throbber
Proc. Nall. Acad. Sci. USA
`Vol. 84, pp. 7159-7163, October 1987
`Cell Biology
`
`Increased expression of the putative growth factor receptor
`p185I
`R2 causes transformation and tumorigenesis of NIH 3T3 cells
`ROBERT M. HUDZIAK*, JOSEPH SCHLESSINGERt, AND AXEL ULLRICH*
`*Department of Developmental Biology, Genentech, Inc., 460 Point San Bruno Boulevard, South San Francisco, CA 94080; and tBiotechnology Research
`Center, Meloy Laboratories, 4 Research Court, Rockville, MD 20850
`
`Communicated by Hilary Koprowski, July 13, 1987
`
`ABSTRACT
`The HER2 gene encodes a cell-surface glyco-
`protein with extensive homology to the epidermal growth factor
`receptor. Recently it was found to be amplified in about 30%
`of primary human breast malignancies. In experiments de-
`signed to assess the role of the HER2 gene in oncogenesis, we
`found that overexpression of unaltered HER2 coding sequences
`in NIH 3T3 cells resulted in cellular transformation and
`tumorigenesis.
`
`The HER2 gene encodes a transmembrane glycoprotein with
`extensive structural homology to the human epidermal
`growth factor (EGF) receptor and the chicken oncogene
`v-erbB (1-3). Chromosomal mapping and sequence compar-
`ison strongly suggest that the HER2 gene product and the
`ethylnitrosourea-activated, rat neuroblastoma oncogene neu
`represent species variants of the same polypeptide (4). The
`neu oncogene encodes a 185-kDa cell-surface glycoprotein
`that possesses intrinsic tyrosine-specific kinase activity that
`is likely to be activated by an as yet unidentified ligand (5, 6).
`Comparison of the transforming neu oncogene sequence with
`its normal rat protooncogene counterpart suggested that a
`point mutation in the transmembrane domain resulting in
`substitution of a valine residue by glutamate unmasked the
`transforming potential of this putative growth factor receptor
`(7). Analogously, structural alterations have converted nor-
`mal genes coding for the receptors for macrophage colony-
`stimulating factor type 1 and EGF into v-fms (8) and v-erbB
`(9) oncogenes, respectively.
`Southern analysis of primary human tumors and estab-
`lished tumor-derived cell lines revealed amplification and in
`some cases rearrangement of the EGF receptor gene. Am-
`plification was particularly apparent in squamous carcinomas
`(10, 11) and glioblastomas (12). The HER2 gene was also
`found to be amplified in a human salivary gland adenocarci-
`noma (3), a mammary gland carcinoma (2), and a gastric
`cancer cell line (13). Recently, Slamon et al. (14) demon-
`strated that about 30% of primary human breast carcinoma
`tumors contained an amplified HER2 gene. Although a few
`sequence rearrangements were detected, in most tumors
`there were no obvious differences between amplified and
`normal HER2 genes. Furthermore, amplification of the
`HER2 gene correlated significantly with the prognosis of the
`disease and the probability of relapse.
`To investigate the significance of the correlation between
`overexpression and cellular transformation as it has been
`observed for protooncogenes c-mos (15) and c-Ha-rasl (16),
`we employed a HER2 expression vector and a selection
`scheme that permitted sequence amplification after transfec-
`tion of mouse NIH 3T3 cells. We report here that amplifi-
`cation of the unaltered HER2 gene in NIH 3T3 cells leads to
`overexpression ofp185HER2 as well as cellular transformation
`and tumor formation in athymic mice. These findings, in
`
`combination with the results of Slamon et al. (14), suggest
`that mere amplification of the HER2 gene and resulting
`overexpression of its product may play a crucial role in the
`genesis and development of some types of human cancer.
`
`MATERIALS AND METHODS
`Expression Plasmids. The mammalian expression vector
`CVN (17) contained expression units for mouse dihydrofolate
`reductase (DHFR) cDNA (18) and the bacterial neomycin
`phosphotransferase (neo) gene (19), both under simian virus
`40 early promoter control. Transcription of a 4.4-kilobase-
`pair Sal I-Dra I HER2 fragment containing the full-length
`HER2 coding region (1) was driven by the Rous sarcoma
`virus (RSV) long terminal repeat promoter (LTR). The
`poly(A) site was provided by the 3' untranslated sequence of
`the hepatitis B virus surface antigen gene (20). The control
`CVN plasmid was identical but lacked cDNA sequences
`downstream from the RSV LTR.
`Cell Culture. NIH 3T3 cells were cultured in a 1:1 mixture
`of Dulbecco's modified Eagle's medium and Ham's nutrient
`mixture F-12 supplemented with glutamine (2 mM), penicillin
`(100 units/ml), streptomycin (100 ,g/ml), and 10% HyClone
`(Logan, Utah) calf serum in a humidified incubator under 5%
`CO2 in air atmosphere.
`Transfections and Amplification. Plasmid DNA was intro-
`duced into mammalian cells by the calcium phosphate
`coprecipitation method (21). Half-confluent plates of cells (60
`mm) were exposed to 5 ,ug of plasmid DNA in 1 ml of
`precipitate for 6-8 hr. After a 20% (vol/vol) glycerol shock
`(22), the cells were fed with nonselective medium. Two days
`later, they were passaged into selective medium containing
`Geneticin (G418) at 400 ,ug/ml.
`Clones were picked using glass cloning cylinders with
`petroleum jelly for the bottom seal. Colonies arising from
`transfected cells selected for growth in G418 were picked,
`expanded, and subcultured into medium containing 7%
`dialyzed fetal bovine serum in place of 10% calf serum and the
`appropriate concentration of methotrexate for plasmid am-
`plification (23). The dialysis step removes trace amounts of
`purines and pyrimidines present in serum that decrease the
`efficiency of the methotrexate selection. To apply selective
`pressure, stepwise increasing concentrations of methotrex-
`ate were used with a final concentration of 400 nM. To avoid
`enriching for spontaneously transformed cells, cells were
`kept subconfluent. An additional control was to amplify the
`CVN neo-DHFR vector without the HER2 cDNA insert in
`the NIH 3T3 recipient cell line.
`Immunoprecipitations and Labeling. The G-H2CT17 anti-
`body recognizing the C-terminal 17 amino acids of HER2 was
`prepared in rabbits using a synthetic peptide conjugated with
`soybean trypsin inhibitor.
`
`The publication costs of this article were defrayed in part by page charge
`payment. This article must therefore be hereby marked "advertisement"
`in accordance with 18 U.S.C. §1734 solely to indicate this fact.
`
`Abbreviations: DHFR, dihydrofolate reductase; R, resistance; EGF,
`epidermal growth factor; RSV, Rous sarcoma virus; LTR, long
`terminal repeat.
`
`7159
`
`PETITIONER'S EXHIBITS
`
`Exhibit 1047 Page 1 of 5
`
`

`
`7160
`
`Cell Biology: Hudziak et al.
`
`Proc. Natl. Acad. Sci. USA 84 (1987)
`
`Cells were harvested by trypsinization and counted by
`Coulter Counter, and 1.5 x 106 cells were plated per 60-mm
`culture dish. After 36 hr, the cells were lysed at 4°C with 0.4
`ml of HNEG buffer per plate (50 mM Hepes, pH 7.5/150 mM
`NaCI/1 mM EGTA/10% glycerol) containing 1.0% Triton
`X-100 detergent and 1 mM phenylmethylsulfonyl fluoride.
`After 10 min, 0.8 ml of lysis dilution buffer (HNEG buffer
`with 1% bovine serum albumin and 0.1% Triton X-100) was
`added to each plate and the extracts were pelleted at 12,000
`x g for 5 min.
`HER2 antibody was added to the cell extracts, which were
`then incubated at 4°C for 2 hr; this was followed by incubation
`with protein A-Sepharose beads for 20 min and three washes
`with 1 ml of HNEG buffer with 0.1% Triton X-100.
`Autophosphorylation reactions were carried out at 4°C in 50
`Al of HNEG wash buffer with 5 mM MnCl2 and 3 ,Ci of
`[y-32P]ATP (Amersham, 5000 Ci/mmol; 1 Ci = 37 GBq) for
`20 min. Proteins were separated on 7.5% NaDodSO4/poly-
`acrylamide gels and analyzed by autoradiography.
`Transformation Assays. The efficiency of colony formation
`in soft agar (24) was determined by plating 25,000 cells in 3
`ml of 0.2% agar (Difco, "purified") over 4 ml of 0.4% agar in
`a 60-mm dish. After 2-4 weeks, colonies of about 100 cells or
`more were counted.
`The plating efficiency of cell lines (25) in 1% calf serum was
`determined by plating equal numbers of cells into 100-mm
`plates with either 10% or 1% calf serum. After 2-3 weeks, the
`plates were stained with crystal violet and colonies were
`counted.
`Mouse Tumorigenicity Assays. Athymic (nu/nu) mice were
`obtained from Charles River Breeding Laboratories. Control
`NIH 3T3 and NIH 3T3/CVN cells and experimental HER2-
`3400 cells were harvested by trypsinization and counted with
`a Coulter Counter. They were then collected by low-speed
`centrifugation and resuspended in ice-cold phosphate-buff-
`ered saline to either 2.5 x 106, 5.0 x 106, or 1.0 x 107 cells
`per ml. Animals were injected subcutaneously with 0.1-ml
`volume of the cell suspensions. Tumor occurrence and size
`were monitored twice weekly.
`
`RESULTS
`For expression ofHER2 sequences in NIH 3T3 cells, a cDNA
`coding for the entire 1255-amino acid polypeptide (1) was
`placed under transcriptional control of the RSV LTR. Tran-
`scriptional termination signals and a poly(A) site were pro-
`vided by 3' sequences of the hepatitis virus surface antigen
`gene (20). In addition, the expression vector contained the
`neo resistance (neoR) gene, which confers cellular resistance
`to the aminoglycoside antibiotic G418 (18) and therefore
`allows selection of primary transfectants, as well as the
`DHFR gene for methotrexate resistance, which was used to
`amplify transfected DNA sequences under selective pres-
`sure. Both drug resistance genes were under simian virus 40
`early promoter transcriptional control. Bacterial plasmid
`sequences, including an origin of replication and the gene for
`ampicillin resistance, allowed replication of the entire expres-
`sion plasmid in Escherichia coli.
`The transforming activity of HER2 sequences was initially
`tested using a conventional NIH 3T3 cell focus-formation
`assay. Under conditions that resulted in about 104 foci per ,g
`of a v-fms viral construct, we were unable to detect any
`HER2-transforming activity. Because of the recently report-
`ed finding that about 30% of mammary carcinomas contain
`amplified HER2 gene sequences without apparent sequence
`rearrangements (14), we investigated whether amplification
`of an unaltered HER2 gene could transform mouse fibro-
`blasts. NIH 3T3 cells were transfected with the pCVN/HER2
`construct. An identical plasmid missing the HER2 expression
`module was used as a control. Four independent primary
`
`Assay for growth in soft agar and 1% calf serum of
`Table 1.
`HER2 primary and amplified cell lines
`
`Plating
`efficiency
`Soft agar
`HER2 gene copies
`in 1% calf
`colonies,
`per haploid
`serum, %
`Cell line
`no.
`genome, no.
`NIH 3T3/CVN
`0.27
`0
`0
`NIH 3T3/CVN400
`0
`0
`0
`3
`1
`3
`HER2-1
`HER2-14m
`38
`424
`60
`1.4
`0
`2
`HER2-3
`HER2-34m
`11.7
`836
`55
`HER2-4
`0.2
`0
`4
`HER2-44
`49
`376
`90
`0.6
`0
`1
`HER2-B3
`HER2-B34w
`50.2
`373
`131
`Two control lines were used. The first one was a NIH 3T3 line
`transfected with a plasmid containing only the neo and DHFR genes.
`The second control line contained the neo-DHFR plasmid and was
`amplified to resistance to 400 mM methotrexate. HER2 gene copy
`numbers were determined using a human DNA standard and
`densitometer scanning of Southern hybridization autoradiograms.
`Equal cell numbers (25,000) were plated in soft agar and colonies
`were counted after 2-4 weeks. The plating efficiency in 1% calf
`serum is relative to the number of colonies arising when an equal
`aliquot was simultaneously plated in medium containing 10% calf
`serum.
`
`G418-resistant clones (HER2-1, HER2-3, HER2-4, HER2-
`B3) were isolated. Cell lines containing amplified HER2
`coding sequences were generated from these parental clones
`by culturing the cells in gradually increasing concentrations
`of methotrexate up to 400 nM (HER2-14m, HER2-3400,
`HER2-4400, HER2-3B4w). Southern hybridization analysis of
`parental and amplified cell lines demonstrated that the HER2
`cDNA copy number increased from 1-4 to 55-131 per haploid
`genome (Table 1).
`To test whether gene amplification resulted in overexpres-
`sion of the HER2 gene product, cell lysates were im-
`munoprecipitated with an antibody against the C-terminal 17
`amino acids of the HER2 sequence. As shown in Fig. 1,
`substantially increased levels of the p185 HER2 gene product
`were found in amplified cell lines relative to their parental
`G418R transfectants. The parental cells had a normal mor-
`phology that was indistinguishable from NIH 3T3 cells.
`However, amplified cells had the typical refractile, spindle-
`1 234 5 6 789
`
`..'
`
`O
`
`200-
`
`116-
`
`Quantitation ofp185HER2 in four primary, unamplified cell
`FIG. 1.
`lines and lines derived from them by amplification to resistance to 400
`nM methotrexate. Lane 1, neo-DHFR control; lanes 2 and 3, HER2-1
`parent and amplified lines; lanes 4 and 5, HER2-3 parent and
`amplified lines; lanes 6 and 7, HER2-4 parent and amplified lines;
`lanes 8 and 9, HER2-B3 parent and amplified lines. Positions of the
`size markers myosin and j8-galactosidase are indicated in kDa.
`
`PETITIONER'S EXHIBITS
`
`Exhibit 1047 Page 2 of 5
`
`

`
`Cell Biology: Hudziak et al.
`
`Proc. Natl. Acad. Sci. USA 84 (1987)
`
`7161
`
`FIG. 2.
`Morphology of NIH 3T3 cells transfected with HER2
`expression construct. (1) NIH 3T3 cells transfected with the control
`neo-DHFR plasmid. (2) NIH 3T3 cells with the neo-DHFR plasmid
`amplified to resistance to 400 mM methotrexate. (3) Primary G418R
`cell line HER2-1, an unamplified clone expressing low levels ofp185.
`(4) The same clone amplified to 400 mM methotrexate resistance.
`(x50.)
`
`shaped appearance oftransformed cells and grew in irregular,
`piled-up clumps (Fig. 2).
`NIH 3T3 cell lines with an amplified HER2 gene and high
`levels of HER2 gene expression also displayed other char-
`acteristics associated with a transformed phenotype. As
`shown in Table 1 and Fig. 3, these cells all formed colonies
`in soft agar and were able to grow at low density at low serum
`concentration. In contrast, primary transfectants did not
`grow under these conditions. The primary transfectants did,
`however, grow to a higher saturation density than the
`parental NIH 3T3 cells.
`The correspondence between a transformed phenotype
`and HER2 gene amplification and overexpression was inde-
`pendently confirmed by directly selecting for transformed
`cells and then analyzing the resulting clones. For this purpose
`the parental cell line HER2-3, which contains about two
`copies of the HER2 expression construct, was cultured in
`medium containing a low concentration of fetal calf serum
`(0.5%); control cells containing the expression vector without
`HER2 coding sequences (pCVN) were cultured in parallel.
`After 5 weeks, a few colonies appeared in the control culture
`and roughly 10-fold more colonies appeared in dishes con-
`taining the HER2-3 cell line. These colonies appeared to be
`morphologically transformed and were subsequently ana-
`lyzed for HER2 overexpression. As shown in Fig. 4, three
`individual clones as well as a pool of the remaining colonies
`had elevated levels of p185HER2 compared with the original
`parental G418-resistant HER2-3 cell line. In addition, there
`
`Anchorage-independent growth of HER2-transformed
`FIG. 3.
`cells in soft agar. Cells were plated in 0.2% soft agar over a 0.4% agar
`lower layer. After 3 weeks the plates were photographed at 40x
`magnification using a Nikon microscope with phase-contrast optics.
`(Upper) Control untransformed NIH 3T3 cells. (Lower) Anchorage-
`containing the HER2
`independent growth of the cell line HER2-34
`expression plasmid and amplified to resistance to 400 mM methotrex-
`ate. (X25.)
`
`was a 26-fold increase in the number ofcells plating in 100 nM
`methotrexate in the selected cells compared with the parental
`cells, implying that the unselected but linked DHFR gene had
`also been coamplified.
`The tumorigenicity of cell lines with a high HER2 cDNA
`copy number was tested in nude mice by subcutaneous
`
`1 2 3 4 5 6
`
`wast -185
`
`Quantitation of p185 in the primary line HER2-3 and cell
`FIG. 4.
`lines selected for growth in 0.5% calf serum. HER2-encoded protein
`was immunoprecipitated and labeled. Lanes 1-3, three colonies
`picked and expanded from plates after selection for growth in 0.5%
`serum; lane 4, the starting cell line, HER2-3; lane 5, a pool of 0.5%
`serum-selected colonies of HER2-3; lane 6, a clone derived from a
`G418R control line selected for growth in 0.5% calf serum. The
`position expected for a protein of apparent molecular mass of 185
`kDa is indicated.
`
`PETITIONER'S EXHIBITS
`
`Exhibit 1047 Page 3 of 5
`
`

`
`7162
`
`Cell Biology: Hudziak et al.
`
`Table 2.
`
`Tumorigenicity testing of cell lines
`
`Cell line
`NIH 3T3
`
`NIH 3T3/CVN
`
`HER2-34m
`
`Mice with tumors/
`Cells injected, no.
`mice injected
`0.25 x 106
`0/6
`0/6
`0.5 x 106
`0/6
`1.0 x 106
`0/6
`0.25 x 106
`0.5
`x 106
`0/6
`1.0 x 106
`0/6
`0.25 x 106
`5/5
`0.5 x 106
`6/6
`1.0 X 106
`5/5
`The cell line HER2-3 and two control lines, NIH 3T3 cells and a
`NIH 3T3 line containing the control plasmid, were injected subcu-
`taneously at three different dosages into nude mice. The time after
`injection before tumors became visible was dose related: 22 days
`(average) for 1 x 106 cells, 28 days (average) for 0.5 x 106 cells, and
`34 days (average) for 0.25 X 106 cells.
`
`injection ofthree different cell numbers per animal. As shown
`in Table 2, the overexpressing cell line HER2-34m was
`strongly tumorigenic at all dosages tested, whereas the
`control cell lines, NIH 3T3 cells and NIH 3T3 cells
`transfected with CVN vector without the HER2 insert, were
`both negative under the same conditions. Necropsy of three
`mice with well-established tumors failed to identify any
`metastasis. Cell lines reestablished from three excised tu-
`mors still expressed the G418R phenotype, were resistant to
`methotrexate, and expressed high levels of pl85HER2 (not
`shown).
`
`DISCUSSION
`Amplification of the HER2 gene has been reported in a few
`primary human tumors (2, 3) and tumor-derived cell lines (13,
`26). Recently, Slamon et al. (14) found that the HER2 gene
`is amplified in 30% of primary breast tumors, a common
`human malignancy that affects about 7% of all American
`females. Notably, only 3/189 tumors surveyed showed any
`gross rearrangement of the HER2 gene. To assess the role of
`the HER2 gene in the neoplastic process, we characterized
`the transforming potential ofthe HER2 gene in an in vitro cell
`culture system.
`Expression of the full-length cDNA in NIH 3T3 cells did
`not lead to transformation as determined by a standard
`focus-forming transfection assay. However, HER2 overex-
`pression caused by gene amplification transformed these
`cells. Colonies that survived methotrexate selection were
`morphologically transformed and exhibited loss of contact
`inhibition. Such cells also grew in soft agar and would grow
`in 1% calf serum. Furthermore, cells transformed by HER2
`were tumorigenic in athymic mice.
`Selection of cells transfected with the HER2 cDNA for
`growth in low serum provided independent evidence that
`high-level expression of an unaltered HER2 gene product
`caused cellular transformation. Since DNA introduced into
`mammalian cells by transfection is more labile than genomic
`DNA (23, 27), we reasoned that selection for a property
`demonstrated by pharmacologically amplified HER2 lines-
`namely, growth in low serum-might lead to amplification or
`other changes resulting in overexpression of p185HER2.
`Clones derived from an unamplified HER2 line by this selection
`procedure appeared morphologically transformed and exhibit-
`ed elevated levels of p185HER2.
`Taken together, the characteristic morphological changes,
`results of in vitro and in vivo transformation and tumorigenic-
`ity assays, and the elevated levels of p185HER2 in cells
`selected for a transformed phenotype imply that high-level
`expression of HER2 results in transformation of NIH 3T3
`
`Proc. Natl. Acad. Sci. USA 84 (1987)
`
`cells. Another member ofthe tyrosine kinase gene family was
`recently found to be amplified 4- to 8-fold in spontaneously
`arising foci of NIH 3T3 cells (28). Amplification of the met
`gene appears to be a frequent event and, similar to HER2
`amplification in mammary carcinomas, is rarely accompa-
`nied by gross rearrangements. These findings and the dis-
`covery of amplified EGF receptor genes in primary human
`tumors (10-12) suggest that overexpression of other growth
`factor receptor genes will also lead to transformation and
`tumorigenesis. Whether susceptibility to spontaneous ampli-
`fication is caused by characteristics of the genetic loci for
`HER2, met and EGF receptor, or cell-specific selective
`advantages caused by receptor overexpression remains to be
`investigated.
`is not yet clear whether transformation of cells
`It
`overexpressing a growth factor receptor gene is dependent on
`paracrine or autocrine stimulation by the appropriate ligand.
`For HER2, this question cannot yet be addressed since its
`ligand has not been identified. In the case of the EGF
`receptor, however, we were able to demonstrate that primary
`human tumors and tumor-derived cell lines frequently ex-
`press mRNAs for both the receptor and transforming growth
`factor type a ligand (29). The finding that mere overexpres-
`sion of an intact growth factor receptor may subvert normal
`cellular growth control mechanisms and lead to tumorigenic
`growth provides new potential for diagnostic approaches and
`therapeutic strategies for treatment of human malignancies.
`We express appreciation for the superb technical assistance of
`Thomas Dull in constructing the HER2 expression plasmids and Bill
`Lagrimas for help with the mouse tumorigenicity assays. We are
`grateful to Jeanne Arch for her patience and skill in typing this
`manuscript and to Dr. Suzanne Pfeffer for her valuable editorial
`comments.
`Coussens, L., Yang-Feng, T. L., Liau, Y.-C., Chen, E., Gray,
`1.
`A., McGrath, J., Seeburg, P. H., Libermann, T. A., Schles-
`singer, J., Francke, U., Levinson, A. & Ullrich, A. (1985)
`Science 230, 1132-1139.
`King, C. R., Kraus, M. H. & Aaronson, S. A. (1985) Science
`229, 974-976.
`Semba, K., Kamata, N., Toyoshima, K. & Yamamoto, T.
`(1985) Proc. Natl. Acad. Sci. USA 82, 6497-6501.
`Schechter, A. L., Hung, M.-C., Vaidyanathan, L., Weinberg,
`R., Yang-Feng, T. L., Francke, U., Ullrich, A. & Coussens,
`L. (1985) Science 229, 976-978.
`Akiyama, T., Sudo, C., Ogawara, H., Toyoshima, K. &
`Yamamoto, T. (1986) Science 232, 1644-1646.
`Stem, D. F., Heffernan, P. A. & Weinberg, R. A. (1986) Mol.
`Cell. Biol. 6, 1729-1740.
`Bargmann, C. I., Hung, M.-C. & Weinberg, R. A. (1986) Cell
`45, 649-657.
`Sherr, C. J., Rettenmier, C. W., Sacca, R., Roussel, M. F.,
`Look, A. T. & Stanley, E. R. (1985) Cell 41, 665-676.
`Ullrich, A., Schlessinger, J. & Waterfield, M. D. (1984) Na-
`ture (London) 307, 521-527.
`Hunts, J., Ueda, M., Ozawa, S., Abe, O., Pastan, I. &
`Shimizu, N. (1985) Gann 76, 663-666.
`Cowley, G., Smith, J. A., Gusterson, B., Hendler, F. &
`Ozanne, B. (1984) in Cancer Cells, eds. Levine, A., Vande
`Woude, G., Topp, W. & Watson, J. D. (Cold Spring Harbor
`Laboratory, Cold Spring Harbor, NY), pp. 5-10.
`Libermann, T. A., Nusbaum, H. R., Razon, N., Kris, R.,
`Lax, I., Soreq, H., Whittle, N., Waterfield, M. D., Ullrich, A.
`& Schlessinger, J. (1985) Nature (London) 313, 144-147.
`Fukushige, S.-I., Matsubara, K.-I., Yoshida, M., Sasaki, M.,
`Suzuki, T., Semba, K., Toyoshima, K. & Yamamoto, T.
`(1986) Mol. Cell. Biol. 6, 955-958.
`Slamon, D. J., Clark, G. M., Wong, S. G., Levin, W. J.,
`Ullrich, A. & McGuire, W. L. (1987) Science 235, 177-182.
`Blair, D. G., Oskarsson, M., Wood, T. G., McClements,
`W. L., Fischinger, P. J. & Vande Woude, G. G. (1981) Sci-
`ence 212, 941-943.
`Pulciani, S., Santos, E., Long, L. K., Sorrentino, V. &
`Barbacid, M. (1985) Mol. Cell. Biol. 5, 2836-2841.
`
`10.
`
`11.
`
`12.
`
`13.
`
`14.
`
`15.
`
`16.
`
`2.
`
`3.
`
`4.
`
`5.
`
`6.
`
`7.
`
`8.
`
`9.
`
`PETITIONER'S EXHIBITS
`
`Exhibit 1047 Page 4 of 5
`
`

`
`Cell Biology: Hudziak et al.
`
`Proc. Nati. Acad. Sci. USA 84 (1987)
`
`7163
`
`17.
`
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`23.
`
`Riedel, H., Schlessinger, J. & Ullrich, A. (1987) Science 236,
`197-200.
`Alt, F. W., Kellems, R. D., Bertino, J. R. & Schimke, R. T.
`(1978) J. Biol. Chem. 253, 1357-1370.
`Colbere-Garapin, F., Horodniceanu, F., Kourilsky, P. &
`Garapin, A.-C. (1981) J. Mol. Biol. 150, 1-14.
`Simonsen, C. C. & Levinson, A. D. (1983) Mol. Cell. Biol. 3,
`2250-2258.
`Graham, F. L. & van der Eb, A. J. (1973) Virology 52,
`456-467.
`Frost, E. & Williams, J. (1978) Virology 91, 39-50.
`Kaufman, R. J. & Sharp, P. A. (1982) J. Mol. Biol. 159,
`601-621.
`
`24.
`
`25.
`
`26.
`
`27.
`
`28.
`
`29.
`
`Tooze, J. (1973) The Molecular Biology of Tumor Viruses
`(Cold Spring Harbor Laboratory, Cold Spring Harbor, NY).
`van der Hoorn, F. A. & Muller, V. (1985) Mol. Cell. Biol. 5,
`2204-2211.
`Kraus, M. H., Popescu, N. C., Amsbaugh, S. C. & King,
`C. R. (1987) EMBO J. 6, 605-610.
`Jolly, D. J., Willis, R. C. & Friedmann, T. (1986) Mol. Cell.
`Biol. 6, 1141-1147.
`Cooper, C. S., Tempest, P. R., Beckman, M. P., Heldin,
`C.-H. J. & Brookes, P. (1986) EMBO J. 5, 2623-2628.
`Derynck, R., Goeddel, D. V., Ullrich, A., Gutterman, J. U.,
`Williams, R. D., Bringman, T. S. & Berger, W. H. (1987)
`Cancer Res. 47, 707-712.
`
`PETITIONER'S EXHIBITS
`
`Exhibit 1047 Page 5 of 5

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