CA1205026A - Expression of polypeptides in yeast - Google Patents

Expression of polypeptides in yeast

Info

Publication number
CA1205026A
CA1205026A CA000396832A CA396832A CA1205026A CA 1205026 A CA1205026 A CA 1205026A CA 000396832 A CA000396832 A CA 000396832A CA 396832 A CA396832 A CA 396832A CA 1205026 A CA1205026 A CA 1205026A
Authority
CA
Canada
Prior art keywords
yeast
dna
expression vector
promoter
strain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
CA000396832A
Other languages
French (fr)
Inventor
Ronald A. Hitzeman
Benjamin D. Hall
Franklin E. Hagie, Iv
Gustav Ammerer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Washington
Genentech Inc
Original Assignee
University of Washington
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22895764&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA1205026(A) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by University of Washington, Genentech Inc filed Critical University of Washington
Application granted granted Critical
Publication of CA1205026A publication Critical patent/CA1205026A/en
Expired legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/57581Thymosin; Related peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/62Insulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/655Somatostatins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/8215Microorganisms
    • Y10S435/911Microorganisms using fungi
    • Y10S435/94Saccharomyces
    • Y10S435/942Saccharomyces cerevisiae

Abstract

ABSTRACT

EXPRESSION OF POLYPEPTIDES IN YEAST

In order to make a yeast strain capable of expressing a desired polypeptide (e.g. leukocyte interferon D) which it does not normally make or use, it is transformed with an expression vector. This is obtained by producing a transfer vector which has respective origins of replication and phenotypic selection genes derived from both bacteria and yeast, and inserting into it DNA fragments: (i) coding fox the desired polypeptide; and (ii) comprising a yeast-derived promoter. The insertion is effected so that 'start' and 'stop' signals are provided, and these and the promoter can control expression of the polypeptide, and so that the replication origins and phenotypic selection genes of the transfer vector are still functional. Thus the expression vector and its precursors can replicate, and their presence can be recognised, both in yeast and bacteria. Bacteria are used for amplification. The yeast-derived phenotypic selection gene may complement a mutation carried by the strain to be transformed.

Description

EXPRESSION OF POLYPEPTIDES IN YEAST

FIELD OF THE INVENTION

This invention relates to the production, via recombinant DNA
technology, of useful polypeptides in Saccharomyces cerevisiae (yeast), and to the means and methods of such productionO

BACKGROUND OF THE INVENTION .

The publications and other materials referred to herein to illuminate the background of the invention and, in particular cases, to provide additional detail respecting its practice are, for convenience, numerically referenced hnd grouped in the appended bibliography.

Recombinant DNA Technology With the advent of recombinant DNA technology~ the controlled microbial production of an enormous variety of useful polypeptides has become possible. Already in hand are bacteria modified by this technology to permit the production of such polypeptide products as %~ ~

somatostatin (1), the component A and B chains of human insulin (1), human proinsulin (2), th~nos-in alpna 1 (3), human growth hormone (4), human (5) and hybrid (6) leukocyte and fibroblast (7) interferons, as well as a number of other products. The continued application of techniques already in hand is expected in the future to permit bacterial production of a host of other useful polypeptide products, including other hormones, enzymes, immunogens useful in the prepardtion of vaccines, immune modulators and antibodies for diagnos-tic and drug-targeting applications.

The workhorse of recombinant DNA technology is the plasmid, a non-chromosomal loop of double-stranded DNA found in bacteria and other microbes, oftentimes in multiple copies per cell. Included in the information encoded in the plasmid DNA is that required to reproduce the plasmid in daughter cells (i.e., an "origin of replication") and, ordinarily one or more selection characteristics such as, in the case of bacteria, resistance to antibiotics, which permit clones of the hos-t cell containing the plasmid of interest to be recosni~ed and pre-ferentially grown under selective conditions. The utility of plasmids lies in the fact that they can be specifically cleaved by one or another restriction endonuclease or "restriction enzyme", each of which recognizes a different site on the plasrnid DNA. Thereafter heterologous genes or gene fragments may be inserted into the plasmid by endwise joining at the cleavage site or at reconstructed ends adjacent to the cleavage site.
DNA recombination is performed outside the cell, but the resulting "recombinant" plasmid can be introduced into it by a process known as transformation and large quantities of the heterologus gene-containing recombinant plasmid are then obtained by growing the transformant.
Moreover, where the gene is properly inserted with reference to portions of the plasmid which govern the transcription and translation of the encoded DNA in-formation~ the resulting expression vehicle can be used to actually produce the polypeptide sequence for which -the inserted gene codes, a process referred to as expression.

Expression is initiated in a region kno~n as thé promoter which is recognized by and bound by RNA polymer~se. The polymerase travels along the DNAg transcribing the in-formation contained in the coding strand from its ~' to 3' end into messenger RNA which is in turn translated into a polypeptide having ~he amino acid sequence for which the DNA codes. Each amino acid is er.coded by a nucleotide triplet or "codon" within what may for present purposes be referred to as the "structural gene", i.e., that part which encodes the amino acid sequence of the expressed product.
After binding to the promoter, the RNA polymerase, transcribes a 5' leader region of messenger. RNA, then a translation initiation or "start signal" (ordinarily ATG, which in the resulting messenger RNA becomes AUG), then the nucleotide codons within -the structural gene itsel-f.
So-called stop codons are transcribed at the end of the structural gene whereafter the polymerase may form an additional sequence of messenger RNA which, because of the presence of the stop signal, will remain untranslated by the ribosomes. Ribosomes bind to the binding site provided on the messenger RNA, and themselves produce the encoded polypeptideg beginning at the translation start signal and ending at -the previously mentioned stop signalO The resulting ~roduct may be obtained by lysing the host cell and recovering the product by appropriate purification from other microbial protein or, in particular instances9 possibly by purification from the fermentation medium into which the product has been secreted.
Plasmids employed in genetic manipulations involved in the construction of a vehicle suita~le for the expression of a useful polypeptide product are referred to as DNA trans-fer vectors, Thusg emplo~ying restriction enzymes and associated technology, gene fragments are ordered within the plasmid in in vitro manipulationsg then amplified in vivo in the transforrnant microbes into which the resulting, recombinant plasmid has been 'transferred'. A "DNA expression vector"
comprises not only a structural gene intended for expression but also a promoter and associated controls for effec-ting expression from the structural gene. Both transfer and expression vectors include origins of 360~G/3 replicatjon Transfer vectors must and expression vectors may also include one or more genes for phenotypic selection of transformant colonies.

Thus far, the useful products of e~pression from recomblnant nenes have fallen into two categories. In the first, a polypeptide having the amino acid sequence o-f a desired end product is expressed directly, as in the case of human growt,l hormone and the interferons referred to above.
In the second, the product of expression is a fusion protein which includes not only the an~ino acid sequence of the desired end product but also one or more additional lengths of superfluous pro-tein so arranged as to permit subsequent and speci~ic cleavage away of the superfluous protein and so as to yield the desired end product. Thus, cyanogen bromide cleavage at methionine residues has yielded somatostatin9 thymosin alpha 1 and the component A and B chains of human insulin from fusion proteins; e~zymatic cleavage at defined residues has yielded beta endorphin (8).

A "biocompetent polypeptide", as that term is used herein, refers to a product exhibiting bioactivity akin to tha-t of a polypeptide innately produced within a living organism for a physiological purpose, as well as to intermediates which can be processed into such polypeptides, as by cleavage away of superfluous protein, folding, combination (as in the case of the A and B chains of human insulin), etc.

Sacch _omyces cerevisiae The cells of Saccharomyces cerevisiae, or yeast, are, like those of mammalian organisms, eukaryotic in nature as distinguished from the prokaryotic nature of bacteria. Wi-th regard to mechanisms for the expression of genetic information, eukaryotes are distinguished from bacteria by:

(1) chromosomes which are organized in 140 base pair units, each containing two molecules each of histones ~12A~ H2~, H3, and H4.
(2) Transcription of the protein-encoding gene by the alpha-amanitin sensitive RNA polymerase II.
(3) Post transcriptional addition of Gppp and po1ya~en~lic acid to -the 5' and 3' termini of mRNA molecules.
(4) Transport of newly completed mRNA from the nuclei ~nere they are transcribed to the cytoplasm where they are translated.
(S) Some but not all eukaryotic genes contain intervening sequences (introns) which make them non-colinear with the corresponding mature mRNA
molecule. The initial transcription products of these genes contain -the intron sequence which is spliced out subsequently in the formation of a finished mRNA ~olecule.

The nucleotide sequences of all eukaryotic cells are transcribed, processed, and then translated in the context described above. There are reasons to believe that expression of eukaryotic genes may proceed with greater efficiency in yeast than in E. coli because yeast is a eukaryote cell.
A number of workers have previously expressed, or attempted to express, foreign genes in yeast trans-Formants. Thus, attempted expression from a fragment comprising both a promoter and structural gene for rabbit globin is reported (9) to have yielded partial mRNA
transcripts, seemingly unaccompanied either by translation in-to protein or maturation (intron elimination) of the message. A gene coding for Drosophila GAR tranformylas (yeast ADE8), an enzyme in the adenine synthesis pathway, is reported to have been expressed under the control of its own promoter (10). A number of yeast proteins have hitherto been expressed in yeast via recombinant plasmids (see, eg., 12). In the .
experiments~ as in the Ade-8 case earlier discussed, express;on occurred under the selective pressure of genetic complementation. Thus, each expression product was required for grow-th of the host s-trains employed mutants whose chromosomal DNA was defec-tive in the structura1 gene(s) from which expression occurred.

The availability of means for the production in yeast of proteins of choice could provide significant advantages relative to the use o~
bacteria for the production of polypeptides encoded by recombinant DNA.
Yeast has been employed in 'large scale fermentations for centuries, as compared to tne relatively recent advent of large scale E. coli fermentation. Presently, yeast can be grown to higher densities than bacteria, and is readily adaptable to continuous fermentation processing. Many critical functions of the organism, e.g., oxidative phosphorylation, are located within organelles, and hence not exposed to the possible deleterio~s effects of the organism's overproduction of foreign proteins. As a eukaryotic organism, yeast may prove capable of glycosylating expression products ~here important to enhanced bioactivity. Again9 it is possible that as eukaryotic organisms, yeast cells will exhibit the same codon preferences as higher organisms, tending toward more efficient production of expression products from mammalian genes or from complementary DNA (cDNA) obtained by reverse transcription from, e.g., mammalian messenger RNA. Until the present invention9 however, at~empts to produce biocompetent expression products o-ther than those required for cellular growth have proven largely unsuccessful.

BRIEF SUMMARY OF THE INVENTION

The present invention provides DNA expression vectors capable~ in transformant strains of yeast, of expressing biologica'l'ly competent (preferably pharmacologically active) polypeptides under the contro'l of genetically distinct yeast promoters9 the polypeptides being ordinarily exogenous to yeast and other than those required for growth of the transformant. The invention also provides DNA transfer vectors for the transformation of yeast strains with genes encoding biocompetent polypeptides, as well as novel yeast organisms and cultures -thereof incorporating such vectors and methods for the formation of the same.
The structural genes incorporated in the expression vec-tors and transformant organisms of the invention are under the control of genetically distinct yeast promoters7 i.e., promoters different from those evolutionarily associated with the subject structural genes.

The manner in which these and other objects ~nd advantages of the invention are obtained wlll become apparent from the detailed description which follows, and from the accompanying drawings in which:

Figure 1 schematically illustra-tes the construction of a DNA transfer vector having a single_Eco RI restriction site for the subsequent insertion of a yeast promoter and comprising both bacterial and yeas-t orisins of replication and selection phenotypes;

Figure 2 schematically illustrates the construction of alcohol dehydrogenase promoter fragments for insertion into the -transfer vector of Figure 1;

Figure 3 illustrates the coding strand sequence and end points (904, 9069 etc.) of a series of yeast promoter fragments made by digestion with the exonuclease Bal 31; and attachment of EcoRI molecular recombinational linkers.

Figure 4 schematically illustrates the insertion of yeast promoter fragments into the transfer vector of Figure 1 and subsequent insertion (in two orientations~ of a str-uctural gene encoding human leukocyte interferon D.

In the Figures and throughout, the letters A, T, C and G respectively connote the nucleotides containing the bases adenine, thymine, cytosine and yuanine. Only the coding strands of plasmids and gene fragments are depicted. Though obviously not to scale, the representa-tions of plasmids depict the relative position of restriction enzyme cleavaye sites ("Eco RI", "HindIII" etc.) and o-ther functions such as tetracycline resistance . . .
("Tcr") and ampicillin resistance ("Apr").

~2~ 2~i DESCRIPTION OF PREFERKED EMBODI,`~lENTS

Preferred embodiments of the invention are obtained by bringing an exogenous gene under the control of a yeast promoter carried by a plasmid suitable for the transformation of yeast. Essentially any yeast strain suited for -the selection of transformants may be employecl. In order to achieve direct expression of the desired end product or an intermediate therefor~ rather than a fusion comprising portions o-f the yeast protein whose expression is con-Erolled by the promo-ter in wild-type strains, the parental plasmid is resected toward the promoter in the direction opposite that of transcription, so as to excise the ATG triplet which initiates translation of mRNA encoding the yeast protein referred to.

An ordinarily exogenous gene, with its associated start signal, may then be inserted at the endpoint of the resection, and thus positioned for direct expression under the control of the yeast promoter. This and other aspects of the invention are illustrated in the description of preferred embodiments which follow.

_ T~IODS_ Materials: All DNA restriction and metabolism enzymes were purchased from New England Biolabs except for exonuclease Bal 31 and bacterial alkaline phosphatase, which were obtained from Be-thesda Research Laboratories. DNA restriction enzyme and metabolic enzymes were used in conditions and bu-ffers described by their respective manufacturers. ATP
and the deoxynucleoside triphosphates dATP, dGTP, dCTP and dTTP were purchased from PL Biochemicals. Eco RI9 Bam ~ ind III and Xho I
linkers were obtained from Collaborative Research, Inc. [~_32p] was obtained from New England Nuclear Corp.

D~A Preparation and Transformati~n: Purification of covalently closed _ circular plasmid DNAs from E, coli (13) and yeast (1~) plus the transformation of E. coli (15) was as previously described.
Transformation of yeast was as described by Hsiao and Carbon (16) with the exception that 1.2 M Sorbitol was used instead of 1.0 M Sorbitol. E.
Coli miniscreens were as described by (17).

Strains and Media: ~. co11 strain JA 300 (thr 1euB6 thi thyA
trpC1117 hsdM _sdR strR) (18) was used to select for plasmids containing functional ~ gen. E. coli K~12 strain 294 (ATCC

no. 31446 28 Oct. 78)(19) was used for all othex bacterial transformationO Yeast strains RH218 having the genotype ( a trpI gal2 suc2 ma1 CuPI) (20) and GM-3C-2 (~ leu 2-3, 1eu 2-112, trp 1-1, his 4-519, ~ 1, cyp 3-1) (21 were used for yeast transformations. Yeast strain RH 21~ has been desposited without restriction ~n the Pimerican Type Culture Collection, ATCC No. 44076. on 8 Dec. 80.
M9 (minimal medium) with 0.25 percent casamino acids (CAA) and LB
(rich medium) were as described by Miller (22) with the addition of 20 ~g/ml ampicillin (Sigma) after media is autoclaved and cooled. Yeast were grown on the following media: YEPD contained 1 percent yeast extract, 2 percent peptone and 2 percent glucose ~3 percent Difco agar YNB+CAA contained 6.7 grams of yeast nitrogen base (without amino acids) (YNB) (Difco)9 10 mg of adenine, 10 mg of uracil, 5 grams CAA, 20 grams glucose and ~30 grams agar per liter. The selection of ADH promoter active fragments occurred on YEPGE plates containing 3 percent glycerol and 2 percent ethanol substituted for glucose in the YEPD formula.
Leucine prototrophy was determined on plates containing 6.7 gms YNB, 20 gms glucose, 50 mgs histidine and 50 mgs trytophan and 30 gms Difco agar per L.

~ Identi _cation of ADH Promoter Deletions: pY9T6 was digested with Sau3A then run on a preparative 1 percent agarose gel. The 1600 bp fragment containing the ~DH promoter region ;Yas Cllt from the gel, electroeluted then purified on a diethylamino cellulose (DE52~ Wha-tman) ~608~l9 column before ethanol precipitation. Fragment D~ as resuspended in DNA
Polymerase I (Klenow fragment) buffer supplemented with the ~our deoxyribonucleoside triphosphates in a final concentration of ~0 IIM.
Polymerase I was added and tlle thirty-mirlute room ternperature reaction was termindted by ethanol precipitatioll of the DNA. An equal molar amount of BamHI and HindlII linker was added to the resuspended Sau3A
fragment so th~t each linker was in a 20:1 molar excess to the large DNA
fragment. T4 DNA ligase was added and the 12 hour reaction occured at 12 degrees centigrade. After ethanol precipitation and resuspension in the appropriate bu~fer,-tlle DNA was diges-ted wi-th BamHI, then HindIII.
The now larger promoter-containing fragment was purified away from -the unattached linkers by passage through a 10ml sizing column before ethanol precipitation. This DNA fragment was then ligated in pBR322 previously isolated as missing the HindIII-to-Ba~HI restriction fragment. E. coli strain RR1 was transFormed to ampicillin resistance using part of this ligation mix. After quick screen analysis of a number of recombinant plasmids, pJD221 which had the insert with the HindIII linker added to the end o-F the fragment closest to the ATG of the ADH structural gene was isolated by plasmid preparation.
p~D221 was linearized with HindIII and the resulting fragment than successively treated with exonuclease III and S1 nuclease. The ends of these deleted p'lasmids were then made blunt using the Klenow fragment of DNA Pol~vmerase I (see procedure above). After ethanol precipi-tation the ends o-f the DNA were ligated with XhoI linkers in a 12 hour reaction mixture. After digestion of resulting ligation mix with XhoI, plasmid solution was run in a 0O5 percent preparative agarose gel. DNA bands were cut from the gel~ electroluted, then passed through a DE52 colurnn before ethanol precipitation. Linear plasmid was circularized using T4 DNA Ligase. The resulting ligation mix was used to transform E. coli strain RR1 to ampicillin resistance. All such colonies were pooled together. The resulting single plasmid pool was cut with Xhol and Baml-lI, then run on a preparative 0.7 percent agarose gel. The 1500bp bands containing the ADH promo-ter region were cut froln the ge'l, eléc-troeluted 3~0~G/10 ~hen passed through a DE52 column before ethanol precipitdtion and liyation into the vector pYecycI~x+I. This plasmid had previously been isolated from an agarose gel as having lost the XhoI to BamHI restriction fragment described in the Figure. The resulting ligation WdS used to transform E. coli strairl RRI to am~icillin rusistance Colonies were mixed for preparation of a plasmid pool which was then used to transform yeast strain G~l-3C-2 to leucine prototrophy. Plasmids ~ere then isolate~
from leucine proto~rophs able to grow on glycerol plates. One plasmid, pACE 301, was found to contain a deletion ex~ending toward the ATG of the ADH1 structural gene, ~eaving intact the first five triplets o-F the structural gene and the AC of the ACC of Thr6 (Fig. 2b). This plasmid was digested with XhoI then treated with exonuclease Bal31 for 15 and 30 seconds (two differen-t aliquots). Resulting plasmids were pooled, ethanol precipitated and then treated with DNA Polymerase I (reaction described above) so that all DNA ends were made blunt. EcoRI linkers were then added to the DNA solution and ligation allowed to proceed for 12 hours. After digestion wi-th EcoRI and BamHI, ligation mix was run on a preparative agarose gel. A DNA band about 1500 bp in size was cut from the gel, electroeluted then passed through a sizing column before ethanol precipitation. This DNA was then ligated into the linear pBR322 DNA
previously isolated as missing the EcoRI-to-BamHI restriction Fragment.
Th1s ligation mix was used to transForm E. coli strain 294 to ampicillin resistance. Plasmids isolated from these colonies are referred to as the pGBn plasmid series.

3608G/ll l~iniscreen an~lysis of a numl)cr of different recolnDlnant plasmi~s from the pG~n plasmi~ series indicated that nine particular pldsmi(ls had small Bal 31 generatcd deletions toward the AD~I promoter region through the ATC of the ADH struc-tural gene. All nine plasmids were digested with EcoRI, then end labele~ by incuhation ~lith (c,32P)dATP and nNA
polymerase I (conditions as ~escribed above). After ethanol precipitation, seven plasmids were digested with AluI then electrophoresed on a 20 percent acrylamide - urea sequencing gel.
32p _ labelled plasmid DNAs from pGB904 and pGB906 were cut with BamHI
then run on a preparati~e gel. Labelled fragrnents containing the ADH
promoter region were excised from the gel, electroluted, passed through a DE52 column before ethanol precipitation. These two resuspended fragments (from plasmids pGB904 and pGB906 ) were then subjected to the G~A and T+C sequence specific degradation reactions describe~ by Maxam and Gilbert (procedure 11 and 12 respectively (2~)). These sequencing reaction products were electrophoresed along with labeled fragments from pGB905, pGB914, pGB917, pGB919 and pGB921 on the thin 20 percent acrylamide sequencing gel (described in the sequencing reference).
Autoradiography was as described. This procedure allowed the determination of the extent of deletion of ADH promoter region as this region had previously been sequenced using all four Maxam-Gilbert sequencing reactions (J. Bennetzen, Ph.D Thesis, University of Washington, 1980).

Expression Vector Construction: 10 ~9 oF YRp7 (24-26) ~as digested ~ith _ EcoRI. Resulting sticky DNA ends were made blunt using DNA Polymerase I
~Klenow fragment). Vector and insert were run on 1 percent agarose ~SeaKem) gel, cut from the gel~ electroeluted and 2X extracted w;th equal volumes of chloroform and phenol before ethanol precipitation. The resulting blunt end DNA molecules were then ligated -together in a final volume of 50 ~l for 12 hours at 12C. This ligation mix was then used to transform E. coli strain JA300 to ampicillin resistance and tryptophan prototrophy. Plasmids containing the TRPI gene in both orientations were isolated. pFRW1 had the TRPI gene in the sarne oricntation d5 YRp7 ~hile pFRW2 had the TRPI gene in the opposite orientation.
10 ~9 of pFRW1 and 10 ~9 of YRp7 werc digested witl~ HlndIII then run in separate lanes on a 1 percént agarose gel. The large HindIII fragment fronl the pFR',~1 lane and the small frag,nent fr~m the YRp7 lar,e were eluted from the gel, extracted with phenol and chloroform, ethanol precipitated, then ligated for 12 hours at 15C in a final volume of S0 ~l. This ligate mix was used to transform JA300 to tryptophan prototrophy and ampicillin resistance. Plasmid (pFRL4) containing a single EcoRI site was then purified. _.
The pGBm plasmid series was digeste~ with P,amHI and EcoRI then run on a 1 percent agarose gel. The ~ 1500 bp promoter containing fragment from each lane was cut from the gel, electroeluted, then purified on a 10 ml diethylamino cel'lulose (Whatman) column before ethanol precipitation.
20 ~g of pFRL4 was digested with BamHI and EcoRI then run on a 1 percent agarose gel. The large (- 5kb) fragment was Cllt from the gel, electroeluted, 2X extracted with phenol and chloroform before ethanol precipitation. 3 ~g of this fragment was then separately ligated with each of the promoter containing fragments for 12 hours at 15~C in 50 ~l ligation mix. E. coli K-12 strain 294 was transformed with the ligation , .
mix to ampicillin resistance and plasmids frorrl each of these different transformation mixtures were purified (pFRPn plasmid genes).
10 ~g of pLeIF D (5) was digested with EcoRI then run on a 6 percent acrylamide gel. The 560 bp leukocyte interFeron D gene was cut from the gel, electroeluted and 2X extracted with phenol/chloroform before e-thanol precipitation. This interferon gene was then ligated into the unique EcoRI site in the pFRPn plasmids previously cut with EcoRI and treated with bacterial alkaline phosphatase. These vectors were then used For BglII restriction analysis and yeast transformations.

Interferon Assay: Extracts of yeast were assayed for interferon by comparison with interferon standards by the cytopathic eFfect (CPE) inhibition assay (27). Yeast extracts were prepared as follows: Five ml 0~

cultures :Yere grown in Y~ CAA until reaching A6~o-1-2. Cells ~ere collected by centrifugdtion then resuspended in 600 ~1 of 1.2 M sorbitol, 10 ml~ KH2P0~, pH=6.8 and 1 percent zymolyase 60,000 then incubated at 30C for 30 min. Spheroplasts were pelleted at 3000 xg for 10 min., then resuspended in 150 ~1 of 7 M guanidine hydrochlGr;de plus l,nl~
phenylmetllylsulfonylfouride (P~lSF). Extracts were diluted 1,000 fold in PBS buffer ~20 m~1 NaH2P04, pH=7.4, 150 m~ NaCl, 0.5 percent BSA) immediately before the assay.

RESULTS

Construction of a Vec-to _ or Insertion of a Series _ Pror,loter Fragments and for Insertion of A Gene to be Exp essed To design a plasmid vector for autonomous replica~ion in yeast, i~ is necessary to have both an origin o~ replication and a gene present for selection in yeast. Furthermore, the plasmid must contain a bacterial plasmid origin of replication and a means of selection in bacteria (e.g., an antibiotic resistance gene). With these requirements a plasmid can be constructed and modified ln Yitro using recombinant DNA techniques, amplified in bacteria, preferably E. _oli, and finally transformed into yeast.
Such a vector is shown in Fig. 1 and is designated YRp7 (24 - 26).
It contains a chromosomal origin of replication from yeast (arsl) as well as the TRPl gene which codes for N-(5'-phosphoribosyl)-anthranilate isomerase (23). The TRPl yeast gene can complernent (allow for growth in the absence of tryptophan) trpl mutations in yeast (e.g., RH218a see Methods) and can also complement the _rpC1117 mutation of E. coli (e.g.
JA300) (1~). The plasmid is pBR322 (29) based so it also permits growth and selection in E. coli using antibiotic resistance selectionO
Since it was necessary to clone into this vector BamHI/EcoRI
restriction fragments containillg a yeas-t promoter, it proved convenient to first remove one EcoRI site from the vector. This was done as shown in Fig. 1. The vector YRp7 was cut with EcoRI followed by filling in of the sticky EcoRI ends of both fragments with Klenow DNA polymerase I.
The fragments were then blunt end ligated and the resulting DNA was used to transform E. coli JA300 to Trp and ampicillin resistance (ApR~.
In such a ~Jay plasmid pFRW1 was isolated with both EcoRI sites removed.
One EcoRI site was then restored to the plasmid in order that an EcoRI/BamHI fragment could be later cloned into the vector. This was done by cutting both YRp7 and pFR~1 with HlndIII followe(l by the isolation of the fragments indicated. When the small HindIII frdgll)ent o~

YRp7 w~s put tog~tll~r witl~ the lar~e ~lindlII fragment of pFR~,Il pFRL4 was obtained. It ~as selected for in E. coli JA300 using Trp+ and ApR
phenotypes.

Construction of Yeast Alcoho1 Deh~(lrogen~lse (ADH) Promoter Frayrnents Sînce it is not known ~/hether certain specific se4uences in the leader region preceding structural genes are required for Ri~A polyrmerase lI binding or what DNA is necessary for ribosome recogni-tion (ribosome binding sites) of the mR~A promoter fragments from the ADH gene (ADCI) were obtained as described in Fig. 2.
The first step was to show that the 5'-leader D~A sequence of the ADH
gene could be used to express another structural gene from yeast without its leader sequence ( YC1). Thus a plasmid which can complement a cycl mutation in yeast can be used to isolate the ADH promoter fragment that will result in cycl expression. This promoter fragment could then be used to express other eukaryotic genes (eg. the Leukocyte Interferon D
gene).
As shown in Fig. 2~ pY9T6 containing the ADC1 locus (Bennetzen supra) was cut ~ith Sau3A to isolate the 5'-flanking sequence of the ADH
gene on an approximately 1600 bp fragment. The ATG translation start for the ADH coding sequence is shown with the A at position ~1 and transcription goes from left to righ-t as shown. This fragment was blunt ended using Klenow DNA polymerase I followed by a ligation with a mixture of BamHI and HindIII linkers. After cutting with BamHI and H_ndIII the ~ragments were ligated with the large BamHI/HindIII fragrnent of pBR322.
The ligation products were used to transform E. coli to ApR and the desired pJD221 was isolated from a transformant colony using a standard miniscreen procedure (see Methods). pJD221 was Cllt with tlindIII and then w;th exonuclease III and S1 nuclease to remove base pairs toward but not through the ATG of the ADH structural gene.
This procedure also removes base pairs in the opposite direction (toward the EcoRI site) at approxilnately the same rate. The reaction was designed so as to not remove the ATG o~ ADH since the ATG of CYC1 was not present in the fragment to be e~pres,ed under ADI~ prom~ter control.
Therefore a complclllentation of ~yc1 yeast would rcquire d functional AOH1-CrC1 fusion protein.
The end-deleted products ue,Ye treated with Klenow D~ polymerase I to ensure blunt ends followed by ti~e addition of XhoI linkers by blunt end ligation. After X I cutting a circular plasmid ~as regenerated by ligation. Plasnids containing ~2l-determined, properly s~zed EcoRI ~ to - BamHI restriction fragments were digested ~lith XhoI, then ligated Witil the 'large XhoI - to - BamHI restriction fragment of plasmid p`(ecyc 1 ~x~1. After amplific~tion in E. coli RR1 resulting plasmids were used to transform a cycl c _ leu1 yeast strain to leucine prototrophy on minimal glucose plates. Growing coloni~s were patched onto glycerol/ethanol plates. Yeast able to grol~ on ,uch plates require -the presence of functional cytochrome-c protein. This can only occur on this plasmid if fragments containing ADH promot~r deletions can initiate (in the correct reading frame) translation of the cytochrome c-codiny region. Plasmid pACF301 was isolated from one sr~ch transformant. The junction between ADHl and CYC1 is shown at the bottom of Fig. 2b. Six amino acid codons from the ADH sequence were pres~nt ~ith 3 new amino acid codons due to the Xho~ linker, and the rest rapresented the CYC1 structur'al gene. Thus the ADH promoter fragment is e~pressing a -fusion gene product that produces a phenotypically acti~ CYC1 gene fusion product.
In the construction of a yeast expression plasmid, it is desirable that the ATG codon of the non-yeast gene to be expressed be the one belonging to the same non-yeast gene rather than a vector ATG which would lead to the synthesis of an undesired fusion protein. Therefore, it proved appropriate -to remove n~cleotides through the ATG o-f the ADil promoter fragment by another s ries of deletions and supply a new translation start signal with the gene to be expressed. Since the functionality of upstream DNA sequence (-1 to -1500) during -the expression process is not ~no~n, it was desirable to remove as little sequence as possible upstream ~rom the ATG and to try different fragments lacking both the initially pr~,ent ATG and ~arious amounts of addi-tional DliA sequence.
17/35gO(;

These additional promo~er fragments were isolated as shown in Fig.
2b. pACF301 was CUt with Xhol and Bal31. After blunt-ending, a~dition of EcoRI linker BamHI/EcoRI cutting and siz;ng fragments; the correct size class of fragments were ligated with EcoRI/BamHI-cut pBR322.
Specific 'r'ecloned ADI~ promoter fragments were isolated from plas~ids from various E. coli ApR transformants.
Fig. 3 sho~s the DNA sequences of the transcribed strand of ~ of the resulting, variously sized and numbered promoter fragments. The numbered lines sho~ where the right end of the fragment ends and where the EcoRI
linker sequence begins.
The erds of fra~ments 904 and 906 wPre exactly determined by sequencing. The EcoRI sticky ends of these fragments were labelled with Klenow DI~A polymerase using ~-32P-dATP. A sequencing gel was used'to read from the A's into the linker through the junction. The other 6 fragment ends were approximated to within about 1-2 base pairs ~y labelling as above, cutting with AluI, followed by sizing on ~he same 'denaturing gel.

Construction of Plasmids that Express Biologically Active Leukocyte _ _nterferon D in Yeast In order to optimize for successful expression of LeIF D in yeast9 eight dif~erent promoter fragments (Fig. 3~ wer ligated into the pFRL4 vector as shown in Fig. 4.
The vector was designed to have ADH promoter transcription in the same direction as TRP1 gene transcription (31). Since the LeIF D gene was to be inserted in the EcoRI site and was not known to contain proper 3' termination and processing sequences for yeast recognition9 ~he TRP1 gene flanking sequence was ali~ned $o perform these funct~onsO
The resulting pFRPn series (~here n ~s the promoter fragmen~ number3 was obtained as shown~ The preferred embodiment of these pFRP6 in a transformant strain of E coli ~94,has been deposited 24/F~/81 in the American Type Culture Collection (ATCC no. 31814~. These vectors were cut with E RI, alkaline phosphakase treaked (to avoid premature recircularizdtion) and lig~ted ~itll th~ EcoRI LclF D g~n~ fr~ment. The ATG of this gene imme(lidtely follo~ls the C of the EcoRI linker ~G~ATTCATG) dS sho~n (Fig. 4).
Ampicillin resistant transform~nts of E. coli K-12 strain 294 were screened to find plasmids containing bo-th orientdtions OT~ the LelF D
fragment (pFRSn series--n refers to screening number). Orien-tations ~ere determined by agarose gel electrophoresis using ~II digestion ~Ihich cuts both in the vector and in the LelF ~ gene as sho~n.
Three of the plasrnids demonstra-ted unpredicted restriction patterns.pFRS7 and pFR~35 have an extra BglII fragment at 560 bp. This results from having two fragTilents of LeIF D in line with ADH
transcription. pFRS16 has no proper orientation fragmen-t but has a 1700 bp fragment which apparently resulted from the ligation of -two vector fragments togetller (two TRP1 containing "tails" together) with one LeIF D
fragment in between two "h2ads" containing ADH prornoter fragments. Thus in this ligation product the interferon gene is in the proper orientation for expression by one of the ADH promoter fragments.

Evidence for Leukocyte Interferon D Expression in Yeast First the E. coli 294 strains containing the various plasmids (pFRSn) were grown and extracts prepared (see Methods). No interferon activity was observed in tlle extracts using the cytopathic efFect inhibition assay (see ilethods). Ho~eYer when plasmids were purified and used for yeast transformations by selection of TRP~ phenotype using yeast RH218 (trpl mutation) all plasmids WitTl orientation I produced interferon activity in yeast while no plasmids ~ith orientation Il produced interferon.
Table 1 shows the results of interferon assays which measure antiYiral activity effects on VSV virus challenge of T~DBK tissue culture cells (see l~lethods). Seven of the promoter fragments definitely express the LeIF D gene wilen the gene is in the proper orientation (1). This is demonstrated by comparing units/(ml of extract) for the orientation I
plasmids with the orientation II plasmids. All orientation Il plasmids expresse(J <1~00 units/(ml of extract), a value 1 to ~ percent of the values for orientation I plasmids ~actually back~round values ~re probably much lower than this since the 1900 value is a function of the assay procedure).

~ C~ ~ O O O O O O O O ~ r--a) r~ O O O O O O O O O r--r~ r- C ~= O O O O O O O O O O
r-- r (~ ~~ ~ O O C~J O O ~ O ~L) ,_ U L~ l L~ r--I 1~ ~ a a ... ~ ~ ~

¦ C ~ J ~ _ L ~t ~t ~t ~ ~ O ~ ~ O ~ 1-- ~

(_) r-- S ~r Cl~ O t~ D ~1 0C) ~-- C~ ~) Lr) v--~ ~ a ~ r~ 4_ r~ ~ a~
O 3 a r--I 1~) 1~) ~_ r~ C~ r-1 ~--I C~J C~J t~> ~ ~t C~l (~) t~ ~ ~lJ ~ Ci. ~ ~r r~1 ~ 4-- ,_ . O ~ L~) ~ ~
n~ O c ~ ~ ~ ~
X O ~( d- ~ r-- O D Lt) NC) a~ O r~ U) ~ C~> ~S
a)r-- O -I J Oo~i ~ Lri C~ t~') t~-) ~t C~ C a.J Cl- ~ ~ r-- ~r- r r-- r . ~r ~ ~ ~ ~ c V E
aJ a.l a) ~ ) r.
::~ r-- V) ~1 ~ I ~ . r-- .a l~ r-- ~ r- V~

r~ ~t 1-~ Li ¢ O ~ l O C~l ~1 ~_ ~) C~ ~ E 3 y r-~ ~ X ~ ~ r- ~~

Vr~ r-- ~ ~ LD 0;~ LD r~l Lr~ C~ O Vl ~S O 3 C C~ U') ~~
o;l~ v o ~ ~ Ln ~ Ln C~ l ~ ~ rc~ ~ 4~ 3 E
4_C ~ X 4_ E~ ~ ~r I ~ r~ E ~r-C~ ~,~ t O O O O O O O O V _ _ = = r- C ~IJ L0 Il) ~) ~
O ~ r~ Ln r~ Ln O ~ V ~r- ~ r C,l ~r- r- X

o ~ r ~r- 1~ C~ V E 3 ,, '~S O C~ ) ~ L dC~l LO r-l r ~ ) ~ O O =5 +-1 ~) L ~1) C O

u) I~ . C~~ ~, o~ c~, (Y: ~ C E - V ~ ~) O ~ ~ r~ ;~
Cl. ~ L ~ ~ _ ~ ~ CL ~ L:L ~-1 r- ~ O C V

V C rU~ ~ O u~ _C-- E ~
E 1~ ~ ~) O D Ln r ~1O O r-l O r-l C\l rU) O ~) r~ ~ a) S ~ I:JI C) v E

5_ ~O c ~~ c r~J ~ aC~ C ~ ~O ~
I ~ ~ C~ V ~ aJ O ~[ r-- d a ~ O L~
~r- ~) ~ ~ w ~ ~ O a:) E O ~s O r O ~ N S~ ~ V L) c ~

Since arsl (chromosolnal ori~in of replication)-cont~inin~ pldsmid, have previously been sho~n to be unstable and lost in a high percentage of the cells even under selective maintenance pressure (2~, 25), the percent of cells containing the plasmid at thc time of extrac~
preparation ~as measurcld. This was done by plating diluted cultures on plates with and without tryptophan. The results of this demonstra-te that the plasmid is some~hat unstable in yeast ~though not in bacteria), but can be maintained by gro~th under selective pressure. These results also are evidence for the presence of the plasmid, since RH218 (trpl) yeast do not gro~ on plates witbou-t tryptophan and since a revertant to TRP+
would plate ~i-th equal efficienc~ on plates wit'n and without tryptophan.
Furtherlnore, the percentages of cells containing plasmid are similar comparing yeast with orientation I and II plasmids. This suggests that the production of interferon in the yeast cell does not result in increased instability of the plasnid due to interferon toxicity to the cell.
The fact that all the promoter fragments express interferon when up to 32 bp are removed upstream from the ATG suggests that the DNA sequence in this region is relatively unimportant in transcription and translation. These results also suggest that precise spacing between the promoter and the ATG may be relatively unimportant for expression in yeast.
In addition, Table 1 shows molecules/cell values which are very much higher than the 10,000 molecules/cell observed for interferon D
expression in E. coli on a high copy plasmid with a strong promoter (trp promoter) ~32). Assessment of this extreme difference (up to 18 fold) in molecules per cell should recognize that the yeast cell volume is probably 2 orders of magnitude higher -than that of E. coli, however, the amount of expression froln only 1-2 copies of the yeast plasmid versus the high copy number of plasmids producing interferon in Eo coli is dramatic.

Colipirison of the Size of Interferon ~ro-luced in Yeast verslis E. coli Since the intcrfcron gene u,es its own ATC-initiation codon ancl since the alcohol dehydrogenase ATG has been removed in the construction, one would expect to find thdt the interferon expressed in yeast is the ~ame size as the interferon in E. coli (32). SDS-polyacrylamide gel electrophoresis was accordingly done on an E. coli extract containing interferon D versus a yeast extract containing interferon D. Af-ter running the gel, two lanes containing yeast ex-tract versus E. coli extract were simultaneously sliced. The slices were put into assay dilution buffer and left at 4~C for 3 days. Interferon assays were then perfo~Amed to compare sizes of the peptides. Both appear to be about 20,000 daltons, the size expected for interferon D. However, there does appear to be a slight difference in the molecular weigh-ts, with yeast interferon D being about 7 percent larger, possibly owing to glycosylation. Despite the size difference, the products of yeast expression exhibited interferon activity (Table 1).

The preceding data cledrl~ demons-trdtes that a yedst 5'-flanking D~A
sequence, without the translation star~ signal of tlle struc~ur~ ne, can efficiently promote the expression of an inserted mamr,lalian or other structural gene for a biocompetent polypeptide, and do so without the aid of selective pressure for the product of expression (i.e., the e~pression product is not re~uired for cell growth).
The availability of yeast promoter-containing plasmids (pFRPn series) having both yeast and bacterial phenotypical genes and origins of replication, and a site downstream from the promoter convenient for the insertion of translati~on start- and stop-bearing structural genes permits the creation of D,~A expression vectors for a wide variety of polypeptides. Thus, into such a site may be inserted, for example, structural genes for both normal (5) and hybrid (6) human leukocyte interferons, fibroblast interferon (7), somatostatin or the A or B chains of human insulin (1), human proinsulin (2), thymosin alpha 1 (3), human growth hormone ~4) and, indeed, virtually any other biocompetent polypeptide.
Following expression, product may be extracted and purified as in the case of bacterial expression, mutatis mutandis.
It will be appreciated that the invention is not limited in its appl;cat;on to the particular expression vector exemplified above. For example, use of the so-called t\~o micron origin of replication would provide additional stability, rnaking unnecessary resort to selective pressure for maintenance o-f the plasmid in the yeast cell, particularly if the host strain is [CIR-~, i.e., contains normal two micron plasmid (33). Such an expression vector would be stable in yeast in the rich medium ordinarily best for large scale fermentations. At the same time, use of the two micron origin of replication could significantly increase plasmid copy number in each cell.
Stability of the expression vector in yeast may also be enhanced by inclusion within the plasmid of a yeast centromere (34), an element involved in maintenance of the yeast chromosome. The resulting plasmid will behaYe as a minichromosome, such that selective pressure will not be .

required during growth or maintenance of thc pl~smid. As many as 17 different yeast centrolneres h~ve been identified ~o the present date.
Transcription ter,ninators other than that preserlt on the TRPI gene may be employed, e.g., other 3'-flanking sequences from yeast such as the 3'-flan~ing sequence contained on a Hlnc II-Ba~HL fragment of -the ADH 1 gene.
Optimiza-tion may also result from alteration of the sequence be-t~leen the yeast promoter fragment and the inserted gene fragment. Thus, an A
(adenine base) is found at posi-tion -3 (the third base before the translation start sigral) of all twenty different mRNA-coding yeast genes heretofore sequenced. A variety of means (eg., use o-F linkers) for includins such an element in the plasmids of the invention will appear to those skilled in the art.
Of course, promoters other than the ADH promoter exemplified above may be employed in variants oF the invention. For example, the promoter of the yeast 3-phosphoglycerate kinase gene may be employed, doubtless increasing expression levels significantly over those observed for the ADH system. Again, one or more of the promoters for yeast glyceraldehyde-3-phosphate dehydrogenase may be employed. This system is nonfunctional in the absence of glucose, but induced 200-fold in its presence, and could accordingly be employed for fine control of expression.
From the foregoing, it will be apparent that the invention provides new means for the expression oF valuable polypeptides. In particu'lar instances, efficiency of expression relative to that in recombinant bacteria may result from the different codon usage patterns as between yeast and bacteria, such that eukaryotic genes may be better expressed in yeast. The yeast expression systems of the invention may also provide advantage in the glycosylation of biocompetent polypeptides, an ability bacteria lack. The glycosylation system of yeast is very similar to that of higher eukaryotes, and glycosylation may prove to have profound effects on the functions of proteins.

As ~.Yill be dpparent to those skilled in t~e art in the light of the foregoing discussion, the inverltion is not to ~e limited to the preferred embodiments thereof ex~mplified above, but rather only to the la,/ful scope uf the appended claims.

* * *

The contribu-tions of certain of the coinventors hereof arose in the course of work funded in part by the United States Department of Health and Human Services.

BIBL~GRAPHY

1. United States Patent application Ser. ~o. 90,980 filed Nov. 5, 1979 by Ys~aisi9nee G~nentech, Inc. (Published as EPA Publication ~.
001930).
2. ~nited States patent application Ser. No. 222,044, Atty. Dkt. No.
1980/109, filed Jan. 2, 1981 by D. Goeddel et al.,assignee Grenentech, Inc. (Published as Sures et al, S~ience 208,57 (1980).).
3. United States Patent application Ser. NoO 125,685, filed Feb. 28, 1980, by Grea e _ ., asignee Genentech, Inc. (Published as Wetzel et al., Biochemistry 19, 6096 (1980).
4. United States patent application Ser. No. 55126, filed July 5, 1979 by D. Goeddel et al.,assignee Genentech, Inc. (Published as EP~ Public~
ation No. 0022242).
5. United States patent application Ser. No. 205,578, filed Nov. 10, 1980 by D. Coeddel et al.,agsi~nees Genentech, Inc., and Hoffin~nn-La Roche, Inc. (Published as Goeddel et al., Nature 287,411 (1980).
6. IJnited States patent application Ser. No. , Atty. Dkt. No.
100/42, filed Feb. 23, 1981 by D. Goeddel et al.,a5sign~e G~lentech, Inc. (Published as Weck et al., Nucleic Acids Research 9, 6153 (1981).
7. United States patent application Ser. No. 190,799, filed Sept. 25, 1980 by D. Goeddel~assignee Genentech, Inc. (Publi~hed as Goeddel et al., Nucleic Acids Research 8, 4057 ~1980)~
8. Shine et al. Nature 285, 456-461 ~1980).
9. Beggs, J.D., van den Berg, J., van Ooyen, A., and Weissmann, C. Nature 283, 835840 (1980).
10. Henikoff, S., Tatchell, K., Hall, B.D. and NasN~th, K.A. Nature 289, 33-37 (1981).
11.
12. Carbon, J., Clarke, L., Chinault, C., Ratzkin, B., and Wal~, A. (1978) in Biochemi_try_and Genetics of Yeast (Bacila, M. Horecker, B.L. and Sboppani, A.O.M., eds) pp. 425-443, Academ~c Press, New York.
13. Clarke, L: and Carbon, J. Cell 9, 91-99 (1976).
14. Davis, R~W., Thomas, M., Cameron, J., St. John, T.P. Schererl S., and Padgett, R.A. Methods in En~ymolo~y, Vol. 65, Part 1, p. 404-411.

3~
15. Clark> L. and Carbon, J. PNAS 72, 4361-4365 (1975).
16. Hsiao, C.-l. and Carbon, J., Proc. Natl. Acad. Sci. USA 76, 3~29-3833 (1979).
17. Birnboim, ~I.C., and Doly, J. _cleic Acids Res. 7, 1513~1523 (1979).
18. ilitzeman, R.A., Clarke, L. àl~d Carboll, J. Jourral of Blolog_cal Ch~mistry 255, 12073-12030 (1980).
19. Backman, K., Ptashne, M., and Gilbert, W., Proc. Natl. hcad. Scl.
USA 73, 4174-4178 (1976).
20. Miozzari, G., Nei~eiberger, P., and Hutter, R. Journal oF
Bacteriology 134~..48-59 /(1978).
21. Faye, G., Leung, D.W., Tachell, K., Hall, B.D., and Smith, ~. Proc.
Natl. Acad. Sci. USA, in press (1981).
22. I~liller, J.H. xperiments in ~olecular Genetics, pp. 431-433,-Cold Spring Harbor Laboratory, Cold Spring Harborg NY.
23. Maxam, A.M., and Gilbert, ~. Methods ln Enzymol. 65, 490-565 (1980).
24. Stinchcomb, D.T., Struhl, K., and Davis, R.~. Nature 282, 39-43 ~1979~.
25. Ki ngsman, A.J., Clarke, L., Mortimer, R., and Carbon, J. Gene 7, 141-153 (1979).
26. Tschumper~ G., and Carbon, J. Gene 10, (1980).
27. Stewart, W.E. Il The Interferon System (Springer, N~w York, 1979).
28. De~oss, J.A. Biochem. Biophys. Res. Commun. 18, 850-857 (1965).
29. Bolivar, F., Rodriguez, R.L.g Green, P.Y., Betlach, M.C., Heyneker, H.L., Boyer, H.ll., Crosa, Y.H., and Falkow, S. Gene 2~ 95-113 ~1977).
30. Williamson, Y.M., Bennetzen, J., Young, E.T., Nasmyth, K.~ and Hall, B.D. Nature 283, 214-216 (1980).
31. Tschumper, G., and Carbon, J. Gene (Amst.) 10, 157-166 (1980).
32. Goeddel, D.Y. et al. Nature 287, 411-416 (1980).
33. Beggs, J.D. Nature 275, 104-109 ~1978).
34. Clarke, L.~ and Carbon, J. _roc. Natl. Acad. Sci. USA 77, 2173~2177 (1980).

28/35~0~

Claims (12)

CLAIMS:
1. A method of forming a transformant of a given yeast strain, which transformant is capable of expressing a biocompetent polypeptide ordinarily exogenous to yeast and not required for growth of said strain, which comprises:
a) providing a DNA transfer vector having bacterial and yeast origins of replication and genes for phenotypic selection of both bacterial and yeast moieties transformed with said genes;
b) providing. a DNA fragment comprising a structural gene encoding said polypeptide;
c) providing a DNA fragment comprising a yeast promoter genetically distinct from said structural gene;
d) inserting the fragments of steps (b) and (c) into said transfer vector together with appropriately positioned translation start and stop signals for said structural gene to form an expression vector in which said structural gene is under the control of said promoter while maintaining said origins of replications and genes for phenotypic selection; and e) transforming said strain with the resulting expression vector.
2. The method of Claim 1 which additionally comprises provision of a transcription terminator between the 3' end of said coding strand and said origins of replication in the direction in which the structural gene is transcribed.
3. The method of Claim 1 wherein the DNA fragment of step (b) also comprises translation start and stop codons respectively adjacent to the 5' and 3' ends of the coding strand of said structural gene.
4. The method of Claim 3 wherein the bacterial selection phenotype encodes antibiotic resistance and wherein the yeast selection phenotype complements a mutation carried by the yeast strain to be transformed.
5. The method of Claim 4 wherein the yeast strain trans-formed is strain RM218.
6. The method of Claim 3 wherein the amino acid sequence of said polypeptide is selected to correspond to the amino acid sequences of polypeptides selected from the group consisting of the normal and hybrid human interferons, human proinsulin, the A and s chains of human insulin, human growth hormone, somato-statin and thymosin alpha 1.
7. A DNA expression vector including origins of replication and capable, in a transformant strain of yeast, of expressing a biologically competent polypeptide ordinarily exogenous to yeast under the control of a genetically distinct yeast promoter, said polypeptide being unrequired for growth of the transformant and prepared by the following method:
a) providing a DNA transfer vector having bacterial and yeast origins of replication and one or more genes for phenotypic selection of transformant bacteria or yeast;
b) providing a DNA fragment comprising a structural. gene encoding said polypeptide;
c) providing a DNA fragment comprising a yeast promoter genetically distinct from said structural gene; and d) inserting the fragments of steps (b) and (c) into said transfer vector together with appropriately positioned transla-tion start and stop signals for said structural gene to form an expression vector in which said structural gene is under the control of said promoter, while maintaining said yeast origin of replication.
8. A DNA expression vector according to Claim 7 having a transcription terminator between the 3' end of the structural gene encoding said polypeptide and the origin(s) of replication of said vector, in the direction in which said gene is transcribed.
9. A DNA expression vector according to Claim 8 which com-prises one or more genes for phenotypic selection of yeast transformants.
10. The DNA expression vector of Claim 9 which additionally comprises one or more genes for phenotypic selection in bacteria.
11. The DNA expression vector of Claim 10 wherein the phenotypic basis for selection in bacteria is a gene encoding antibiotic resistance and wherein the phenotype for selection in yeast complements a mutation carried by a yeast strain suitable for transformation with the expression vector.

12. The expression vector of claim 8 wherein the amino acid sequence of said polypeptide is selected to correspond to the amino acid sequences of polypeptides selected from the group consisting of normal and hybrid human interferons, human proinsulin, the A and B chains of human insulin, human growth hormone, somatostatin and thymosin alpha 1.

13 . The transformed yeast strain of the process of Claim 1.

14. A yeast strain transformed with the DNA expression vector of Claim 7.

15. A yeast strain transformed with the DNA expression vector of
Claim 12.
CA000396832A 1981-02-25 1982-02-23 Expression of polypeptides in yeast Expired CA1205026A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23791381A 1981-02-25 1981-02-25
US237,913 1981-02-25

Publications (1)

Publication Number Publication Date
CA1205026A true CA1205026A (en) 1986-05-27

Family

ID=22895764

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000396832A Expired CA1205026A (en) 1981-02-25 1982-02-23 Expression of polypeptides in yeast

Country Status (10)

Country Link
US (4) US5919651A (en)
EP (1) EP0060057B1 (en)
JP (2) JPH0757190B2 (en)
AT (1) ATE27000T1 (en)
AU (1) AU553885B2 (en)
CA (1) CA1205026A (en)
DE (1) DE3276241D1 (en)
MY (1) MY101599A (en)
NZ (1) NZ199722A (en)
ZA (1) ZA821079B (en)

Families Citing this family (145)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI77877C (en) * 1979-04-20 1989-05-10 Technobiotic Ltd Process for the preparation and purification of human Le-shaped interferon protein.
US4460685A (en) * 1980-05-29 1984-07-17 New York University Method of enhancing the production of human γ Interferon
EP0062971B1 (en) * 1981-03-27 1990-03-07 Imperial Chemical Industries Plc Genetically modified microorganisms
JPS58146281A (en) * 1981-10-19 1983-08-31 Suntory Ltd Genetic engineering using karyota as host cell
US4775622A (en) * 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
DE3379665D1 (en) * 1982-05-19 1989-05-24 Unilever Nv Yeast of the genus kluyveromyces modified for the expression of preprothaumatin or its various allelic and modified forms or their maturation forms
GB2125047B (en) * 1982-08-09 1986-02-19 Ciba Geigy Ag Yeast hybrid vectors and their use for the production of polypeptides
IL69202A (en) * 1982-09-08 1991-08-16 Smith Kline Rit Surface antigen polypeptides of hbv virus produced in yeast and the preparation thereof
JPH0716432B2 (en) * 1982-10-20 1995-03-01 サントリー株式会社 Method for producing peptide using yeast high expression vector plasmid
DE3382547D1 (en) * 1983-01-12 1992-05-27 Chiron Corp SECRETORIC EXPRESSION IN EUKARYOTS.
NO172595C (en) * 1983-02-07 1993-08-11 Battelle Memorial Institute PROCEDURE AND EXPRESSION PLASMID FOR THE PREPARATION OF HEMAGGLUTININ UNDER CONTROL OF DROSOPHILA HSP PROMOTER
US4876197A (en) * 1983-02-22 1989-10-24 Chiron Corporation Eukaryotic regulatable transcription
CA1341302C (en) * 1983-02-22 2001-10-09 Rae Lyn Burke Yeast expression systems with vectors having gapdh or pyk promoters and synthesis of foreign protein
US4661454A (en) * 1983-02-28 1987-04-28 Collaborative Research, Inc. GAL1 yeast promoter linked to non galactokinase gene
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPS60501140A (en) * 1983-04-22 1985-07-25 アムジエン Secretion of exogenous polypeptides by yeast
US7198919B1 (en) 1983-04-25 2007-04-03 Genentech, Inc. Use of alpha factor sequences in yeast expression systems
ZA843275B (en) * 1983-05-05 1984-12-24 Hoffmann La Roche Novel vectors for interferon expression
WO1984004539A1 (en) * 1983-05-19 1984-11-22 Transgene Sa Production of catechol 2,3-oxygenase by means of yeasts, plasmide for the implementation thereof and application
DE3480006D1 (en) * 1983-05-19 1989-11-09 Unilever Nv Improvements in the expression of newly introduced genes in yeast cells
US4870008A (en) * 1983-08-12 1989-09-26 Chiron Corporation Secretory expression in eukaryotes
FR2562090B2 (en) * 1984-03-27 1988-04-08 Transgene Sa GLYCOSYLATED RABIES ANTIGENIC PROTEIN
FR2552776B1 (en) * 1983-10-03 1986-05-09 Transgene Sa VECTORS FOR THE EXPRESSION OF AN ANTIGENIC PROTEIN OF RABIES IN EUKARYOTIC CELLS AND THEIR APPLICATION TO THE PREPARATION OF A VACCINE
WO1985001516A1 (en) * 1983-10-03 1985-04-11 Transgene S.A. Vectors for the expression of an antigenic protein of rabies in eucaryotic cells and application thereof to the preparation of a vaccine
US5639639A (en) * 1983-11-02 1997-06-17 Genzyme Corporation Recombinant heterodimeric human fertility hormones, and methods, cells, vectors and DNA for the production thereof
PH23920A (en) * 1983-12-20 1990-01-23 Cetus Corp Glucoamylase cdna
GB8334261D0 (en) * 1983-12-22 1984-02-01 Bass Plc Fermentation processes
EP0154576A1 (en) * 1984-02-06 1985-09-11 INTERFERON SCIENCES, INC a Delaware Corporation Hybrid nucleic acid sequences having tandemly arranged genes
US4626505A (en) * 1984-02-24 1986-12-02 E. I. Du Pont De Nemours And Company Selectable markers for yeast transformation
US4745057A (en) * 1984-05-18 1988-05-17 Eli Lilly And Company Method, vectors and transformants for high expression of heterologous polypeptides in yeast
DK58285D0 (en) * 1984-05-30 1985-02-08 Novo Industri As PEPTIDES AND MANUFACTURING AND USING THEREOF
US5683688A (en) 1984-05-31 1997-11-04 Genentech, Inc. Unglycosylated recombinant human lymphotoxin polypeptides and compositions
SE8403929L (en) * 1984-07-31 1986-02-21 Sten Gatenbeck RECOMBINANT DNA MOLECULE, TRANSFORMED MICROORGANISMS AND PROCEDURE FOR PENICILLIN V-AMIDAS PREPARATION
US4752473A (en) * 1984-10-12 1988-06-21 The Regents Of The University Of California Expression of glycosylated human influenza hemagglutinin proteins
US7273695B1 (en) * 1984-10-31 2007-09-25 Novartis Vaccines And Diagnostics, Inc. HIV immunoassays using synthetic envelope polypeptides
CA1341482C (en) 1984-10-31 2005-05-10 Paul A. Luciw Process for preparing fragments of aids-associated retroviruses
US7285271B1 (en) 1984-10-31 2007-10-23 Novartis Vaccines And Diagnostics, Inc. Antigenic composition comprising an HIV gag or env polypeptide
US4812405A (en) * 1986-02-18 1989-03-14 Phillips Petroleum Company Double auxotrophic mutants of Pichia pastoris and methods for preparation
JPS6447380A (en) * 1987-08-19 1989-02-21 Agency Ind Science Techn Steroid-oxidizing yeast strain
US7442525B1 (en) 1987-12-24 2008-10-28 Novartis Vaccines And Diagnostics, Inc. Method for expressing HIV polypeptides
US5580734A (en) * 1990-07-13 1996-12-03 Transkaryotic Therapies, Inc. Method of producing a physical map contigous DNA sequences
DK0574402T3 (en) 1990-11-26 1998-05-18 Chiron Corp Expression of PACE in Host Cells and Methods for Using Them
FR2686899B1 (en) * 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5629167A (en) 1994-04-19 1997-05-13 Biocine S.P.A. Detection of antibodies against Chlamydia trachomatis pgp3 antigen in patient sera by enzyme-linked immunosorbent assay
ATE171728T1 (en) * 1994-07-29 1998-10-15 Teijin Ltd TRYPSIN TYPE ENZYMES, NUCLEIC ACID SEQUENCE CODING THEREFOR AND METHOD FOR PRODUCING THE ENZYME
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US6197545B1 (en) 1996-09-25 2001-03-06 Vanderbilt University Genetically engineered yeast with modified signal peptidase complex
MX9605082A (en) 1996-10-24 1998-04-30 Univ Autonoma De Nuevo Leon Genetically modified methylotrophic yeasts for human growth hormone secretion production.
ATE476508T1 (en) 1997-11-06 2010-08-15 Novartis Vaccines & Diagnostic NEISSERIAL ANTIGENS
WO1999036544A2 (en) 1998-01-14 1999-07-22 Chiron S.P.A. Neisseria meningitidis antigens
WO1999040434A1 (en) 1998-02-04 1999-08-12 Invitrogen Corporation Microarrays and uses therefor
GB9808932D0 (en) 1998-04-27 1998-06-24 Chiron Spa Polyepitope carrier protein
EP2261349A3 (en) 1998-05-01 2012-01-11 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
US7081360B2 (en) * 1998-07-28 2006-07-25 Cadus Technologies, Inc. Expression of G protein-coupled receptors with altered ligand binding and/or coupling properties
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
WO2000066741A2 (en) 1999-04-30 2000-11-09 Chiron S.P.A. Conserved neisserial antigens
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
GB9916529D0 (en) 1999-07-14 1999-09-15 Chiron Spa Antigenic peptides
ES2541830T3 (en) 1999-10-29 2015-07-27 Novartis Vaccines And Diagnositics S.R.L. Neisseria antigenic peptides
US6232074B1 (en) * 1999-12-10 2001-05-15 Complegen, Inc. Functional gene array in yeast
EP2275129A3 (en) 2000-01-17 2013-11-06 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins
US20060269575A1 (en) * 2000-02-08 2006-11-30 Allergan, Inc. Botulinum toxin pharmaceutical compositions formulated with recombinant albumin
US8632785B2 (en) * 2000-02-08 2014-01-21 Allergan, Inc. Clostridial toxin pharmaceutical composition containing a gelatin fragment
US20030118598A1 (en) * 2000-02-08 2003-06-26 Allergan, Inc. Clostridial toxin pharmaceutical compositions
US7780967B2 (en) 2000-02-08 2010-08-24 Allergan, Inc. Reduced toxicity Clostridial toxin pharmaceutical compositions
ES2275992T5 (en) * 2000-02-08 2011-05-18 Allergan, Inc. BOTULIN TOXIN PHARMACEUTICAL COMPOSITIONS.
EP1274720A4 (en) 2000-04-12 2004-08-18 Human Genome Sciences Inc Albumin fusion proteins
US20050100991A1 (en) * 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
IL152634A0 (en) 2000-05-04 2003-06-24 Univ Yale High density protein arrays for screening of protein activity
US7598055B2 (en) * 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
US7449308B2 (en) 2000-06-28 2008-11-11 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US7625756B2 (en) 2000-06-28 2009-12-01 GycoFi, Inc. Expression of class 2 mannosidase and class III mannosidase in lower eukaryotic cells
ATE309385T1 (en) 2000-06-28 2005-11-15 Glycofi Inc METHOD FOR PRODUCING MODIFIED GLYCOPROTEINS
US8697394B2 (en) * 2000-06-28 2014-04-15 Glycofi, Inc. Production of modified glycoproteins having multiple antennary structures
US7863020B2 (en) 2000-06-28 2011-01-04 Glycofi, Inc. Production of sialylated N-glycans in lower eukaryotes
US6737262B1 (en) * 2000-07-11 2004-05-18 Robert I. Bolla Animal feed containing polypeptides
JP2002057727A (en) * 2000-08-10 2002-02-22 Hitachi Ltd Semiconductor integrated circuit and optical communication module
CA2425303A1 (en) 2000-10-27 2002-05-02 John Telford Nucleic acids and proteins from streptococcus groups a & b
US7083787B2 (en) * 2000-11-15 2006-08-01 Globeimmune, Inc. Yeast-dendritic cell vaccines and uses thereof
US6861237B2 (en) 2000-11-30 2005-03-01 Novo Nordisk A/S Production of heterologous polypeptides in yeast
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
GB0107661D0 (en) 2001-03-27 2001-05-16 Chiron Spa Staphylococcus aureus
US20050054051A1 (en) * 2001-04-12 2005-03-10 Human Genome Sciences, Inc. Albumin fusion proteins
US7507413B2 (en) * 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
DE10123980B4 (en) * 2001-05-17 2008-01-10 Tesa Ag final insulation
NZ546711A (en) 2001-12-12 2008-06-30 Chiron Srl Immunisation against chlamydia trachomatis
EP1463752A4 (en) * 2001-12-21 2005-07-13 Human Genome Sciences Inc Albumin fusion proteins
EP2277888A3 (en) 2001-12-21 2011-04-27 Human Genome Sciences, Inc. Fusion proteins of albumin and erythropoietin
EP1572720A4 (en) 2002-05-24 2008-12-24 Nps Allelix Corp Method for enzymatic production of glp-2(1-33) and glp-2-(1-34) peptides
US20040018581A1 (en) * 2002-07-24 2004-01-29 Pharmacia And Upjohn Company Method for discovering substances for inhibiting enzymes
EP2279746B1 (en) 2002-11-15 2013-10-02 Novartis Vaccines and Diagnostics S.r.l. Surface proteins in neisseria meningitidis
CN104147597A (en) * 2002-12-16 2014-11-19 全球免疫股份有限公司 Yeast-based vaccines as immunotherapy
US7439042B2 (en) * 2002-12-16 2008-10-21 Globeimmune, Inc. Yeast-based therapeutic for chronic hepatitis C infection
US8343502B2 (en) * 2002-12-16 2013-01-01 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
DE60335602D1 (en) * 2002-12-18 2011-02-17 Roche Diagnostics Gmbh Recombinant deoxyribonuclease I from bovine pancreas with high specific activity
EP1433842B1 (en) 2002-12-18 2011-01-05 Roche Diagnostics GmbH Recombinant bovine pancreatic desoxyribonuclease I with high specific activity
DE60319333D1 (en) * 2002-12-20 2008-04-10 Roche Diagnostics Gmbh Heat labile deoxyribonuclease I variants
CA2513213C (en) 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
US7332299B2 (en) 2003-02-20 2008-02-19 Glycofi, Inc. Endomannosidases in the modification of glycoproteins in eukaryotes
EP1460425A1 (en) * 2003-03-17 2004-09-22 Boehringer Mannheim Gmbh Deglycosylated enzymes for conjugates
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
US20040248323A1 (en) 2003-06-09 2004-12-09 Protometrix, Inc. Methods for conducting assays for enzyme activity on protein microarrays
AU2004203649B2 (en) 2003-08-12 2006-01-12 F. Hoffmann-La Roche Ag Thermostable Taq polymerase fragment
EP1704234B1 (en) 2003-11-21 2012-01-18 NPS Pharmaceuticals, Inc. Production of glucagon like peptide 2 and analogs
ATE455849T1 (en) * 2003-12-05 2010-02-15 Hoffmann La Roche RECOMBINANT CARBOXYPEPTIDASE B AND ITS PURIFICATION
GB0329681D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Gene expression technique
GB0329722D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Modified plasmid and use thereof
EP2460533A3 (en) * 2004-10-18 2014-01-08 Globeimmune, Inc. Yeast-based therapeutic for chronic hepatitis C infection
AU2005317828A1 (en) 2004-12-23 2006-06-29 Novozymes Delta Limited Gene expression technique
JP4690787B2 (en) * 2005-06-14 2011-06-01 株式会社日立製作所 Microorganism detection chip, microorganism detection system, and microorganism detection method
EP1900813A4 (en) 2005-06-24 2009-08-05 Teijin Pharma Ltd Novel physiological substance nesfatin, substance relevant thereto, and use of the substances
CN101262883A (en) * 2005-07-11 2008-09-10 环球免疫公司 Compositions and methods for eliciting an immune response to escape mutants of targeted therapies
WO2007031187A1 (en) * 2005-09-14 2007-03-22 Sanofi-Aventis Deutschland Gmbh Cleavage of precursors of insulins by a variant of trypsin
CN101313216A (en) * 2005-09-22 2008-11-26 普洛茨股份有限公司 Glycosylated polypeptides produced in yeast mutants and methods of use thereof
US20070172503A1 (en) 2005-12-13 2007-07-26 Mycologics,Inc. Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines
WO2007133835A2 (en) * 2006-03-27 2007-11-22 Globeimmune, Inc. Ras mutation and compositions and mehods related thereto
US20100015168A1 (en) 2006-06-09 2010-01-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
JP5570819B2 (en) * 2007-02-02 2014-08-13 グローブイミューン,インコーポレイテッド Composition comprising yeast and method for growing yeast
AU2008216418A1 (en) * 2007-02-09 2008-08-21 Medimmune, Llc Antibody libray display by yeast cell plasma membrane
EP2134358A1 (en) * 2007-03-19 2009-12-23 Globeimmune, Inc. Compositions and methods for targeted ablation of mutational escape of targeted therapies for cancer
CN101835801B (en) 2007-08-08 2014-09-10 诺维信生物制药丹麦公司 Transferrin variants and conjugates
US8728489B2 (en) 2008-09-19 2014-05-20 Globeimmune, Inc. Immunotherapy for chronic hepatitis C virus infection
DK2393834T3 (en) 2009-02-06 2013-06-24 Novozymes Biopharma Dk As Process of purification
SG2014012918A (en) 2009-02-11 2014-04-28 Novozymes Biopharma Dk As Albumin variants and conjugates
CN102458458B (en) 2009-04-17 2016-11-16 全球免疫股份有限公司 Combination immunotherapy compositions against cancer and method
WO2011032119A1 (en) 2009-09-14 2011-03-17 The Regents Of The University Of Colorado Modulation of yeast-based immunotherapy products and responses
EA201290897A1 (en) 2010-03-11 2013-03-29 Иммьюн Дизайн Корп. VACCINES FOR PANDEMIC FLU
EP2380905A1 (en) 2010-04-19 2011-10-26 Thrombotargets Europe, S.L. Phospholipid-enriched vesicles bearing tissue factor having haemostatic activities and uses thereof
ES2696551T3 (en) 2010-12-17 2019-01-16 Globeimmune Inc Compositions and methods for the treatment or prevention of human adenovirus-36 infection
EA028659B1 (en) 2011-02-12 2017-12-29 Глоубиммьюн, Инк. Yeast-based therapeutic for treating chronic hepatitis b
EP3238731B1 (en) 2011-03-17 2021-02-17 Globeimmune, Inc. Yeast-brachyury immunotherapeutic compositions
JP2014523878A (en) 2011-06-14 2014-09-18 グローブイミューン,インコーポレイテッド Yeast-based compositions and methods for the treatment or prevention of hepatitis D virus infection
WO2013025972A1 (en) 2011-08-17 2013-02-21 Globeimmune, Inc. Yeast-muc1 immunotherapeutic compositions and uses thereof
SG11201408652SA (en) 2012-06-26 2015-01-29 Biodesix Inc Mass-spectral method for selection, and de-selection, of cancer patients for treatment with immune response generating therapies
RU2679806C2 (en) 2013-03-19 2019-02-13 Дзе Юнайтед Стейтс Оф Эмерика, Эз Репрезентед Бай Дзе Секретэри, Дипартмент Оф Хелт Энд Хьюман Сервисиз Yeast-based immunotherapy for chordoma
EP2978449B1 (en) 2013-03-26 2021-09-08 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human immunodeficiency virus infection
WO2015031778A2 (en) 2013-08-30 2015-03-05 Globeimmune, Inc. Compositions and methods for the treatment or prevention of tuberculosis
KR102409372B1 (en) 2014-04-11 2022-06-16 글로브이뮨 Yeast-Based Immunotherapy and Type I Interferon Sensitivity
US10106579B2 (en) 2014-06-12 2018-10-23 Ra Pharmaceuticals, Inc. Modulation of complement activity
EP3250230B9 (en) 2015-01-28 2022-02-23 RA Pharmaceuticals, Inc. Modulators of complement activity
US11065313B2 (en) 2015-08-03 2021-07-20 Globeimmune, Inc. Modified yeast-brachyury immunotherapeutic compositions
DK3389692T3 (en) 2015-12-16 2020-03-30 Ra Pharmaceuticals Inc MODULATORS OF COMPLEMENT ACTIVITY
MX2019006527A (en) 2016-12-07 2019-08-01 Ra Pharmaceuticals Inc Modulators of complement activity.
US10837031B2 (en) 2017-05-10 2020-11-17 Baymedica, Inc. Recombinant production systems for prenylated polyketides of the cannabinoid family
EP3934751A1 (en) 2019-03-08 2022-01-12 Mevion Medical Systems, Inc. Collimator and energy degrader for a particle therapy system

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4237224A (en) * 1974-11-04 1980-12-02 Board Of Trustees Of The Leland Stanford Jr. University Process for producing biologically functional molecular chimeras
CA1114295A (en) * 1977-11-02 1981-12-15 John H. Growdon Process and composition for treating disorders by administering choline or a compound that dissociates to choline
FI783366A (en) * 1977-11-08 1979-05-09 Genentech Inc FOERBAETTRAT FOERFARANDE FOER MIKROBIPOLYPEPTIDUTTRYCK
US4563424A (en) * 1979-11-05 1986-01-07 Genentech, Inc. Method and means for somatostatin protein conjugate expression
US4366246A (en) * 1977-11-08 1982-12-28 Genentech, Inc. Method for microbial polypeptide expression
US4356270A (en) * 1977-11-08 1982-10-26 Genentech, Inc. Recombinant DNA cloning vehicle
CH662128A5 (en) * 1977-11-08 1987-09-15 Genentech Inc RECOMBINATION CLONING SUPPORT.
FR2441659A1 (en) * 1978-11-14 1980-06-13 Anvar NOVEL HYBRID PLASMIDS AND MICROORGANISMS CONTAINING THEM
FR2480779B2 (en) * 1979-08-30 1986-07-18 Anvar VECTOR CONTAINING A NUCLEOTIDE SEQUENCE OF THE SURFACE ANTIGEN OF HEPATITIS B VIRUS AND METHOD FOR MANUFACTURING AN IMMUNOGENIC MOLECULE USING THE SAME
LU88258I2 (en) * 1978-12-22 1994-02-03
AU542264B2 (en) * 1979-06-01 1985-02-14 G.D. Searle & Co. Plasmid vectors
FR2458585A1 (en) * 1979-06-07 1981-01-02 Pasteur Institut Genetically engineered proteins for nutrition - prepd. from ovalbumin
US4342832A (en) * 1979-07-05 1982-08-03 Genentech, Inc. Method of constructing a replicable cloning vehicle having quasi-synthetic genes
US4431739A (en) * 1979-11-05 1984-02-14 Genentech, Inc. Transformant bacterial culture capable of expressing heterologous protein
US4425437A (en) * 1979-11-05 1984-01-10 Genentech, Inc. Microbial polypeptide expression vehicle
GB2068969B (en) * 1980-02-05 1983-07-27 Upjohn Co Gene expression
JPS57500408A (en) * 1980-02-25 1982-03-11
JPS56138154A (en) * 1980-02-28 1981-10-28 Genentech Inc Desacetylthymosin-alpha-1 and manufacture
CH651308A5 (en) * 1980-07-01 1985-09-13 Hoffmann La Roche INTERFERON AND THEIR PRODUCTION.
FR2490675B1 (en) * 1980-09-25 1985-11-15 Genentech Inc MICROBIAL PRODUCTION OF HUMAN FIBROPLASTER INTERFERON
NZ199391A (en) * 1981-01-02 1985-12-13 Genentech Inc Chimeric polypeptides comprising a proinsulin sequence,and preparation by recombinant dna technique;production of human insulin
US4666847A (en) * 1981-01-16 1987-05-19 Collaborative Research, Inc. Recombinant DNA means and method
US4769238A (en) * 1981-08-04 1988-09-06 The Regents Of The University Of California Synthesis of human virus antigens by yeast
US4462199A (en) * 1981-08-13 1984-07-31 Xerox Corporation Rotary inserter
DK368882A (en) * 1981-08-25 1983-02-26 Alan John Kingsman EXPESSIONS VECTORS
AU561343B2 (en) 1981-10-19 1987-05-07 Genentech Inc. Human immune interferon by recombinant dna

Also Published As

Publication number Publication date
US5856123A (en) 1999-01-05
ATE27000T1 (en) 1987-05-15
US5618676A (en) 1997-04-08
NZ199722A (en) 1985-12-13
US5919651A (en) 1999-07-06
JPS57159489A (en) 1982-10-01
EP0060057A1 (en) 1982-09-15
AU553885B2 (en) 1986-07-31
DE3276241D1 (en) 1987-06-11
MY101599A (en) 1991-12-17
US5854018A (en) 1998-12-29
JPH09182585A (en) 1997-07-15
AU8065782A (en) 1982-09-02
EP0060057B1 (en) 1987-05-06
ZA821079B (en) 1983-04-27
JPH0757190B2 (en) 1995-06-21

Similar Documents

Publication Publication Date Title
CA1205026A (en) Expression of polypeptides in yeast
EP0329203B1 (en) Yeast expression systems with vectors having pyk promoters, and synthesis of foreign protein
EP0123811B1 (en) The use of the gal 1 yeast promoter
Vernet et al. A family of yeast expression vectors containing the phage f1 intergenic region1
CA1340837C (en) Expression and secretion of heterologous proteins by yarrowia lipolytica transformants
EP0945515A2 (en) Production of proteins using homologous recombination
EP0286424A1 (en) Yeast vector
JPH03143385A (en) Yeast cell
GB2107718A (en) Human immune interferon
US4935350A (en) Materials and methods for controlling plasmid copy number and stability
EP0142268A1 (en) Saccharomyces cerevisiae possessing gene coding for interleukin-2 polypeptide and method for producing interleukin-2 using the yeast
EP0118393A2 (en) Methods and compositions for expression of competent eukaryotic gene products
WO1988002779A1 (en) Composite yeast vectors
AU671801B2 (en) Gene manipulation and expression using genomic elements
US5646037A (en) Yeast vectors
USRE40940E1 (en) Yeast promoter and use thereof
EP0339567A1 (en) Expression of Hepatitis B PreS 2 Protein in Methylotrophic Yeasts
AU707657B2 (en) Process for the production of proteins
EP0245479B1 (en) Shortened phosphoglycerate kinase promoter
WO1986000637A1 (en) Yeast cloning vehicle
EP0214638A2 (en) Yeast promoters
NO854864L (en) MANUFACTURING A MULTIPLE COPPER YET VECTOR.
WO1986000638A1 (en) Yeast cloning vehicle
JPH0391486A (en) New gene, vector, transformant using thereof and use of same transformant

Legal Events

Date Code Title Description
MKEX Expiry