throbber
Oncogene (1997) 14, 2099 ± 2109
`1997 Stockton Press All rights reserved 0950 ± 9232/97 $12.00
`
`A subclass of tumor-inhibitory monoclonal antibodies to ErbB-2/HER2
`blocks crosstalk with growth factor receptors
`
`Leah N Klapper1, Nora Vaisman1, Esther Hurwitz1, Ronit Pinkas-Kramarski2, Yosef Yarden2 and
`Michael Sela1
`
`Departments of 1Immunology and 2Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
`
`ErbB-2 is an orphan receptor that belongs to a family of
`tyrosine kinase receptors for either epidermal growth
`factor
`(EGF) or Neu di€erentiation factor
`(NDF/
`neuregulin). Because overexpression of
`the
`erbB-2
`proto-oncogene is frequently associated with an aggres-
`sive clinical course of certain human adenocarcinomas,
`the
`encoded
`protein
`is
`an
`attractive
`target
`for
`immunotherapy. Indeed, certain monoclonal antibodies
`(mAbs) to ErbB-2 e€ectively inhibit tumor growth in
`animal models and in clinical trials, but the underlying
`mechanism is incompletely understood. To study this
`question, we generated a large battery of mAbs to
`ErbB-2, that were classified epitopically. Whereas most
`antibodies
`stimulated
`tyrosine
`phosphorylation
`of
`ErbB-2,
`their anti-tumor e€ect correlated with its
`accelerated
`endocytic
`degradation. One
`group
`of
`tumor-inhibitory mAbs (Class II mAbs) was elicited by
`the most antigenic site of ErbB-2, and inhibited in trans
`binding of NDF and EGF to their direct receptors. The
`inhibitory e€ect was due to acceleration of
`ligand
`dissociation, and it resulted in the reduction of
`the
`ability of ErbB-2 to transactivate the mitogenic signals
`of NDF and EGF. These results identify two potential
`mechanisms of antibody-induced therapy: acceleration of
`ErbB-2 endocytosis by homodimerization and blocking of
`heterodimerization between ErbB-2 and growth factor
`receptors.
`
`Keywords: signal transduction; tyrosine kinase; onco-
`gene; Neu di€erentiation factor; epidermal growth
`factor; adenocarcinoma
`
`Introduction
`
`The identification of tumor associated antigens (TAA)
`accessible on human cancer cells, hailed immunother-
`apeutic approaches relying on specific recognition of
`neoplasms (Hellstrom and Hellstrom, 1989). Extensive
`e€orts have indeed been invested in examining the
`plausibility
`of
`anti-TAA monoclonal
`antibodies
`(mAbs)
`in the treatment of human malignancies
`proving promising in laboratory and clinic (Gold-
`enberg, 1993). Protooncogene-encoded growth factor
`receptors are putative targets for such recognition-
`dependent
`therapy, due to their suggested role in
`pathological proliferation of cells (Aaronson, 1991).
`ErbB-2, a receptor-like tyrosine kinase, has been
`repeatedly implicated in cell
`transformation (Hynes
`
`Correspondence: M Sela
`Received 23 October 1996; revised 10 January 1997; accepted 10
`January 1997
`
`and Stern, 1994; Stancovski et al., 1994). Amplification
`of the corresponding gene and overexpression of the
`protein itself were observed in 20% to 30% of
`adenocarcinomas of
`the breast
`(King et al., 1985;
`Slamon et al., 1987, 1989), ovary (Slamon et al., 1989),
`lung (Kern et al., 1990) and stomach (Park et al.,
`1989). Causative relationships between the cellular
`ErbB-2 content and the tumor’s proliferative capacity
`and aggressiveness have been supported by di€erent
`lines of evidence. When overexpressed in mouse
`fibroblasts, the human gene conferred a transformed
`phenotype in vitro and tumorigenesis in vivo (Di Fiore
`et al., 1987; Hudziak et al., 1987). Consistently,
`receptor overexpression is considered a predictor of
`poor survival and short time to relapse (Slamon et al.,
`1987, 1989). Direct interference with the transforming
`potential of ErbB-2 has thus become a subject of great
`interest. Antibodies directed against the extracellular
`domain of either a mutated version of the rodent
`homolog of this receptor-like molecule, or against the
`human wild-type protein have been shown to confer
`inhibitory, as well as stimulatory, e€ects on tumor
`growth in vivo (Drebin et al, 1986; Fendly et al., 1990;
`Hudziak et al., 1989; Stancovski et al., 1991). More-
`over, a murine antibody capable of such growth
`inhibition has been recently humanized and tested in
`a phase II clinical trial, resulting in anti-tumor activity
`in patients with ErbB-2-overexpressing metastatic
`breast cancers (Baselga et al., 1996).
`Although the potential therapeutic use of anti-ErbB-2
`mAbs is presently acknowledged and intensely exam-
`ined, the molecular mechanisms underlying these e€ects
`are not well understood. Accelerated down-regulation
`of the receptor has been suggested to mediate antibody
`inhibition of cell transformation (Hudziak et al., 1989;
`van Leeuwen et al., 1990). However, the ability of
`mAbs to induce receptor internalization showed only
`partial
`correlation with anti-tumorigenic
`activity
`(Harwerth et al., 1992; Hurwitz et al., 1995). An
`obstacle in the understanding of mAb-mediated e€ects
`is the possibility that ErbB-2 has a direct ligand, that
`has not yet been completely characterized (Dougall et
`al., 1994). Activation of receptor tyrosine kinases is
`dependent on receptor dimerization induced by the
`binding of specific ligands (Yarden and Schlessinger,
`1987). However, ErbB-2 may participate in signal
`transduction even in the absence of a direct ligand,
`because it
`forms heterodimeric complexes with its
`family members, namely ErbB-1 (EGF receptor) and
`the two NDF/neuregulin receptors, ErbB-3 and ErbB-4
`(Goldman et al., 1990; Pinkas-Kramarski et al., 1996;
`Riese et al., 1995; Wada et al., 1990). Hierarchical
`recognition of the dimerization partner o€ers ErbB-2 a
`key role in the determination and mediation of
`
`IMMUNOGEN 2035, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`

`

`

`for antibody binding to a cell surface-expressed ErbB-
`2. A dozen of new mAbs was generated, and analysed
`together with a panel of five mAbs that we previously
`described (Stancovski et al., 1991). The new mAbs were
`assayed for their ability to a€ect
`the tumorigenic
`growth in nude mice of
`the N87 human gastric
`carcinoma cell line overexpressing the ErbB-2 protein.
`Nine di€erent mAbs, or saline as control, were injected
`intraperitoneally into groups of six mice, on days 3, 7
`and 10 after tumor inoculation. Figure 1 depicts tumor
`progression in the presence of
`four representative
`mAbs. The tumorigenic growth of N87 cells was
`inhibited by 85%, 61% and 82% in nude mice
`injected with mAbs L26, L140 and L431, respectively.
`These values
`correspond to mean of
`inhibition
`measured at five time points throughout a period of
`46 days post inoculation. Antibody L87 showed no
`e€ect on the growth of tumors in vivo, presumably due
`to its relatively low a(cid:129)nity (data not shown). Because
`previous studies suggested that di€erent regions on the
`extracellular domain of ErbB-2 mediate mAb-depen-
`dent e€ects on tumor growth (Bacus et al., 1992) we
`examined the dependency of
`tumor
`inhibition on
`specific immunogenic determinants by performing a
`reciprocal binding assay (Figure 2). Several mAbs were
`radiolabeled and their binding to N87 cells was
`determined in the presence of all other antibodies to
`enable classification of the mAbs. Thus, antibody L431
`was e(cid:129)ciently displaced by the strong tumor-inhibitory
`mAb N12 (Stancovski et al., 1991), but not by any
`other antibody (Figure 2A). Therefore, these mAbs
`were classified into the same group, denoted Class I. In
`a similar manner we could categorize mAbs L26, L96,
`and L288 into a second group, denoted Class II
`(Figure 2B) and mAb L140 into Class
`III,
`that
`comprises only a single antibody. Antibody L87 could
`be displaced by antibodies
`from several groups,
`although it could not fully displace its own binding
`
`2100
`
`Tumor-inhibitory antibodies to ErbB-2
`LN Klapper et al
`
`signaling and resultant cellular fate (Tzahar et al.,
`1996). The importance of ErbB-2 as a transregulator
`expands in light of reported coexpression of ErbB
`family receptors in malignant cells (Gullick, 1990;
`Lemoine et al., 1992), as well as by its ability to
`reconstitute
`the aberrant
`tyrosine kinase activity
`characteristic of ErbB-3 (Pinkas-Kramarski et al.,
`1996; Riese et al., 1995; Sliwkowski et al., 1994).
`Heterodimer formation between ErbB-2 and ErbB-1 is
`responsible for synergistic growth signals in cells that
`co-overexpress the two receptors (Kokai et al., 1989).
`A similar synergy was observed upon co-overexpres-
`sion of ErbB-2 and ErbB-3 (Alimandi et al., 1995;
`Wallasch et al., 1995), probably due to the extremely
`high mitogenic potential of the corresponding receptor
`heterodimer (Pinkas-Kramarski et al., 1996). Selective
`suppression of ErbB-2 expression at the cell surface by
`means of retention in the endoplasmic reticulum (Beerli
`et al., 1994), demonstrated that this molecule can act as
`a shared signaling subunit of both EGF- and NDF-
`receptors (Graus-Porta et al., 1995; Karunagaran et al.,
`1996)
`that augments and prolongs
`signaling by
`deceleration of
`the
`rate of
`ligand dissociation
`(Karunagaran et al., 1996).
`The ability of ErbB-2 to serve as a pan ErbB
`auxiliary receptor
`subunit
`implies a versatility of
`mechanisms by which the receptor is involved in
`transformation. Hence, attempts to inhibit malignan-
`cies
`should consider
`the
`transacting potential of
`ErbB-2,
`in addition to its presumed ability to act
`through
`homodimer
`formation. Our
`study
`has
`addressed the possibility that mAbs directed against
`ErbB-2 might exert at
`least part of
`their tumor-
`inhibitory
`e€ects
`via
`interference with receptor-
`receptor interactions. A battery of antibodies directed
`against the extracellular domain of ErbB-2 has been
`generated and classified into groups according to
`specific epitope recognition. Several classes of tumor-
`inhibitory mAbs that accelerate cellular degradation of
`ErbB-2 were identified.
`Interestingly, one class of
`tumor-inhibitory antibodies partially reduced cellular
`binding of both NDF and EGF. Consistent with an
`ability to interfere with receptor crosstalk, these mAbs
`also reduced the trans-stimulatory e€ect of ErbB-2 on
`growth signals. We suggest that anti-ErbB-2 antibodies
`can inhibit
`cancer not only by
`impeding
`the
`homodimer-dependent activity, but also by blocking
`heterodimer formation and receptor crosstalk. This
`implies wider than currently accounted-for mechan-
`isms
`that
`can be utilized for
`the designing of
`therapeutically e(cid:129)cient anti-ErbB-2 inhibitors.
`In
`addition, our
`results may be
`relevant
`to the
`mechanism by which EGF-like ligands recruit ErbB-
`2 into receptor heterodimers.
`
`Results
`
`Classification of anti-ErbB-2 monoclonal antibodies
`
`To study the mechanistic basis of tumor inhibition by
`certain mAbs to ErbB-2, we extended our antibody
`repertoire by employing an exhaustive immunization
`protocol. Essentially, mice were immunized with a
`recombinant
`extracellular domain of
`the human
`protein and the resulting hybridomas were screened
`
`Figure 1 Inhibition of tumor growth by representative mAbs to
`ErbB-2. Athymic mice received a subcutaneous injection of
`36106 N87 human gastric cancer cells that overexpress ErbB-2.
`Three, 7 and 10 days later monoclonal antibodies (a total dose of
`1 mg per animal) were injected intraperitoneally, and tumor
`volumes were measured at the end of the indicated time periods.
`Phosphate-bu€ered saline- (PBS-) injected mice were used for
`control (closed circles). The following mAbs were used: L26
`(circles), L87 (rhombuses), L140 (triangles) and L431 (squares).
`Bars represent standard deviations for groups of five mice. The
`experiment was repeated three times with each of the mAbs and
`yielded similar results
`
`IMMUNOGEN 2035, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Tumor-inhibitory antibodies to ErbB-2
`LN Klapper et al
`
`(Figure 2D). This characteristic of mAb L87 is in
`accordance with its weak precipitating ability of the
`native ErbB-2 protein and is reinforced by the ability
`of mAb L87 to recognize the denatured protein (data
`not
`shown). The
`low a(cid:129)nity of mAb L87 to
`conformationally-intact ErbB-2 could underlie
`the
`observed pattern of displacement by a wide variety of
`mAbs. Several additional mAbs (e.g., L151, L242,
`L219, N28 and N29) were found to react with distinct
`antigenic determinants of ErbB-2,
`indicating multi-
`plicity of antigenic sites, of which site II is apparently
`the most e(cid:129)cient. Anti-ErbB-2 antibody classification
`is summarized in Table 1.
`
`Tumor inhibition correlates with mAb-induced receptor
`internalization but not with kinase activation
`
`Upon binding of certain mAbs, ErbB-2 has been
`shown to undergo internalization (Drebin et al., 1985)
`in a pathway shared by other growth factor receptors,
`when induced by ligands and antibodies (Sorkin and
`Waters, 1993). Several
`lines of evidence indicate a
`correlation between tumor-inhibitory activity of mAbs
`and their ability to accelerate ErbB-2 uptake and
`down-regulation (Hurwitz et al., 1995; Tagliabue et
`al., 1991). To test
`the applicability of
`such a
`correlation to the new inhibitory mAbs, we studied
`
`2101
`
`Figure 2 Analyses of competitive antibody binding to ErbB-2. The ability of unlabeled mAbs to displace a cell surface-bound
`125I-mAb was used as a measure for the degree of antigenic overlap. The following radiolabeled mAbs were used: L431 (A), L288
`(B), L140 (C) and L87 (D). The labeled mAbs were added to the medium of N87 cells growing in 96-well culture dishes, in the
`presence of the same unlabeled antibody (closed circles). Alternatively, unlabeled mAbs representing the di€erent classes were used:
`Class I (squares, N12 in A and C, L431 in B and D), II (circles, L26), III (triangles, L140) and IV (rhombuses, L87 in C). Following
`1 h of incubation at 228C, the monolayers of cells were washed three times with PBS and solubilized in an alkaline solution. The
`results are presented as the mean+s.d. of duplicates. The experiments were repeated thrice
`
`Antibody
`class
`
`Antibody
`
`Tumor growth
`inhibition (%)a
`
`Receptor
`internalizationb
`
`Receptor
`phosphorylationc
`
`Ligand binding inhibitiond
`EGF
`NDF
`
`Table 1
`
`+ N
`
`D + + + – +
`
`+
`
`+ N
`
`D + + +
`
`+++
`+++
`++
`
`+ +
`
`+
`
`– + –
`
`82
`86
`85
`60
`74
`
`– 6
`
`I I I
`
`I
`II
`II
`III
`IV
`
`V V
`
`L431
`N12
`L26
`L96
`L288
`L87
`L140
`1
`L151
`40
`I
`L219
`49
`ND
`VII
`L242
`61
`+++
`740*
`VIII
`N28
`–
`IX
`N29
`88
`ND
`aAverage tumor volume inhibition of five time points, as percentage of control. bReceptor internalization was determined by cell surface protein
`labeling, as described in text and Figure 3 and its strength is expressed in correlation of the appearance of early (+), intermediate (++), and
`late (+++) degradation products. cExtent of induction of tyrosine phosphorylation of ErbB-2 protein in D2 cells was determined according to
`the assay in Figure 4. dInhibition of EGF and NDF binding to T47D cells was determined as described in Figure 5. *Stimulatory antibody
`
`– – + + N
`
`D – –
`
`ND
`
`– – – –
`
`– N
`
`D + + + – – N
`
`D
`ND
`
`– – –
`
`IMMUNOGEN 2035, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`thereby excluding
`absence of other ErbB proteins,
`transphosphorylation e€ects that widely occur within
`the ErbB family (Pinkas-Kramarski et al., 1996).
`Lysates of antibody-treated cells,
`immunoblotted for
`the detection of proteins phosphorylated on tyrosine
`residues, demonstrated the dependency of ErbB-2
`phosphorylation on mAb bivalency (Figure 4, upper
`panel). Tumor-inhibitory mAbs from both Class I and
`Class
`II
`(L431 and L26,
`respectively)
`caused a
`comparable
`extent of phosphorylation,
`that was
`absent when the monovalent antibody fragments
`(Fab) were used. Antibody L140, a moderate cancer
`e€ector,
`exerted maximal
`elevation of
`receptor
`phosphorylation, that was higher than the e€ect of
`the tumor-stimulatory mAb, N28 (Stancovski et al.,
`1991). This suggests that the ability of antibodies to
`a€ect the extent of ErbB-2 phosphorylation reflects
`only their capacity to form ErbB-2 homodimers and as
`implied by Table 1 it may be independent of the long-
`term biological activity in vivo. Consistent with its low
`binding a(cid:129)nity, mAb L87 could not mediate a
`phosphorylation signal upon the receptor.
`
`Class II mAbs inhibit ErbB-2 interactions with
`ErbB-familiy counterparts
`
`The engagement of ErbB-2 in hetero-complexes with
`other ErbB-family tyrosine kinases, has been shown to
`augment both binding and signaling of EGF and NDF
`when associated with their respective receptors (Graus-
`Porta et al., 1995; Karunagaran et al., 1996; Kokai et
`al., 1989; Sliwkowski et al., 1994). To examine whether
`anti-ErbB-2 mAbs can interfere with heterodimer
`formation, we used ligand a(cid:129)nity as an indicator of
`ErbB-2 involvement in ligand binding complexes. This
`
`Fab431
`
`L431
`
`Fab26
`
`L26
`
`–
`
`6B11
`
`L140
`
`L87
`
`N28
`
`–
`
`190 —
`
`190 —
`
`Figure 4 Antibody-induced stimulation of ErbB-2 phosphoryla-
`tion on tyrosine residues. ErbB-2-expressing 32D cells (denoted
`D2 cells) were incubated for 15 min at 378C with the indicated
`mAbs at 20 mg/ml, or with their respective monovalent fragments
`(Fab, 20 mg/ml). Whole cell
`lysates were then prepared and
`subjected to gel electrophoresis. The gel-resolved proteins were
`transferred to a nitrocellulose filter that was blotted with an
`antibody to phosphotyrosine and detected with a secondary
`antibody. Incubation in the absence of antibody (lane labeled –)
`or with an isotope matched control mAb (antibody 6B11) were
`used for control. The location of a marker protein is indicated in
`kDa
`
`Tumor-inhibitory antibodies to ErbB-2
`LN Klapper et al
`
`their e€ect on the internalization of membrane-bound
`ErbB-2. The internalization assay used has not been
`previously
`applied to ErbB-2,
`and it
`included
`biotinylation of the surface-exposed protein, followed
`by exposure to the various mAbs. Molecules that
`underwent
`internalization
`escaped
`a
`subsequent
`digestion with pronase, that was applied extracellula-
`rily and were thus visualized by streptavidin detection.
`A 20 min-long incubation in the presence of mAbs
`from Class I (represented by L431) revealed a band of
`approximately 85 kDa, that represents a relatively late
`degradation product of ErbB-2 (Figure 3). A similar
`result was obtained upon incubation with the tumor-
`inhibitory mAb L242 (comprising a single-mAb
`group), whereas antibodies from Class II caused the
`appearance of the apparently early proteolytic product
`of 120 kDa, implying a slower degradation pathway.
`Internalization of ErbB-2 by mAb L140 (Class III), a
`moderate inhibitory mAb, resulted in both proteolytic
`products and a residual
`intact
`receptor
`(185 kDa
`protein band). Antibody L87 (Class VI), a non-
`inhibitory antibody, induced no detectable internaliza-
`tion of ErbB-2, an e€ect that was shared with the
`control IID2 antibody against a-fetoprotein. Because
`the size of the protein recovered by the internalization
`assay presumably reflects the rate of internalization,
`the results presented in Figure 3 suggest a dependency
`of tumor inhibition on the ability of the mAbs to
`internalize the receptor (Table 1). Of note, however, is
`the relatively slow endocytic processing that was
`induced by Class II mAbs (L26 and L288), implying
`that
`their
`strong
`anti-tumor
`e€ect depends on
`additional activities.
`tumor-
`It has been previously reported that
`inhibitory e€ects of anti-ErbB-2 mAbs only partially
`correlate with modulations of
`the phosphotyrosine
`content of the receptor (Kumar et al., 1991; Stancov-
`ski et al., 1992). To examine the relationship between
`tumor inhibition and stimulation of ErbB-2 phosphor-
`ylation, we selected a model cellular system of 32D
`myeloid cells that ectopically express ErbB-2 in the
`
`ID2
`
`L140
`
`26
`
`– L
`
`L242
`
`L431
`
`L288
`
`L87
`
`199 —
`
`120 —
`87 —
`
`Figure 3 E€ect of mAbs on degradation of ErbB-2. Cell surface
`proteins of confluent monolayers of N87 cells (10 cm dishes) were
`labeled with biotin as described under Materials and methods.
`The cells were then exposed for 30 min at 48C to the indicated
`mAbs (at 40 mg/ml). The monolayers were thereafter incubated
`for 20 min at 378C in order to allow receptor internalization and
`degradation. At the end of this incubation, the monolayers were
`transferred back to 48C, treated for 30 min with pronase to digest
`surface-exposed proteins and cell lysates were prepared. ErbB-2
`proteins that escaped hydrolysis by pronase were visualized by
`immunoprecipitation with the NCT antibody, that was followed
`by gel
`electrophoresis,
`transfer
`to nitrocellulose filter and
`detection with horseradish peroxidase-labeled streptavidin. Note
`that only internalized receptor is visualized. For control we
`incubated the cells in the absence of mAb (lane labeled -) or in the
`presence of an irrelevant mAb (antibody IID2). The locations of
`marker proteins are
`indicated in kilodaltons
`(kDa). The
`experiment was repeated twice
`
`2102
`
`IMMUNOGEN 2035, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`I
`

`

`Tumor-inhibitory antibodies to ErbB-2
`LN Klapper et al
`
`2103
`
`Figure 5 E€ect of mAbs to ErbB-2 and their Fab fragments on receptor binding of NDF and EGF. (A) Monolayers of T47D cells
`growing in 48-well dishes were incubated for 2 h at 48C with either 125I-EGF or 125I-NDF (each at 10 ng/ml) in the presence of
`increasing concentrations of the following mAbs that represent four di€erent classes of mAbs: L431 (squares), L26 (circles), L140
`(triangles), and L87 (rhombuses). Unbound radiolabeled ligand was then removed by washing and cell-associated radioactivity
`determined. (B) Experiments were conducted as in (A) to compare ligand binding in the presence of the Fab fragment of mAb L26
`(closed circles) to the binding in the presence of the whole mAb (open circles). Antibody N28 (open crosses) and its Fab fragment
`(closed crosses) and antibody L140 (open triangles) served as controls. Each data point represents the average and standard
`deviation (bars) of duplicate determinations after subtraction of the non-specific ligand binding. The experiment was performed
`thrice
`
`including
`lines,
`assay was performed on several cell
`N87 that
`expresses ErbB-1, ErbB-2 and ErbB-3
`receptors and the human T47D breast cancer cell line,
`that expresses all four ErbB proteins. Figure 5A depicts
`the results of binding analyses of radiolabeled NDF
`and EGF,
`in the presence of representative mAbs
`directed against di€erent ErbB-2 epitopes.
`It
`is
`important to note that none of the antibodies cross-
`reacted with other ErbB family members. Antibody
`L26, as well as other Class II mAbs, were able to
`displace up to 74% and 42% of cell-bound EGF and
`NDF, respectively. This phenomenon was not char-
`acteristic of mAbs
`capable of
`recognizing other
`receptor determinants (e.g., mAbs L431, L87 and
`L140, Figure 5A), suggesting that the epitope bound
`by mAbs from Class II is involved in the formation of
`heterodimers. This hypothesis was further supported by
`the inhibition of EGF and NDF binding to T47D cells
`by monovalent fragments of antibody L26 (Figure 5B).
`Fab fragments of L26 could inhibit the binding of both
`ligands, to an extent similar to that of the whole mAb,
`whereas the Fab of an antibody incapable of ligand
`binding inhibition (N28) could not. It is worth noting
`that all our ligand binding analyses were performed at
`48C,
`in order to exclude di€erences due to ErbB-2
`
`internalization. Similar results were obtained with the
`N87 cell
`line (data not shown). To directly test the
`prediction that Class II mAbs inhibit ligand binding by
`interfering with the formation of ErbB-2-containing
`heterodimers, we covalently labeled each receptor with
`a radiolabeled ligand, and analysed coprecipitation of
`the a(cid:129)nity labeled receptor with ErbB-2. The results of
`this experiment, that was performed with N87 cells, are
`shown in Figure 6. Evidently, both monomeric and
`dimeric receptor species were precipitated by anti-
`ErbB-2 antibodies. However, the presence of mAb L26
`during the a(cid:129)nity labeling reaction significantly
`reduced coprecipitation with ErbB-2. The e€ect was
`larger with EGF than with NDF, consistent with the
`results of the ligand displacement assay (Figure 5A),
`and it was not
`induced by a control non-relevant
`antibody (6B11). However, all mAbs to ErbB-2 slightly
`reduced the e(cid:129)ciency of a(cid:129)nity labeling, especially
`with EGF, probably due to aspecific masking of
`primary amino groups.
`To further study the mechanism underlying anti-
`body-induced inhibition of ligand binding we measured
`the e€ect of mAb L26 on the a(cid:129)nity of EGF and NDF
`to their receptors (Figure 7A). Scatchard analyses
`revealed a minimal e€ect on the number of binding
`
`IMMUNOGEN 2035, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`sites, but a significant reduction in ligand a(cid:129)nities
`upon co-incubation with a Class II mAb: threefold for
`EGF and twofold for NDF. Because ErbB-2-contain-
`ing heterodimers are characterized by a relatively slow
`rate of ligand association (Karunagaran et al., 1996),
`we tested the prediction that an acceleration in ligand
`dissociation was responsible for the observed inhibitory
`e€ect of mAb L26. The kinetics of EGF and NDF
`release were determined in the presence or absence of
`the L26 mAb. Evidently,
`the mAb significantly
`increased the rates of dissociation of both ligands
`from their cellular binding sites (Figure 7B). Taken
`together, the results of ligand binding analyses imply
`that mAbs belonging to Class
`II are capable of
`interfering with the stability of dimers formed between
`ErbB-2 and its family members, namely EGF- and
`NDF-receptors.
`
`Class II mAbs inhibit transactivation of growth-
`regulatory signals by ErbB-2
`
`Prolonged binding of NDF and EGF achieved by the
`presence of ErbB-2 is thought to augment growth-
`regulatory signals. Indeed, ErbB-2 has the ability to
`potentiate the proliferative e€ect of EGF and to
`
`Tumor-inhibitory antibodies to ErbB-2
`LN Klapper et al
`
`EGF
`
`6B11
`
`L26
`
`L431
`
`125 I-LIGAND:
`
`NDF
`
`6B11
`
`L26
`
`L151
`
`mAb:
`
`2104
`
`D
`
`M
`
`D s
`
`M
`
`— 180
`
`— 180
`
`Figure 6 The e€ect of mAbs on a(cid:129)nity labeling and co-
`immunoprecipitation of NDF- and EGF-receptors with ErbB-2.
`Monolayers of confluent N87 cells were incubated for 2.5 h with
`10 ng/ml of 125I-labeled NDF-b1177 – 246 or EGF, in the presence
`of mAb L26 (5 and 25 mg/ml, left and right panels respectively),
`L151 (5 mg/ml), or L431 (25 mg/ml), as indicated. A control mAb,
`6B11, was also added (5 and 25 mg/ml,
`left and right panels
`respectively). The cell monolayers were washed with PBS and
`then subjected to a(cid:129)nity labeling for 25 min with BS3 (1 mM),
`followed by cell lysis. After clearance of cell debris, the detergent-
`solubilized lysates were subjected to separate immunoprecipitation
`reactions with NCT, a rabbit polyclonal antiserum directed to the
`C-terminus of
`the ErbB-2 protein.
`Immunocomplexes were
`resolved by gel electrophoresis on a 6.5% acrylamide gel
`followed by autoradiography. Note that monomeric receptor is
`coprecipitated representing ligand bound receptor that is probably
`involved in heterodimers
`
`Figure 7 Demonstration of mAb-induced changes in NDF and EGF binding. (A) Reduction of ligand a(cid:129)nity. Monolayers of
`T47D cells were incubated for 2 h at 48C with increasing concentrations of 125I-NDF or 125I-EGF in the presence (open circles) or
`absence (closed circles) of mAb L26 (50 mg/ml). Unbound ligand removal was followed by cell lysis and radioactivity monitoring.
`Non-specific ligand binding was determined in the presence of a 100-fold excess of the unlabeled ligand, and was subtracted from
`the total amount of ligand that bound at each concentration. The results are presented as Scatchard plots and as saturation curves
`(insets). Each data point represents an average of a duplicate, and the whole experiment was repeated thrice. (B) Acceleration of
`ligand dissociation. T47D cells were first incubated for 2 h at 48C with radiolabeled EGF or NDF (each at 20 ng/ml) as indicated.
`Thereafter, the ligands were removed and the cells incubated with an unlabeled ligand (200 ng/ml) and in the presence (open circles)
`or absence (closed circles) of mAb L26 (20 mg/ml). Cell-bound as well as released radioactivity were then followed as a function of
`time of incubation at 48C. The results are expressed as the ratio between the amount of ligand that was bound at time t (Bt) and the
`initially bound ligand (B0) and they represent the average and standard deviation of duplicates. The experiments were repeated twice
`
`IMMUNOGEN 2035, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`s
`s
`s
`s
`

`

`reconstitute mitogenic signals transmitted by NDF
`activation of ErbB-3 (Pinkas-Kramarski et al., 1996).
`To examine the e€ect of heterodimer destabilization by
`antibody L26 on signal transmission by growth factors,
`we measured the synthesis of DNA in cells expressing
`the appropriate
`receptor
`combinations. 32D cells
`expressing a combination of ErbB-2 with either
`ErbB-1 (denoted D12 cells) or with ErbB-3 (D23
`cells) showed a decreased potency of the mitogenic
`e€ects exerted by EGF and NDF, respectively, when
`examined in the presence of mAb L26. Consistent with
`the lower ligand a(cid:129)nity exhibited in the presence of
`antibody L26, the dose-response curves of EGF- and
`NDF-induced DNA synthesis shifted to higher ligand
`concentrations. Furthermore,
`the maximal
`signal
`attained declined in the presence of mAb L26 (Figure
`8). It is of relevance to note that signals transmitted by
`EGF were adjusted by the mAb to a greater extent
`than those of NDF, reflecting the di€erences exhibited
`in antibody displacement of each ligand (Figure 5A).
`In conclusion, our results suggest
`that
`the tumor-
`inhibitory Class II mAbs can interfere with the ability
`of ErbB-2 to form the superior heteromeric signaling
`complexes with NDF- and EGF-receptors.
`
`Figure 8 Antibody-induced trans-inhibition of EGF- and NDF-
`mediated mitogenic e€ects. 32D myeloid cells that ectopically
`express a combination of ErbB-2 with ErbB-1 (D12 cells, upper
`panel) or a combination of ErbB-2 with ErbB-3 (D23 cells, lower
`panel) were deprived of serum factors and interleukin-3. The cells
`were plated at a density of 56105 cells/ml in media containing
`increasing concentrations of the indicated growth factors in the
`presence (open circles) or absence (closed circles) of mAb L26
`[3H-methyl]thymidine (1 mCi/ml) was added and its
`(50 mg/ml).
`incorporation into DNA determined 18 h later. The results are
`presented as the extent of induction over the control untreated
`cells, and are the average and standard deviation (bars) of four
`determinations. The experiments were repeated thrice
`
`Tumor-inhibitory antibodies to ErbB-2
`LN Klapper et al
`
`Discussion
`
`2105
`
`Understanding the molecular mechanisms underlying
`antibody-induced retardation of
`tumor growth is
`essential not only for optimal selection of mAbs for
`therapy, but it may also help explain the relatively high
`transforming potential of ErbB-2 in comparison with
`other receptor tyrosine kinases. In the present study we
`used a large collection of mAbs to ErbB-2,
`in an
`attempt
`to correlate specific intrinsic activities of
`various mAbs with their tumor-inhibitory potential.
`This analysis, summarized in Table 1, led us to the
`following conclusions: (i) The extracellular domain of
`ErbB-2 contains multiple (47) non-overlapping anti-
`genic sites that are capable of tumor inhibition. (ii) The
`intrinsic
`abilities of mAbs
`to induce
`endocytic
`degradation (Figure 3), to elevate tyrosine phosphor-
`ylation of ErbB-2 (Figure 4), and to increase DNA
`synthesis in factor-dependent cells (Pinkas-Kramarski
`et al., 1996 and data not shown) are independent of a
`specific antigenic determinant. However, all
`three
`e€ects are strictly dependent on antibody bivalency
`(Hurwitz et al., 1995 and data not shown), implying
`their association with ErbB-2 homodimers. (iii) All
`mAbs that are directed to the most antigenic site of
`ErbB-2 (Class II mAbs) are able to inhibit crosstalk
`with EGF- and NDF-receptors. Unlike other intrinsic
`mAb actions, this e€ect is independent of antibody
`bivalency,
`suggesting an association with ErbB-2-
`containing heterodimers.
`It is clear from our results with antibodies L140
`(Figures 1 and 4) and N28 (Stancovski et al., 1991) that
`stimulation of ErbB-2 phosphorylation is not coupled
`to the e€ects on tumor growth. This conclusion is
`consistent with previous reports, including the partial
`agonist activity of
`the tumor-inhibitory mAb 4D5
`(Kumar
`et al., 1991). On the other hand, our
`experiments with the new set of mAbs to ErbB-2
`provide further support for the possibility that tumor
`inhibition is correlated with e(cid:129)cient antibody-induced
`endocytic degradation of ErbB-2. Consistent with the
`observed variation in the extent of ErbB-2 degradation
`(Figure 3),
`the less e(cid:129)cient mAbs L87 and L140,
`respectively exhibited no or weak tumor-inhibitory
`e€ects, and the monovalent fragments of the strong
`tumor-inhibitory mAbs lost both internalizing and
`growth-inhibitory activities
`(data not
`shown). A
`linkage between tumor
`retardati

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