EP0669981A1 - Interferon tau compositions and methods of use - Google Patents

Interferon tau compositions and methods of use

Info

Publication number
EP0669981A1
EP0669981A1 EP93924958A EP93924958A EP0669981A1 EP 0669981 A1 EP0669981 A1 EP 0669981A1 EP 93924958 A EP93924958 A EP 93924958A EP 93924958 A EP93924958 A EP 93924958A EP 0669981 A1 EP0669981 A1 EP 0669981A1
Authority
EP
European Patent Office
Prior art keywords
seq
interferon
ifnt
cells
polypeptide
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.)
Withdrawn
Application number
EP93924958A
Other languages
German (de)
English (en)
French (fr)
Inventor
Fuller Warren Center For Animal Biotechn. Bazer
Howard Marcellus Dept. Of Microbiology Johnson
Carol Hanlon Department Of Microbiology Pontzer
Troy Lee Department Of Animal Science Ott
Gino Van Heeke
Kazuhiko The Women's Research Institute Imakawa
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 Florida
Women's Research Institute
Original Assignee
University of Florida
Women's Research Institute
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 University of Florida, Women's Research Institute filed Critical University of Florida
Priority to EP03006878A priority Critical patent/EP1360962A3/en
Publication of EP0669981A1 publication Critical patent/EP0669981A1/en
Withdrawn legal-status Critical Current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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/249Interferons
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/61Fusion polypeptide containing an enzyme fusion for detection (lacZ, luciferase)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • C07K2319/75Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor containing a fusion for activation of a cell surface receptor, e.g. thrombopoeitin, NPY and other peptide hormones

Definitions

  • the present invention relates to interferon- ⁇ compo- sitions and methods of use.
  • Mullis, K.B. U. S. Patent No. 4,683,202, issued 28 July 1987.
  • Mullis, K.B. et al., U. S. Patent No. 4,683,195, issued 28 July 1987.
  • ovine trophoblast protein- one (oTP-1)
  • the protein oTP-1 was shown to inhibit uterine secretion of prostaglandin F 2 -alpha, which causes the corpus luteum on the ovary to undergo physiological and endocrinological demise in nonpregnant sheep (Bazer, et al., 1986).
  • oTP-1 has antiluteolytic bio- logical activity.
  • the primary role of oTP-1 was assumed to be associated with the establishment of pregnancy.
  • oTP-1 was subsequently found to (i) exhibit limited homology (50-70%) with interferon alphas (IFN ⁇ ) of vari- ous species (Imakawa, et al . , 1987), and (ii) bind to a Type I interferon receptor (Stewart, et al . , 1987).
  • oTP-1 has several features that distinguish it from IFN ⁇ including the following: oTP-l's role in reproductive biochemistry (other interferons are not known to have any role in the biochemical regulation of reproductive cycles), oTP-l's cellular source — trophoblast cells (IFN ⁇ is derived from lymphocytes cells) , oTP-l's size — 172 amino acids (IFN ⁇ is typically about 166 amino acids) , and oTP-1 is weakly inducible by viruses (IFN ⁇ is highly inducible by viruses) .
  • interferon-tau interferon-tau
  • T trophoblast
  • the interferons have been classified into two dis ⁇ tinct groups: type I interferons, including IFN ⁇ , IFN/3, and IFN ⁇ (also known as IFN ⁇ ll) ; and type II interferons, represented by IFN7 (reviewed by DeMaeyer, et al . ) .
  • IFN ⁇ interferon-tau
  • IFN ⁇ 's has been shown to inhibit various types of cellular proliferation. IFN ⁇ 's are especially useful against hematologic malignancies such as hairy-cell leukemia (Quesada, et al . , 1984). Further, these pro ⁇ teins have also shown activity against multiple myeloma, chronic lymphocytic leukemia, low-grade lymphoma, Kaposi's sarcoma, chronic myelogenous leukemia, renal- cell carcinoma, urinary bladder tumors and ovarian can- cers (Bonnem, et al . , 1984; Oldham, 1985).
  • interferons and interferon receptors have also been investigated (Benoit, et al . , 1993). IFN ⁇ 's are also useful against various types of viral infections (Finter, et al . , 1991).
  • Alpha interfe ⁇ rons have shown activity against human papillomavirus infection, Hepatitis B, and Hepatitis C infections (Finter, et al., 1991; Kashima, et al . , 1988; Dusheiko, et al., 1986; Davis, et al . , 1989).
  • IFN ⁇ 's have been limited by their toxicity: use of interferons in the treatment of cancer and viral disease has resulted in serious side effects, such as fever, chills, anorexia, weight loss, and fatigue (Pontzer, et al., 1991; Oldham, 1985). These side effects often require (i) the interferon dosage to be reduced to levels that limit the effectiveness of treatment, or (ii) the removal of the patient from treat ⁇ ment. Such toxicity has reduced the usefulness of these potent antiviral and antiproliferative proteins in the treatment of debilitating human and animal diseases.
  • the present invention includes a method of inhibiting tumor cell growth.
  • cells are contacted with interferon-T (IFN ⁇ ) at a concen ⁇ tration effective to inhibit growth of the tumor cells.
  • IFN ⁇ interferon-T
  • IFNT can be obtained from a number of sources including cows, sheep, and humans. Two embodiments include the IFNT presented as either SEQ ID NO:2 or SEQ ID NO:4.
  • a number of tumor cells can be targeted for growth inhibi ⁇ tion by IFNr, including but not limited to the following: human carcinoma cells and steroid-affected tumor cells (e .g. , mammary tumor cells).
  • the present invention also includes a method of inhibiting viral replication in a cell.
  • RNA e.g., feline leukemia virus, human immunodeficiency virus, or Hepatitis C Virus
  • DNA e.g., Hepatitis B Virus
  • the human IFNT molecules of the present invention can also be used in a method of enhancing fertility in a female mammal.
  • an amount of human IFNT effective to enhance fertility of a female mammal is administered, typically in a pharmaceutically acceptable carrier.
  • Exemplary of such human IFNT molecules are the protein sequences presented as SEQ ID NO:4 and SEQ ID NO:12.
  • the present invention also includes an isolated nucleic acid which encodes a human interferon- ⁇ . Exem ⁇ plary of such nucleic acid molecules are SEQ ID NO:3 and SEQ ID NO:11. Further, the invention includes expression vectors for the expression of human IFNT.
  • the expression vector includes (a) a nucleic acid containing an open reading frame that encodes human interferon- ⁇ ; and (b) regulatory sequences effective to express said open reading frame in a host cell.
  • the regulatory se ⁇ quence may include sequences useful for targeting or secretion of the IFNT polypeptide: such sequences may be endogenous (such as the normally occurring IFNT leader sequences, see SEQ ID NO:11) or heterologous (such as a secretory signal recognized in yeast or bacterial expres ⁇ sion systems) .
  • regulatory sequences may also include, 5' to said nucleic acid sequence, a promoter region and an ATG start codon in- frame with the interferon- ⁇ coding sequence, and 3' to said coding sequence, a translation termination signal followed by a transcription termination signal.
  • the nucleic acid in the expression vector may be selected from, for example, SEQ ID NO:l, SEQ ID NO:3 and SEQ ID NO:ll.
  • the present invention in ⁇ cludes a recombinantly produced human interferon- ⁇ pro- tein.
  • One exemplary sequence for such a protein is given as SEQ ID NO:4.
  • Human IFNT may also contain a carboxy terminal extension (such as the sequence presented as SEQ ID NO:12) .
  • the invention includes a method of recombinantly producing interferon- ⁇ .
  • a recombinant expression system containing an open reading frame (ORF) having a polynucleotide sequence which encodes a human interferon- ⁇ polypeptide, where the vector is designed to express the ORF in the host is introduced into suitable host cells. The host is then cultured under conditions resulting in the expression of the ORF sequence.
  • ORF open reading frame
  • the human IFNT sequences discussed above are examples of suitable human IFNT polypeptides. Examples of polynucle ⁇ otide coding sequences are SEQ ID NO:3 and SEQ ID NO:11.
  • the invention further includes expression systems useful for the expression of interferon- ⁇ polypeptides. Typically these systems include a host capable of sup ⁇ porting expression of an open reading frame in a selected expression vector, and the selected expression vector containing an open reading frame (ORF) having a polynu- cleotide sequence which encodes a human interferon-T polypeptide.
  • ORF open reading frame
  • Exemplary of sequences that can be utilized in such expression systems are SEQ ID NO:4, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19 and SEQ ID NO:20.
  • the invention also includes isolated interferon- ⁇ polypeptides. These polypeptides are derived from the interferon-T amino acid coding sequence and are between about 15 and 172 amino acids in length. Such polypep- tides can be selected, for example, from the sequences presented as SEQ ID NO:2 and SEQ ID NO:4. Exemplary IFNT-derived polypeptides include, but are not limited to, the following: SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:10, SEQ ID NO:15, SEQ ID NO:17, and SEQ ID NO:20. In another embodiment, the invention includes a method of blocking the binding of alpha-interferon to a cell having an alpha-interferon receptor.
  • the cell is contacted with an interferon-T polypeptide at a concentration effective to allow the binding of inter- feron-T to each alpha-interferon receptor.
  • the cells, having the IFNT polypeptide bound to the receptor, are then exposed to alpha-interferon (IFN ⁇ ) .
  • IFN ⁇ alpha-interferon
  • the IFNT poly ⁇ peptides and IFN ⁇ -derived polypeptides described above are examples of IFNT polypeptides useful in this method.
  • the invention includes a method of blocking the binding of interferon-T to a cell having a interfer ⁇ on-T receptor. In this method, the cell is contacted with an interferon-T-derived polypeptide (e .g.
  • polypeptide is at a concentration effective to allow the binding of the polypeptide to each interferon- -receptor.
  • the cells are then exposed to interferon-T.
  • the invention also includes purified antibodies that are immunoreactive with human interferon-T.
  • the antibod- ies may be polyclonal or monoclonal.
  • Exemplary IFNr polypeptide antigens include, but are not limited, to the following: SEQ ID NO:4, SEQ ID NO:15, SEQ ID NO:17 and SEQ ID NO:20.
  • the present invention also includes the following: interferon- ⁇ -derived polypeptides that have anti-tumor (i .e . , anti-proliferative) activity; interferon-T-derived polypeptides that have anti-viral activity; and hybrid ⁇ - interferon molecules in which the toxicity portion of native IFN ⁇ has been replaced by analogous sequences from IFNT.
  • Figure 1 presents the nucleic acid coding sequence of a synthetic gene of OVIFNT designed to include 19 unique restriction enzyme sites spaced evenly throughout the coding sequence.
  • Figure 2 shows the cloning strategy used for making a synthetic gene encoding OVIFNT.
  • Figure 3 shows a comparison of the predicted protein sequences of a human interferon-T gene and an ovine interferon-T gene. Divergent amino acids are indicated by presentation of the alternative amino acid on the line below the nucleic acid sequences.
  • Figure 4 presents data demonstrating that both
  • Figure 5 presents data demonstrating that rHuIFN ⁇ is cytotoxic and OVIFNT is not.
  • results of one of three replicate experiments are presented as mean % viability ⁇ SD.
  • Figure 6 presents the sequences of polypeptides derived from the IFNT sequence.
  • Figure 7 presents the complete nucleic acid and amino acid sequence of an OVIFNT sequence.
  • Figure 8 presents data supporting the lack of cyto- toxicity, relative to IFN ⁇ , when IFNT is used to treat peripheral blood mononuclear cells.
  • Figure 9 shows the results of treatment of a human cutaneous T cell lymphoma line, HUT 78, with IFNT.
  • Figure 10 shows the results of treatment of a human T cell lymphoma line, H9, with IFNT.
  • Figure 11A presents data for the peptide inhibition, relative to FIV (feline immunodeficiency virus) replica ⁇ tion, of polypeptides derived from OVIFNT with whole OVIFNT.
  • Figure 11B presents data for the peptide inhibi ⁇ tion, relative to HIV (human immunodeficiency virus) replication, of polypeptides derived from OVIFNT with whole OVIFNT.
  • Figure 12 presents data demonstrating the inhibition of the antiviral activity of IFNT by IFN -derived pep ⁇ tides.
  • Figure 13 presents data demonstrating the inhibition by IFNT-derived peptides of OVIFNT antiviral activity.
  • Figure 14 presents data demonstrating the inhibition by IFNT-derived peptides of bovine IFN ⁇ antiviral activi ⁇ ty.
  • Figure 15 presents data demonstrating the inhibition by IFNT-derived peptides of human IFN ⁇ antiviral activi ⁇ ty.
  • Figure 16 presents data evaluating the lack of inhibition by IFNT-derived peptides of bovine IFN7 anti ⁇ viral activity.
  • Figure 17 presents data demonstrating the anti-IFN ⁇ - derived peptide antisera inhibition of the antiviral activity of IFNT.
  • Figure 18 presents data demonstrating the anti-IFN ⁇ - derived peptide antisera inhibition of the binding of radiolabeled IFNT to cells.
  • SEQ ID N0:1 is the nucleotide sequence of a synthet- ic gene encoding ovine interferon-T (OVIFNT) . Also shown is the encoded amino acid sequence.
  • OVIFNT ovine interferon-T
  • SEQ ID NO:2 is an amino acid sequence of a mature OVIFNT protein.
  • SEQ ID NO:3 is a synthetic nucleotide sequence encoding a mature human interferon-T (HUIFNT) protein.
  • SEQ ID NO:4 is an amino acid sequence for a mature HUIFNT protein.
  • SEQ ID NO:5 is the amino acid sequence of fragment 1-37 Of SEQ ID NO:2.
  • SEQ ID NO:6 is the amino acid sequence of fragment 34-64 Of SEQ ID NO:2.
  • SEQ ID NO:7 is the amino acid sequence of fragment 62-92 of SEQ ID NO:2.
  • SEQ ID NO:8 is the amino acid sequence of fragment 90-122 Of SEQ ID NO:2.
  • SEQ ID NO:9 is the amino acid sequence of fragment 119-150 of SEQ ID NO:2.
  • SEQ ID NO:10 is the amino acid sequence of fragment 139-172 of SEQ ID NO:2.
  • SEQ ID NO:11 is the nucleotide sequence of a natural HUIFNT gene with a leader sequence.
  • SEQ ID NO:12 is the predicted amino acid coding sequence of the SEQ ID NO:11.
  • SEQ ID NO:13 is a 25-mer synthetic oligonucleotide according to the subject invention.
  • SEQ ID NO:14 is a 25-mer synthetic oligonucleotide according the subject invention.
  • SEQ ID NO:15 is the amino acid sequence of fragment 1-37 of SEQ ID NO:4.
  • SEQ ID NO:16 is the amino acid sequence of fragment 34-64 of SEQ ID NO:4.
  • SEQ ID NO:17 is the amino acid sequence of fragment 62-92 of SEQ ID NO:4.
  • SEQ ID NO:18 is the amino acid sequence of fragment 90-122 of SEQ ID NO:4.
  • SEQ ID NO:19 is the amino acid sequence of fragment 119-150 of SEQ ID NO:4.
  • SEQ ID NO:20 is the amino acid sequence of fragment 139-172 of SEQ ID NO:4.
  • Interferon-T refers to any one of a family of inter ⁇ feron proteins having the following characteristics: (i) anti-luteolytic properties; (ii) anti-viral properties; (iii) anticellular proliferation properties; (iv) 45-68% amino acid homology with ⁇ -Interferons and greater than 70% amino acid homology to the sequence presented as SEQ ID NO:2. Interferon-T can be isolated from a number of mammalian sources as described below.
  • An interferon-T polypeptide is a polypeptide having between about 15 and 172 amino acids derived from an interferon-T amino acid coding sequence, where said 15 to 172 amino acids are contiguous in native interferon- . Such 15-172 amino acid regions can also be assembled into polypeptides where two or more such interferon-T regions are joined that are normally discontinuous in the native protein.
  • Ovine interferon- ⁇ (OVIFNT) is a major conceptus secretory protein produced by the embryonic trophectoderm during the critical period of maternal recognition in sheep.
  • One isolate of mature OVIFNT is 172 amino acids in length (SEQ ID NO:2).
  • the cDNA coding sequence con ⁇ tains an additional 23 amino acids at the amino-terminal end of the mature protein (Imakawa, et al . , 1987).
  • the coding sequence of this OVIFNT isolate is presented as Figure 7.
  • OVIFNT Conceptuses were collected from pregnant sheep and cultured in vitro in a modified Minimum Essential Medium as described previously (Godkin, et al . , 1982). Conceptuses were collected on various days of pregnancy with the first day of mating being described as Day 0. IFNT was purified from conceptus culture medium essentially as described by Vallet, et al . , (1987) and Godkin, et al. (1982).
  • IFNT The homogeneity of IFNT was assessed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS- PAGE; Maniatis, et al . ; Ausubel, et al . ) . Determination of protein concentration in purified IFNT samples was performed using the bicinchoninic (BCA) assay (Pierce Chemical Co., Rockford, IL; Smith, et al . , 1985).
  • BCA bicinchoninic
  • a homologous protein to OVIFNT was isolated from cows (bIFN ⁇ ; Helmer, et al., 1987; Imakawa, et al . , 1989) .
  • OVIFNT and BOIFNT (i) have similar functions in maternal recognition of pregnancy, and (ii) share a high degree of amino acid and nucleotide sequence homology between mature proteins.
  • the nucleic acid sequence homology between OVIFNT and BOIFNT is 76.3% for the 5' non-coding region, 89.7% for the coding region, and 91.9% for the 3' non-coding region.
  • the amino acid sequence homology is 80.4%.
  • Example 1 describes the reproductive functions of OVIFNT.
  • OVIFNT and recombinant human ⁇ -2-Interferon (rHuIFN ⁇ 2 ) were infused into uterine lumen of ewes at a variety of concentrations.
  • the life span of the corpus luteum was assessed by examination of interestrous inter ⁇ vals, maintenance of progesterone secretion, and inhibi ⁇ tion of prostaglandin secretion (Davis, et al . , 1992 ) .
  • Comparison of the data resulting from these examinations demonstrated a considerable lengthening of the inter ⁇ estrous interval when IFNT is administered at lOO ⁇ g/day and no meaningful effect when rHuIFN ⁇ is administered.
  • Example 2 The antiviral properties of interferon-T at various stages of the reproductive cycle were also examined (Example 2) .
  • Conceptus cultures were established using conceptus obtained from sheep at days 12 through 16 of the estrus cycle. Antiviral activity of supernatant from each conceptus culture was assessed. Culture superna ⁇ tants had increasing antiviral activity associated with advancing development of the conceptus up to Day 16 post estrus.
  • IFNT Recombinant Production of IFNT.
  • Recombinant IFNT was produced using bacterial and yeast cells.
  • the amino acid coding sequence for OVIFNT was used to generate a corresponding DNA coding sequence with codon usage optimized for expression in E. coli
  • Example 3 The DNA coding sequence was synthetically constructed by sequential addition of oligonucleotides. Cloned oligonucleotides were fused into a single polynu- cleotide using the restriction digestions and ligations outlined in Figure 2. The polynucleotide coding sequence had the sequence presented as SEQ ID NO:l.
  • this synthetic coding sequence can be placed in a number of bacterial expression vectors: for example, lambda gtll (Promega, Madison WI) ; pGEX (Smith, et al . ) ; pNH vectors (Strata- gene, La Jolla CA) .
  • Cloning of the IFNT synthetic poly ⁇ nucleotide into a modified pIN III omp-A expression vector is described in Example 3. Production of the IFNT protein was induced by the addition of IPTG. Soluble recombinant IFNT was liberated from the cells by sonica- tion or osmotic fractionation.
  • the protein can be further purified by standard methods, including size fractionation (column chromatog- raphy or preoperative gel electrophoresis) or affinity chromatography (using, for example, anti-IFN ⁇ antibodies (solid support available from Pharmacia, Piscataway NJ) . Protein preparations can also be concentrated by, for example, filtration (A icon, Danvers, Mass.).
  • the synthetic IFNT gene was also cloned into the yeast cloning vector pBS24Ub (Example 4; Sabin, et al . ; Ecker, et al . ) . Synthetic linkers were constructed to permit in-frame fusion of the IFNT coding sequences with the ubiquitin coding sequences in the vector. The re- suiting junction allowed in vivo cleavage of the ubiqui ⁇ tin sequences from the IFNT sequences.
  • the recombinant plasmid pBS24Ub-IFN ⁇ was transformed into the yeast S. cerevisiae .
  • Transformed yeast cells were cultured, lysed and the recombinant IFNT (r-IFN ⁇ ) protein isolated from the cell lysates.
  • the amount of r-IFN ⁇ was quantified by radioimmuno- assay. Microsequencing of the purified r-IFN ⁇ was car ⁇ ried out. The results demonstrated identity with native IFNT through the first 15 amino acids. The results also confirmed that the ubiquitin/IFN ⁇ fusion protein was correctly processed in vivo.
  • Recombinant IFNT obtained by this method exhibited antiviral activity similar to the antiviral activity of IFNT purified from conceptus-conditioned culture medium.
  • yeast vectors can be used in the practice of the present invention including, but are not limited to, vectors with regulatable expression (Hitzeman, et al . ; Rutter, et a . ; Oeda, et al.) .
  • the yeast transformation host is typically Saccharomyces cerevisiae, however, other yeast suitable for transformation can be used as well (e .g. , Schiz ⁇ saccharomyce ⁇ pombe) .
  • the DNA encoding the IFNT polypeptide can be cloned into any number of commercially available vectors to generate expression of the polypeptide in the appropriate host system.
  • These systems include the above described bacterial and yeast expression systems as well as the following: baculovirus expression (Reilly, et al . ; Beames, et al . ; Clontech, Palo Alto CA) ; and expression in mammalian cells (Clontech, Palo Alto CA; Gibco-BRL,
  • polypeptides can be expressed as fusion proteins or as native proteins.
  • a number of features can be engineered into the expression vectors, such as leader sequences which promote the secretion of the expressed sequences into culture medium.
  • the recombinantly produced polypeptides are typically isolated from lysed cells or culture media. Purification can be carried out by methods known in the art including salt fractionation, ion exchange chromatography, and affinity chromatography. Immunoaffinity chromatography can be employed, as described above, using antibodies generated based on the IFNT polypeptides.
  • B. Human Interferon-T can be employed, as described above, using antibodies generated based on the IFNT polypeptides.
  • DNA was screened for sequences homologous to inter- feron-T (Example 5) .
  • Several sequences that hybridized with the OVIFNT CDNA probe were identified.
  • Several clones containing partial sequences of human interferon- ⁇ were then isolated (Example 6) .
  • Two synthetic 25-mer oligonucleotides, corresponding to sequences from the OVIFNT CDNA (Imakawa, et al . , 1987) were synthesized. These primers were employed in amplification reactions using DNA derived from the following two cDNA libraries: human term placenta and human term cytotrophoblast.
  • the resulting amplified DNA fragments were electrophoreti- cally separated and a band containing an IFNT amplifica ⁇ tion product was isolated.
  • the product was subcloned and the inserted amplification product sequenced using the dideoxy termination method.
  • Example 7 describes the isolation of a full-length human IFNT gene.
  • High molecular weight DNA was isolated from peripheral blood mononuclear cells (PBMCs) and size- fractionated. Fractions were tested for the presence of IFNT sequences using polymerase chain reaction: DNA molecules from fractions that tested amplification posi- tive were used to generate a subgenomic library in ⁇ gtll.
  • This subgenomic library was plated and hybridized with an OVIFNT CDNA probe (Example 7A) . Approximately 20 clones were identified that hybridized to the probe. Plaques corresponding to the positive clones were pas- saged, DNA isolated and analyzed by amplification reac- tions using OVIFNT primers. Of these twenty plaques, six plaques generated positive PCR signals. The phage from these six clones were purified and the inserts sequenced. One of the inserts from one of these six clones was used as a hybridization probe in the following screening.
  • Recombinant phage from the ⁇ gtll subgenomic library were screened using the hybridization probe just de ⁇ scribed (Example 7B) .
  • Three clones giving positive hybridization signals were isolated and the inserts sequenced.
  • the resulting nucleic acid sequence informa ⁇ tion is presented as SEQ ID NO:11 and the predicted protein coding sequence is presented as SEQ ID NO:12.
  • the predicted mature protein coding sequence is presented as SEQ ID NO:4.
  • Comparison of the predicted protein sequences (Fig ⁇ ure 3) of the human interferon-T gene (SEQ ID NO:4) and the ovine interferon-T gene demonstrates the levels of sequence homology and divergence at the amino acid level.
  • the human IFNT sequences presented as SEQ ID NO:12 and SEQ ID NO:11, and primers and probes derived there ⁇ from, can be used as specific probes to detect isolates of further human IFNT coding sequences and/or pseudo- genes. Further, there may be more than one isofor of the IFNT protein and more than one coding sequence per species.
  • the specific nucleic acid probes used in the practice of the present invention and antibodies reactive with the IFNT polypeptides of the present invention may be useful to isolate unidentified variants of interferon- T in mammals, according to the methods of the invention disclosed herein. 2. Characterization of the Expression of Interfe- ron- ⁇ in Human Tissues.
  • Human placental cDNA libraries and an ovine cDNA library were analyzed by hybridization to the OVIFNT CDNA probe (Example 8) .
  • This DNA hybridization analysis sug ⁇ gested that the IFN ⁇ -signals from human cDNA libraries were approximately 1/100 of the signal obtained using the ovine cDNA library.
  • OVIFNT CDNAS constitute around 0.4% of the ovine cDNA library. Accordingly, the abundance of human cDNAs responding to the OVIFNT probe appears to be low, at least in the term placenta from which the cDNA libraries were generated.
  • HUIFNT mRNA The presence of HUIFNT mRNA in human term placenta and amniocytes were also analyzed. The results suggest the presence of human IFNT mRNA in the feto-placental annex. The aminocytes also expressed the messages corre ⁇ sponding to OVIFNT primers and human probe, suggesting that the expression of IFNT mRNA is not limited to the term placenta.
  • a RT-PCR analysis for the presence of HUIFNT was applied to the total cellular RNA isolated from human adult lymphocytes: the results suggest that IFNT mRNA exists in lymphocytes.
  • interferon-T The expression of interferon-T in human tissue was also examined using in situ hybridization (Example 9) .
  • Sections from four healthy, different term and first tri ⁇ mester human placentas were examined. This analysis em ⁇ ployed a cDNA probe derived from the OVIFNT CDNA sequen ⁇ ces (Example 9B) . In situ hybridization was performed using an anti-sense RNA probe. In three separate experi ⁇ ments, specific hybridization was observed in all term and first trimester placental tissues.
  • First trimester placental villi (composed of an outer layer of syncytiotrophoblast, an underlying layer of cytotrophoblast, and a central stromal region with various types of mesenchymal cells) displayed the highest transcript level of IFNT in the cytotrophoblast cells. Less intense but detectable levels were present in both the syncytiotrophoblast and stromal cells. A similar pattern of transcript expression was demonstrated in the placental villi of term tissue but the level of signal detection was low. First trimester extravillous tropho- blasts displayed the highest amount of message and stained positive when present in the maternal blood spaces.
  • the present results demonstrate that the human IFNT gene is highly expressed in early placental tissues by migrating extravillous trophoblasts, but is also ex ⁇ pressed in villous syncytiotrophoblasts, cytotropho- blasts, and various stromal cells. These results demon ⁇ strate the detection of IFNT transcripts in human preg ⁇ nancy tissues, and IFNT expression in the villous cyto- trophoblasts as well as the extravillous trophoblast of first trimester placenta.
  • IFNT Antiviral Properties of Interferon-T.
  • the antiviral activity of IFNT has been evaluated against a number of viruses, including both RNA and DNA viruses.
  • IFNT had a higher specific antivi ⁇ ral activity than either rBoIFN ⁇ or rBoIFN ⁇ (Example 10, Table 3) .
  • IFNT has potent antiviral activity with limited cytotoxic effects.
  • Highly purified OVIFNT was tested for anti- retroviral and cytotoxic effects on peripheral blood lymphocytes exposed to feline AIDS and human AIDS retro- viruses (Bazer, F.W., et al., (1989)).
  • This feline AIDS lentivirus produces a chronic AIDS-like syndrome in cats and is a model for human AIDS (Pederson, et al . , 1987).
  • Replication of either virus in peripheral blood lympho ⁇ cytes (PBL) was monitored by reverse transcriptase (RT) activity in culture supernatants over time.
  • RT reverse transcriptase
  • RNA-dependent DNA polymerase RT activity was assayed in FIV- and HIV-infected feline and human PBL cultures treated with IFNT (Example 11) . Replication of FIV was reduced to about one-third of control values when cells were cultured in the presence of IFNT. Addition of OVIFNT produced a rapid, dose-dependent decrease in reverse transcriptase (RT) activity (Example 11, Table 4). While concentrations as low as 0.62 ng/ml of IFNT inhibited viral replication, much higher concentrations (40 ng/ml) having greater effects on RT-activity were without toxic effects on the cells.
  • RT reverse transcriptase
  • IFNT appeared to exert no cytotoxic effect on the cell ⁇ hosting the retrovirus. This was true even when IFNT was present at 40 ng per ml of culture medium. This concentration of IFNT is equivalent to about 8,000 anti ⁇ viral units of alpha interferon, when IFNT is assayed for its ability to protect Madin-Darby bovine kidney cells from lysis by vesicular stomatitis virus as described by Pontzer, et al . (1988).
  • IFNT was also tested for activity against HIV repli ⁇ cation in human cells.
  • Human peripheral lymphocytes, which had been infected with HIV were treated with vary ⁇ ing concentrations of IFNT (Example 12) .
  • Replication of HIV in peripheral blood lymphocytes was monitored by reverse transcriptase activity in culture supernatants over time. Over a range of concentrations of IFNT pro- prised significant anti-HIV effects (Example 12, Table 5).
  • a concentration of only 10 ng/ml resulted in over a 50% reduction in RT activity after only six days;
  • a concen ⁇ tration of 500 ng/ml resulted in a 90% reduction in RT activity within 10 days.
  • there was no evidence of any cytotoxic effects attributable to the administra ⁇ tion of IFNT (Example 12, Table 5).
  • IFNT antiviral effects of IFNT against HIV were evaluated by treating human PBMC cells with various amounts of either recombinant IFNT or recombinant human IFN ⁇ 2 at the time of infection with HIV (Example 18) .
  • the data from these experiments (Example 18, Table 12) sup ⁇ port the conclusion that, at similar concentrations, IFN ⁇ and IFNT are effective in reducing the replication of HIV in human lymphocytes.
  • treatment of cells with IFN ⁇ 2 resulted in cytoxicity, whereas no such cytotoxity was observed with treatment using IFNT, even when IFNT was used at much higher concentrations. No cytotoxicity was observed using IFNT, even when IFNT was used at 200 times the dosage of interferon-alpha II.
  • the hepatocyte was examined for the effects of IFN ⁇ and IFNT on hepatospeci- fic mRNA production (Example 18) .
  • IFNT is an effective antiviral agent against a wide variety of viruses, in ⁇ cluding both RNA and DNA viruses.
  • IFNT D. Antiproliferative Properties of IFNT.
  • the effects of IFNT on cellular growth have also been examined.
  • anti-cellular growth activity was examined using a colony inhibition assay (Example 13) .
  • Human amnion (WISH) or MDBK cells were plated at low cell densities to form colonies originating from single cells. Dilutions of interferons were added to triplicate wells and the plates were incubated to allow colony formation.
  • IFNT inhibited both colony size and number in these assays.
  • IFNT was more effective at inhibiting cell proliferation of the human cell line (WISH) than human IFN ⁇ .
  • the antiproliferative activity of IFNT was dose-dependent. High concentrations of IFNT stopped proliferation, while cell viability was not impaired.
  • IFNT appears to inhibit progress of cells through S phase. These results demonstrate the antiproliferative effect of IFNT, and underscore its low cytotoxicity.
  • the antiproliferative effects of IFNT were also studied for rat and bovine cell lines (Example 14) .
  • the rate of 3 H-thymidine incorporation was used to assess the rate of cellular proliferation.
  • IFNT The antiproliferative activity and lack of toxicity of IFNT was further examined using a series of human tumor cell lines (Example 15) .
  • a variety of human tumor cell lines were selected from the standard lines used in NIH screening procedure for antineoplastic agents (Pontzer, C.H., et al., (1991)). At least one cell line from each major neoplastic category, was examined.
  • HL-60 human promyelocytic leukemia
  • H9 human T cell lymphoma HUT 78 human cutaneous T cell lymphoma;
  • MCF7 human breast adenocarcinoma As above, the antiproliferative activity was evalu ⁇ ated by measuring the rate of 3 H-thymidine incorporation into cells which havt* been treated with IFNT. Signifi ⁇ cant differences between treatments were assessed by an analysis of variance followed by Scheffe's F-test. Cell cycle analysis was performed by flow cytometry.
  • IFNT inhibition of MCF7 (breast adenocarcinoma) proliferation demonstrated that IFNT reduced MCF7 proliferation in a dose-dependent manner.
  • a 50% reduction in 3 H-thymidine was observed with 10,000 units/ml of IFNT (Example 15, Table 8) . This cell line had previously been found to be unresponsive to anti- estrogen treatment.
  • IFNT and IFN ⁇ were equally efficacious antitumor agents.
  • SK-MEL-28 inhibition of proliferation by IFN ⁇ was accomplished by a 13% drop in viability, while IFNT was not cytotoxic.
  • IFNT is equal or preferable to IFN ⁇ as an antineoplastic agent against human tumors.
  • IFNT exhibits antiproliferative activity against human tumor cells without toxicity and is as potent or more potent than human IFN ⁇ .
  • Clinical trials of the IFN ⁇ 2s have shown them to be effective antitumor agents (Dianzani, F., 1992; Krown, 1987).
  • One therapeutic advantage of IFNT as a therapeutic is the elimination of toxic effects seen with high doses IFN ⁇ s.
  • IFNT IFNT-induced tumors like Kaposi's sarcoma (associated with HIV infec- tion) where the antineoplastic effects of IFNT are cou ⁇ pled with IFNT ability to inhibit retroviral growth.
  • B16-F10 is a syngeneic mouse transplantable tumor selected because of its high incidence of pulmonary metastases (Poste, et al . , 1981). Interferon treatment was initiated 3 days after the introduction of the tumor cells. The in vivo administration of IFNT dramatically reduced B16-F10 pulmonary tumors. Thus, IFNT appears to be an effica- cious antineoplastic agent in vivo as well as in vitro . III. Interferon-T Polypeptide Fragments. Protein Modeling and Protein Modifications.
  • IFNT activity has been examined using six overlapping synthet ⁇ ic peptides corresponding to the entire OVIFNT sequence ( Figure 6) .
  • the corresponding polypeptides derived from the ovine IFNT sequence are presented as SEQ ID NO:15 to SEQ ID NO:20.
  • Three peptides representing amino acids l- 37, 62-92 and 139-172 have been shown to inhibit IFNT antiviral activity (Example 17) .
  • the peptides were effective competitors at concentrations of 300 ⁇ M and above.
  • the two synthetic peptides OVIFNT(1-37) and OVIFNT(139-172) also blocked OVIFNT anti-FIV and anti-HIV activity (Example 17; Figures 11A and 11B) . While both peptides blocked FIV RT activity, only the C-terminal peptide, OVIFNT(139-172) , appeared to be an efficient inhibitor of vesicular stomatitis virus activity on the feline cell line, Fc9.
  • the above data taken together suggest that the C- terminal regions of type I interferons may bind to common site on the type I interferon receptor, while the N- terminal region may be involved in the elicitation of unique functions.
  • portions of the IFNT interferon molecule may be used to substitute regions of interferon alpha molecules.
  • the region of an interferon alpha molecule that is responsi ⁇ ble for increased cytotoxicity, relative to IFNT treat- ment can be identified by substituting polypeptide regions derived from IFNT for regions of an interferon alpha molecule.
  • substitutions can be carried out by manipulation of synthetic genes (see below) encoding the selected IFNT and interferon alpha molecules, coupled to the functional assays described herein (such as, antivi ⁇ ral, antiproliferative and cytoxicity assays) .
  • IFNT The antiproliferative activity of IFNT (Example 17, Table 11) involved a further region of the molecule, since IFNT(119-150) was the most effective inhibitor of OvIFNT-induced reduction of cell proliferation. This results suggests that the region of the molecule primari ⁇ ly responsible for inhibition of cell growth is the IFNT(119-150) region.
  • This region of the IFNT molecule may be useful alone or fused to other proteins (such as serum albumin, an antibody or an interferon alpha poly- peptide) as an antineoplastic agent.
  • a conjugated pro ⁇ tein between an N-terminal peptide derived from human interferon- ⁇ and serum albumin was shown to have anticel ⁇ lular proliferation activity (Ruegg, et al .
  • CD circular dichroism
  • the final modeling step was to apply the IFN/3 x-ray crystallographic coordinates of the IFN/3 carbon backbone to the IFNT sequence.
  • the functionally active domains of IFNT, identified above, were localized to one side of the molecule and found to be in close spatial proximity. This is consistent with multiple binding sites on IFNT interacting simultaneously with the type I IFN receptor.
  • the three dimensional modeling data coupled with the function data described above, provides the ability to introduce sequence variations into specific regions of IFNT to generate enhancement of selected functions (e.g., antiviral or anticellular proliferation) or the ability to substitute a region(s) of selected function into other interferon molecules (e .g. , antiviral, antineoplastic, or reduced cytotoxicity) .
  • Example 3 The construction of a synthetic gene for OVIFNT is described in Example 3. Briefly, a consensus amino sequence was back-translated using optimal codon usage for E. coli . The sequence was edited to include 20, unique, restriction sites spaced throughout the length of the construct. This 540 base pair synthetic gene se ⁇ quence was divided into 11 oligonucleotide fragments. Individual fragments were synthesized and cloned, either single or double stranded, into either pTZ 19R, pTZ 18R or pBluescript, amplified and fused. The synthetic OVIFNT construct was then cloned into a modified pIN-IIl- ompA expression vector for expression in bacteria and also cloned into a yeast expression plasmid. A similarly constructed human IFNT synthetic gene (SEQ ID NO:3) has been designed, constructed and expressed in yeast cells.
  • SEQ ID NO:3 A similarly constructed human IFNT synthetic gene (SEQ ID NO:3) has been designed, constructed and expressed in yeast cells.
  • OVIFNT synthetic gene in yeast allowed over production of recombinant IFNT in S. cerevisiae : large quantities (5-20 mg/1) of recom ⁇ binant IFNT can be purified from soluble yeast extract . using sequential ion exchange and molecular sieve chroma ⁇ tography. Recombinant IFNT purified in this fashion exhibited potent antiviral activity (2 to 3 X 10 8 units/mg) similar to native OVIFNT.
  • the synthetic gene construct facilitates introduc ⁇ tion of mutations for possible enhancement of antitumor (anticellular proliferative) and antiviral activities. Further, the disparate regions of the molecule responsi- ble for different functions allows for separate manipula ⁇ tion of different functions. For example, two deletion mutants, OVIFN (1-155) and OVIFNT(1-166) , have been constructed to examine the role of carboxy terminal sequences in IFNr molecules. Additional mutant IFNT molecules have been con ⁇ structed to identify residues critical for antiprolifera ⁇ tive activity. For example, one particular residue, Tyr 123 has been implicated in the anticellular proliferative activity of IFN ⁇ (Mclnnes, et al . , 1989). The equivalent of Tyr 123 in IFNT is contained within peptide
  • OVIFNT(119-150) this polypeptide inhibits OVIFNT and human IFN ⁇ antiproliferative activity. Mutations con ⁇ verting Tyr 123 to conservative (Trp) and nonconservative (Asp) substitutions have been generated, as well as mutant sequences having deletion of this residue. The codon for Tyr 123 is located within an Sspl site; elimi ⁇ nation of this site has been used for screening. The antiproliferative activity of these mutant IFNT is evalu- ated as described herein.
  • Synthetic peptides can be generated which correspond to the IFNT polypeptides of the present invention. Synthetic peptides can be commercially synthesized or prepared using standard methods and apparatus in the art (Applied Biosystems, Foster City CA) .
  • oligonucleotide sequences encoding peptides can be either synthesized directly by standard methods of oligonucleotide synthesis, or, in the case of large coding sequences, synthesized by a series of clon- ing steps involving a tandem array of multiple oligonu ⁇ cleotide fragments corresponding to the coding sequence (Crea; Yoshio et al . ; Eaton et al . ) . Oligonucleotide coding sequences can be expressed by standard recombinant procedures (Maniatis et al . ; Ausubel et al . ) . The biological activities of the interferon- ⁇ poly ⁇ peptides described above can be exploited using either the interferon-T polypeptides alone or conjugated with other proteins (see below) .
  • the present invention includes interferon-T or interferon-T-derived polypeptides cova ⁇ lently attached to a second polypeptide to form a fused, or hybrid, protein.
  • the interferon-T sequences making up such fused proteins can be recombinantly produced inter ⁇ feron-T or a bioactive portion thereof, as described above.
  • the polypeptides presented as SEQ ID NO:10 and SEQ ID NO:20 may be advantageously fused with a soluble peptide, such as, serum albumin, an antibody (e .g. , specific against an virus-specific cell surface antigen) , or an interferon alpha polypeptide.
  • a soluble peptide such as, serum albumin, an antibody (e .g. , specific against an virus-specific cell surface antigen)
  • interferon alpha polypeptide include (i) replacing toxici- ty-associated regions of interferon- ⁇ with the interfer ⁇ on-T regions SEQ ID NO:5 and SEQ ID NO:15, and (ii) fusion proteins containing the interferon-T regions SEQ ID NO:9 and SEQ ID NO:19 as anticellular proliferation agents.
  • the fused proteins of the present invention may be formed by chemical conjugation or by recombinant tech ⁇ niques.
  • the interferon- ⁇ and second selected polypeptide are modified by conventional coupling agents for covalent attachment.
  • serum albumin is derivatized with N- succinimidyl-S-acetyl thioacetate (Duncan) , yielding thiolated serum albumin.
  • the activated serum albumin polypeptide is then reacted with interferon-T derivatized with N-succinimidyl 3-(2-pyridyldithio) propionate (Cum ⁇ ber) , to produce the fused protein joined through a disulfide linkage.
  • recombinant interferon-T may be prepared with a cysteine residue to allow disul- fide coupling of the interferon-T to an activated ligand, thus simplifying the coupling reaction.
  • An interferon- ⁇ expression vector, used for production of recombinant interferon-T can be modified for insertion of an inter ⁇ nal or a terminal cysteine codon according to standard methods of site-directed mutagenesis (Ausubel, et al . ) .
  • a fused protein is prepared recombi ⁇ nantly using an expression vector in which the coding sequence of a second selected polypeptide is joined to the interferon-T coding sequence.
  • human serum albumin coding sequences can be fused in-frame to the coding sequence of an interferon-T polypeptide, such as, SEQ ID NO:9.
  • the fused protein is then expressed using a suitable host cell.
  • the fusion protein may be purified by molecular-sieve and ion-exchange chromatogra- phy methods, with additional purification by polyacryl- amide gel electrophoretic separation and/or HPLC chroma ⁇ tography, if necessary.
  • interfer- on-T-containing fusion proteins may be prepared.
  • One variation on the above fusion is to exchange positions of the interferon-T and selected second protein molecules in the fusion protein (e.g., carboxy terminal versus amino terminal fusions) .
  • internal portions of a native interferon-T polypeptide for example, amino acid regions of between 15 and 172 amino acids
  • two or more such interferon- T portions are contiguous that are normally discontinuous in the native protein.
  • Fusion proteins containing the polypeptide antigens of the present invention fused with the glutathione-S- transferase (Sj26) protein can be expressed using the pGEX-GLI vector system in E. coli JM101 cells.
  • the fused Sj26 protein can be isolated readily by glutathione substrate affinity chromatography (Smith) . Expression and partial purification of IFNT proteins is described in (Example 20) , and is applicable to any of the other soluble, induced polypeptides coded by sequences de- scribed by the present invention.
  • Insoluble GST (sj26) fusion proteins can be puri ⁇ fied by preparative gel electrophoresis.
  • IFN ⁇ -?-galactosidase fusion proteins can be isolated as described in Example 19.
  • an expression vector such as the lambda gtll or pGEX vectors described above, containing IFNT coding sequences and expression control elements which allow expression of the coding regions in a suitable host.
  • the control elements gener ⁇ ally include a promoter, translation initiation codon, and translation and transcription termination sequences, and an insertion site for introducing the insert into the vector.
  • the DNA encoding the desired polypeptide can be cloned into any number of vectors (discussed above) to generate expression of the polypeptide in the appropriate host system. These recombinant polypeptides can be expressed as fusion proteins or as native proteins.
  • a number of features can be engineered into the expression vectors, such as leader sequences which promote the secretion of the expressed sequences into culture medium.
  • Recombinantly produced IFNT, and polypeptides derived therefrom are typically isolated from lysed cells or culture media. Purification can be carried out by meth ⁇ ods known in the art including salt fractionation, ion exchange chromatography, and affinity chromatography. Immunoaffinity chromatography can be employed using antibodies generated against selected IFNT antigens.
  • the invention includes specific antibodies directed against the polypeptides of the present invention. Typically, to prepare antibodies, a host animal, such as a rabbit, is immunized with the purified antigen or fused protein antigen.
  • Hybrid, or fused, proteins may be generated using a variety of coding sequences derived from other proteins, such as ⁇ - galactosidase or glutathione-S-transferase.
  • the host serum or plasma is collected following an appropriate time interval, and this serum is tested for antibodies specific against the antigen.
  • Example 20 describes the production of rabbit serum antibodies which are specific against the IFNT antigens in a Sj26/IFN ⁇ hybrid protein. These techniques can be applied to the all of the IFNT molecules and polypeptides derived therefrom.
  • the gamma globulin fraction or the IgG antibodies of immunized animals can be obtained, for example, by use of saturated ammonium sulfate or DEAE Sephadex, or other techniques known to those skilled in the art for produ ⁇ cing polyclonal antibodies.
  • purified protein or fused protein may be used for producing monoclonal antibodies.
  • the spleen or lymphocytes from a animal immunized with the selected polypeptide antigen are removed and immortalized or used to prepare hybridomas by methods known to those skilled in the art (Harlow, et al . ) . Lym ⁇ phocytes can be isolated from a peripheral blood sample.
  • Epstein-Barr virus can be used to immortalize human lymphocytes or a fusion partner can be used to produce hybridomas.
  • Antibodies secreted by the immortalized cells are screened to determine the clones that secrete antibodies of the desired specificity, for example, by using the ELISA or Western blot method (Ausubel et al . ) .
  • Experi ⁇ ments performed in support of the present invention have yielded four hybridomas producing monoclonal antibodies specific for ovine IFNT have been isolated.
  • Antigenic regions of polypeptides are generally relatively small, typically 7 to 10 amino acids in length. Smaller fragments have been identified as anti- genie regions. Interferon-T polypeptide antigens are identified as described above. The resulting DNA coding regions can be expressed recombinantly either as fusion proteins or isolated polypeptides.
  • amino acid sequences can be conve- niently chemically synthesized (Applied Biosystems, Foster City CA) .
  • Antigens obtained by any of these methods may be directly used for the generation of anti ⁇ bodies or they may be coupled to appropriate carrier molecules. Many such carriers are known in the art and are commercially available (e .g. , Pierce, Rockford IL) .
  • Antibodies reactive with IFNT are useful, for exam ⁇ ple, in the analysis of structure/function relationships.
  • IFNT bears some similarity to the IFN ⁇ family based on structure and its potent antiviral prop ⁇ erties, the IFN ⁇ s do not possess the reproductive proper ⁇ ties associated with IFNT. Also, recombinant bovine IFN ⁇ has little or no effect on interestrous interval compared to IFNT (Davis, et al., 1992).
  • IFNT has some structural simi ⁇ larities to other interferons, it has very distinctive properties of its own: for example, the capability of significantly influencing the biochemical events of the estrous cycle.
  • the human IFNT of the present invention can be used in methods of enhancing fertility and prolonging the life span of the corpus luteum in female mammals as generally described in Hansen, et al . , herein incorporated by reference. Further, the human interferon-T of the pres ⁇ ent invention could be used to regulate growth and devel ⁇ opment of uterine and/or fetal-placental tissues. The human IFNT is particularly useful for treatment of hu- mans, since potential antigenic responses are less likely using such a same-species protein.
  • IFNT The antiviral activity of IFNT has broad therapeutic applications without the toxic effects that are usually 40 associated with IFN ⁇ s. Although the presence of IFNr in culture medium inhibited reverse transcriptase activity of the feline immunodeficiency virus (Example 11) , this is not due to a direct effect of IFNT on the FIV. Rath- 5 er, IFNT appears to induce the host cell to produce a factor(s) which is inhibitory to the reverse transcrip ⁇ tase of the virus.
  • Formulations comprising the IFNT compounds of the present invention can be used to inhibit viral replica ⁇ tion.
  • the human IFNT of the present invention can be any suitable IFNT of the present invention.
  • the human interferon-T is particularly useful for treatment of humans, since potential antigenic
  • IFNT exhibits potent anticellular activity. IFNT can also be used to inhibit cellular growth without the 30 negative side effects associated with other interferons which are currently known. Formulations comprising the IFNT compounds of the subject invention can be used to inhibit, prevent, or slow tumor growth.
  • IFNT can suppress estrogen receptor numbers. Therefore, IFNT can be used in the treatment or prevention of estrogen-dependent tumors.
  • IFNT appears to interact with the Type I IFN recep ⁇ tor via several epitopes on the molecule, and these regions either separately or in combination may affect distinct functions of IFNT differently.
  • polypeptides of the present invention are useful for the selective inhibition of binding of interferons to the interferon receptor. Specifically, as described herein, certain of the disclosed peptides selectively inhibit the antiviral activity of IFNT while others inhibit the antiproliferative activity. Combinations of these peptides could be used to inhibit both activities.
  • these peptides despite binding to the interferon recep- tor and blocking IFNT activity, these peptides do not, themselves, elicit the antiviral or antiproliferative activity.
  • polypeptides can be used as immuno- regulatory molecules when it is desired to prevent immune responses triggered by interferon molecules.
  • These peptides could be used as immunosuppressants to prevent, for example, interferon-mediated immune responses to tissue transplants.
  • Other types of interferon mediated responses may also be blocked, such as the cytotoxic effects of alpha interferon.
  • IFNT proteins can be formulated according to known methods for preparing pharmaceutically useful composi- tions. Formulations comprising interferons or interfer- on-like compounds have been previously described (for example, Martin, 1976) . In general, the compositions of the subject invention will be formulated such that an effective amount of the IFNT is combined with a suitable carrier in order to facilitate effective administration of the composition.
  • compositions used in these therapies may also be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills,, powders, liquid solutions or suspensions, liposomes, suppositories, injectable, and infusible solutions. The preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions also preferably include conventional pharma- ceutically acceptable carriers and adjuvants which are known to those of skill in the art.
  • the compositions of the invention are in the form of a unit dose and will usually be administered to the patient one or more times a day.
  • IFNT IFNT, or related polypeptides
  • IFNT may be administered to a patient in any pharmaceutically acceptable dosage form, including intravenous, intramuscular, intrale- sional, or subcutaneous injection.
  • compo ⁇ sitions and methods used for other interferon compounds can be used for the delivery of these compounds.
  • IFNT interferon-containing interferon
  • IFNT can be administered at rates from about 5 x 10 4 to 20 x 10 6 units/day to about 500 x 10 6 units/day or more. In a preferred embodiment, the dosage is about 10 6 units/day. High doses are preferred for systemic admin ⁇ istration. It should, of course, be understood that the compositions and methods of this invention may be used in combination with other therapies.
  • a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administra ⁇ tion, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained. When the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • compositions of the subject invention can be administered through standard procedures to treat a variety of cancers and viral diseases including those for which other interferons have previously shown activity. See, for example, Finter, et al . (1991); Dianzani, et al . (1992); Francis, et al . (1992) and U.S. Patent Nos. 4,885,166 and 4,975,276.
  • the compositions of the subject invention have unique features and advantages, including their ability to treat these conditions without toxicity.
  • disorders of the skin can be treated intralesionally using IFNT, wherein formulation and dose will depend on the method of administration and on the size and severity of the lesion to be treated.
  • Preferred methods include intradermal and subcutaneous injection. Multiple injec ⁇ tions into large lesions may be possible, and several lesions on the skin of a single patient may be treated at one time.
  • the schedule for administration can be deter ⁇ mined by a person skilled in the art. Formulations designed for sustained release can reduce the frequency of administration.
  • Systemic Treatment is essentially equivalent for all applications. Multiple intravenous or subcutaneous doses are possible, and in the case of implantable methods for treatment, formulations designed for sustained release are particularly useful. Patients may also be treated using implantable subcutaneous portals, reservoirs, or pumps.
  • Regional treatment with the IFNT polypeptides of the present invention is useful for treatment of cancers in specific organs.
  • Treatment can be accomplished by intra- arterial infusion.
  • a catheter can be surgically or angiographically implanted to direct treatment to the affected organ.
  • a subcutaneous portal, connected to the catheter, can be used for chronic treatment, or an im ⁇ plantable, refillable pump may also be employed.
  • T4 DNA Iigase, T4 polynu- cleotide kinase, Tag DNA polymerase, and calf intestinal phosphatase were purchased from New England Biolabs (Beverly, MA) or Promega Biotech (Madison, WI) : these reagents were used according to the manufacturer's in ⁇ struction.
  • a "SEQUENASE DNA II" sequencing kit was used (United States Biochemical Corporation, Cleveland OH) .
  • Immunoblotting and other reagents were from Sigma Chemical Co. (St. Louis, MO) or Fisher Scientific (Needham, MA) . Nitrocellulose filters are obtained from Schleicher and Schuell (Keene, NH) .
  • Synthetic oligonucleotide linkers and primers are prepared using commercially available automated oligonu- cleotide synthesizers (e .g. , an ABI model 380B-02 DNA synthesizer (Applied Biosystems, Foster City, CA)) .
  • custom designed synthetic oligonucleotides may be purchased, for example, from Synthetic Genetics (San Diego, CA) .
  • cDNA synthesis kit and random priming labeling kits are obtained from Boehringer-Mannheim Biochemical (BMB, Indianapolis, IN) .
  • Oligonucleotide sequences encoding polypeptides can be either synthesized directly by standard methods of oligonucleotide synthesis, or, in the case of large coding sequences, synthesized by a series of cloning steps involving a tandem array of multiple oligonucleo ⁇ tide fragments corresponding to the coding sequence (Crea; Yoshio et al.; Eaton et al.). Oligonucleotide coding sequences can be expressed by standard recombinant procedures (Maniatis et al.; Ausubel et al.).
  • peptides can be synthesized directly by standard in vitro techniques (Applied Biosystems, Foster City CA) .
  • Recombinant human IFN ⁇ (rHuIFN ⁇ ) and rBoIFN7 was obtained from Genentech Inc. (South San Francisco, CA) .
  • the reference preparation of recombinant human IFN ⁇ (rHuIFN ⁇ ) was obtained from the National Institutes of Health: rHuIFN ⁇ is commercially available from Lee Biomolecular (San Diego, CA) . All tissue culture media, sera and IFNs used in this study were negative for endotoxin, as determined by assay with Limulus amebocyte lysate (Associates of Cape Cod, Woods Hole, MA) at a sensitivity level of 0.07 ng/ml.
  • HRP horseradish peroxidase conjugated goat anti- human antiserum
  • the reagent consists of 50 mL 0.05 M citric acid, pH 4.2, 0.078 mL 30% hydrogen peroxide solution and 15 mg ABTS. 0.1 mL of the substrate was added to each well, then incubated for 30 min at room temperature. The reaction was stopped with the addition of 0.050 mL 5% SDS ( /v) . The relative absorbance is determined at 410 nm.
  • EXAMPLE 1 Reproductive Functions of IFNT The effect of interferon-T on the lifespan of the corpus lutem was examined. IFNT was infused into uterine lumen of ewes at the concentrations given in Table 1. Recombinant human IFN ⁇ (rHuIFN ⁇ ) was infused at similar concentrations. In addition, control animals, which received co .trol pro ⁇ teins, were also used. The life span of the corpus luteum was assessed by examination of interestrous inter ⁇ vals, maintenance of progesterone secretion, and inhibi ⁇ tion of prostaglandin secretion (Davis, et al . , 1992 ) .
  • VSV vesicular stoma ⁇ titis virus
  • One antiviral unit caused a 50% reduction in de ⁇ struction of the monolayer, relative to untreated MDBK cells infected with VSV (control plates) .
  • Specific activities were further evaluated using normal ovine fibroblasts (Shnf) in a plaque inhibition assay (Lang- ford, et al . , 1981). A minimum of three samples were examined at each time point, and each sample was assayed in triplicate. The results presented in Table 2 are expressed as mean units/ml.
  • the amino acid coding sequence for OVIFNT (Imakawa, et al . , 1987) was used to generate a corresponding DNA coding sequence with codon usage optimized for expression in E. coli .
  • Linker sequences were added to the 5' and 3' ends to facilitate cloning in bacterial expression vec ⁇ tors.
  • the nucleotide sequence was designed to include 19 unique restriction enzyme sites spaced evenly throughout the coding sequence ( Figure 1) .
  • the nucleotide sequence was divided into eleven oligonucleotide fragments ranging in sizes of 33 to 75 bases.
  • oligonucleotides were synthe ⁇ sized on a 380-B 2-column DNA synthesizer (Applied Bio- sytems) and cloned single- or double-stranded into one of the following vectors: "pBLUESCRIPT + (KS)" (Stratagene, LaJolla, CA) , pTZ18R (Pharmacia, Piscataway, NJ) , or pTZ19R (Pharmacia, Piscataway, NJ) cloning vectors.
  • KS SK
  • the vectors were transformed into E. coli K. strain "XLl-BLUE" (recAl endAl gyrA96 thi hsdR17 (r , m ⁇ +) supE44 relAl ⁇ - (lac), ⁇ F', proAB, lac'Z ⁇ Ml ⁇ , Tnl0(tet R ⁇ ) which is commercially available from Stratagene (LaJolla, CA) .
  • Transformed cells were grown in L broth supplemented with ampicillin (50 ⁇ g/ml) . Oligonucleotide cloning and fusion was performed using standard recombinant DNA techniques.
  • Cloning vectors were cut with the appropriate re ⁇ striction enzymes to insert the synthetic oligonucleo ⁇ tides.
  • the vectors were treated with calf intestine alkaline phosphatase (CIP) to remove terminal phosphate groups.
  • Oligonucleotides were phosphorylated and cloned, as either single- or double-stranded molecules, into the appropriate vector using T4 DNA Iigase. When single- strands were introduced into cloning vectors, the second strand was completed by the bacterial host following transfection.
  • CIP calf intestine alkaline phosphatase
  • oligonucleotides were first annealed with their synthetic complementary strand then ligated into the cloning vector. E. coli K12 strains SB221 or NM522 were then transformed with the ligation. E. coli strain GM119 was used for cloning when the methylation-sensitive Stul and Clal restriction sites were involved. Restriction analyses were performed on isolated DNA at each stage of the cloning procedure. Cloned oligonucleotides were fused into a single polynucleotide using the restriction digestions and ligations outlined in Figure 2.
  • Oligonucleotide-contain- ing-DNA fragments were typically isolated after electro- phoretic size fractionation on low-melting point agarose gels (Maniatis, et a .; Sambrook, et al . ; Ausubel, et al . ) .
  • the resulting IFNT polynucleotide coding sequence spans position 16 through 531: a coding sequence of 172 amino acids.
  • the nucleotide sequence of the. final polynucleotide was confirmed by DNA sequencing using the dideoxy chain termination method.
  • the full length Stul/Sstl fragment (540 bp; Figure 2) was cloned into a modified pIN III omp-A expression vector and transformed into a competent SB221 strain of E. coli .
  • cells carrying the expression vector were grown in L-broth containing ampicillin to an OD (550 nm) of 0.1-1, induced with IPTG for 3 hours and harvested by centrifugation. Soluble recombinant IFNT was liberated from the cells by sonication or osmotic fractionation.
  • EXAMPLE 4 Expression of IFNT in Yeast The synthetic IFNT gene, synthesized in Example 3, was flanked at the 5' end by an Stul restriction site and at the 3' end by a Sacl restriction site.
  • Two oligonucleotide primers (SEQ ID NO:13 and SEQ ID NO:14) were used to attach linkers to the synthetic IFNr gene using polymerase chain reaction.
  • the linker at the 5' end allowed the placement of the synthetic IFNr gene in correct reading with the ubiquitin coding sequence present in the yeast cloning vector pBS24Ub (Chiron Corp., Emeryville, CA) .
  • the linker also constructed a ubiquitin-IFN ⁇ junction region that allowed in vivo cleavage of the ubiquitin sequences from the IFNT se ⁇ quences.
  • the 5' oligonucleotide also encoded a Sa ⁇ ll restriction endonuclease cleavage site.
  • the 3' oligonu ⁇ cleotide contained a Stul cleavage site.
  • the vector carrying the synthetic IFNT gene (Example 3) was isolated from E. coli strain "XLl-BLUE" by the alkaline lysis method. Isolated vector was diluted 500- fold in 10 mM Tris, pH 8.0/1 mM EDTA/10 mM NaCl.
  • the PCR reaction was performed in a 100 ⁇ l volume using Tag DNA polymerase and primers SEQ ID NO:13/SEQ ID NO:14.
  • the amplified fragments were digested with -Stul and -Sacll. These digested fragments were ligated into the Sacll and Smal sites of "pBLUESCRIPT+(KS) .”
  • the resulting plasmid was named pBSY-IFN ⁇ .
  • the DNA sequence was verified using double stranded DNA as the template.
  • Plasmid pBSY-IFN ⁇ was digested with Sacll and -BcoRV and the fragment containing the synthetic IFNT gene was isolated.
  • the yeast expression vector pBS24Ub (Sabin, et al . ; Ecker, et al . ) was digested with Sail . Blunt ends were generated using T4 DNA polymerase.
  • the vector DNA was extracted with phenol and ethanol precipitated (Sam- brook, et al . , 1989).
  • the recovered linearized plasmid was digested with Sacll, purified by agarose gel electro- phoresis, and ligated to the Sa ⁇ II--EcoRV fragment isolat ⁇ ed from pBSY-IFN ⁇ .
  • the resulting recombinant plasmid was designated pBS24Ub-IFN ⁇ .
  • the recombinant plasmid pBS24Ub-IFN ⁇ was transformed into E. coli .
  • Recombinant clones containing the IFNT insert were isolated and identified by restriction enzyme analysis. Plasmid DNA from clones containing IFNr coding sequences was used for transformation of S. cerevisiae (Rothstein, 1986) . Transformation mixtures were plated on uracil omission medium and incubated for 3-5 days at 30°C. Colonies were then streaked and maintained on uracil and leucine omission medium (Rothstein, 1986) .
  • IFNT Recombinant IFNT was visualized specifically by immunoblotting with monoclonal antibody or polyclonal antiserum against ovine IFNT upon electrotransfer of the fractionated cell extract to "NYTRAN" paper (Rothstein, 1986) .
  • pBS24-IFNr was grown for 24 hours at 30°C in 5 x uracil and leucine omission medium containing 8% glucose. This culture was then diluted 20-fold in YEP medium containing 1% glucose and further incubated for another 24-36 hours.
  • Cells were harvested by centrifugation, washed in 50 mM Tris, pH 7.6,/l mM EDTA and resuspended in wash buffer containing 1 mM PMSF. The cells were lysed using a Bead- beater apparatus (Biospec Products, Bartlesville, OK) . The lysate was spun at 43,000 x g for 20 minutes. The supernatant fraction was recovered and subjected to the purification protocol described below.
  • HMW DNA High molecular weight DNA was isolated from these cells (Sa brook, et al . , 1989). Two 10 ⁇ g samples of HMW DNA were digested with the restriction endonucleases Ifindlll or Pstl (Promega) for 2 hours at 37°C, and the DNA fragments electrophoretically separated in a 0.8% agarose gel (Bio-Rad, Richmond, CA) at 75 volts for 8 hours.
  • the DNA fragments were trans- ferred onto a nylon membrane (IBI-International Biotech ⁇ nologies, Inc., New Haven, CT) .
  • the membrane was baked at 80°C for 2 hours and incubated at 42°C for 4 hours in the following prehybridization solution: 5 x SSC (l x SSC is 0.15 M NaCl and 0.15 M sodium citrate), 50% vol/- vol formamide, 0.6% (wt/vol) SDS, 0.5% (wt/vol) nonfat dry milk, 20 mM Tris-HCl (pH 7.5), 4 mM EDTA, and 0.5 mg/ml single stranded herring sperm DNA (Promega) .
  • the filter was then incubated in a hybridization solution (5 x SSC, 20% vol/vol formamide, 0.6% (wt/vol) SDS, 0.5% (wt/vol) nonfat dry milk, 20 mM Tris-HCl (pH 7.5), 4 mM EDTA, and 2 x 10 8 cpm/ml 32 P-labelled OVIFNT cDNA (Imakawa, et al., 1987)) for 18 hours at 42°C.
  • a hybridization solution 5 x SSC, 20% vol/vol formamide, 0.6% (wt/vol) SDS, 0.5% (wt/vol) nonfat dry milk, 20 mM Tris-HCl (pH 7.5), 4 mM EDTA, and 2 x 10 8 cpm/ml 32 P-labelled OVIFNT cDNA (Imakawa, et al., 1987)
  • the filter was washed at 42°C for 15 minutes with 2 x SSC and 0.1% (wt/vol) SDS and exposed to X-ray film (XAR, Eastman Kodak, Rochester, NY) at -80°C for 48 hours in the pres ⁇ ence of an intensifying screen.
  • X-ray film XAR, Eastman Kodak, Rochester, NY
  • Two synthetic oligonucleotides (each 25-mer) , corre ⁇ sponding to the sequence 231 to 255 (contained in SEQ ID NO:13) and 566 to 590 (contained in SEQ ID NO:14) of OVIFNT CDNA (numbering relative to the cap site, Imakawa, et al . , 1987) were synthesized. These primers contained, respectively, cleavage sites for the restriction endonu ⁇ cleases Pstl and -EcoRI. SEQ ID NO:13 was modified to contain the EcoRI site, which begins at position 569.
  • DNA was isolated from approximately 1 x 10 5 plaque forming units (pfu) of the following two cDNA libraries: human term placenta (Clontech, Inc. , Palo Alto, CA) and human term cytotrophoblast (Dr. J.F. Strauss, University of Pennsylvania, Philadelphia PA) .
  • the DNA was employed in polymerase chain reaction (PCR) amplifications (Mul ⁇ lis; Mullis, et al . ; Pekin Elmer Cetus Corp. Norwalk CT) .
  • PCR polymerase chain reaction
  • Amplification reactions were carried out for 30 cycles (45°C, lm; 72°C, 2m; 94°C, lm) (thermal cycler and re ⁇ agents, Perkin Elmer Cetus) using primers SEQ ID NO:13/SEQ ID NO:14.
  • Amplification products were electrophoretically separated (100 volts in a 1.5% agarose gel (Bio-Rad)) and transferred onto a nylon membrane (IBI) .
  • the membrane was baked at 80°C for 2 hours and prehybridized and hybridized with 32 P-labelled OVIFNT CDNA as described above.
  • the membrane was washed in 5 x SSC/0.1% (wt/vol) SDS for 5 minutes at 42°C and in 2 x SSC/0.1% (wt/vol) SDS for 2 minutes at 42°C. It was then exposed at -80°C to "XAR" (Eastman Kodak) X-ray film for 24 hours in the presence of an intensifying screen. An amplification product that hybridized with the labelled probe DNA was detected.
  • PCR was performed again as directed above. Ampli ⁇ fied products were digested with the restriction endonu- cleases .EcoRI and Pstl (Promega) for 90 minutes at 37°C. The resulting DNA fragments were electrophoretically separated as described above and the band containing the IFNT amplification product was excised from the gel. DNA fragments were recovered by electroelution, subcloned into EcoRI /Pstl digested-dephosphorylated plasmid pUCl9 and transformed into E. coli strain JM101 (Promega) by calcium chloride method (Sambrook, et al . , 1989). The plasmids were isolated and the inserted amplification product sequenced using the dideoxy termination method (Sanger, et al . , 1977; "SEQUENASE" reactions. United
  • Plaques that hybridized to the probe were further analyzed by PCR using the OVIFNT primers described above. Six plaques which generated positive PCR signals were purified. The phage DNA from these clones was isolated and digested with .EcoRI restriction endonuclease. The DNA inserts were subcloned into pUC19 vectors and their nucleotide sequences determined by dideoxy nucleotide sequencings.
  • Recombinant phage from the ⁇ gtll subgenomic library were propagated in E. coli Y1080 and plated with E. coli
  • the plates were overlaid with duplicate nitrocellulose filters, which were hybridized with a 32 P-labelled probe from one of the six human IFNT CDNA clones isolated above. Three clones giving positive hybridization sig ⁇ nals were further screened and purified.
  • the phage DNAs were isolated, digested with -EcoRI, subcloned into pUC19 vector and sequenced. The three clones yielded sequence information for over 800 bases relative to cap site (clones were sequenced in both orientations) .
  • the nucle ⁇ ic acid sequence information is presented as SEQ ID NO:11 and the predicted protein coding sequence is presented as SEQ ID NO:12. Comparison of the predicted mature protein sequence (SEQ ID NO:12) of this gene to the predicted protein sequence of OVIFNT is shown in Figure 3.
  • HUIFNT mRNA The presence of HUIFNT mRNA in human term placenta and amniocytes (26 weeks, 2 million cells) was analyzed by using reverse transcriptase-PCR (RT-PCR) method (Clon ⁇ tech Laboratories, Palo Alto CA) .
  • RT-PCR reverse transcriptase-PCR
  • tcRNA Total cellular RNA isolated from human placenta, amniocytes and ovine conceptuses were reverse transcribed using the primer SEQ ID NO:14.
  • the primer SEQ ID NO:13 was then added to the reaction and polymer ⁇ ase chain reaction carried out for 40 cycles.
  • the PCR products were size fractionated on agarose gels and transferred to filters.
  • the DNA on the filters was hybridized with.
  • the results of these analyses demonstrate the presence of human IFNT mRNA in the feto-placental annex. •
  • the amino- cytes also expressed the messages corresponding to OVIFNT primers and human probe.
  • HUIFNT was applied to the tcRNA isolated from human adult lymphocytes. A densitometric analysis revealed that IFNT mRNA exists in lymphocytes.
  • OVIFNT CDNA is base #81-665; Figure 7
  • pBS New England Biolabs
  • Several pBS clones were isolated, subcloned, and their nucleotides sequenced. From this clone a 3' fragment (bases #425-736) was excised using the restriction endon ⁇ ucleases NlalV and -EcoRI and subcloned into the tran ⁇ scription vector pBS. This vector was designated pBS/OVIFNT.
  • an antisense cRNA probe was synthesized by in vitro tran ⁇ scription (Sambrook, et al., 1989) using T 7 RNA polymerase (Stratagene) .
  • a trace amount of 3 H-CTP was used in the transcription reaction.
  • dUTP labeled with digoxigenin was incorporated into the cRNA and yield was estimated through TCA precipitation and scin ⁇ tillation counting.
  • In situ hybridization was performed using the anti- sense RNA probe, as described by Lawrence, et al . (1985) with the following modifications.
  • Deparaffinized and hydrated sections were prehybridized for 10 minutes at room temperature in phosphate buffered saline (PBS) containing 5 mM MgCl 2 .
  • Nucleic acids in the sections were denatured for 10 minutes at 65°C in 50% formamide/2 x SSC.
  • Sections were incubated overnight at 37°C with a hybridization cocktail (30 ⁇ l/slide) containing 0.3 ⁇ g/ml digoxigenin-labelled cRNA probe and then washed for 30 minutes each at 37°C in 50 formamide/1 x SSC. Final washes were performed for 30 minutes each at room temper ⁇ ature in 1 x SSC and 0.1 x SSC.
  • the sections were blocked for 30 minutes with 0.5% Triton X-100 (Sigma) and 0.5% non-fat dry milk.
  • Hybridization signal was detected using purified sheep antidioxigenin Fab fragments conjugated to alkaline phosphatase (Boehringer-Mannheim) . After unbound anti ⁇ body was removed, nitroblue tetrazolium/5-bromo-4-chloro- 3-indolyl-phosphate substrate (Promega) and levamisole (Bector Laboratories, Burlingame, CA) were added for signal detection via colorimetric substrate generation. The tissues were counterstained in methyl green (Sigma) , dehydrated, and mounted.
  • tissue sections were pretreated with 100 ⁇ g/ml of pancreatic RNaseA (Sigma) for 30 min ⁇ utes at 37°C.
  • the RNase was inactivated on the slide with 400 units of RNase inhibitor (Promega) .
  • the slides were then washed twice in 250 ml of PBS/5 mM MgCl 2 .
  • tRNA (Sigma) was substituted for the digoxigenin probes.
  • First trimester placental villi composed of an outer layer of syncytiotrophoblast, an underlying layer of cytotrophoblast, and a central stromal region with vari- ous types of mesenchymal cells, displayed the highest transcript level of IFNT in the cytotrophoblast cells. Less intense but detectable levels were present in both the syncytiotrophoblast and stromal cells. A similar pattern of transcript expression was demonstrated in the placental villi of term tissue but the level of signal detection was low. First trimester extravillous tropho ⁇ blast displayed the highest amount of message and stained positive when present in the maternal blood spaces.
  • Antiviral Activity of IFNr The relative specific activity of OVIFNT, purified to homogeneity, was evaluated in antiviral assays.
  • the antiviral assays were performed essentially as described above in Example 2. Specific activities are expressed in antiviral units/mg protein obtained from antiviral assays using either Madin-Darby bovine kidney (MDBK) cells or sheep normal fibroblasts (Shnf) . All samples were as ⁇ sayed simultaneously to eliminate interassay variability.
  • MDBK Madin-Darby bovine kidney
  • Scnf sheep normal fibroblasts
  • IFNT had a higher specific activity than either rBoIFN ⁇ or rBoIFN7 (Table 3) .
  • the NIH standard prepara ⁇ tion of rHuIFN ⁇ had a similar specific activity, while a commercial preparation of rHuIFN ⁇ exhibited low specific antiviral activity. Comparable relative antiviral activ ⁇ ity was demonstrated using either bovine or ovine cells.
  • OVIFNT Highly purified OVIFNT was tested for anti-retrovi- ral and cytotoxic effects on feline peripheral blood lymphocytes exposed to the feline immunodeficiency retro- virus.
  • This lentivirus produces a chronic AIDS-like syndrome in cats and is a model for human AIDS (Pederson, et al . , 1987). Replication of the virus in peripheral blood lymphocytes is monitored by reverse transcriptase activity in culture supernatants over time. The data from these assays are presented in Table 4.
  • IFNT HIV Infected Human Peripheral Lymphocytes
  • Human peripheral blood lymphocytes which had been infected with HIV (Crowe, et al . ) , were treated with varying concentrations of OVIFNT.
  • Replica ⁇ tion of HIV in peripheral blood lymphocytes was monitored by reverse transcriptase activity in culture supernatants over time. Reverse transcriptase activity was measured essentially by the method of Hoffman, et al . The data from these assays are presented in Table 5.
  • concentrations of OVIFNT pro ⁇ resulted significant antiviral effects.
  • a concentration of only 10 ng/ml resulted in over a 50% reduction in RT activity after only six days.
  • a concentration of 500 ng/ml resulted in a 90% reduction in RT activity within 10 days.
  • IFNT IFNT-induced cellular growth
  • WISH Human amnion
  • MDBK MDBK cells
  • HMEM fetal bovine serum
  • FBS fetal bovine serum
  • Various dilu ⁇ tions of interferons were added to triplicate wells, and the plates were incubated for 8 days to allow colony formation. Colonies were visualized after staining with crystal violet, and counted.
  • Cell cycle analysis was performed with HMEM containing 0.5% "spent" media for an additional 7 days. WISH cells were used without being synchronized.
  • IFNT activity For examination of IFNT activity, cells were re-plated at 2.5 x 10 5 cells/well in HMEM with 10% FBS in 6 well plates. Various dilutions of IFNT alone or in combination with peptides were added to achieve a final volume of 1 ml. Plates were incubated at 37°C in 5% Co 2 for 12, 15, 18, 24, or 48 hours. Cells were treated with trypsin, collected by low speed centrifugation and washed. The cell pellet was blotted dry and 250 ⁇ l of nuclear staining solution (5 mg propidium iodide, 0.3 ml NP40 and 0.1 gm sodium citrate i 100 ml distilled H 2 0) was added to each tube.
  • nuclear staining solution 5 mg propidium iodide, 0.3 ml NP40 and 0.1 gm sodium citrate i 100 ml distilled H 2 0
  • the tubes were incubated at room temperature. After 10 minutes, 250 ⁇ l of RNase (500 units/ml in 1.12% sodium citrate) was added per tube and incubated an additional 20 minutes. Nuclei were filtered through 44 ⁇ m mesh, and analyzed on a FACStar (Becton Dickinson, Mountain View, CA) using, the DNA Star 2.0 software.
  • RNase 500 units/ml in 1.12% sodium citrate
  • Ovine IFNT inhibited both colony size and number.
  • Ovine IFNT was more effective than human IFN ⁇ on the human cell line; thus, it is very potent in cross-species activity. Its activity was dose-dependent, and inhibition of proliferation could be observed at concentrations as low as l unit/ml. Concentrations as high as 50,000 units/ml (units of antiviral activity/ml) stopped proliferation, while cell viability was not impaired.
  • Rat (MtBr7 .c5) or bovine kidney (MDBK) cells were seeded in phenol red-free DME-F12 medium supplemented with 3% dextran-coated charcoal stripped Controlled Process Serum Replacement 2 (CPSR 2, Sigma) and 5% dex ⁇ tran-coated charcoal stripped fetal, bovine serum (FBS) . After attaching for approximately 15-18 hours, the cells were washed once with serum-free DME-F12 medium.
  • CPSR 2 Controlled Process Serum Replacement 2
  • FBS bovine serum
  • the medium was replaced with phenol red-free DME-F12 medium supplemented with 3% stripped CPSR2, 1% stripped FBS ("3/1" medium) or 3/1 medium containing OVIFNT at various units of antiviral activity as determined in the vesicu ⁇ lar stomatitis virus challenge assay for interferons (Example 2) .
  • Media containing a similar dilution of buffer undiluted buffer - 10 mM Tris, 330 mM NaCl, [TS]), in which the OVIFNT was dissolved was used for controls.
  • the antiproliferative activity of OVIFNT on human tumor cell lines was evaluated by measuring the rate of 3 H-thymidine incorporation into cells which have been treated with OVIFNT.
  • Adherent tumor lines were plated at 2.5 X 10 5 cells/well in 1 ml in 6-well plates. They received interferon treatments as just described, but were tryp- sinized prior to counting. Significant differences between treatments were assessed by an analysis of variance followed by Scheffe's F-test. Cell cycle analysis was performed by flow cyto ⁇ metry using propidium iodide.
  • Human MCF7 breast adenocarcinoma cells were seeded from logarithmically growing cultures in phenol red-free DME-F12 medium supplemented with 3% dextran-coated char ⁇ coal stripped CPSR and 5% dextran-coated FBS. After attaching for approximately 15-18 hours, the cells were washed once with serum-free DME-F12 medium. The medium was replaced with phenol red-free DME-F12 medium supple ⁇ mented with 3% stripped CPSR2, 1% stripped FBS ("3/1" medium) or 3/1 medium containing OVIFNT at the indicated number of units of antiviral activity as determined in the vesicular stomatitis virus challenge assay for inter ⁇ ferons.
  • TCA trichloroacetic acid
  • OVIFNT was able to substantially reduce the rate of 3 H- thymidine incorporation in the human carcinoma cell line. This demonstrates the efficacy of OVIFNT in inhibiting tumor cell proliferation, in particular, mammary tumor cell proliferation.
  • Figure 4 shows that both OVIFNT and IFN ⁇ were able to drastically reduce growth of HL-60 cells.
  • the growth reduction for each compound exceeded 60% for each concentration tested.
  • OVIFNT caused an approximately 80% reduction in growth while IFN ⁇ caused a 100% reduction in growth.
  • the data presented in Figure 4 reveal, that a substantial factor in the ability of IFN ⁇ to reduce growth was its toxic effect on the cells.
  • the toxicity of IFN ⁇ resulted in less than 25% of the cells remaining viable.
  • nearly 100% of the cells remained viable when OVIFNT was applied at 10,000 units/ml.
  • Figure 5 presents data demonstrating that rHuIFN ⁇ is cytotoxic.
  • results of one of three repli ⁇ cate experiments are presented as mean % viability ⁇ SD.
  • the T cell lymphoma cell line H9 was slightly less sensitive to the antiproliferative effects of the IFNs than the tumor cell lines described above. Results of one of three replicate experiments are presented in Figure 10 as mean % growth reduction ⁇ SD. While rHuIFN ⁇ was not toxic to the H9 cells, it failed to inhibit cell division significantly at any of the concentrations examined. In contrast, OVIFNT was observed to reduce H9 growth by approximately 60% ( Figure 10) . Thus, only OVIFNT is an effective growth inhibitor of this T cell lymphoma.
  • B16-F10 is a syngeneic mouse transplantable tumor selected because of its high incidence of pulmonary metastases (Poste, et al . , 1981). Interferon treatment was initiated 3 days after the introduction of the tumor cells. Each mouse received 100 ⁇ l of either PBS alone, PBS containing l X 10 s units of OVIFNT, or PBS containing 1 X 10 s units of recombinant murine IFN ⁇ (MuIFN ⁇ ) , i.v. per day for 3 consecutive days.
  • mice were sacrificed at 21 days and the lungs were preserved in 10% buffered formalin.
  • the frequency of pulmonary metastases were compared between control mice (PBS) , OVIFNT-treated mice, and MuIFN ⁇ -treated mice.
  • the results of these in vivo administrations demonstrated that OVIFNT dramatically reduced B16-F10 pulmonary tu ⁇ mors. These results support the use of IFNT as an effi ⁇ cacious antineoplastic agent in vivo.
  • Overlapping synthetic peptides were synthesized corresponding to the entire IFNT sequence ( Figure 6) . Average hydropathicity values were calculated by taking the sum of the hydropathy values for each amino acid divided by the total number of amino acids in each se ⁇ quence. Hydropathy values were taken from Kyte, et al . (1982) . These peptides were of approximately the same molec ⁇ ular weight but differed slightly in overall hydrophili- city. Despite this difference, all peptides were anti ⁇ genic as demonstrated by the production of rabbit anti ⁇ sera with titers greater than 1:3,000 as assessed by ELISA (Harlow, et al . ) .
  • the peptides were used to inhibit the antiviral activity (Example 2) of OVIFNT and rBoIFN ⁇ .
  • the results of this analysis are presented in Figure 12: 1 mM N- and C-terminal peptides both effectively blocked the antivi- ral activity of OVIFNT using MDBK cells.
  • the peptide OVIFNT (119-150) showed minimal inhibitory activity.
  • the OVIFNT (34-64) and (90-122) peptides had no apparent inhibitory activity.
  • Figure 13 are expressed as the percent of control antivi ⁇ ral activity: that is, in the absence of any competing peptide.
  • Data presented are the means of 6 replicate experiments. The data demonstrate that inhibition by OVIFNT (1-37), (62-92), (119-150), and (139-172) were significantly different than OVIFNT (34-64) and (90-122) at 10- 3 M and 3 x 10' 3 M. OVIFNT (139-172) was signifi ⁇ cantly different than all other peptides at 10' 3 M. Sig ⁇ nificance was assessed by analysis of variance followed by Scheffe's F test at p ⁇ 0.05. Thus, OVIFNT (1-37) (62-92), (119-150), and (139-172), in particular (139- 172), may represent receptor binding regions for IFNr.
  • OVIFNT ⁇ bovine IFN ⁇ antiviral activity
  • Monolayers of Madin Darby bovine kidney cells were incu ⁇ bated with 40 units/ml bovine IFN ⁇ in the presence or absence of various concentrations of OVIFNT peptides.
  • the results are presented in Figure 14 and are expressed as the percent of control antiviral activity in the absence of OVIFNT peptides.
  • the data presented are the means of 4 replicate experiments.
  • the results indicate that inhibition by OVIFNT (62-92), (119-150), and (139- 172) were significantly different from OVIFNT (1-37) , (34-64) and (90-122) at 10' 3 M.
  • OVIFNT (139-172) was significantly different than OVIFNT (1-37), (34-64) and (90-122) at 3 x 10" 3 M. Significance was assessed by analysis of variance followed by Scheffe's F test at p ⁇ 0.05. Thus, OVIFNT (62-92), (119-150), and (139-172), in particular (139-172) , may represent common receptor binding regions for IFNT and bovine IFN ⁇ .
  • OVIFNT may represent a common receptor binding region for IFNT and various IFN ⁇ (s).
  • the OVIFNT peptides described above appear to have no effect on the antiviral activity of IFN7.
  • Peptide inhibition of bovine IFN7 antiviral activity was evaluat ⁇ ed as follows. Monolayers of Madin Darby bovine kidney cells were incubated with 40 units/ml bovine IFN gamma in the presence or absence of various concentrations of OVIFNT peptides. Results are expressed as the percent of control antiviral activity in the absence of OVIFNT peptides. The data are presented in Figure 16 and are the means of 3 replicate experiments. There were no significant differences among peptides as assessed by analysis of variance followed by Scheffe's F test at p ⁇ 0.05.
  • RT Reverse transcriptase activity
  • FIV- infected FET-1 cells (1 X 10 6 /ml)
  • HIV-infected HPBL l X 10 6 /ml
  • Control cultures received no OVIFNT.
  • OVIFNT was used at 100 ng/ml
  • peptides were used at 200 ⁇ M.
  • Data from a representative experiment are expressed as cpm/ml culture supernatant and are presented for FIV infected cells, Figure 11A, and HIV infected cells, Figure 11B.
  • Antipep ⁇ tide antisera inhibition of OVIFNT antiviral activity was evaluated as follows. Monolayers of MDBK cells were incubated with 20 units/ml of OVIFNT in the presence a 1:30 dilution of either preimmune sera or antisera to each of the OVIFNT peptides described above. In Figure 17 the data from duplicate experiments are presented as the mean percent inhibition of OVIFNT antiviral activity produced by antipeptide antisera relative to the appro ⁇ priate preimmune sera ⁇ standard error. Significant differences were assessed by analysis of variance fol ⁇ lowed by Scheffe's F test at p ⁇ 0.05.
  • the IFNT binding assay was carried out as follows.
  • IFNT Five ⁇ g of IFNT was iodinated for 2 minutes with 500 ⁇ Ci of Na 125 I (15 mCi/ ⁇ g; Amersham Corporation, Arlington Heights, IL) in 25 ⁇ l of 0.5 M potassium phosphate buff ⁇ er, pH 7.4, and 10 ⁇ l of chloramine-T (5 mg/ml) (Griggs, et al . , 1992). The specific activity of the iodinated protein was 137 ⁇ Ci/ ⁇ g. For binding assays, monolayers of MDBK cells were fixed with paraformaldehyde and blocked with 5% nonfat dry milk.
  • Antisera to IFNT (119-150) , which inhibited binding of OVIFNT to receptor, also reversed the OVIFNT antipro- liferative effect.
  • IFNT 139-172
  • Several other peptides notably IFNT (139-172) , reversed the OVIFNT antiproliferative effect, but to a lesser extent.
  • EXAMPLE 18 Further Analysis of the Cellular and Anti-Viral Effects of IFNT A. HIV Anti-Viral Effects.
  • the antiviral effects of IFNT against HIV were evaluated by treating human PBMC cells with various amounts of either recombinant ovine IFNT (r-OvIFN ⁇ ) or recombinant human IFN ⁇ 2 at the time of infection with HIV. Drug was present throughout the experiment. At day 7 and day 14, p24 production was determined (by ELISA (Wang, et al . , 1988, 1989) and compared to a zero drug control. The results of this analysis are presented in Table 12.
  • IFNT are effective in reducing the replication of HIV in human lymphocytes.
  • B. In vitro Cytotoxicity Test in PBMCs Human PBMCs were seeded at 5 x 10 5 cells/ml. Cells were stimulated at day 0 with 3 ⁇ g/ml PHA. Cells were treated with recombinant human IFN ⁇ 2A (at concentrations of 10, 100, 1,000 and 10,000 units/ml) and IFNT (at concentrations of 2.6, 26, 260, 2,600, 26,000, 260,000, and 2,600,000 units/ml) in 200 ⁇ l/wells (4 replicates of each concentration using 96 well flat bottom plates) . Control cultures were given no interferons. After 4 days of incubation, cells were pulsed for 9 hours using 3 H- thymidine at 1 uCi/well. The cells were harvested and the incorporation of labeled thymidine into DNA was determined ( Figure 8) .
  • HepG2-T14 is a human cell that was derived from liver cells transfected with Hepatitis B
  • HBV Virus The cell line semi-stably produces HBV virus: over time the cell line's production of HBV intracellular DNA and secreted virus decreases.
  • the cells are pre-treated with deAZA-C (5-azacytidine; Miyoshi, et al . ) to induce production of the virus. Treatment was for 2-3 days and the amount of induction was about a factor of two.
  • the cells were then treated with either the IFN ⁇ and IFNT at levels of 0, 5,000, 10,000, 20,000 and 40,000 units per ml. All levels of either IFN ⁇ or IFNT reduced DNA pro ⁇ duction by about a factor of 2 compared to the no drug control.
  • the hepatocyte cell line HepG2-T14 (described above) was examined for the effects of IFN ⁇ and IFNT on hepato ⁇ specific mRNA production.
  • Cells were incubated in con ⁇ centrations of IFN ⁇ or IFNT at 0, 5,000, 10,000, 20,000, and 40,000 units per ml.
  • the messenger RNAs for the hepatocyte specific proteins Apo E and Apo Al were de ⁇ tected by hybridization analysis (Sambrook, et al . ; Maniatis, et al . ) using probes specific for these two mRNA's (Shoulders, et al . , and Wallis, et al . ) .
  • the IFNT coding sequence (e .g. , Figure 7) is cloned into the polylinker site of lambda gtll.
  • the IFNT coding sequence is placed in-frame with the amino terminal ⁇ - galactosidase coding sequences in lambda gtll.
  • Lysogens infected with gtll/IFN ⁇ are used to inoculate 500 ml of NZYDT broth.
  • the culture is incubated at 32°C with aeration to an O.D. of about 0.2 to 0.4, then brought to 43°C quickly in a 43°C water bath for 15 minutes to induce gtll peptide synthesis, and incubated further at 37°C for 1 hour.
  • the cells are pelleted by centrifuga ⁇ tion, suspended in 10 ml of lysis buffer (10 mM Tris, pH 7.4 containing 2% "TRITON X-100" and 1% aprotinin added just before use.
  • the resuspended cells are frozen in liquid nitrogen then thawed, resulting in substantially complete cell lysis.
  • Non-solubilized material is removed by centrifugation.
  • the clarified lysate material is loaded on the Sepharose column, the ends of the column closed, and the column placed on a rotary shaker for 2 hrs. at room temperature and 16 hours at 4°C. After the column set ⁇ tles, it is washed with 10 ml of TX buffer.
  • the fused protein i ⁇ eluted with 0.1 M carbonate/bicarbonate buff ⁇ er, pHlO. Typically, 14 ml of the elution buffer is passed through the column, and the fusion protein is eluted in the first 4-6 ml of eluate.
  • the eluate containing the fusion protein is concen ⁇ trated in "CENTRICON-30" cartridges (Amicon, Danvers, Mass.).
  • the final protein concentrate is resuspended in, for example, 400 ⁇ l PBS buffer. Protein purity is ana ⁇ lyzed by SDS-PAGE.
  • the purified fused pro ⁇ tein is injected subcutaneously in Freund's adjuvant in a rabbit. Approximately 1 mg of fused protein is injected at days 0 and 21, and rabbit serum is typically collected at 6 and 8 weeks.
  • the IFNT coding sequence (e . g. , Figure 7) is cloned into the pGEX vector (Boyer, et al . ; Frangioni, et al . ;
  • the pGEX vector (Smith, et al . ) was modified by in ⁇ ertion of a thrombin cleavage sequence in-frame with the glutathi- one-S-transferase protein (GST — sj26 coding sequence) .
  • This vector is designated pGEXthr.
  • the IFNT coding sequence is placed in-frame with the ⁇ j26-thrombin coding sequences (Guan, et a . ; Hakes, et al . ) .
  • the IFNT coding sequence insert can be generated by the polymerase chain reaction using PCR primers specific for the insert.
  • the IFNT fragment is ligated to the linearized pGEXthr vector.
  • the ligation mixture is transformed into E. coli and ampicillin resistant colonies are selected. Plasmids are isolated from the ampicillin resistant colonies and analyzed by restriction enzyme digestion to identify clones containing the IFNT insert (vector desig ⁇ nated pGEXthr-IFNT) .
  • E. coli strain XL-I Blue is transformed with pGEXthr-IFNT and is grown at 37°C overnight. DNA i ⁇ prepared from randomly-picked colonies.
  • the presence of the insert coding sequence is typically confirmed by (i) restriction digest mapping, (ii) hybridization screening u ⁇ ing labelled IFNT probes (i.e.. Southern analysis), or (iii) direct DNA sequence analysis.
  • a pGEXthr-IFNT clone is grown overnight. The over- night culture is diluted 1:10 with LB medium containing ampicillin and grown for one hour at 37°C. Alternative ⁇ ly, the overnight culture is diluted 1:100 and gro-- to OD of 0.5-1.0 before addition of IPTG (isopropylth - ⁇ - galacto ⁇ ide) . IPTG (GIBCO-BRL, Gaithersburg MD) is added to a final concentration of 0.2-0.5 mM for the induction of protein expre ⁇ ion and the incubation i ⁇ typically continued for 2-5 hour ⁇ , preferably 3.5 hours.
  • Bacterial cells are harvested by centrifugation and resuspended in 1/100 culture volume of MTPBS (150 mM NaCl, 16 mM Na 2 HP0 4 , 4 mM NaH 2 P0 4 ) . Cells are lysed by lysozyme, sonication or French press, and lysate ⁇ cleared of cellular debri ⁇ by centrifugation.
  • MTPBS 150 mM NaCl, 16 mM Na 2 HP0 4 , 4 mM NaH 2 P0 4
  • the fusion proteins are partially purified over a glutathione agarose affinity column as de ⁇ cribed in detail by Smith, et a .
  • 100 ml culture ⁇ are grown overnight.
  • the culture ⁇ are dilut ⁇ ed to 1 liter, and the cell ⁇ grown another hour at 37°C.
  • Expression of the fusion proteins is induced using IPTG.
  • the induced cultures are grown at 37°C for 3.5 hours.
  • Cells are harvested and a ⁇ onicator u ⁇ ed to lyse the cells.
  • Cellular debris is pelleted and the clear lysate loaded onto a glutathione "SEPHAROSE" column. The column is washed with several column volumes.
  • the fusion pro ⁇ tein is eluted from the affinity column with reduced glutathione and dialyzed.
  • the IFNT can be liberated from the hybrid protein by treatment with thrombin.
  • the ⁇ j26 and IFNT fragment ⁇ of the hybrid protein can then be ⁇ eparated by ⁇ ize fractionation over column ⁇ or on gel ⁇ .
  • the IFNT portion of the hybrid pro- tein is released from the column by treatment with throm ⁇ bin (Guan, et al . ; Hakes, et al . ) .
  • the purified Sj26/IFN ⁇ fused protein i ⁇ injected subcutaneously in Freund's adjuvant in a rabbit. Approx ⁇ imately 1 mg of fused protein is injected at day ⁇ 0 and 21, and rabbit ⁇ erum is typically collected at 6 and 8 weeks.
  • a second rabbit is similarly immunized with purified Sj26 protein obtained from control bacterial lysate. Minilysates from the following bacterial cultures are prepared: (1) KM392 cells infected with pGEXthr and pGEXthr containing the IFNT insert; and (2) cells infect ⁇ ed with lambda gtll containing the IFNT insert.
  • the minilysates and a commercial source 8-galactosidase are fractionated by SDS-PAGE, and the bands transferred to nitrocellulose filters for Western blotting (Sambrook, et al . ; Ausubel, et al . ) .
  • serum from control (Sj26) rabbits is immunoreactive with each of the Sj.26 and Sj26 fused protein antigens.
  • Serum from the animal immunized with Sj26/IFN ⁇ fused protein is reactive with all Sj-26 and beta-gal fusion proteins containing IFNT coding ⁇ equence ⁇ , indicating the presence of specific immunoreaction with the IFNT antigen. None of the sera are expected to be immunoreactive with beta-galactosi- dase.
  • Anti-IFNT antibody present in the sera from the ani ⁇ mal immunized with the Sj26/IFN ⁇ is purified by affinity chromatography (u ⁇ ing immobilized recombinantly produced IFNT a ⁇ ligand, e ⁇ sentially as described above in Example 12 for the anti-beta-galactosidase antibody) .
  • ADDRESSEE Law Offices of Peter J. Dehlinger
  • B STREET: 350 Cambridge Ave., Suite 300
  • TTGTTGGAAC CATGTAGAAC CGGTTTGCAC CAACAATTGG ACAACTTGGA TGCATGTTTG 30 GGTCAAGTTA TGGGTGAAGA AGACTCTGCT CTCGGGAGAA CCGGTCCAAC GCTAGCTTTG
  • Gin Glu Ala Gin Ala lie Ser Val Leu His Glu Met Leu Gin Gin Ser 50 55 60
  • MOLECULE TYPE protein
  • MOLECULE TYPE DNA (genomic)
EP93924958A 1992-10-30 1993-10-19 Interferon tau compositions and methods of use Withdrawn EP0669981A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP03006878A EP1360962A3 (en) 1992-10-30 1993-10-19 Interferon tau composition and methods of use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US96989092A 1992-10-30 1992-10-30
US969890 1992-10-30
PCT/US1993/010016 WO1994010313A2 (en) 1992-10-30 1993-10-19 Interferon tau compositions and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP03006878A Division EP1360962A3 (en) 1992-10-30 1993-10-19 Interferon tau composition and methods of use

Publications (1)

Publication Number Publication Date
EP0669981A1 true EP0669981A1 (en) 1995-09-06

Family

ID=25516125

Family Applications (2)

Application Number Title Priority Date Filing Date
EP03006878A Withdrawn EP1360962A3 (en) 1992-10-30 1993-10-19 Interferon tau composition and methods of use
EP93924958A Withdrawn EP0669981A1 (en) 1992-10-30 1993-10-19 Interferon tau compositions and methods of use

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP03006878A Withdrawn EP1360962A3 (en) 1992-10-30 1993-10-19 Interferon tau composition and methods of use

Country Status (8)

Country Link
EP (2) EP1360962A3 (zh)
JP (1) JPH08505047A (zh)
KR (1) KR100357768B1 (zh)
CN (1) CN1090510A (zh)
AU (1) AU689450B2 (zh)
CA (1) CA2148119A1 (zh)
TW (2) TW585911B (zh)
WO (1) WO1994010313A2 (zh)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6372206B1 (en) 1989-03-02 2002-04-16 University Of Florida Orally-administered interferon-TAU compositions and methods
US5705363A (en) * 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
US5906816A (en) * 1995-03-16 1999-05-25 University Of Florida Method for treatment of autoimmune diseases
US5648241A (en) * 1989-09-15 1997-07-15 The General Hospital Corporation Conjugate vaccine against group B streptococcus
US5939286A (en) * 1995-05-10 1999-08-17 University Of Florida Hybrid interferon tau/alpha polypeptides, their recombinant production, and methods using them
US6204022B1 (en) 1996-04-12 2001-03-20 Pepgen Corporation And University Of Florida Low-toxicity human interferon-alpha analogs
KR100464531B1 (ko) * 1996-04-12 2005-03-07 유니버시티 오브 플로리다 잡종인터페론조성물및그것을사용하는방법
FI104363B (fi) * 1997-05-19 2000-01-14 Timo Kalevi Korpela Immunosuppressanttien ja interferonien farmakologisten keskinäisten vaikutusten parantaminen lisäaineilla
US6833256B1 (en) 1999-06-22 2004-12-21 University Of Maryland Interferon tau mutants and methods for making them
EP1156771A2 (en) * 1999-06-22 2001-11-28 The University Of Maryland Interferon tau mutants and methods for making them
CN1376165A (zh) * 1999-08-27 2002-10-23 佛罗里达大学 抑制lgE产生的物质和方法
US7105154B2 (en) * 2000-07-19 2006-09-12 Pepgen Corporation Method of treatment using interferon-tau
AU2001277055A1 (en) * 2000-07-19 2002-01-30 Pepgen Corporation Composition for treatment of and method of monitoring hepatitis c virus using interferon-tau
US7083782B2 (en) 2000-07-19 2006-08-01 Pepgen Corporation Method of treatment using interferon-tau
US7431920B2 (en) 2000-07-19 2008-10-07 Pepgen Corporation Method of treating IL-10 deficiency
JP2006213597A (ja) 2000-07-19 2006-08-17 Pepgen Corp インターフェロン−タウを用いるc型肝炎ウイルスの処置のための組成物およびモニタリングの方法
WO2003016472A2 (en) 2001-08-12 2003-02-27 Pepgen Corporation Hybrid interferon/interferon tau proteins, compositions and methods of use
TW200518768A (en) * 2003-11-17 2005-06-16 Pepgen Corp Methods for treatment of obesity and for promotion of weight loss
US20070025963A1 (en) * 2005-07-27 2007-02-01 Chih-Ping Liu Methods for reduction of scar tissue formation
DE602006016469D1 (de) 2005-09-27 2010-10-07 Biovet Inc Mittel zur behandlung von reproduktionsstörungen
WO2008021487A1 (en) * 2006-08-18 2008-02-21 Pepgen Corporation Combination treatment method with interferon-tau
CN101244261B (zh) * 2008-03-10 2010-09-15 山东大学 一种含未复性重组蛋白的生物制剂及其制备方法与应用
CN113969285B (zh) * 2021-11-09 2023-05-23 上海市农业科学院 重组表达的羊干扰素-τBB8基因及其制备方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989008706A1 (fr) * 1988-03-18 1989-09-21 Institut National De La Recherche Agronomique (Inr Isoformes de la trophoblastine, nouveaux interferons constitues par lesdites isoformes, leurs procedes d'obtention et leurs applications
EP0367063A1 (en) * 1988-10-26 1990-05-09 The Curators Of The University Of Missouri Isolation and cloning of complementary DNA for gene coding of bovine trophoblast protein-1
WO1990009806A2 (en) * 1989-03-02 1990-09-07 University Of Florida Composition for the inhibition of tumors and for the non-cytotoxic inhibition of replication of viruses

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989008706A1 (fr) * 1988-03-18 1989-09-21 Institut National De La Recherche Agronomique (Inr Isoformes de la trophoblastine, nouveaux interferons constitues par lesdites isoformes, leurs procedes d'obtention et leurs applications
EP0367063A1 (en) * 1988-10-26 1990-05-09 The Curators Of The University Of Missouri Isolation and cloning of complementary DNA for gene coding of bovine trophoblast protein-1
WO1990009806A2 (en) * 1989-03-02 1990-09-07 University Of Florida Composition for the inhibition of tumors and for the non-cytotoxic inhibition of replication of viruses

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
C.H. PONTZER ET AL: "Antiproliferative activity of a pregnancy recognition hormone, ovine trophoblast protein-1", CANCER RESEARCH, vol. 51, October 1991 (1991-10-01), pages 5304 - 5307, XP002058354 *
G. ABOAGYE-MATHIESEN ET AL: "Isolation,purification and biochemical characterization of human placental interferons by tandem high-performance affinity chromatography", PREPARATIVE BIOCHEMISTRY, vol. 22, no. 2, 1992, pages 105 - 121, XP002058355 *
G. CHARPIGNY ET AL: "High homology between a trophoblastic protein (trophoblastin) isolated from ovine embryo and alpha-interferons", FEBS LETTERS., vol. 228, February 1988 (1988-02-01), AMSTERDAM NL, pages 12 - 16, XP002058352 *
IMAKAWA K ET AL: "INTERFERON-LIKE SEQUENCE OF OVINE TROPHOBLAST PROTEIN SECRETED BY EMBRYONIC TROPHECTODERM", NATURE, vol. 330, 26 November 1987 (1987-11-26), pages 377 - 379, XP002010876 *
M. CHARLIER ET AL: "Cloning and expression of cDNA encoding ovine trophoblastin: its identity with a class-II alpha interferon", GENE., vol. 77, 1989, AMSTERDAM NL, pages 431 - 348, XP002058353 *
See also references of WO9410313A3 *

Also Published As

Publication number Publication date
CA2148119A1 (en) 1994-05-11
CN1090510A (zh) 1994-08-10
EP1360962A2 (en) 2003-11-12
AU689450B2 (en) 1998-04-02
KR100357768B1 (ko) 2003-04-07
TW391983B (en) 2000-06-01
TW585911B (en) 2004-05-01
AU5444994A (en) 1994-05-24
JPH08505047A (ja) 1996-06-04
WO1994010313A2 (en) 1994-05-11
EP1360962A3 (en) 2004-02-04
KR950704489A (ko) 1995-11-20
WO1994010313A3 (en) 1994-10-27

Similar Documents

Publication Publication Date Title
US6174996B1 (en) Hybrid interferon τ/type I interferon polypeptides
AU689450B2 (en) Interferon tau compositions and methods of use
EP0914435B1 (en) Hybrid interferon compositions and methods of use
FI100972B (fi) Pesäkkeitä stimuloivaa tekijä-1:tä (CSF-1) koodaava DNA-sekvenssi ja m enetelmä CSF-1-proteiinin valmistamiseksi
KR100223255B1 (ko) 개량된 알파 인터페론 조성물 및 인간 말초혈 백혈구로 부터의 제조 방법
JP3507507B2 (ja) 非免疫原性ペプチドを介して結合されたインターフェロン―αと免疫グロブリンとのハイブリッド
Pontzer et al. Structure/function studies with interferon tau: evidence for multiple active sites
CA2164811A1 (en) Super proteins including interferons and interleukins
US5104650A (en) Uses of recombinant colony stimulating factor-1
US20030017550A1 (en) DNA sequences encoding fusion proteins comprising IFN-beta and TM-alpha1
AU706762B2 (en) Interferon tau compositions and methods of use
WO2004022088A1 (en) Interferon and immunoglobulin fc fragment hybrid
KR100464531B1 (ko) 잡종인터페론조성물및그것을사용하는방법
US7235232B2 (en) Interferon alpha hybrids
WO2000006596A2 (en) INTERFERON HYBRIDS- alpha
CA2495480A1 (en) Interferon and immunoglobulin fc fragment hybrid

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19950529

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU NL PT SE

17Q First examination report despatched

Effective date: 19960314

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20051201