throbber
American journal ofPathology, Vol, 151, No, 6, December 1997
`Copyright © American Society for Investigative Pathology
`
`Short Communication
`
`Neutralizing Antibodies against Epidermal Growth
`Factor and ErbB-2/neu Receptor Tyrosine Kinases
`Down-Regulate Vascular Endothelial Growth Factor
`Production by Tumor Cells in Vitro and in l/ivo
`
`Angiogenic Implications for Signal Transduction Therapy of
`Solid Tumors
`
`Alicia M. Viloria Petit,*T Janusz Rak,*
`Mien-Chie Hung,t Patricia Rockwell,§
`Neil Goldstein,§ Brian Fendly,", and
`Robert S. Kerbel“r
`
`From the Division of Cancer Biology Research,‘ Sunnybrook
`Health Science Centre, and the Department ofMedical
`Biophysicsfi University of Toronto, Toronto, Ontario, Canada; the
`Department of Tumor Biology/,1: University of Texas, MD.
`Anderson Cancer Center, Houston, Texas,- ImClone Systems, Inc,5
`New York, New York; and Genentech, Inc.,'| South San
`Francisco, California
`
`The overexpression in tumor cells of (proto)-onco-
`genie receptor tyrosine kinases such as epidermal
`growth factor receptor (EGFR) or ErbBZ/neu (also
`known as HER-2) is generally thought to contribute to
`the development of solid tumors primarily through
`their effects on promoting uncontrolled cell prolifer-
`ation. However, agents that antagonize the function
`of the protein products encoded by these (prom)-
`oncogenes are known to behave in viva in a cytotoxic-
`like manner. This implies that such oncogenes may
`regulate critical cell survival functions, including an-
`giogenesis. The latter could occur as a consequence of
`regulation of relevant growth factors by such onco-
`genes. We therefore sought to determine whether
`EGFR or ErbBZ/neu may contribute to tumor angio-
`genesis by examining their effects on the expression
`of vascular endothelial cell growth factor (VEGF)/vas-
`cular permeability factor (VPF), one of the most im-
`portant of all known inducers of tumor angiogenesis.
`We found that in vitro treatment of EGFR-positive
`A431 human epidermoid carcinoma cells, which are
`
`known to be heavily dependent on VEGF/VPF in vivo
`as an angiogenesis growth factor, with the C225 anti-
`EGFR neutralizing antibody caused a dose-dependent
`inhibition of VEGF protein expression. Prominent
`suppression of VEGF/VPF expression in viva , as well
`as a significant reduction in tumor blood vessel
`counts, were also observed in established A431 tu-
`mors shortly after injection of the antibody as few as
`four times into nude mice. Transformation of NIH 3T3
`fibroblasts with mutant ErbBZ/neu, another EGFR-
`like oncogenic tyrosine kinase, resulted in a signifi-
`cant induction of VEGF/VPF, and the magnitude of
`this effect was further elevated by hypoxia. Moreover,
`treatment of ErbBZ/neu-positive SKBR-3 human
`breast cancer cells in vitro with a specific neutralizing
`anti-ErbBZ/neu monoclonal antibody (4D5) resulted
`in a dosedependent reduction of VEGF/VPF protein
`expression. Taken together, the results suggest that
`oncogenif properties of EGFR and ErbBZ/neu may, at
`least in part, be mediated by stimulation of tumor
`angiogenesis by up-regulating potent angiogenesis
`growth factors such as VEGF/VPF. These genetic
`changes may cooperate with epigenetic/environmen-
`tal efi'ects such as hypoxia to maximally stimulate
`VEGF/VPF expression. Therapeutic disruption of
`EGFR or ErbBZ/neu protein function in viva may
`
`
`Supported by Medical Research Council of Canada (CA—41223) and
`National Institutes of Health, US, Public Health Service (CA-58880).
`Accepted for publication September 22, 1997.
`Address reprint requests to Dr, Robert S. Kerbel. Division of Cancer
`Biology Research, Research Building, 8-218, Sunnybrook Health Science
`Centre, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5. R. S,
`Kerbel is a Terry Fox Scientist of the National Cancer Institute of Canada,
`supported by the Canadian Cancer Society,
`Genentech 2077
`Celltrion v. Genentech
`lPR2017-01122
`
`1523
`
`Genentech 2077
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`Viloria Petit et al
`1524
`AJP December 1997, Vol. 151, No. 6
`
`therefore result in partial suppression of angiogene-
`sis, a feature that could enhance the therapeutic in-
`dex of such agents in vivo and endow them with
`anti-tumor effects, the magnitude of which may be
`out of proportion with their observed cytostatic ef-
`fects in monolayer tissue culture.
`(AmJ Pathol 1997,
`151:1523—1530)
`
`One of the major cellular changes that accompanies
`tumor development and progression is overexpression of
`proto—oncogenic protein receptor tyrosine kinases. such
`as the epidermal growth factor receptor (EGFR) or the
`ErbB2/neu (also known as HER-2).1 Given their increased
`expression in many types of solid tumors, and location at
`the external cell surface, there has been considerable
`interest in developing and utilizing agents that block in a
`relatively selected way the signaling function of these
`receptor tyrosine kinases?3 These agents include spe-
`cific neutralizing monoclonal antibodies, such as 4D5
`and C225, which block the human ErbB2/neu and
`EGFRs, respectively. Both are now being evaluated in
`early-phase clinical trials, either alone or in combination
`with various cytotoxic anti-cancer chemotherapeutic
`drugs with which they may synergize.4
`As with most other agents that act as inhibitors of
`signal transduction, such blocking antibodies are gener-
`ally viewed as cytostatic drugs. This is based on the
`observation that they usually appear to lack any obvious
`cytotoxic properties when tested against relevant tumor
`cell targets grown in monolayer tissue culture. Despite
`this, there are situations in which these antibodies or
`agents appear to exert therapeutic effects against estab-
`lished human solid tumors in preclinical animal models,
`the magnitude of which are strongly suggestive of the
`involvement of a cytotoxic, or cytotoxic-like, effect. A
`similar discrepancy has been noted with respect to pro—
`tein farnesyltransferase inhibitors of mutant RAS onco-
`proteins.5 For example, the maximal anti-tumor effect of
`the C225 monoclonal anti-EGFR antibody against A431
`human epidermoid carcinoma cells in culture is approx-
`imately 35% growth inhibition, with no cytotoxicity.6 Nev-
`ertheless, injections of this antibody into nude mice har-
`boring established A431 xenografts can result in total
`regression of the tumors within a relatively short period of
`time.6 A similar in vitro/in vivo therapeutic discrepancy has
`been noted with the 4D5 anti-ErbBZ/neu antibody.7 The
`extent of these in vivo therapeutic effects are all the more
`surprising given the usual physiological and pharmaco-
`kinetic problems, such as antibody delivery into solid
`tumors, that exist in vivo but not in vitro to limit the thera-
`peutic potential of many anti-cancer agents.a These ob-
`servations suggest that the antibodies may acquire anti-
`tumor mechanisms of action in vivo not normally detected
`in monolayer tissue culture, such as, for example, anti-
`body-dependent cell—mediated cytotoxicity. Perhaps an
`even more appealing possibility is inhibition of tumor
`angiogenesis.
`The notion that signal transduction inhibitor drugs may
`function as anti-angiogenic agents, which we first put
`forward in the context of the effects of protein farnesyl—
`
`transferase inhibitors of mutant RAS oncoproteins,9 is
`based on the hypothesis that oncogene/proto—oncogene-
`mediated signal transduction pathways may up-regulate
`the expression of one or more growth factors that function
`as stimulators of angiogenesis."-10 One such factor is
`vascular endothelial growth factor (VEGF), also known as
`vascular permeability factor (VPF), which is currently re-
`garded as the major angiogenesis stimulator for most
`types of human cancers.11 indeed, of the many known
`inducers of VEGFNPF, two of the most potent are epider-
`mal growth factor (EGF) and transforming growth factor
`(TGF)-ar‘2'13 which are ligands for the EGFR. Hence,
`anti—EGFR antibodies might be expected to suppress
`VEGFNPF expression both in vitro and, possibly, in vivo. If
`so, this could result in an anti-angiogenic effect that might
`even lead to tumor regression, given that even partial
`VEGF withdrawal can lead to destruction of newly formed
`immature vessels, such as those found in neonatal reti-
`nas14 and solid tumors,15 in addition to inhibiting the
`development of new blood vessels.
`The purpose of the present study was to evaluate
`whether EGFR or ErbBZ induces or up-regulates VEGF/
`VPF expression and,
`if so, whether pharmacological or
`genetic blockade of either EGFR or ErbBZ/neu receptor
`kinases can lead to a suppression of VEGFNPF produc-
`tion, both in vitro and in vivo, using appropriate receptor—
`bearing murine or human target tumor cells. The phar-
`macological approach was undertaken using monoclonal
`neutralizing antibodies specific for either receptor kinase.
`Our decision to focus on VEGFNPF as an overall surro-
`
`gate marker of angiogenesis was based on several con-
`siderations, namely, 1) its ubiquity as a tumor angiogen-
`esis factor,11 2) that relatively small reductions (two— to
`threefold) in VEGFNPF can lead to unexpectedly pro-
`found suppressions of developmental16 and tumor angio-
`genesis,17 and 3) that it is known to be a major angio-
`genesis growth factor for the A431 human squamous
`carcinoma,18 which was the tumor selected for our in vivo
`therapy studies reported here, The results we obtained
`showed that these agents can indeed suppress tumor
`VEGFNPF expression, both in cell culture and in vivo, and
`therefore raise the possibility that these agents may have
`an anti-angiogenic component as part of their mode of
`anti-tumor action in vivo.
`
`Materials and Methods
`
`Cell Lines and Culture Conditions
`
`The human epidermoid carcinoma cell line, A431, was
`obtained from the American Type Culture Collection
`(ATCC, Rockville, MD) and maintained as a monolayer
`culture in Dulbecco’s minimal essential medium (DMEM)
`supplemented with 10% fetal bovine serum (FBS; Gibco-
`BRL, Grand Island, NY). This cell line is known to over-
`express EGFR. The human breast adenocarcinoma cell
`line SKBR-3, which overexpresses ErbB2/neu, came from
`two independent sources, namely, the ATCC and as a gift
`from Dr. Peter Taylor (lnstituto Venezolano de Investiga—
`ciones Cienti’ficas IVIC, Caracas, Venezuela). These cells
`
`

`

`were maintained in DMEM/F12 medium supplemented
`with 10% FBS (Gibco-BRL). B10411. cells are trans-
`formed NIH 3T3 fibroblasts generated by transfection
`with a mutant neu (the rat homologue of
`the human
`ErbB-2) oncogene originally identified in a rat neuro(g|io-
`)blastoma.19 This cell line and its parental NIH 3T3 coun—
`terpart were cultured in DMEM/F12 supplemented with
`10% FBS.
`
`Antibodies
`
`The neutralizing anti-EGFR monoclonal antibody C225,
`originally described by Kawamoto et al,20 was produced
`by lmClone Systems (New York, NY). Murine monoclonal
`antibody against the extracellular domain of the human
`ErbBZ/neu receptor 4D53 and the anti-VEGF antibody
`A461 were produced by Genentech (South San Fran-
`cisco, CA).
`
`Measurement of Human and Mouse VEGF
`
`Protein Levels in Conditioned Medium (EL/SA)
`
`Commercially available human or mouse VEGF ELISA
`kits (R&D Systems, Minneapolis, MN) were used to quan-
`titate the levels of VEGF in conditioned medium obtained
`from A431 and SKBR-3 or NIH 3T3 and B10411. cells,
`respectively, according to the manufacturer’s instruc-
`tions. Briefly, cells were plated at a density of 105 (A431
`and SKBR-S) or 75 X 103 (NIH 3T3 and B104.1.1.)cel|s/
`0.5 ml/well in a 24-well plate and allowed to reach near
`confluency, at which point the growth medium was re-
`placed with fresh assay medium containing 0225, 4D5
`monoclonal antibodies, or nonspecific lgG (as a control),
`FBS and, where indicated, 100 umol/L CoClg. Condi—
`tioned medium was collected after 24, 36, or 48 hours,
`cellular debris removed by centrifugation, and medium
`kept at —70°C until VEGF quantitation was undertaken.
`Cell number was determined immediately after medium
`recovery using a Coulter Counter ZM (Coulter Electron-
`ics, Luton, UK). Cobalt chloride was used to mimic hy-
`poxic conditions in cell culture.21
`
`Northern Blotting
`
`Approximately 108 cells were used for the extraction of
`polyadenylated mRNA by a standard SDS-oligodeoxy—
`thymidylic acid method. The RNA was resolved on 1%
`agarose gel containing 6.6 moI/L formaldehyde,
`trans—
`ferred to Zeta Probe (Bio-Rad, Hercules, CA) membrane,
`and hybridized at 65°C with a 32P-labeled cDNA probe
`containing 200-bp human VEGF sequence common for
`all four known VEGFNPF isoforms (a gift from Dr. Brygida
`Berse and Dr. Harold Dvorak, Beth Israel Hospital, Bos-
`ton, MA). The membranes were autoradiographed after
`the transfer, and the intensity of the 3.7- and 4.5-kb
`VEGFNPF signal was evaluated.
`
`1525
`VEGF Down-Regulation by Anti-HER-2 MAb
`AjP December 1997, Vol. 151, No. 6
`
`Evaluation of the in Vivo Anti-Tumor Activity of
`
`the 0225 Anti-EGFR Antibody
`
`A431 cells were cultured to semiconfluency in their ap-
`propriate growth medium. The cultures were harvested
`by brief trypsin/EDTA (GlBCO-BRL) treatment, washed in
`serum-free medium, resuspended at
`the appropriate
`density in PBS, and then inoculated subcutaneously
`(3.0.) at a density of 106/02 ml into athymic 8-week-old
`nu/nu BALB/c mice. After 15 days (when tumors reached
`a size of 200 to 300 mm3), treatment was initiated by
`injecting intraperitoneally 1 mg. of C225 monoclonal an-
`tibody per mouse every other day, for a total of four
`injections. Control mice were injected with PBS. Tumors
`were measured periodically, and tumor volume (mms)
`was calculated by using the standard formula a2 X b/2,
`where a is the width and b is the length of the horizontal
`tumor perimeter. The experiment was terminated after 1
`week, when tumors were removed and immediately fro-
`zen in ornithine carbamyl transferase compound (Tissue-
`Tek) or formalin fixed for immunohistochemistry. Similarly,
`as a control in vivo experiment, mice bearing established
`A431 tumors were treated with two injections, 3 days
`apart, of anti-VEGF A4.6.1 antibody (300 jig/mouse or 0.2
`ml of PBS), and tumor tissue was processed for immu—
`nohistochemistry.
`
`Immunohistochemistiy and Blood Vessels
`
`The formalin-fixed, paraffin-embedded specimens of in-
`dividual control or antibody—treated tumors were sec-
`tioned and processed for standard immunohistochemical
`staining. Anti-VEGF rabbit polyclonal antibody A-20 (San-
`ta Cruz Biotechnology, Santa Cruz, CA) was used at a
`1:200 dilution in combination with a proper secondary
`antibody from the Histostain—SP kit (Zymed Laboratories,
`San Francisco, CA) and 3-amino—9-ethyl carbazole (AEC)
`chromogen to reveal antigen as a red signal. Anti-Ki67
`rabbit polyclonal antibody NCLki67p (Novocastra Labo—
`ratories, New Castle, UK) was used at a 1:1000 dilution.
`The color reaction was developed by using an anti-rabbit
`secondary antibody, the Vectastain ABC kit (Vector Lab-
`oratories, Burlingame, CA) and diaminobenzidine tetra-
`hydrochloride (DAB, Pierce, Rockford,
`IL) as a chromo—
`gen to obtain brown coloration. Blood vessel staining was
`performed on unfixed frozen sections by endothelial cell
`labeling with GSI Iectin from Griffonla simplicifolia as pre—
`viously described.22
`
`Statistical Analysis
`
`The Mann-Whitney nonparametric test for unpaired data
`was used to evaluate the immunohistochemistry results.
`All P values represent two-sided tests of statistical signif-
`icance. The analyses were performed using the Graph—
`PAD lnStat program version 1.14 (GraphPAD Software,
`Inc., San Diego, CA).
`
`

`

`more immunodetectable VEGF. Thus, although hypoxic
`conditions, as expected. can induce VEGF in nontrans-
`formed cells, the magnitude of this effect is remarkably
`enhanced in the same cells when they contain an acti-
`vated oncogene. This observation reinforces the notion
`that genetic and epigenetic factors cooperate to bring
`about an angiogenic switch during malignant transforma-
`tion and tumor progression.26-27'30
`
`Down-Regulation of VEGF Production in SKBR-
`
`3 Human Breast Cancer Cells upon Treatment
`
`with 4D5 (Anti-ErbBZ/neu) Antibody
`
`The aforementioned results suggest that through phar-
`macological blockade of the ErbBZ/neu protein it should
`be possible to suppress VEGFNPF expression. We de-
`cided to test this hypothesis using mouse neutralizing
`monoclonal anti-human ErbB2/neu antibodies and rele-
`vant human tumor cells. We could not use the neu-trans-
`
`fected variants of NIH 3T3 cells for such experiments as
`the antibody does not react against rodent neu. In human
`breast cancer cells, endogenous overexpresslon of the
`ErbBZ/neu oncogene is frequently associated with tumor
`progression, and the neutralizing anti-ErbBZ/neu anti-
`body known as 4D5 (a specific monoclonal antibody
`against the human but not rat extracellular domain of
`ErbBZ/neu) has been shown to possess significant anti-
`tumor properties in vivo in preclinical models.7 We there-
`fore employed SKBR-S cells to assess whether treatment
`with this antibody is able to suppress VEGF production in
`the case of a natural. human, ErbB2/neu-transformed
`breast cancer cell line. This indeed appears to be the
`case as shown in Figure 18. Although untreated SKBRS
`cells were found to secrete appreciable quantities of
`VEGF (approximately 1000 pg/ml/105 cells) into the con-
`ditioned media. increasing concentrations of 4D5 anti-
`body brought about a dose-dependent decrease in
`VEGF production, which reached 50% at the concentra-
`tion of 50 lug/ml antibody, indicating that indeed ErbBZ/
`neu activity can be at least one significant factor regulat-
`ing VEGFNPF expression in these cells.
`
`Down—Regulation of VEGF Production in A431
`
`Cells by Anti-EGFR Antibody C225 in Vitro
`
`The tumorigenic transformed phenotype of A431 human
`epidermoid carcinoma cells is thought to be dependent,
`at least in part, on overexpresslon of EGFR. As monoclo-
`nal antibody C225, directed against the EGFR, has been
`shown to inhibit growth of established A431 tumors in
`vivo, we hypothesized that down-regulation of VEGF and
`ultimately tumor angiogenesis itself might be a contribut-
`ing factor in causing such an anti-tumor effect. As a first
`step toward testing this hypothesis we first treated A431
`cells in vitro with increasing concentrations of the C225
`antibody and measured secretion of VEGF into condi-
`tioned media as well as expression of VEGF mRNA.
`Figure 2A shows that VEGF production was inhibited by
`the antibody treatment
`in a dose-dependent manner.
`
`Viloria Petit et al
`1526
`AjP December 1997, Vol. 151, No. 6
`
`A
`
`1400
`
`NIH3T3
`1200
`
`Q E
`
`so
`a.
`
`
`
`
`
`(pg/ml)
`VEGFproduction
`
`
`v E
`
`‘5=
`'8
`a
`E
`L;
`
`1000
`800
`
`600
`400
`200
`
`
`2
`
`0
`
`0
`
`FBS (%)2 0
`CoClz(uM): 0
`
`4D5 (pg/ml): 0
`
`0.78
`
`3.13
`
`12.5
`
`50.0
`
`Figure 1. VEGF production by cells transformed with ErbBZ/ neu oncogene,
`A: Up-regulation of VEGF immunoreactivity in conditioned medium of
`ErbBZ/neu-transformed NIH 3T3 cells (B104.1.1). VEGF production is stim-
`ulated by serum and hypoxia (CoClz) treatment in both 8104.11 and control
`NIH 3T3 cells, but maximal levels in the case of the former cells are up to
`100-fold higher. B: Dose-dependent down-regulation of VEGF production by
`SKBR-3 human breast carcinoma cells after 48 hours of treatment with 4D5
`(anti-ErbBZ/neu) antibodyi Error bars, SD.
`
`Results
`
`Up-Regulation of VEGF Production in
`Fibroblasts Transformed with ErbB2/neu
`
`Oncogene
`
`Transformation of NIH 3T3 fibroblasts with a rat onco-
`
`genic mutant of the ErbBZ/neu receptor results in acqui-
`sition of a tumorigenic phenotype23 for which expression
`of angiogenic properties is presumably an absolute re—
`quirement.24 Therefore we decided to examine whether
`expression of VEGF, a potent angiogenic growth factor
`frequently regulated by oncogenic proteins such as mu—
`tant ras,9'25‘28 is also up-regulated in the case of fibro-
`blastic cell line B104.1.1 transformed with ErbB2/neu on-
`
`cogene.‘9"“9 Figure 1A shows the comparative analysis
`of VEGF protein production by B104.1.1 cells and their
`nontransformed parental NIH 3T3 cells under different
`culture conditions. In confirmation of the results of Grugel
`et al,25 VEGF secretion into conditioned media by NIH
`3T3 cells was virtually undetectable in the absence of
`serum.
`In striking contrast. B104.1.1 cells produced an
`abundance of this angiogenic growth factor. Exposure of
`parental NIH 3T3 cells to serum (2 to 10%) and hypoxia
`(100 umol/L CoCl2) resulted in secretion of measurable
`amounts of VEGF; however,
`the corresponding condi-
`tioned media of B104.1.1 cells contained up to 100—fold
`
`

`

`1527
`VEGF Down-Regulation by Anti-HER-2 MAb
`AjP December 1997, Vol. 151, No. 6
`
`
`
`A
`
`
`
`
`VEGF production (mean)
`-Thymidine
`C225
`
`(pg/ml) " ~
`.
`_ in2‘!-
`'
`874 pg/ml
`
`696 pg/ml
`
`583 pg/ml
`
`524 pg/ml
`
`509 pg/ml
`
`120
`
`140
`
`
`
` ”2"“:WM
`
`100
`80
`0
`20
`40
`6f]
`Percent of control
`
`275
`250
`
`421 pg/ml
`l c225 injections
`
`
`
`
`
`Tumorsize(%increase)
`
`225
`175
`200
`150
`125
`100
`50
`25
`0
`
`[17%;
`
`.,
`
`l
`l
`0
`
`f
`l
`2
`
`r
`
`
`I/-
`'W' *Control
`l
`’
`,, o.
`, Treated / /
`a
`/;
`/‘
`
`F:
`l
`‘77
`l
`l
`'T"
`4
`
`,
`g
`
`l
`6
`
`"
`
`_,
`
`_"— l
`8
`
`Days of treatment
`
`Although control A431 cells produced significant quanti—
`ties of VEGF (800 to 900 pg/mI/1O5 cells), the amount of
`this cytokine produced by their counterparts treated for
`24 hours with 62.5 ng/ml C225 antibody was approxi-
`mately 50% lower (400 pg/ml/105 cells). Corresponding
`measurements of [3H]thymidine incorporation indicated
`that the suppressive effect of the antibody on A431 cell
`proliferation was on the order of 35% (Figure 2A),
`in
`confirmation of previous results.6 The down-regulation of
`VEGF production by C225 antibody is likely to operate at
`the level of gene transcription, given that treatment with 1
`fig/ml C225 antibody resulted in a 50% decrease in
`expression of VEGF mRNA (Figure 2B).
`
`Down-Regulation of VEGF Production and
`
`Inhibition of A431 Tumor Growth by C225
`
`Antibody in Vivo
`
`To further investigate the potential role of the C225 antibody
`as a possible anti-angiogenesis agent in vivo, we examined
`the effect of this antibody on the growth of established A431
`tumors and their in situ VEGF production. Transient therapy
`consisting of only four injections of C225 antibody (spaced
`2 days apart) into A431 tumor-bearing mice resulted in
`appreciable (treated to control ratio = 33%) inhibition of
`tumor growth (Figure 20). As expected, staining for expres-
`sion of proliferation-associated Ki67 antigen revealed a pro—
`nounced withdrawal of tumor cells from the cell cycle when
`the mice were exposed to C225 (Figure 3, E and F; Table 1).
`
`GAPDH
`
`Figure 2. The effect of treatment with C225 anti-EGFR antibody
`on growth of A431 tumor cells in vim: and in viva and their
`VEGF production. A: Dose-dependent inhibitory effect of C225
`treatment on VEGF production and growth of A431 cells in who.
`B: Down—regulation of VEGF mRNA expression in A431 cells
`treated for 24 hours with 1 [Lg/ml C225 antibody. Bottom panel:
`loading control (GAPDH). C: Growth—inhibitory effect of C225
`antibody on in viva growth of established A431 tumors in nude
`mice after transient (l—week) treatment (time 0 corresponds to
`day 15 of tumor growth; error bars, SD; n 2 6 mice).
`
`Such an effect may be either direct, as reported earlier, or
`secondary to inhibition of angiogenesis, as treatment of
`A431 tumors with anti—VEGF neutralizing antibody A4.6.1
`also resulted in a decrease in numbers of Ki67-positive
`tumor cells, especially in the innermost part of the tumor
`(34% t 4.6 positive cells in control versus 15% t 3.1 in
`treated mice; P = 0.0004; data not shown). Therefore, it is
`possible that a similar anti-angiogenic effect would conceiv-
`ably contribute to the therapeutic efficacy of C225 treat-
`ment, at least in the case of A431 tumors. Figure 3, A and B,
`shows the results of
`immunohistochemical staining for
`VEGF in C225-treated and control A431 tumor sections.
`
`Control tumors are highly positive for VEGF protein. This is
`particularly true for clusters of large tumor cells that are
`negative for Ki67, suggesting that the main source of VEGF
`in this case are nondividing rather than proliferating tumor
`cells.
`In contrast, tumors treated with C225 antibody are
`largely negative for VEGF staining with the exception of
`weakly positive clusters of the aforementioned large tumor
`cells. Consistent with this pattern, a twofold reduction in the
`average number of blood vessels in treated versus control
`tumors was also observed (Figure 3, C and D; Table 1).
`These in vivo results appear to corroborate those obtained in
`vitro.
`
`Discussion
`
`Many inhibitory signal transduction agents that disrupt
`the transforming functions of oncogenes and (proto)—
`
`

`

`Viloria Petit et al
`1528
`AJP December 1997, Vol. 151, No. 6
`
`Control
`
`C225 treated
`
`VEGF
`
`
`
`
`Griffonialectin
`
`
`Figure 3. Immunohistochemical evaluation of VEGF expression and cellular proliferation in A431 tumors. A431 tumor—bearing mice were treated with anti-EGFR
`(C225) antibody (B, D, and F) or with PBS (A, C, and E). VEGF down-regulation is apparent in C225-treated A431 tumors (compare A and B), Blood vessel
`numbers were reduced in treated versus control mice (compare C and D), Consistent with an antiproliferative effect in viva, Ki67 positivity was lower after C225
`antibody treatment (compare E and F). Bar, 20 pm. See Table 1 and Materials and Methods for details.
`
`oncogenes lack overt cytotoxic properties when tested
`against tumor cells in culture?"32 Nevertheless, they can
`bring about impressive cytotoxic—like effects against es-
`tablished solid tumors in various preclinical models?"6
`These agents include the anti-EGFR and anti-ErbBZ/neu
`monoclonal neutralizing antibodies used in the studies
`reported here?7 Such a discrepancy can be explained,
`at least in part, by postulating that these agents may exert
`an anti-angiogenic effect in vivo by down-regulating one
`or more angiogenesis growth factors,
`including VEGF/
`VPF. Clearly, the therapeutic benefits of such an anti-
`
`angiogenic effect would not be apparent in cell-culture-
`based drug testing or screening protocols.
`We chose to evaluate whether overexpression of the
`EGFR, and the EGFR-related receptor tyrosine kinase
`ErbBZ/neu, stimulate VEGF/VPF production in solid tu-
`mors as various neutralizing monoclonal antibodies to the
`proteins encoded by these (proto)—oncogenes have been
`used extensively in preclinical therapeutic models and
`have now entered early-phase clinical trials.4 Other meth-
`ods of targetting proto-oncogene function in tumors are
`under active investigation, eg, gene therapy”34 and
`
`

`

`Table 1.
`
`Immunohistochemical Analysis of Ki67, VEGF, and
`Blood Vessel Staining on Paraffin-Embedded and
`Frozen Sections from A—431 Tumors Grown on
`Nude Mice
`
`
`Blood vessel
`Ki67
`
`Treatment
`VEGF
`counts : SD
`(% 1 SD)
`
`30 t 7.1 (6)
`24.2 t 4.7
`(+++)
`Control (PBS)
`
`
`
`10.3 : 4.1(+)C225 MAb 9 x 3.7 (7)
`VEGF cytoplasmic staining was assessed semiquantitatively by
`assigning a score based on color
`intensity produced by AEC red
`chromogen: —_ negative; +. slightly positive; ++, moderately positive;
`+++, highly positive.
`Percentages of positive cells and blood vessel counts were
`determined on a minimum of 10 high-power (40X) fields (100 cells/field
`in the case of Ki67) per slide and two slides per sample. Two-tailed P
`value was <0.0001 in both cases. See Materials and Methods for
`details. Numbers of tumors evaluated are indicated in parentheses.
`
`synthetic receptor antagonists.35 Our results, considered
`as a whole, strongly implicate the EGFR and ErbB2/neu
`as inducers of VEGFNPF and, hence, by extension, tu-
`mor angiogenesis. For example,
`in the case of EGFR,
`treatment of the human epidermoid carcinoma A431 cell
`line with the C225 monoclonal anti-EGFR antibody re-
`sulted in a dose-dependent inhibition of VEGFNPF ex-
`pression at both the mRNA and protein levels. Signifi-
`cantly, this decline in VEGFNPF was not restricted to in
`vitro treatments; four injections of C225 antibody into
`nude mice with an established A431 human epidermoid
`carcinoma xenograft resulted in an obvious down-regu—
`lation of VEGFNPF expression, which accompanied the
`tumor
`growth
`inhibition.
`Similar
`conclusions were
`reached in the context of ErbB2/neu. Thus, transfection of
`a transforming-competent, mutant ErbB2/neu oncogene
`into mouse NIH 3T3 cells resulted in a dramatic up—
`regulation of VEGFNPF expression, and neutralizing an-
`tibodies to human ErbB2/neu could cause a detectable
`suppression of VEGFNPF expression in SKBR-S breast
`cancer cells, which are known to be VEGFNPF -positive
`and overexpress ErbB2/neu. These results may be perti-
`nent to the prognostic link recently established between
`tumor angiogenesis and c-erbB2/neu expression in na-
`sopharyngeal cancer.36
`One obvious question our findings raise is whether it is
`reasonable to propose that an anti-tumor effect could result.
`at least in part, by inhibition of angiogenesis when the
`degree of VEGFNPF suppression induced by the antibody
`treatments is in the range of twofold in vitro and perhaps
`slightly more in vivo. For several reasons, we feel the answer
`is yes. First, studies in VEGF knockout mice have shown that
`disruption of only a single VEGF allele, equivalent to 50%
`reduction of VEGF protein, is sufficient to block vasculogen-
`esis,and angiogenesis to such an extent that embryos die
`between days 11 and 12 gestation.16 This remains an un-
`precedented finding. Second,
`induced suppression of
`VEGF protein expression in a human glioblastoma by only
`threefold, assessed by using antisense genetic methods,
`can almost completely obliterate the tumorigenic ability of
`such variant cells in nude mice.17 Third, it is clearly proba-
`ble that the antibody treatments we studied would suppress
`the expression of some additional growth factors known to
`be pro-angiogenic, eg, basic fibroblast growth factor, inter—
`
`1529
`VEGF Down-Regulation by Anti-HER-2 MAb
`AjP December 1997, Vol. 151, No. 6
`
`leukin-8, TGF-a, and TGF—B, as shown by Ciardiello et al.37
`This would clearly enhance the potential anti-angiogenic
`activity of the antibody treatments. Nevertheless, in the case
`of A431 squam0us carcinoma cells,
`it has already been
`shown that their growth in mice can be almost completely
`blocked by inhibiting the function of the flk—1
`(VEGFR2)
`mouse endothelial cell receptor for VEGF.18 Thus, even if
`the EGFR antibody treatment inhibited VEGF expression in
`vivo in A431 cells by approximately 50%, and did not sup-
`press any other angiogenic growth factor, it would still be
`reasonable to postulate that an anti-angiogenic effect could
`ensue from such a treatment. It is therefore not surprising
`that we also found a significant reduction in the extent of
`tumor vascularity in the A431 tumors removed from nude
`mice after only four treatments with the C225 antibody
`spaced 2 days apart, a finding consistent with our anti-
`angiogenesis hypothesis. The extent of reduction in blood
`vessel counts,
`in the range of twofold,
`is in line with the
`results of others, eg, Cheng et al,17 who observed a three-
`fold reduction in such counts in tumors obtained from injec-
`tion of VEGF antisense transfected cells where the growth of
`such tumors was profoundly suppressed in vivo.
`Finally. our results could be very important to the issue of
`the therapeutic index that can be attained by anti—tumor
`agents such as anti-EGFR or anti-ErbBZ/neu neutralizing
`antibodies. This is because, unlike cell proliferation, angio-
`genesis is not normally a prominent physiological process
`in healthy humans, with the exceptions of corpus luteum
`development in females. Thus,
`if the effect of such anti-
`tumor agents is mediated through suppression of tumor
`angiogenesis in addition to inhibition of tumor cell growth,
`the therapeutic index would be increased. This could help
`explain why these agents can exert anti-tumor effects in vivo
`that seem out of proportion With their generally modest
`anti-tumor effects in monolayer cell culture.
`It should be
`noted that such an in vitro/in vivo discrepancy may also be
`due to a pro-apoptotic effect mediated by these anti-tumor
`agents on tumor cells growing in a multicellular and/or an-
`chorage-independent context, as opposed to monolayer
`context. Indeed, genetic or pharmacological disruption of
`mutant ras genes or RAS proteins can lead to a high degree
`of apoptosis of mutant ras-transformed cells growing in
`three-dimensional culture38 or anchorage independently39
`but not in monolayer cell culture, where only an anti-prolif-
`erative effect is observed.
`
`Acknowledgments
`
`We thank Lynda Woodcock, Mina Viscardi, and Cassan-
`dra Cheng for their excellent secretarial help. The gift of
`the anti-ErbBZ/neu (4D5) antibody from Genentech is
`gratefully acknowledged.
`
`References
`
`1. Salomon DS, Brandt R. Ciardiello F, Normanno N: Epidermal growth
`factor-related peptides and their receptors in human

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