throbber
Proc. NatI. Acad. Sci. USA
`Vol. 77, No. 6, pp. 3567-3570, June 1980
`Genetics
`
`Transformation of mammalian cells with an amplifiable
`dominant-acting gene
`(animal cell vectors/methotrexate resistance/gene amplification)
`M. WIGLER*, M. PERUCHO*, D. KURTZ*, S. DANAt, A. PELLICERt, R. AXELt, AND S. SILVERSTEINt
`*Cold Spring Harbor Laboratories, P.O. Box 100, Cold Spring Harbor, New York 11724; and tInstituteof Cancer Research, Columbia University,
`701 West 168th Street, New York, New York 10032
`Communicated by J. D. Watson, March 10, 1980
`
`We have transferred a mutant hamster gene
`ABSTRACT
`coding for an altered dihydrofolate reductase to wild-type cul-
`tured mouse cells by using total genomic DNA from metho-
`trexate-resistant Chinese hamster ovary A29 cells as donor. By
`demonstrating the presence of hamster gene sequences in
`transformants we have provided direct evidence for gene
`transfer. Transformants selected for increased resistance to
`methotrexate contain increased amounts of the newly trans-
`ferred gene. We have used this mutant dhfr gene to introduce
`the Escherichia coli antibiotic resistance plasmid pBR322 into
`animal cells. Amplification of the dhfr sequences results in
`amplification of the pBR322 sequences as well. The use of this
`gene may allow the introduction and amplification of virtually
`any genetic element in various new cellular environments.
`The ability to transfer purified genes into cultured cells provides
`a unique opportunity to study the function and physical state
`of exogenous genes in new cellular environments. The devel-
`opment of systems for DNA transfer in animal cells originated
`with the lytic transfection of cells by using purified viral DNA
`(1, 2) and progressed to the stable transfer of viral transforming
`functions to appropriate recipient cells (3). Subsequently, viral
`genes from the herpesviruses coding for the biochemically se-
`lectable marker thymidine kinase (TK) (4-6) were transferred
`to enzyme-deficient mutant cells. Restriction fragments of
`herpes simplex virus type 1 encoding TK were isolated (6) and
`subsequently cloned into bacterial plasmids (7). Through the
`use of this selectable marker, virtually any gene can now be
`introduced into recipient cells (8, 9); however, these cells must
`be tk- mutants. Other potential selection systems are available,
`and several laboratories have recently demonstrated the
`DNA-mediated transfer of cellular genes coding for selectable
`markers such as TK (10), adenine phosphoribosyltransferase
`(11) and hypoxanthine phosphoribosyltransferase (12, 13).
`Dominant mutant cellular genes coding for drug resistance
`in principle could serve as generalized biochemical vectors for
`wild-type cells. Cultured mammalian cells are exquisitely
`sensitive to the folate antagonist methotrexant (Mtx). Mtx-
`resistant cell lines have been identified in three categories: (i)
`cells with decreased uptake of this drug (14, 15); (ii) cells that
`produce inordinately high levels of dihydrofolate reductase
`(DHFR) (16, 17); and (iii) cells with structural mutations which
`lower the affinity of DHFR for Mtx (18). When they were ex-
`amined, cells producing high levels of DHFR were found to
`contain increased copy numbers of the dhfr gene (gene am-
`plification) (19). An interesting Mtx-resistant variant cell line
`(A29) has been identified that synthesizes increased amounts
`of a mutant DHFR with decreased affinity for Mtx (18). We
`have used genomic DNA from this cell line to transfer the
`
`The publication costs of this article were defrayed in part by page
`charge payment. This article must therefore be hereby marked "ad-
`vertisement" in accordance with 18 U. S. C. §1734 solely to indicate
`this fact.
`
`mutant dhfr gene to wild-type Mtx-sensitive cells. Exposure
`of Mtx-resistant transformed cells to increasing levels of Mtx
`selects for cells that have amplified the transferred gene.
`
`MATERIALS AND METHODS
`Cell Culture. Mouse Ltk- aprtV cells (11) and NIH 3T3 cells
`(20) (the latter generously provided by R. A. Weinberg) were
`maintained in Dulbecco's modified Eagle's medium containing
`10% calf serum and antibiotics (growth medium). Chinese
`hamster ovary (CHO) cells and A29 cells (18), Mtx-resistant
`CHO derivatives (generously provided by L. Siminovitch), were
`maintained in growth medium supplemented with 3 times the
`usual concentration of nonessential amino acids. A29 cells were
`grown in the presence of Mtx at 20 ug/ml.
`Extraction, Restriction Endonuclease Digestion, and Li-
`gation of DNA. High molecular weight DNA was extracted
`from cultured cells as described (9). DNA was analyzed by
`electrophoresis in 0.3% agarose gels with restriction fragments
`of herpes simplex virus DNA as markers. Only DNA whose
`molecular weight average was 35 X 106 or greater was used for
`transformation experiments. CsCI/ethidium bromide-purified
`form I DNA of the Escherichia colt plasmid pBR322 was iso-
`lated from cultures of E. «ft strain HB101 (21). A29 DNA and
`pBR322 were mixed at 3:1 mass ratios, completely digested with
`restriction endonuclease Sal I (under conditions recommended
`by supplier, Bethesda Research Labs), extracted once with
`aqueous buffer-saturated phenol/chloroform/isoamyl alcohol
`25:24:1 (vol/vol), and once with chloroform/isoamyl alcohol,
`24:1, and precipitated with ethanol. DNA was resuspended and
`ligated with T4 DNA ligase (Bethesda Research Laboratories,
`Rockville, MD) at 100 ,gg of DNA and 3 units of ligase per ml
`at 40C for 24 hr in the buffers recommended by the supplier.
`The ligation product was reextracted with phenol/chloro-
`form/isoamyl alcohol and precipitated with ethanol.
`Transformation and Selection. Ltk- aprF cells and NIH
`3T3 cells were transformed with genomic DNA by the calcium
`phosphate coprecipitation method (2) as described (11). All
`DNAs were sterilized by ethanol precipitation and resuspended
`in 1 mM Tris-HCl/1 mM EDTA, pH 7.9. For tk+ transforma-
`tion, cells were exposed to hypoxanthine/aminopterin/thy-
`midine selective medium as described (10). Transformants
`resistant to Mtx were select~d in growth medium containing
`either 0.1 or 0.2 jqg of Mtx per ml with the same feeding
`schedule as for tk selection. Afte 2-3 weeks, colonies were
`isolated from individual dishes with cloning cylinders to ensure
`that each transformant arose from an independent event. In
`transformation with ligated DNAs, no more than 1 ,ug of
`pBR322 DNA was added to 106 cells per dish because higher
`
`Abbreviations: DHFR, dihydrofolate reductase; Mtx, methotrexate
`(amethopterin); TK, thymidine kinase; CHO, Chinese hamster ovary;
`kb, kilobase.
`
`3567
`
`Merck Ex. 1035, pg 1114
`
`

`

`Proc. Natl. Acad. Sci. USA 77 (1980)
`
`\
`
`\
`
`I
`
`(
`
`I )
`
`K
`T
`
`I.
`
`*_..
`._..._.
`*Ee_.....:
`_I___j
`:s_b
`. . t _
`
`,
`
`1i
`
`I.
`
`si
`
`*.
`
`s_ ^
`; =
`L
`*; _ s_ w * w
`. .,$.
`.b
`' [
`.s v
`,_ _ w w s w t . w
`w W
`|
`
`._
`
`w _* *__._
`.; -
`- -
`
`*
`
`w
`
`3i
`
`Chinese hamster dhfr sequences are present in mouse
`FIG. 1.
`cells. Mouse Ltk- aprt- cells were transformed to Mtx resistance and
`their DNAs were examined for the presence of CHO sequences by
`molecular hybridization to 32P-labeled pdhfr-21 DNA. The hybrid-
`ization profiles of 20 ,ug of HindIII-cleaved DNA from dhfr-amplified
`mouse (lane M) and the dhfr-amplified CHO line A29 (lane H) are
`shown along with the HindIII patterns from four Mtx-resistant cell
`lines derived after transformation and selection at 0.1 ,ug of Mtx per
`ml (lanes A, D, G, and J). Each ofthese cell lines was also grown at Mtx
`levels of 1 ,ug/ml (lanes B, E, H, and K) and 10 pg/ml (lanes C, F, I, and
`L) and scored for amplification of dhfr sequences.
`
`intensity of unamplified dhfr. A series of additional bands were
`observed whose molecular weights were identical to those of
`restriction endonuclease-cleaved hamster dhfr gene. The 17.9-,
`7.9-, and 1.4-kb bands observed in hamster DNA are diagnostic
`for the presence of the hamster dhfr gene and were present in
`all transformants although in disproportionate intensities. In
`similar studies of NIH 3T3 Mtx-resistant transformants, 12 of
`12 contained bands diagnostic of hamster sequences (data not
`shown).
`In initial experiments, we chose the lowest concentration of
`Mtx (0.1-0.2 ,ug/ml) that would decrease survival of the mouse
`cells to <f0-7. Previous studies (18) suggested that the presence
`of a single mutant dhfr gene can render cells resistant to this
`concentration of Mtx. Comparison of the band intensities of the
`hamster dhfr gene fragments of transformed cell DNA with
`A29 suggest that our transformants contain fewer Mtx-resistant
`hamster genes than do donor A29 cells.
`Amplification of the Transferred dhfr Gene. Initial Ltk-
`aprt- transformants were selected for resistance to relatively
`low levels of Mtx (0.1 ,ug/ml). For each transformant, however,
`it was possible to select cells resistant to increased levels of Mtx
`by exposing mass cultures to successively increasing concen-
`trations of this drug. In this manner, we isolated cultures re-
`sistant to 40 ,ug of Mtx per ml, starting from clones that were
`initially resistant to 0.1 ,ug/ml. We next determined if increased
`resistance to Mtx in these transformants was associated with
`amplification of a dhfr gene and, if so, whether the endogenous
`mouse or the newly transferred hamster gene was amplified.
`DNAs from four independent transformants and their highly
`resistant derivatives were examined by blot hybridization. In
`each instance, enhanced resistance to Mtx was accompanied
`by an apparent increase in the copy number of the hamster
`gene. This is most readily seen by comparing the intensities of
`the 1.4-kb band (Fig. 1). In no instance did we detect amplifi-
`cation of the endogenous mouse dhfr gene. Lastly, different
`lines selected at equivalent Mtx concentrations appeared to
`contain different amounts of the dhfr gene.
`
`3568
`
`Genetics: Wigler et al.
`
`concentrations of pBR322 inhibited transformation. Ltk- aprt-
`DNA was added as carrier in these cases to a final DNA con-
`centration of 20,gg per dish.
`Filter Hybridization. DNA from parental and transformed
`cells was isolated, digested with restriction endonucleases,
`electrophoresed in 0.8% agarose gels, transferred to nitrocel-
`lulose filters, and hybridized as described (9). The probes for
`these experiments were 32P-labeled nick translated pBR322 or
`pdhfr-21, a cloned cDNA copy of mouse DHFR mRNA (22)
`(kindly provided by R. J. Kaufman and R. T. Schimke).
`RESULTS
`Transfer of the Mutant Hamster Dihydrofolate Reductase
`Gene to Mouse Cells. High molecular weight cellular DNA
`was prepared from wild-type Mtx-sensitive CHO cells and from
`A29 cells, and the ability of these DNA preparations to transfer
`either the dhfr gene or the tk gene to tk- mouse cells (Ltk-
`aprt-) or NIH 3T3 cells was tested (Table 1). DNAs from both
`mutant A29 and wild-type CHO cells were competent in
`transferring the tk gene to Ltk- aprtr cells. Mtx-resistant Ltk-
`aprtV colonies were observed after treatment of cells with DNA
`from A29. No colonies were observed in cells treated with
`wild-type CHO DNA. Similarly, there were 40-fold more
`Mtx-resistant 3T3 colonies after treatment of cells with A29
`DNA than after treatment with wild-type CHO DNA. These
`data suggest that treatment of Mtx-sensitive cells with A29 DNA
`resulted in the transfer and expression of a mutant dhfr gene,
`thus rendering these cells insensitive to increased levels of
`Mtx.
`In order to test this hypothesis directly, we demonstrated the
`presence of the hamster dhfr gene in DNA from transformants
`by using the filter hybridization method of Southern (23). A
`mouse dhfr cDNA clone (pdhfr-21) (22) that shares homology
`with the hamster dhfr gene was used as probe in these experi-
`ments. DNAs from A29, from transformants, and from dhfr-
`amplified mouse cells were cleaved with HindIII, electro-
`phoresed in 0.8% agarose gels, and transferred to nitrocellulose
`filters. These filters were hybridized with high-specific activity
`32P-labeled nick translated pdhfr-21 and subjected to autora-
`diography. This procedure visualizes restriction fragments of
`genomic DNA homologous to the dhfr probe. Prominent bands
`were observed at 15, 3.5, and 3 kilobases (kb) for dhfr-amplified
`mouse DNA and at 17, 7.9, 3.7, and 1.4 kb for dhfr-amplified
`hamster DNA (Fig. 1). The restriction profiles of these two
`species were sufficiently different to permit us to detect the
`hamster gene in the presence of an endogeneous mouse
`gene.
`Four Ltk- aprt- cell transformants resistant to Mtx were
`examined in this way (Fig. 1). In each transformed cell line, we
`observed the expected profile of bands resulting from cleavage
`of the endogenous mouse dhfr gene although at the decreased
`
`DNA source
`(CHO cells)
`A29*
`
`Wild type
`
`Transformation data
`Table 1.
`Mtx-resistant
`colonies, no./total
`no. dishes
`56/5t
`161/5*
`0/5t
`4/5$
`Twenty micrograms of DNA was used to transform 106 cells per
`dish. Mtx concentration was 0.2 gg/ml.
`* CHO Pro mtxRIII (18).
`t Ltk- aprt- cells were used as recipients.
`t NIH 3T3 cells were used as recipients.
`
`tk+ colonies,
`no./total no. dishes
`25/5t
`
`30/5t
`
`Merck Ex. 1035, pg 1115
`
`

`

`Genetics: Wigler et al.
`A\
`
`('
`
`D)
`
`A
`
`Proc. Natl. Acad. Sci. USA 77 (1980)
`D)
`G
`C
`13
`
`V
`
`F
`
`3569
`
`m04
`
`_ m
`
`~
`
`__
`
`i
`
`4
`
`I4:I
`
`FIG. 2.
`Presence and amplification of pBR322 sequences in cells
`transformed with A29-pBR322 ligates. Cells were transformed with
`the ligation product of Sal I-cleaved A29 and pBR322 DNAs.
`Transformants were selected initially for resistance to 0.1 ,gg of Mtx
`per ml. After cloning, cultures were exposed to increasing concen-
`trations of Mtx, and DNA was extracted, cleaved with Xba I, and
`analyzed for the presence of pBR322 sequences by filter hybridization.
`Lanes A-D, 10 Ag of DNA from the SS-6 line grown in 0.1, 2, 10, or 40
`;g of Mtx per ml, respectively. Lane E, 50 pg of Pst I-cleaved pBR322
`DNA.
`
`The dhfr Gene as a Generalized Transformation Vector.
`Selectable genes can be used as vectors for the introduction of
`other genetic elements into cultured cells. In previous studies,
`we have demonstrated that cells transformed with the tk gene
`are likely to incorporate other unlinked genes (9). The gener-
`ality of this approach was tested for the selectable marker, the
`mutant dhfr gene. Total cellular DNA (20 Mg) from A29 was
`mixed with 1 jig of HindIll-linearized pBR322 DNA. Recipient
`cells (Ltk- aprt-) were exposed to this DNA mixture and, after
`2 weeks, Mtx-resistant colonies were picked. Genomic DNA
`from transformants was isolated, cleaved with HindIll, and
`analyzed for the presence of pBR322 sequences. Two inde-
`pendent transformants were examined in this way, and multiple
`copies of pBR322 sequences were present in both cases (data
`not shown).
`An alternate approach to generalized transformation involves
`ligation of a nonselectable DNA sequence to a selectable gene.
`Because the mutant dhfr gene is a dominant-acting gene con-
`ferring drug resistance, it can be used as a vector. Furthermore,
`it may be possible to amplify any genetic element ligated to this
`vector by selecting cells resistant to increased levels of Mtx. To
`explore this possibility, restriction endonucleases that do not
`destroy the dhfr gene of A29 were identified by transformation
`assay. One such restriction endonuclease, Sal I, does not destroy
`the transformation potential of A29 DNA. Sal I-cleaved A29
`DNA was therefore ligated to Sal I-linearized pBR322. This
`ligation product was subsequently used in transformation ex-
`periments with Ltk- aprt- cells. Mtx-resistant colonies were
`
`As in Fig. 2. Lanes A, B, and C, 10,ug of DNA from cell
`FIG. 3.
`line SS-1 grown in 0.1, 2, and 40 Mg of Mtx per ml, respectively. Lanes
`D-F, 0lg of DNA from the clone HH-1 grown in 0.1, 2, and 40gg of
`Mtx per ml. Lane G, 50 pg of Pst I-cleaved pBR322 DNA.
`picked and grown into mass culture in the presence of 0.1 ig
`of Mtx per ml. Mass cultures were subsequently exposed to in-
`creasing concentrations of Mtx.
`DNAs were obtained from mass cultures resistant to 0.1, 2,
`10, and 40kg of Mtx per ml and the copy number of pBR322
`and dhfr sequences was determined by blot hybridization. Six
`independent transformed lines were examined in this fashion.
`Five of these lines exhibited multiple bands homologous to
`pBR322 sequences. In four of these transformed clones, at least
`one of the pBR322-specific bands increased in intensity upon
`amplification of dhfr (Figs. 2 and 3). All pBR322 bands present
`in transformant SS-6 at 0.1 Mg/ml continued to increase in in-
`tensity as cells were selected first at 2 ,g/ml and then at 40
`Mig/ml (Fig. 2). We estimate that there was at least a 50-fold
`increase in copy number for pBR322 sequences in this cell line.
`In SS-I (Fig. 3, lanes A, B, and C), two pBR322-specific bands
`were observed in DNA from cells resistant to 0.1 Mug of Mtx per
`ml. These bands increased severalfold in intensity in cells re-
`sistant to 2 ,g/ml. No further increase in intensity was observed,
`however, in cells selected for resistance to 40 ,g/ml. In a third
`cell line, HH-1 (Fig. 3, lanes D, E, and F), two pBR322-specific
`bands increased in intensity upon amplification, whereas others
`remained constant or decreased in intensity. Thus, the pattern
`of amplification of pBR322 sequences we observed in these cells
`was quite varied. Nevertheless, the mutant dhfr gene can be
`used to introduce and subsequently amplify unselected DNA
`sequences in cultured animal cells.
`DISCUSSION
`The potential usefulness of DNA-mediated transformation in
`the study of eukaryotic gene expression depends to a large ex-
`tent on its generality. Cellular genes coding for selectable bio-
`chemical functions have previously been introduced into mu-
`tant cultured cells (10-13). In the present study, we have
`
`Merck Ex. 1035, pg 1116
`
`

`

`3570
`
`Genetics: Wigler et al.
`transferred a dominant-acting Mtx-resistant dhfr gene to
`wild-type cultured cells. In initial experiments, DNA from A29
`cells, a Mtx-resistant CHO derivative synthesizing a mutant
`dhfr, was added to cultures of mouse Ltk- aprt- cells or NIH
`3T3 cells. Mtx-resistant colonies appeared at a frequency of
`about 10 colonies per 5 X 105 cells per 20 ,gg of cellular DNA.
`Upon transformation, fewer colonies were observed with NIH
`3T3 and none with Ltk- cells when DNA obtained from
`wild-type Mtx-sensitive cells was used, although this DNA was
`a competent donor of the tk gene. The Mtx-resistant NIH 3T3
`colonies obtained by using wild-type DNA as donor were not
`significantly above the spontaneous level of resistance (data not
`shown), and these colonies were not studied further. Definitive
`evidence that we effected transfer of a mutant hamster dhfr
`gene was the presence of the hamster gene in mouse transfor-
`mants in blot hybridization experiments. In all transformants
`examined, we observe two sets of restriction fragments ho-
`mologous to a mouse dhfr cDNA clone: a series of bands char-
`acteristic of the endogenous mouse gene and a second series
`characteristic of the donor hamster gene.
`The number of copies of dhfr we observed in our initial
`transformants is low. This observation is consistent with previous
`studies suggesting that a single mutant dhfr gene is capable of
`rendering cells Mtx-resistant under our selective criterion (0.1
`,qg of Mtx per ml) (18). Exposure of these initial Mtx-resistant
`transformants to stepwise increases in drug concentration results
`in the selection of cells with enhanced Mtx resistance. Blot
`analysis indicates that these cells have increased amounts of the
`newly transferred mutant hamster dhfr gene. In no transfor-
`mants have we observed amplification of the endogenous mouse
`gene in response to selective pressure. It is likely that a single
`mutant gene affords significantly greater resistance to a given
`concentration of Mtx than does a single wild-type gene. If the
`frequency of amplification is low, we are merely selecting re-
`sistant variants arising from the minimal number of amplifi-
`cation events. It is also possible that newly transferred genes
`undergo amplification more readily than do endogeneous
`genes.
`We have explored the use of the mutant dhfr gene as a vector
`for the introduction and amplification of nonselectable genetic
`elements into cultured cells. Genomic DNA from A29 cells was
`cleaved with restriction enzymes and ligated to restriction en-
`donuclease-cleaved pBR322 sequences prior to transformation.
`Most resulting transformants contained multiple pBR322 se-
`quences. In many cases, amplification of dhfr genes resulted
`in the concomitant amplification of pBR322. The patterns of
`amplification differed among cell lines. In one transformant,
`all pBR322 sequences amplified with increasing Mtx concen-
`trations. In other transformants, only a subset of the sequences
`amplified. In yet other lines, sequences appeared to be lost or
`rearranged. In some lines, amplification proceeded apace with
`increasing Mtx concentrations up to 40 gg/ml whereas, in
`others, amplification ceased at 2 ,ug/ml. It appears that the
`mechanisms of amplification may be quite varied. Whatever
`mechanisms are responsible for these complex events, it is clear
`that the dhfr amplification unit extends beyond the limits of
`the dhfr gene itself and this can be exploited to control the
`dosage of any gene introduced into cultured cells.
`
`Proc. Natl. Acad. Sci. USA 77 (1980)
`
`2.
`
`3.
`
`4.
`
`5.
`6.
`
`7.
`
`8.
`
`9.
`
`Although we have succeeded in transferring Mtx resistance
`to Ltk- aprt- cells and mouse NIH 3T3 cells, we have not as yet
`had success with various other cell lines. In these instances either
`the cells were poor recipients for DNA-mediated transforma-
`tion or they were already substantially resistant to Mtx. We
`expect that cloning a Mtx-resistant dhfr gene will overcome
`these difficulties and extend the use of this gene as amplifiable
`vector for wild-type cells.
`We thank Drs. Sol Spiegelman and James Watson for their generous
`support and C. Fraser and C. Lama for their excellent technical assis-
`tance. This work was supported by grants from the National Institutes
`of Health to R.A. and S.S. (CA-16346, CA-23767, CA-17477) and a
`Cancer Center Grant to James Watson (CA-13106). S.S. was the re-
`cipient of a Research Career Development Award from the National
`Institutes of Health (CA-00492).
`McCutchan, J. H. & Pagano, J. S. (1968) J. Natl. Cancer Inst. 41,
`1.
`351-356.
`Graham, F. L. & van der Eb., A. J. (1973) Virology 52, 456-
`467.
`Graham, F. L., Abrahams, P. J., Mulder, C., Meijneker, H. L.,
`Warnaar, S. O., deVries, G. A. J., Fiers, W. & van der Eb., A. J.
`(1974) Cold Spring Harbor Symp. Quant. Biol. 39, 637-650.
`Bacchetti, S. & Graham, F. L. (1977) Proc. Natl. Acad. Sci. USA
`74, 1590-1594.
`Maitland, N. J. & McDougal, J. K. (1977) Cell 11, 233-241.
`Wigler, M. H., Silverstein, S., Lee, L. S., Pellicer, A., Cheng, Y.,
`& Axel, R. (1977) Cell 11, 223-232.
`Colbere-Garapin, F., Chousterman, S. Horodniceaunu, F.,
`Kourilsky, P. & Garapin, A.-C. (1979) Proc. Natl. Acad. Sct. USA
`76,3755-3759.
`Mantei, N., Boll, W. & Weissman, C. (1979) Nature (London)
`281,40-46.
`Wigler, M., Sweet, R., Sim, G. K., Wold, B., Pellicer, A., Lacy,
`E., Maniatis, T., Silverstein, S. & Axel, R. (1979) Cell 16, 777-
`785.
`Wigler, M., Pellicer, A., Silverstein, S. & Axel, R. (1979) Cell 14,
`725-731.
`Wigler, M., Pellicer, A., Silverstein, S., Axel, R., Urlaub, G., &
`Chasin, L. (1979) Proc. Natl. Acad. Sci. USA 76, 1373-1376.
`Willecke, K., Klomfass, M., Mirau, R. & Dohmer, J. (1979) Mol.
`Gen. Genet. 170, 179-185.
`Graf, L. H., Jr., Urlaub, G. & Chasin, L. (1979) Somat. Cell
`Genet., 5, 1031-1044.
`Fischer, G. A. (1962) Biochem. Pharmacol. 11, 1233-1234.
`Sirotnak, F. M., Kirita, S. & Hutchinson, D. J. (1968) Cancer Res.
`28,75-80.
`Biedler, J. L., Albrecht, A. M., Hutchinson, D. S. & Spengler, B.
`A. (1972) Cancer Res. 32, 153-161.
`Chang, S. E. & Littlefield, J. W. (1976) Cell 7,391-396.
`Flintoff, W. F., Davidson, S. V. & Siminovitch, L. (1976) Somat.
`Cell Genet. 2, 245-261.
`Schimke, R. T., Kaufman, R. J., Alt, F. W. & Kellems, R. F. (1978)
`Science 202, 1051-1055.
`Shih, C., Shilo, B.-Z., Goldfarb, M. P., Dannenberg, A. &
`Weinberg, R. A. (1979) Proc. Natl. Acad. Sci. USA 76,5714-
`5718.
`Bolivar, F., Rodriguez, R. L., Greene, P. J., Betlach, M. C.,
`Heyneker, H. L., Boyer, H. W., Crosa, J. H. & Falcow, S. (1977)
`Gene 2, 95-113.
`Chang, A. C. Y., Nunberg, J. M., Kaufman, R. J., Erlich, H. A.,
`Schimke, R. T. & Cohen, S. H. (1978) Nature (London) 275,
`617-624.
`Southern, E. M. (1975) J. Mol. Biol. 98,503-517.
`
`10.
`
`11.
`
`12.
`
`13.
`
`14.
`15.
`
`16.
`
`17.
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`
`23.
`
`Merck Ex. 1035, pg 1117
`
`

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