorubicin (100 ,ug/20 g body weight) was given intra-
`peritoneally on days 1 and 2. MAb 225 (1 mg) was given intraperitoneally on day 1 and twice a week thereafter for a total of 10
`doses. Treatment with either doxorubicin alone of MAb alone resulted in a transient inhibition of tumor growth. Doxorubincin.
`in combination with MAb 225, had a pronounced antitumor activity. Arrows show the days on which the treatment was
`administered. (B) Antitumor activity of MAb 528 in combination with doxorubicin (DOXO) on well-established MDA-468 breast
`adenocarcinoma xenografts. The treatment was started when the tumor reached a mean size of 0.2 cm’. A total of nine mice were
`treated in the combination group. Results are given in mean tumor size 1 SE. Doxorubicin (100 pg/20 g body weight) was given
`intraperiteoneally on days 1 and 2. MAb 528 (2 mg) was given intraperiotoneally on day 1 and twice a week thereafter for a total
`of 10 doses. Treatment with either doxorubicin alone or MAb alone resulted in transient inhibition of tumor growth. Arrows
`show the days on which the treatment was administered. (Beselga er al., J Nat Cancer Inst 1993. 85, 1331. with permission.)
`
`tive drug dose and delivery and selectively are addressed.
`Phase I clinical
`trials using these compounds will be
`needed.
`
`The EGF R and its ligands serve as a paradigm for the
`study of receptor-mediated signaling and how growth
`factors affect cellular transformation and proliferation.
`Translating our knowledge about this pathway to the
`treatment of human tumors is becoming a clinical reality,
`particularly employing combined use of cytotoxic chemo-
`therapy and anti—EGFR monoclonal antibodies. Con-
`tinued investigation of the mechanisms through which
`growth factors and their receptors transmit signals to the
`cell nucleus is needed to better understand how these
`
`pathways can be manipulated for the benefit of cancer
`patients.
`
`Acknowledgement: The authors wish to acknowledge Melody
`Owens for her expert assistance in the preparation of this manu-
`script.
`
`REFERENCES
`
`l. Ullrich A. Schlessinger J. Signal transduction by receptors
`with tyrosine kinase activity. Cell l990, 61, 203—2l2.
`2. Schlessinger J, Ullrich A. Growth factor signaling by recep-
`tor tyrosine kinases. Neuron 1992, 9, 383-391.
`3. Lax l, Mitra AK. Ravera C et al. Epidermal growth factor
`
`(EGF) induces oligomerization of soluble, extracellular.
`ligand—binding domain of EGF receptor. J Biol Chem l99l.
`266, 1382843833.
`4. Hurwitz DR, Emanuel SL. Nathan MH et ul. EGF induces
`increased ligand binding affinity and dimerization of soluble
`epidermal growth factor
`(EGF)
`receptor extracellular
`domain. J Biol Chem 1991, 266, 22035-22043.
`5. Greenfield C, Hiles I. Waterfield MD et al. Epidermal
`growth factor binding induces a conformational change in
`the external domain ofits receptor. EMBO J I989, 8, 41 IS——
`4123.
`
`6. Schlessinger J. Signal transduction by allosteric receptor
`oligomerization. Trends Biochem Sm‘ 1988, 13, 443447.
`7. Honnegger AM, Kris RM, Ullrich A, Schlessinger J. Evi-
`dence that autophosphorylation of solubilized receptors for
`epidermal growth factor is mediated by intermolecular
`cross-phosphorylation. Proc Natl Acad Sci USA 1989. 86,
`925-929.
`
`8. Honegger AM. Schmidt A, Ullrich A, Schlessiger J. Evi-
`dence for epidermal growth factor (RGF)-induced inter-
`molecular autophosphorylation of the EGF receptors in
`living cells. Molec Cell Biol l990, [0, 403541044,
`9. Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways
`and transcriptional activation in response to IFNS and
`other extracellular signaling proteins. S(‘i(*)u'('
`l994. 264,
`l4l5——l42l.
`
`10. Zhong Z, Wen Z, Darnell JE Jr. Stat3 and Stat4: Members
`of the family of signal transducers and activators of tran-
`scription. Proc Natl Acad Sci USA 1994. 91, 4806-4810.
`1]. Heim Ml-I. Kerr IM. Stark GR. Darnell Jr JE. Con-
`
`APOTEX EX. 1017-007
`
`

`
`140
`
`12.
`
`13.
`
`14.
`
`15.
`
`16.
`
`17.
`
`I
`
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`
`23.
`
`24.
`
`25.
`
`26.
`
`27.
`
`V. Rusch et al.
`
`tribution of STAT SH2 groups to specific interferon sig-
`naling by the Jak-STAT pathway. Science 1995, 267, 1347-
`1349.
`
`Shuai K, Ziemiecki A, Wilks AF et al. Polypeptide sig-
`nalling to the nucleus through tyrosine phosphorylation of
`Jaka and Stat proteins. Nature 1993, 366, 580-582.
`Qureshi SA, Salditt-Georgiefi" M, Darnell Jr JE. Tyrosine-
`phosphorylated Statl and Stat2 plus a 48-kDa protein all
`Contact DNA in forming interferon-stimulated-gene factor
`3. Proc Natl Acad Sci USA 1995, 92, 3829-3833.
`Zhong Z, Wen Z, Darnell JE Jr. Stat3: A STAT family
`member activated by tyrosine phosphorylation in response
`to epidermal growth factor and interleukin-6. Science 1994,
`264, 95-98.
`Markowitz SD, Molkentin K, Gerbic C, Jackson J , Stellato
`T, Willson JKV. Growth stimulation by coexpression of
`transforming growth factor—oc and epidermal growth factor-
`receptor in normal adenomatous human colon epithelium.
`J Clin Invest 1990, 86, 356-362.
`Rosenthal A, Lindquist PB, Bringman TS, Goeddel DV,
`Derynck R. Expression in rat fibroblasts of a human trans-
`forming growth factor-at cDNA results in transformation.
`Cell 1986, 46, 301-309.
`Riedel H, Massoglia S, Schlessinger J, Ullrich A. Ligand
`activation of overexpressed epidermal growth factor recep-
`tors transforms NIH 3T3 mouse fibroblasts. Proc Natl Acaa’
`Sci USA 1986, 85, 1477-1481.
`Valverius EM, Bates SE, Stampfer MR et al. Transforming
`growth factor on production and epidermal growth factor
`receptor expression in normal and oncogene transformed
`human mammary epithelial cells. Molec Endocr. 1989, 3,
`203-214.
`
`Sandgren EP, Luetteke NC, Palmiter RD, Brinster RL, Lee
`DC. Overexpression of TGF<x in transgenic mice: Induction
`of epithelial hyperplasia, pancreatic metaplasia, and car-
`cinoma of the breast. Cell 1990, 61, 1121-1135.
`Di Marco E, Pierce JH, Fleming TP et al. Autocrine inter-
`action between TGF-oz and the EGF—receptor: quantitative
`requirements for induction of the malignant phenotype.
`Oncogene 1989, 4, 831-838.
`Stampfer MR, Pan CG, Hosoda J, Bartholemew J, Men-
`delsohn J, Yaswen P. Blockage of EGF receptor signal
`transduction causes reversible arrest of normal and immor-
`tal human mammary epithelial cells with synchronous reen-
`try into the cell cycle. Exp Cell Res 1993, 208, 175-188.
`Wu X, Fan Z, Masui H, Rosen N, Mendelsohn J . Apoptosis
`induced by an anti-epidermal growth factor receptor mono-
`clonal antibody in a human colorectal carcinoma cell line
`and its delay by insulin. J Clin invest 1995, 95, 1897-1905.
`Wu X, Rubin M, Fan Z. et al. Involvement of p27’‘"’‘
`in G, arrest mediated by an anti-epidermal growth factor
`receptor monoclonal antibody. Oncogene 1996, 12, 1397-
`1403.
`Fan 2, Lu Y, Wu X, DeBlasio A, Kolf A, Mendelsohn J .
`Prolonged induction of p2 1 C""’“'AF1 /CDK2/PCNA complex
`by epidermal growth factor receptor activation mediates
`ligand-induced A431 cell growth inhibition. J. Cell Biol
`1995, 131, 235-242.
`Fontanini G, Vignati S, Bigini D et al. Epidermal growth
`factor receptor (EGFr) expression in non-small cell lung
`carcinomas correlates with metastatic involvement of hilar
`and mediastinal lymph nodes in the squamous subtype. Eur
`JCancer 1995, 31A, 178-183.
`Neal DE, Bennett MK, Hall RR et al. Epidermal growth-
`factor receptors in human bladder cancer: Comparison of
`invasive and superficial tumours. Lancet 1998, i, 366-368.
`Mukaida H, Toi M, Hirai T, Yamashita Y, Toge T. Clinical
`significance of the expression of epidermal growth factor
`and its receptor in oesophageal cancer. Cancer 1991, 68,
`142-148.
`
`28.
`
`29.
`
`30.
`
`31.
`
`32.
`
`33.
`
`34.
`
`35.
`
`36.
`
`37.
`
`38.
`
`39.
`
`40.
`
`41.
`
`42.
`
`43.
`
`44.
`
`45.
`
`Nicholson S, Richard J, Sainsbury C et al. Epidermal
`growth factor receptor (EGFr); results of a 6 year follow-
`up study in operable breast cancer with emphasis on the
`node negative subgroup. Br J Cancer 1991, 63, 146-150.
`Haeder M, Rotsch M, Bepler G et al. Epidermal growth
`factor receptor expression in human lung cancer cell lines.
`Cancer Res 1988, 48, 1132-1136.
`Damstrup L, Rygaard K, Spang-Thomsen M, Poulsen HS.
`Expression of the epidermal growth factor receptor in
`human small cell lung cancer cell lines. Cancer Res 1992,
`52, 3089-3094.
`Derynck R, Goeddel DV, U

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket