CA1282354C - INTERFERON .alpha. 54 - Google Patents

INTERFERON .alpha. 54

Info

Publication number
CA1282354C
CA1282354C CA000419459A CA419459A CA1282354C CA 1282354 C CA1282354 C CA 1282354C CA 000419459 A CA000419459 A CA 000419459A CA 419459 A CA419459 A CA 419459A CA 1282354 C CA1282354 C CA 1282354C
Authority
CA
Canada
Prior art keywords
ifn
polypeptide
gene
dna
alpha
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 - Fee Related
Application number
CA000419459A
Other languages
French (fr)
Inventor
Michael A. Innis
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.)
Cetus Oncology Corp
Original Assignee
Cetus Corp
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
Application filed by Cetus Corp filed Critical Cetus Corp
Application granted granted Critical
Publication of CA1282354C publication Critical patent/CA1282354C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • 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
    • Y10S435/811Interferon

Abstract

Abstract A new polypeptide, called IFN-.alpha.54, produced by E.coli transformed with a newly isolated and char-acterized human IFN-.alpha. gene is described. The polypep-tide exhibits interferon activities such as antiviral activity, cell growth regulation, and regulation of production of cell-produced substances.

Description

~ ~Z3~

Description Technical Field The invention is in the field of biotech-5 nology. More particularly it relates to a polypeptidehaving interferon (IFN) activity, DNA that codes for the polypeptide, a recombinant vector that includes the DNA, a host organism transformed with the recom-binant vector that produces the polypeptide, pharma-10 ceutical compositions containing the polypeptide, andtherapeutic methods employing the polypeptide.

Background Art IFNs are proteins with antiviral, immuno-modulatory, and antiproliferative activities produced 15 by mammalian cells in response to a variety of indu-cers (see Stewart, W.E., The Interferon System, Springer-Verlag, New YorX, 1979). The activity of IFN
is largely species specific (Colby, C., and Morgan, M.
J., Ann. Rev. Microbiol. 25:333-360 (1971) and thus 20 only human IFN can be used for human clinical studies.
Human IFNs are classiied into three groups, aj ~, and ~, (Nature, 286:110, (1980)). The human IFN-a genes compose a multigene family sharing 85%-95% sequence homology (~Goeddel, D. V., et al, Nature 290:20-27 (1981) Nagata, S., et al, J. Interferon Research 1:333-336 (19~1)). Several of the IFN-a genes have been cIoned and expressed in E.coli (Nagata, S., et 823~

al, ~ature 284:316-320 (19~0); Goeddel, D. V., et al, Nature 287:411-415 (1980); Yelverton, E., et al, Nucleic Acids Research, 9:731-741, 11981); Streuli, M., et al, Proc Nat Acad Sci ~USA), 78:2848-2~52. The 5 resulting polypeptides have been purified and tested for biological activities associated with partially purified native human IFNs and found to possess simi-lar activities. Accordingly such polypeptides are potentially useful as antiviral, immunomodulatory, or 10 antiproliferative agents.
A principal object of the present invention is to provide a polypeptide having interferon activity that is produced by an organism transformed with a newly isolated and newly characterized IFN- gene.
15 This polypeptide is sometimes referred to herein as IFN-54. Other objects of the invention are directed to providing the compositions and hosts that are used to produce this polypeptide and to therapeutic compo-sitions and methods that use this polypeptide as an active ingredient.

Disclosure of the Invention One aspect of the invention is a polypeptide having interferon activity and comprising the amino acid sequence:

CysAspLeuProGln ThrHisSerLeuGly HisArgArfflhrMet MetLeuLeuAlaGln MetArgArgIleSer LeuPheSerCysLeu LysAspArgHisAsp PheArgpheproGln GluGluPheAspGly AsnGlnPheGlnLys AlaGluAlaIleSer ValLeuHisGluVal 25 IleGlnGlnThrPha AsnLeuPheSerThr LysAspSerSerVal AlaTrpAspGluArg LeuLeuAspLysLeu TyrThrGluLeuTyr GlnGlnLeuAsnAsp LeuGluAlaCysVal MetGlnGluVal~rp ValGlyGlyThrPro LeuMetAsnGluAsp SerIleLeaAlaVal ArgLysTyrPheGln ArgIleThrLeuTyr LeuThrGluLysLys Tyr5erprocysAla TrpGluValValArg AlaGluIleMetArg SerPheSerSerSer ArgAsnLeuGlnGlu ArgLeuArgArgLys Glu ' '- . ' , ' , ' ' ' ~' " '"
, ~8~3~

A second aspect of the invention is a DNA
unit or fragment comprising a nucleotide sequence that encodes the above described polypeptide.
A third aspect of the invention is a cloning vehicle or vector that includes the above described DNA.
A fourth aspect of the invention is a host that is transformed with the above described cloning vehicle and that produces the above described polypeptide.
A fifth aspect of the invention is a process for producing the above described polypeptide compri-sing cultivating said transformed host and collecting the polypeptide from the resulting culture.
Another aspect of the invention i5 a pharma-ceutical composition having interferon activity com-prising an effective amount of the above described polypeptide admixed with a phar~aceutically acceptable carrier.
Still another aspect of the invention is a method of providing interferon therapy to a human comprising administering a therapeutically effective amount of the above described polypeptide to the human.

Brief Description of the Drawings Figure 1 is a partial restriction map which shows the two XhoII restriction sites that produce a homologous 260 base pair DNA fragment from the IFN-al and IF~-a2 structural genes. Data for this map are from Streuli, M., et al Science, 209:1343-1347 (1980).
Figure 2 depicts the sequencing strategy used to obtain the complete DNA sequence of the IFN-a54 gene coding region. Bacteriophage mp7:a54-1 . . ~ , ~:

-' ~
:

~8~:354 DNA served as the template for sequences obtained withprimers A, H and F and bacteriophage mp7:a54-2 DNA was the template for sequences obtained with primers E an~
G. The crosshatched area of the gene depicts the 5 region that encodes the 23 amino acid signal peptide and the open box depicts the region that encodes the mature polypeptide. The scale, in base pairs, is numbered with 0 representing the ATG start codon of preinterferon. The arrows indicate the direction and lO extent of sequencing with each primer.
Figure 3 is the nucleotide sequence of the structural gene coding for IFN-a54 including some of the flanking 5'- and 3'- noncoding regions of the gene. The region coding for preinterferon and the 15 mature polypeptide begins with the ATG codon at posi-tion 61 and terminates with the TAA codon at posi-tion 628.
Figure 4 is a partial restriction map o the coding region of the IFN-aS4 gene. The crosshatching 20 represents the region that encodes the 23 amino acid signal peptide and the open box represents the gene coding sequence for the mature polypeptide. The scale, in base pairs, is numbered with 0 representing the ATG start codon of preinterferon.
Figure 5 shows the amino acid sequence of the 23 amino acid signal polypeptide and the 166 amino acid mature IFN-a54 coded for by the gene dep~cted in Figure 3. The 189 amino acid sequence is displayed above the corresponding nucleotide sequence. Amino 3b acid 24, cysteine, is the first amino acid of the mature IFN-a54 protein.
Figure 6 is the DNA sequence of the E. coli trp promoter and the gene of Figure 3 which was inserted between the EcoRI and PvuII sites of the 3~

plasmid pBWll. The amino acid sequence of Figure 5 is written above the corresponding DNA sequence and the location of ~he restriction sites used in the cons-truction of the expression plasmid are indicated.
Figure 7 is a diagram of the expression plasmid, pCS12.

Modes for Carrying Out the Invention In general terms IFN-~54 was made by identi-fying and isolating the IFN-54 gene by screening a library of human genomic DNA with an appropriate IFN-DNA probe, constructing a vector containing the IFN-54 gene, transforming microorganisms with the vector, cultivating transformants that express IFN-54 and collecting IFN-a54 from the culture. A preferred embodiment of this procedure is described below.

DNA Probe Preparation Total cytoplasmic RNA was extracted from human lymphoblastoid cells, Namalwa, which had been induced for IFN production by pretreatment with 5-bromodeoxyuridine and Newfastle Disease Virus (NDV). The poly(A) (polyadenylic acid)-containing messenger RNA (mRNA) was isolated from total RNA by chromatography on oligo(dT)-cellulose (type 3 from Collaborative Research, Aviv, ~1., and Leder, P., Proc Natl Acad Sci (USA), 69:1408-1412, (1972)) and enriched for IFN mRNA by density gradient centrifu-gation on 5~6-20~6 sucrose gradients. Fractions con-taining IFN mRNA were identified by translating the mRNA by microinjecting aliquots of each fraction into Xenopus oocytes and determining the IFN activity of the products of the translations according to a method described by Colman, A., and Morser, J., Cell, 17:517-526 (1979).

, , ' ' ,.

3~

The Namalwa cell human IFN enriched mRNA was used to construct complementary DNA (cDNA) clones in E. coli by the G/C tailing method using the PstI site of the cloning vector pBR322 (Bolivar, F., et al, G _ , 2:95-113 (1977)). A population of transformants containing approximately 50,000 individual cDNA clones was grown in one liter of medium overnight and the total plasmid DNA was isolated therefrom.
The sequences of two IFN-a clones (IFN-al and IFN-2) have been published (Streuli, M., et al, Science, 209:1343-1347 (1980)). Examination of the DNA sequences of these two clones revealed that the restriction enzyme X II would excise a 260 bp frag-ment from either the IFN-al or the IFN-a2 gene (see Figure 1). XhoII was prepared in accordance with the process described by Gingeras, T.R., and Roberts, R.J., J Mol Biol, 118:113-122 (1978).
One mg of the purified total plasmid DNA
preparation was digested with XhoII and the DNA frag-ments were separated on a preparative 6% polyacryl-amide gel. DNA from the region of the gel correspon-ding to 260 bp was recovered by electroelution and recloned by ligation into the BamHI site of the single strand bacteriophage ml3:mp7. Thirty-slx clones were 25 picked at random and the single stranded DNA isolated therefrom and sequenced. I'he DNA sequences of ~our of these clones were homologous to known IFN- DNA
sequences. Clone mp7:a-260, with a DNA sequence iden-tical to IFN-al DNA (Streuli, M. et al, Science, 209:1343-1347 (19~0)) was chosen as a highly specific hybridization probe for identifying additional IFN-a DNA sequences. This clone is hereinafter referred to as the "260 probe."

3Z3~

Screening of Genomic DNA Library In order to isolate other IFN-a gene sequences, a 32P-labelled 260 probe was used to screen a library of human genomic DNA by in situ hybridiza-tion. The human gene bank, prepared by Lawn, R.M., etal, Cell, 15:1157-1174 (1978), was generated by partial cleavage of fetal human DNA with HaeIII and AluI and cloned into bacteriophage ~ Charon 4A with synthetic EcoRI linkers. Approximately 800,000 clones 10 were screened, of which about 160 hybridized with the 260 probe. Each of the 160 clones was further charac-terized by restriction enzyme mapping and comparison with the published restriction maps of 10 chromosomal IFN genes (Nagata, S., et al, J Interferon Research, 1:333-336 (1981))~ One of the clones, hybrid phage ~4A:a54 containing a 16 kb insert, was characterized as follows. A DNA preparation of ~4A:a54 was cleaved with HindIII, BglII, and EcoRI respectively, the frag-ments separated on an agarose gel, transferred to a nitrocellulose filter (Southern, E.M., ~ Mol Biol, 98:503-517 (1977)) and hybridized with 32P-labelled 260 probe. This procedure localized the IFN-aS4 gene to a 3.9 kb EcoRI restriction fragment which was then isolated and recloned, in both orientations, by ligation of the fragment into EcoRI cleaved ml3:mp7.
The two subclones are designated mp7: a54-1 and mp7: a54-2 . The -1 designation indicates that the sinyle-stranded bacteriophage contains insert DNA
complementary to the mRNA (the minus strand) and the -2 designation indicates that the insert DNA is the same sequence as the mRNA (the plus strand).

~ 323~

Sequencing of the IFN-a54 Gene The Sanger dideoxy-technique was used to determine the DNA sequence of the IFN-a54 gene. The strategy employed is diagrammed in Figure 2, the DNA
sequence thus obtained is given in Figure 3, and a partial restriction enzyme map of the IFN-a54 gene is illustrated in Figure 4. Unlike many genes from eukaryotic organisms, but analogous to other IFN
chromosomal genes which have been characterised (Nagata, S., et al, Nature, 287:401-408 (1980), Lawn, R.M , et al, Science, 212:1159-1162 (1981), Lawn, R.M., et al, Nucleic Acids Res., 9:1045-1052 (1981);
Nagata, S., et al, J Interferon Research, 1:333-336 (1981); Lawn, R.M., et al, Proc Natl Acad Sci (USA), 78:5435-5439 (1981)), the DNA sequence of this gene demonstrates that it lacks introns. Homology to protein sequence information from these published IFN-a genes made it possible to determine the correct translational reading frame and thus allowed the entire 166 amino acid sequence of IFN-a54 to be pre~
dicted from the DNA sequence as well as a precursor segment, or signal peptide, of 23 amino acids (Figure 5). The DNA sequence of the IFN-a54 gene and the amino acid sequence predicted therefrom differ sub-stantially from the other known IFN-a DNA and IFN-a amino acid sequences.

Plasmid Preparation and Host Transformation Assembly of the plasmid for direct expres-sion of the IFN-a5~ gene involved replacing the DN~
fragment encoding the 23 amino acid signal polypeptide of preinterferon with a 120 bp EcoRI/Sau3A promoter fragment (E.coli trp promoter, operator, and trp leader ribosome binding site preceding an ATG initia-. , .

.
.

3235~

tion codon) and using the naturally occurring HincIIsite, 139 bp 3'~ of the TAA translational stop codon, to insert the gene into a cloning vehicle derived from the plasmid pBR322. The complete DNA sequence of the 5 promoter and gene fragments inserted between the EcoRI
and PvuII sites of pBR322 is shown in Figure 6 which also shows the exact location of relevant cloning sites. Details of the construction are described below.
The coding region for mature IFN-a54 encom-passes a Sau3~ site between codons for amino acids 2 and 3 and an XbaI site between codons for amino acids 81 and 83. The 239 bp Sau3A to XbaI fragment was iso-lated on a 6% polyacrylamide gel following a S 3A/XbaI double-digest of the 3.9 kb Eco~I genomic fragment. This fragment was ligated to the 120 bp EcoRI/Sau 3A promoter fragment. The promoter fragment contained a synthetic HindIII restriction site, ATG
initîation codon, the initial cysteine codon (TGT) and a Sau 3A "sticky end". The ligation mixture was digested with EcoRI and XbaI to enrich for the desired product and ligated with an EcoRI/XbaI digested vector ragment pBWll (derived from pBR322 w~ich contained unique EcoRI and XbaI restriction sites). The ligation mixture was used to transform E.coli MM294 (Backman, K., et al, Proc Natl Acad Sci (USA), 73:4174-4178 (1976)). The desired correct transforma-tion product, designated pCS10, was identified by restriction enzyme mapping. DN~ from this interme-diate plasmid was prepared, digested with XbaI andPvuII, and the large fragment containing the promoter and the S'-portion of the gene was used as a vector for reconstituting the 3'-end of the gene. ~eferring to the restriction enzyme sites shown in Figure 6, the :

.: . , .: . - .
.

~L~8~S~

397 base pair Xba to HincII fragment encompassing the 3'-cod~n region, the TAA translational stop codon and an additional 139 base pairs of 3'-noncoding sequence was isolated on a 4~ polyacrylamide gel following an 5 XbaI/HlncII double-digest o~ the 3.9 kb genomic EcoRI
fragment. This XbaI to HlncII fragment was ligated to the Xba/PvuII digested pCSlO, the ligation mixture was used to transform E.coli MM294, and correct transfor-mants (3 out of 100 screened) were identified by restriction enzyme mapping. Figure 7 is a diagram of one of the final expression constructs obtained, which is designated pCS12. Other prokaryotic hosts such as bacteria other than E.coli may, of course, be trans-formed with this or other suitable constructs to 15 replicate the IFN-a54 gene and/or to produce IFN-a5~.

C ivation of Transformants_ Bacteria transformed with the IFN-a54 gene may be cultivated in an appropriate growth medium, such as a minimum essential medium, that satisfies the 20 nutritional and other requirements needed to permit the bacteria to grow and produce IFN-a54. If the bac-teria are such that the protein is contained in their cytoplasm, the IFN-a54 may be extracted from the cells by lysing the cells such as by sonication and/or 25 treatment with a strong anionic solubilizing agent such as sodium dodecyl sulfate. Further purification of the extract may be achieved by affinity chroma-tography, electrophoresis, or other protein purifi-cation techniques.
IFN-a54 produced in accordance with the invention is believed to be distinct from the corresponding natlve protein in several respects.

' ' ' ' .

~3Z3~

Firstly, because ~he IFN-a54 gene was expressed by bacterial hosts that utilize N-formyl-methionine and/or methionine to initiate translation, some or all of the bacterially produced IFN-a54 molecules are pre-ceded by an N-formyl-methionine or methionine group.
Some of the N-formyl-methionine or methionine groups could be removed by natural in vivo bacterial cleavage mechanisms. This would result in a mixture of mole-cules, some of which would include an initial N-formyl-methionine or methionine and others that would not. All such IFN-a54 molecules, those containing an initial N-formyl-methionine or methionine, those not containing an N-formyl-methionine or methionine and any mixture thereof, are encompassed by the present invention. Secondly, the amino acid residues of the bacterially produced polypeptide are unsubstituted w~ereas the residues of the native protein may be substituted with sugar groups, ACTH or other moie-ties. Also, native IFN-a extracts consist of mixtures o~ various IFN molecules whereas the bacterially produced IFN-a54 is homogeneous, that is, bacterially produced IFN-a54 does not contain functionally related polypeptides. Accordingly, the invention contemplates producing IFN-a54-containing compositions having bio-logical activity that is attributable solely toIFN-a54 and/or said terminal N-formyl-methionine or methionine derivatives thereof.

Biological Testing of IFN-a54 IFN-a54-containing cell sonicates were tested ln vitro and found to have the following activities: (1) inhibition of viral replication of vesicular stomatitis virus (VSV) and herpes simplex virus-l (HSV-l); (2) inhibition of tumor cell growth, ~' , .
' 23~4 (~) inhibition of colony ~ormation by tumor cells in soft agar; (4) activation of natural killer (NK) cells; (5) enhancement of the level of 2',5'-oligo-adenylate synthetase (2',5'-A); and (6) enhancement of 5 the double-stranded RNA-dependent protein kinase. The sonicates were active in inhibiting viral infection in both human and other mammalian cells such as hamster, monkey, mouse, and rabbit cells.
The tests show that IFN-a54 exhibits anti-lO viral activity against DNA and RNA viruses, cell growth regulating activity, and an ability to regulate the production of intracellular enzymes and other cell-proauced substances. Accordingly, it is expected IFN-a54 may be used to treat viral infections with a 15 potential for interferon therapy such as chronic hepa-titis B infection, ocular, local, or systemic herpes virus infections, influenza and other respiratory tract virus infections, rabies and other viral zoono-ses, arbovirus infections, and slow virus diseases 20 such as Kuru and sclerosing panencephalitis. It may also be useful for treating viral infections in immunocompromised patients such as herpes zoster and varicella, cytomegalovirus, Epstein-Barr virus infec-tion, herpes simplex infections, rubella, and progres-~5 sive multifocal leukoencephalopathy. Its cell growthregulating activity makes it potentially useful for treating tumors and cancers such as osteogenic sar-coma, multiple myeloma, Hodgkin's disease, nodular, poorly differentiated lymp~oma, acute lymphocytic 30 leukemia, breast carcinoma, melanoma, and nasopharyn-geal carcinoma. The fact that IFN-~54 increases protein kinase and 2',5'-oligoadenylate synthetase ~ . .

.

Z35~

indicates it may also increase synthesis of other enzymes or cell-produced substances commonly affected by IFNs such as histamine, hyaluronic acid, prosta-glandin E, tRNA me~hylase, and aryl hydrocarbon hydro-lase. Similarly, it may be useful to inhibit enzymescommonly inhibited by IFNs such as tyrosine amino transferase, glycerol-3-phosphate dehydrogenase glutamine synthetase, ornithine decarboxylase, S-adenosyl-l-methionine decarboxylase, and UDP-N-acetylglucosamine-dolichol monophosp~ate transferase.
The ability of the IFN-a54 to stimulate NK cell activity is indicative that it may also possess other activities such as the abilities to induce macrophage activity and antibody production and to effect cell surface alterations such as changes in plasma membrane density or cell surface charge, altered capacity to bind substances such as cholera toxin, concanavalin A
and thyroid-stimulating hormone, and change in the exposure of surface gangliosides.
Pharmaceutical compositions that contain IFN-a54 as an active ingredient will normally be for-mulated with an appropriate solid or liquid carrier depending upon the particular mode of administration being used. For instance, parenteral formulations are usually injectable fluids that use pharmaceutically and physiologically acceptable fluids such as physio-logical saline, balanced salt solutions, or the like as a vehicle. Oral formulations, on the other hand, may be solid, eg tablet or capsule, or liquid solu-tions or suspensions. IFN-a54 will usually be formu-lated as a unit dosage form that contains in the range of 104 to 107 international units, more usually 106 to 107 international units, per dose.

- ., .
'~,,' ' ,' ' , '.

': ',:
., . ,' : :

35D~

IFN-a54 may be administered to humans in various manners such as orally, intravenously, intra-muscularly, intraperitoneally, intranasally, intra-dermally, and subcutaneously. The particular mode of administration and dosage regimen will be selected by the attending physician taking into account the par-ticulars of the patient, the disease and the disease state involved. For instance, viral infections are usually treated by daily or twice daily doses over a few days to a few weeks, whereas tumor or cancer treatment involves daily or multidaily doses over months or years. IF~-a54 therapy may be combined with other treatments and may be combined with or used in association with other chemotherapeutic or chemo-15 preventive agents for providing therapy against viralinf~ctions, neoplasms, or other conditions against which it is effective. For instance, in the case of herpes virus keratitis treatment, therapy with IFN has been supplemented by thermocautery, debridement and trifluorothymidine t~erapy.
Modifications of the above described mcdes for carrying out the invention, such as, without limitation, use of alternative vectors, alternative expression control systems in the vector, and alter-native host microorganisms and other therapeutic orrelated uses of IFN-a5~ that are obvious to those of ordinary skill in the biotechnology, pharmaceutical, medical and/or reIated fields are intended to be within the scope of the following claims.

, .

:;

Claims (6)

1. A process for preparing a polypeptide having interferon activity and comprising the amino acid sequence:

which process comprises:
(a) inserting into a cloning vector an IFN-.alpha. gene having the nucleotide sequence:

said vector comprising plasmid pCS12;

(b) transforming a host consisting of E. coli with the vector;
(c) cultivating transformants which express the IFN-.alpha. gene; and (d) collecting the polypeptide from the resulting culture.
2. The process of claim 1 wherein at least some of the polypeptide produced has its initial cysteine residue preceded by an N-formylmethionine or methionine group.
3. A polypeptide having interferon activity and comprising the amino acid sequence:

whenever prepared or produced by or significantly derived from the process as defined in claim 1.
4. A DNA unit consisting of a nucleotide sequence which is:

that encodes the polypeptide comprising the amino acid sequence:

5. A cloning vehicle that includes the DNA
unit of claim 4, said cloning vehicle being the plasmid pCS12.
6. A host that is transformed with the cloning vehicle of claim 5 and produces IFN-.alpha. 54, wherein the host is E. coli.
CA000419459A 1982-01-15 1983-01-14 INTERFERON .alpha. 54 Expired - Fee Related CA1282354C (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US33982882A 1982-01-15 1982-01-15
US339,828 1982-01-15
US06/414,051 US4975276A (en) 1982-01-15 1982-09-02 Interferon-alpha 54
US414,051 1982-09-02

Publications (1)

Publication Number Publication Date
CA1282354C true CA1282354C (en) 1991-04-02

Family

ID=26991828

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000419459A Expired - Fee Related CA1282354C (en) 1982-01-15 1983-01-14 INTERFERON .alpha. 54

Country Status (5)

Country Link
US (1) US4975276A (en)
EP (1) EP0099389A1 (en)
CA (1) CA1282354C (en)
IT (1) IT1164862B (en)
WO (1) WO1983002458A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6156199A (en) * 1984-08-27 1986-03-20 Shionogi & Co Ltd Novel human interferon alpha
DE3685044D1 (en) * 1985-02-01 1992-06-04 Ici Plc ANALOG INTERFERON POLYPEPTIDES, METHOD FOR THE PRODUCTION THEREOF AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5028422A (en) * 1986-05-27 1991-07-02 Schering Corporation Treatment of basal cell carcinoma intralesionally with recombinant human alpha interferon
US5676942A (en) * 1992-02-10 1997-10-14 Interferon Sciences, Inc. Composition containing human alpha interferon species proteins and method for use thereof
EP1987845B1 (en) 1997-11-20 2012-03-21 Vical Incorporated Treatment of cancer using cytokine-expressing polynucleotides and compositions therefor.
FR2823220B1 (en) 2001-04-04 2003-12-12 Genodyssee NOVEL ERYTHROPOIETIN (EPO) POLYNUCLEOTIDES AND POLYPEPTIDES
ES2411007T3 (en) 2001-10-10 2013-07-04 Novo Nordisk A/S Remodeling and glycoconjugation of peptides
EP2305311A3 (en) 2001-10-10 2011-07-20 BioGeneriX AG Glycoconjugation of peptides
US20040063912A1 (en) * 2002-03-15 2004-04-01 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
CA2522345A1 (en) 2003-04-09 2004-11-18 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
AU2005209926B2 (en) 2004-02-02 2007-09-06 Ambrx, Inc. Modified human four helical bundle polypeptides and their uses
WO2008137471A2 (en) * 2007-05-02 2008-11-13 Ambrx, Inc. Modified interferon beta polypeptides and their uses
CN101939443B (en) 2008-02-08 2014-01-29 Ambrx公司 Modified leptin polypeptides and their uses

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IN150740B (en) * 1978-11-24 1982-12-04 Hoffmann La Roche
PH21848A (en) * 1980-01-08 1988-03-17 Biogen Nv Dna sequences and recombinant dna molecules
ATE25985T1 (en) * 1980-06-12 1987-04-15 Japan Found Cancer PLASMID.
CH651308A5 (en) * 1980-07-01 1985-09-13 Hoffmann La Roche INTERFERON AND THEIR PRODUCTION.
US4801685A (en) * 1981-08-14 1989-01-31 Hoffmann-La Roche Inc. Microbial production of mature human leukocyte interferon K and L

Also Published As

Publication number Publication date
WO1983002458A1 (en) 1983-07-21
EP0099389A1 (en) 1984-02-01
IT8347560A0 (en) 1983-01-14
IT1164862B (en) 1987-04-15
US4975276A (en) 1990-12-04

Similar Documents

Publication Publication Date Title
CA1282356C (en) INTERFERON .alpha. 76
CA1210715A (en) Multiclass hybrid interferons
US4588585A (en) Human recombinant cysteine depleted interferon-β muteins
USRE33653E (en) Human recombinant interleukin-2 muteins
JP2515308B2 (en) Human immune interferon
US5487984A (en) Processes for producing tumor necrosis factor
US4853332A (en) Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of biologically active proteins
EP0211148B2 (en) Mature human leukozyte interferons, process for their bacterial production, intermediates therefor and compositions containing them
US4959314A (en) Cysteine-depleted muteins of biologically active proteins
US6482613B1 (en) Microbial production of mature human leukocyte interferons
EP0256843A1 (en) Expression of g-csf and muteins thereof and their use
CA1282354C (en) INTERFERON .alpha. 54
CA1282355C (en) INTERFERON .alpha. 61
CA1217440A (en) INTERFERON .alpha. 6L
CA1282353C (en) INTERFERON .alpha. 74
EP0553667A1 (en) Soluble LDL receptor, its production and use
Langer et al. Purification, bacterial expression, and biological activities of the human interferons
BG61060B2 (en) Human immune inteferon
BG60888B2 (en) Human immune interferon

Legal Events

Date Code Title Description
MKLA Lapsed