WO1995028488A1 - NON-SPLICING VARIANTS OF gp350/220 - Google Patents

NON-SPLICING VARIANTS OF gp350/220 Download PDF

Info

Publication number
WO1995028488A1
WO1995028488A1 PCT/US1995/004611 US9504611W WO9528488A1 WO 1995028488 A1 WO1995028488 A1 WO 1995028488A1 US 9504611 W US9504611 W US 9504611W WO 9528488 A1 WO9528488 A1 WO 9528488A1
Authority
WO
WIPO (PCT)
Prior art keywords
thr
amino acid
pro
ser
protein
Prior art date
Application number
PCT/US1995/004611
Other languages
French (fr)
Inventor
Richard Spaete
Winthrop T. Jackman
Original Assignee
Aviron
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
Priority to UA96114355A priority Critical patent/UA47403C2/en
Priority to SI9530569T priority patent/SI0769056T1/en
Priority to CZ19963054A priority patent/CZ292283B6/en
Priority to PL95316941A priority patent/PL181881B1/en
Priority to AT95916984T priority patent/ATE210184T1/en
Priority to HU9602894A priority patent/HU221647B1/en
Priority to JP52710995A priority patent/JP3447743B2/en
Application filed by Aviron filed Critical Aviron
Priority to AU23838/95A priority patent/AU707837B2/en
Priority to DE69524415T priority patent/DE69524415T2/en
Priority to SK1343-96A priority patent/SK283446B6/en
Priority to BR9507473A priority patent/BR9507473A/en
Priority to CA002187908A priority patent/CA2187908C/en
Priority to EP95916984A priority patent/EP0769056B1/en
Priority to DK95916984T priority patent/DK0769056T3/en
Publication of WO1995028488A1 publication Critical patent/WO1995028488A1/en
Priority to NO19964431A priority patent/NO319382B1/en
Priority to FI964186A priority patent/FI118224B/en
Priority to LVP-96-430A priority patent/LV11803B/en
Priority to FI20075338A priority patent/FI20075338A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16211Lymphocryptovirus, e.g. human herpesvirus 4, Epstein-Barr Virus
    • C12N2710/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16622New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • Epstein-Barr virus a member of the herpesvirus group, causes infectious mononucleosis in humans.
  • the disease affects more than 90% of the population.
  • Health analysts estimate the cost of the disease in the United States is 100 million dollars per year.
  • the virus is spread primarily by exchange of saliv: from individuals who shed the virus.
  • Children infected with EBV are largely asym tomatic or have very mild symptoms, while adolescents and adults who become infected develop typical infectious mononucleosis, characterized by fever, pharyngitis, and adenopathy. People who have been infected maintain anti-EBV antibodies for the remainder of their lives, and are thus immune to further infection.
  • EBV vaccine There is no commercially available EBV vaccine.
  • EBV has been shown to transform lymphocytes into rapidly dividing cells and has therefore been implicated in several different lymphomas, including Burkitt's lymphoma and oral hairy leukoplakia. EBV has also been detected in tissue ' samples from nasopharyngeal tumors. Worldwide it is estimated that 80,000 cases of nasopharyngeal cancer occur and it is more prevalent in ethnic Chinese populations.
  • a subunit vaccine uses one or more antigenic proteins from the virus that will elicit an immune response and confer immunity.
  • EBV proteins Two of the more important antigenic EBV proteins are glycoprotein(s) gp350/300 and gp220/200 that form part of the viral membrane envelope and allow virus particles to bind to and enter human target cells by interacting with the cellular membrane protein, CD21. See Nemerow. J. Virology 61:1416(1987 . They have long been singled out as subunit vaccine candidates but difficulties in obtaining antigenically active protein purified from native sources and low yields from recombinantly produced sources have hampered efforts of researcher and vaccine developers. In the literature these proteins are referred to using a variety of molecular weight ranges (350 or 300 kilodaltons (kD) for one of the proteins and 220 or 200 kDs for the other protein).
  • molecular weight ranges 350 or 300 kilodaltons (kD) for one of the proteins and 220 or 200 kDs for the other protein.
  • the gp350 or 300 protein is herein referred to as gp350 protein and the gp220 or 200 protein is herein referred to as gp220 protein. Collectively, both proteins are herein referred to as gp350/220 protein(s).
  • An alternatively spliced, single gene encodes the gp350/220 proteins and results in the generation of gp350 and gp220 mRNA transcripts; no naturally occurring variations in the gp350/220 gene splice sites are known.
  • the gene produces two expression products, the gp350 and gp220 proteins.
  • the open reading frame for the gp350/220 DNA sequence is 2721 base pairs (bp).
  • the entire reading frame encodes the 907 amino acids of g ⁇ 350. See U.S. Patent No. 4,707,358 issued to Kieff (1987).
  • the spliced version of the reading frame covers 2130 bases and translates into gp220 protein, a 710 amino acid sequence.
  • the theoretical molecular weights of gp350 protein and gp220 protein are 95 kD and 70 kD, respectively.
  • the measured molecular weights of expressed gp350 protein and gp220 protein vary but are approximately 350 kilodaltons and 220 kilodaltons (kD), respectively.
  • the extensive glycosylation of the proteins accounts for difference between the predicted and actual molecular weights.
  • both gp350 and gp220 proteins are produced at a molar ratio ranging from about 6: 1 to 1:1. For example, in B95-8 cells, which are persistently infected with EBV, the ratio appears to vary but sometimes approaches the 6: 1 range. See. Miller. Proc. Natl. Acad. Sci. 69:383(19721.
  • gp350/220 proteins have been expressed in rat pituitary, Chinese hamster ovary VERO (African green monkey kidney) cells, as well as in yeast cells. See, Whang, J. Virol. 61 : 1796(1982), Motz, Gene 44:353(1986) and Emini, Virology 166:387(19881.
  • a bovine papillomavirus virus expression system has also been used to make gp350/220 proteins in mouse fibroblast cells. See. Madei. Vaccine 10:777(19921.
  • EBV research relating to gp350/220 heretodate has focused either on obtaining efficient expression of the native gp350/220 sequence or on a modified sequence lacking the transmembrane domain, resulting in a mixture of the two alternate spliced versions of the native or transmembrane lacking protein, or on production of epitopic fragment sequences in ⁇ .actosidase fusion proteins.
  • Partially purified preparations of gp350/220 are known. See, Finerty, J. Gen. Virology 73:449(1992) (recombinantly produced, partially purified). With respect to native gp350/220 protein, in most instances, the purification procedures resulted in inactivating the antigenicity of the protein, making it unacceptable for use in a subunit vaccine. However, highly purified preparations of antigenically active gp350 protein from native (i.e., non-recombinant) sources have been reported in the scientific literature. See, David, J. Immunol. Methods 108:231(19881.
  • splice sites facilitate the processing of pre-mRNA molecules into mRNA.
  • splice sites are required for the efficient accumulation of late mRNA's. Alteration of the 3' and 5' splice sites in polyoma virus transcripts decreased or completely blocked mRNA accumulation. See, Treisman, Nature 292:595(1981).
  • EBV biology and disease is generally reviewed in Straus, Annal of Int. Med. 118:45(1993).
  • a description of the EBV BLLFI open reading frame is found in Baer, Nature 310:207(1984).
  • Descriptions of the Epstein-Barr virus gp350/220 DNA and amino acid sequences are found in articles by Beisel, J. Virology 54:665(19851 and Biggin, EMBO J. 3:1083(1984) and in United States Patent No. 4,707, 358 issued to Kieff, et al. (1987).
  • this invention provides non- splicing variants of the EBV gp350/220 DNA sequence.
  • the DNA sequences of the invention may include an isolated DNA sequence that encodes the expression of homogeneous gp350 protein.
  • the DNA sequence coding for gp350 protein is characterized as comprising the same or substantially the same nucleotide sequence in Figure 1 wherein the native nucleotides at the donor and acceptor splice sites are replaced with non-native nucleotides, and fragments thereof.
  • This DNA sequence may include 5' and 3' non-coding sequences flanking the coding sequence and further include an amino terminal signal sequence.
  • Figure 1 illustrates the non-coding sequences and indicates the end of the putative signal sequence with an asterisk.
  • DNA sequences of this invention may exclude some or all of these flanking or signal sequences.
  • the non-splicing variant DNA sequences of the invention are produced by introducing mutations into the Figure 1 DNA sequence in the donor and acceptor splice sites of the gene encoding gp350/220. This eliminates production of gp220 protein so t at only the gp350 protein is produced.
  • the invention comprises homogeneous gp350 proteins, and methods of making the proteins by expression of the non-splicing variant of EBV gp350/220 DNA sequence in an appropriate prokaiyotic or eukaryotic h ⁇ t cell under the control of suitable expression control sequence.
  • homogeneous means free or substantially free from gp220 protein.
  • homogeneous gp350 protein, recombinantly produced in mammalian or insect cells has not to our knowledge ever been reported in the scientific literature heretofore.
  • homogeneous gp350 proteins, additionally having deletions resulting in a secreted product are provided.
  • deletions comprise either removal of the transmembrane region or removal of the transmembrane region and the remaining C-terminus of gp350.
  • Such additionally modified DNA sequences and the proteins encoded thereby are yet another aspect of this invention.
  • a recombinant DNA molecule comprising vector DNA and a
  • DNA sequence encoding homogeneous gp350 protein The DNA molecule provides the gp350 sequence in operative association with a suitable regulatory sequence capable of directing the replication and expression of homogeneous gp350 in a selected host cell. Host cells transformed with such DNA molecules for use in expressing recombinant homogeneous gp350 are also provided by this invention.
  • the DNA molecules and transformed host cells of the invention are employed in another aspect of the invention, a novel process for producing recombinant homogeneous gp350 protein or fragments thereof.
  • a cell line transformed with a DNA sequence encoding a homogeneous gp350 protein or fragment thereof (or a recombinant DNA molecule as described above) in operative association with a suitable regulatory or expression control sequence capable of controlling expression of the protein is cultured under appropriate conditions permitting expression of the recombinant DNA.
  • the expressed protein is then harvested from the host cell or culture medium by suitable conventional means.
  • the process may employ a number of known cells as host cells; presently preferred are mammalian cells and insect cells.
  • the DNA sequences and proteins of the present invention are useful in the production of therapeutic and immunogenic compounds having EBV antigenic determinants. Such compounds find use in subunit vaccines for the prophylactic treatment and prevention of EBV related diseases, such as mononucleosis, Burkitt 's lymphoma and nasopharyngeal carcinoma. Accordingly, in yet another aspect the invention comprises such therapeutic and/or immunogenic pharmaceutical compositions for preventing and treating EBV related conditions and diseases in humans such as infectitious mononucleosis, Burkett's lymphoma and nasopharyngeal carcinoma.
  • Such therapeutic and/or immunogenic pharmaceutical compositions comprise a immunogenically inducing effective amount of one or more of the homogeneous gp350 proteins of the present invention in admixture with a pharmaceutically acceptable carrier such as aluminum hydroxide, saline and phosphate buffered saline as are known in the art.
  • a pharmaceutically acceptable carrier such as aluminum hydroxide, saline and phosphate buffered saline as are known in the art.
  • a pharmaceutically acceptable carrier such as aluminum hydroxide, saline and phosphate buffered saline as are known in the art.
  • the active ingredient may be administered in the form of a liposome-containing aggregate.
  • such pharmaceutical compositions may be formulated as subunit vaccines for administration in human patients. Patients may be vaccinated with a dose sufficient to stimulate antibody formation in the patient; and revaccinated after six months or one year.
  • a further aspect of the invention therefore is a method of treating EBV related diseases and conditions by administering to a patient, particularly to a human patient, an immunogenically inducing therapeutically effective amount of a homogeneous gp350 protein in a suitable pharmaceutical carrier.
  • Still another aspect of the invention is a method of stimulating an immune response against EBV by administering to a patient an immunogenically inducing effective amount of a homogeneous gp350 protein in a suitable pharmaceutical vehicle.
  • Figure 1 illustrates the DNA and amino acid sequence of gp350/220 (From
  • Figure 2 illustrates construction of gp350 deletion and site directed mutants.
  • the plasmid maps labelled pMDTM and pMSTOP exemplify the non-splicing gp350/220 variants of the invention.
  • section (A) a linear model of the gp350 protein is shown approximately to scale with the encoding clone, BLLF1, below.
  • An N-terminal signal sequence (SS) and the transmembrane domains (TM) are indicated on the protein and important restriction sites are indicated on the gene diagram.
  • the gp350 gene was cloned in two segments, the Hindi ⁇ /Bfal BLSH1 fragment and the Banl/Hindm BLSH2 fragment.
  • SC YT was created using the polymerase chain reaction from the region of BLLF1 indicated.
  • (B) the cloning scheme for pDTM, pSTOP, pMDTM, and pMSTOP is illustrated (plasmids not to scale). The details of the cloning are described in Examples 1 and 2. Plasmid maps are marked with the relevant restriction sites, the cloning vectors used and the gp350 gene fragments. Splice site mutations in pMDTM and pMSTOP are indicated by asterisks.
  • FIG 3 illustrates the results of immunoprecipitation of homogeneous gp350 protein from pMDTM clones as analyzed by SDS-PAGE.
  • samples of labeled tissue culture supernatants (S) and gp350/220 precipitations (Ip) were electrophoresed on 5 % SDS-PAGE (polyacrylamide gel electrophoresis). Location of molecular weight markers are indicated on the left side.
  • Figure 4 illustrates the results of Northern blot analysis of protein from pMDTM clones expressed in CHO cells, as described in Example 3.4
  • compositions and methods comprising cloned EBV DNA sequences encoding non-splicing variants of gp350 protein.
  • non-splicing variants are referred to herein as homogeneous gp350 proteins.
  • gp350/220 gene when the gp350/220 gene is expressed in mammalian cells two gene products are generated, gp350 and gp220, due to RNA splicing of the gene.
  • the invention allows for only one gene product, gp350, to be produced.
  • the invention involves removing some or all of the RNA splice site signals in the gp350 gene and expressing the gene in a suitable host cell.
  • Mutations in the gp350/220 gene were introduced to prevent production of the 220 kD version of the protein when the gp350/220 gene is expressed in mammalian cells. As a result, mRNA transcripts encoding only gp350 are produced. The elimination of gp220 expression by using a gp350/220 gene non- splicing variant will result in increased production of gp350 relative to gp220. Production of gp220 is not essential for production of an effective anti-EBV vaccine because gp350 contains all the potential antigenic sites found on gp220.
  • one aspect of this invention provides a DNA sequence encoding a polypeptide sequence substantially the same as gp350, except that the donor splice site codon encoding amino acid 501 and the acceptor splice site codon encoding amino acid 698 have been modified by replacement of native nucleotides with non-native nucleotides.
  • the native nucleotides are replaced with non-native nucleotides such that the amino acid sequence remains the same.
  • native nucleotides AAGT at the donor splice site (nucleotides 1500 through 1504) and native nucleotides A and T flanking the GG acceptor splice site (nucleotides 2091 and 2094) were replaced with nucleotides GTCA and T and A, respectively. Consequently, the Glutamine at amino acid position 500 and the Serine at position 501 remained the same as a result of this substitution in the donor site. Likewise, the Threonine at amino acid position 697 and the Glycine at position 698 remained the same as a result of the modification in the acceptor site.
  • the invention comprises homogeneous gp350 proteins.
  • the homogeneous gp350 proteins are further characterized by having an amino acid sequence substantially the same as that shown in Figure 1 from amino acids 1 through 907, from amino acids 1 through 862 or from amino acids 1 through 907 and excepting amino acids 863 through 881 , each with or without the N-terminal 18 amino acid signal sequence.
  • analogs of homogeneous gp350 proteins are provided and include mutants in which there are variations in the amino acids sequence that retain antigenic activity and preferably have a homology of at least 80%, more preferably 90% , and most preferably 95%, with the corresponding region of the homogeneous gp350 proteins.
  • Examples include proteins and polypeptides with minor amino acid variations from the amino acid sequence of Figure 1 ; in particular, conservative amino acids replacements.
  • Conservative replacements are those that take place within a family of amino acids that are related in their side chains.
  • Phenylalanine, tryptophan and tyrosine are sometimes classified jointly as aromatic amino acids. For example, it is reasonable to expect that an isolated replacement of a leucine or a similar conservative replacement of an amino acid with a structurally related amino acid will not have a major effect on antigenic activity or functionality.
  • the invention offers the advantag *' simpler purification of gp350. Because gp350 and gp220 have s nilar biochemic properties, gp220 is often co-purified in preparations of gp350. Cells expressing only the non-splicing variant of the gp350/220 gene simplifies protein purification. This will reduce the costs of producing gp350.
  • the invention also makes biochemical characterization of the starting material for gp350 purification easier. Because only one species is present, protein content analysis and amino acid sequence analysis may be performed without accounting for the presence of a second species.
  • the invention additionally offers the advantage of increased gp350 production.
  • Prevention of gp350 gene splicing will shift the cell from dual production of gp350 and gp220 to the production of gp350 alone.
  • the concentrations of gp220 have been estimated to be 30% -100% of the gp350 concentration.
  • gp350 production will be increased by the lack of gp220 production.
  • the DNA sequence of the gp350/220 gene is described by Beisel, J. Virology 54:665(1985) and Biggin, EMBO J. 3:1083(1984) and is illustrated in Figure 1.
  • the gene is an open reading frame of 2721 bases, encoding 907 amino acids and specifying a primary translation product of about 95 kD. The difference between predicted and actual values represents extensive glycosylation of the protein. 591 bases (encoding 197 amino acids) are spliced out to produce gp220.
  • the apparent molecular weight of gp350/220 gene products may also vary depending upon the type of measurement system used, glycosylation site utilization in different cell types, post-translational processing differences or selective gene mutation.
  • gp350/220 gene non-splice site variants vary for the products of different gp350/220 gene non-splice site variants but the term "homogeneous gp350 protein or proteins" encompasses gene products of the non- splicing variant, optionally having additional deletions or mutations such as the C- terminal deletions and/or transmembrane modifications also disclosed herein.
  • the term "gp220 protein” refers to the alternatively spliced gp350/220 gene product with a molecular weight of approximately 220 kD. Splice-sites in the gp350/220 gene were identified by comparison of the gp350/220 gene with consensus donor and acceptor splice sequences based on other genes, predominantly from eukaryotic organisms. The consensus sequences developed by Mount, Nucleic Acids Res. 10:459(1982) from studying the splice sites in other genes are:
  • the bases asterisked above represent bases that appear in 100% of all splice sites (highly conserved). Positions with two bases or one base represent conserved positions (non highlighted positions). The slash indicates the actual site of splicing.
  • the donor splice site occurs after nucleotide 1501 and the acceptor splice occurs after nucleotide 2092, as shown by DNA sequencing (Biggin, EMBQ J. 3:1083(1984)) of the g ⁇ 350/220 gene. (The numbering used herein and in Figure 1 conforms to the numbering in Biggin).
  • the splice site occurs in the corresponding gene region in the Type B strain of EBV (the donor splice site after A 1J01 and the acceptor splice site after G 20 _o).
  • the invention encompasses compositions made using either the A or B strain or another EBV strain's splice site to produce a single species of mRNA from the gp350/220 gene.
  • the DNA sequence of the Type A form of the virus from strain B95-8 was used in the Examples although the DNA sequence of the Type B strain could equally have been used, because the translated gene products of Type A and B strains are 98% identical.
  • the B strain lacks amino acids 507 through 520 and 570 through 576.
  • the type A strain was used because it contains all the possible gp350 antigenic sites.
  • EBV gp350/220 having strain-specific sequences could be used in accordance with the teachings herein to produce EBV strain-specific homogeneous gp350 proteins having immunogenic properties specific to a particular strain and t refore useful in immunogenic and/or therapeutic compositions for the prevention or treatment of strain specific EBV related diseases.
  • Table 1 shows the wild type nucleotide and amino acid sequences of the donor and acceptor splice sites. To prevent RNA splicing of the gp350/220 gene, mutations were introduced into the gp350/220 gene nucleic acid sequence to replace the relevant base pairs of the RNA splice site.
  • At least one of the bases out of the two highly conserved bases framing the donor site or acceptor site should be replaced with nonconserved bases, more preferably at least two highly conserved bases should be mutated to nonconserved bases.
  • Other conserved bases more than two bases away from the splice site, can also be replaced with nonconserved splice site bases to further decrease recognition of the splice site. Both the donor and the acceptor site can be changed to impair splicing mechanisms.
  • both the donor and the acceptor contain at least one change each, in one of the four highly conserved splice site base positions, and more preferably at least two changes in two of the four highly conserved splice site base positions. If one splice site is not mutable due to a desire to maintain the wild-type amino acid sequence then it is preferable to introduce at least two mutations to the other splice site.
  • Mutation at the gp350/220 splice sites may introduce changes into the amino acid sequence of the subsequently expressed gp350 protein.
  • changes should be conservative amino acid substitutions.
  • Conservative substitutions in the amino acid sequence, as opposed to nonconservative changes in the amino acid sequence, will help preserve antigenic sites.
  • Conservative amino acid changes can be made as long as the base change (or base changes) result in a suitable change in the invariant donor/acceptor bases. For example, Gly could be substituted for Ser 50 ⁇ at the donor splice site, using any Gly-specific codons other than GGU (use of GGU would preserve the G nucleotide and would not result in the desired GT replacement in the splice signal).
  • Gly 698 to Ala would be a conservative change, but since all Ala codons start with the highly conserved G nucleotide, this would not result in the desired replacement.
  • Proline also might be a conservative amino acid change, proline would not be used to replace a wild type amino acid because it would result in modification of the tertiary structure of the protein and thereby mask one or more gp350 antigenic sites.
  • Table 1 shows the acceptable conservative amino acid replacements in the wild-type sequences. At the bottom of Table 1 is an example of a mutation with conservative amino acid changes.
  • one aspect of the present invention comprises a non-splicing variant of gp350/220
  • additional mutations of the gp350/220 coding sequence may also be desirable.
  • the membrane spanning region also known as the transmembrane region
  • the membrane spanning region of gp350/220 comprises amino acids 861 (methionine) through 881 (alanine). See, Beisel, J. Virology 54:665(1985).
  • the invention provides non-splicing variants of gp350/220 DNA and/or gp350 homogeneous protein additionally comprising at least one deletion in the transmembrane region of the gp350/220 DNA and/or gp350 homogeneous protein that results in the expression of soluble homogeneous gp350 protein.
  • the C-terminal sequence following the transmembrane domain and comprising amino acids 881 through 907 may also be deleted in whole or in part, as described herein, in accordance with the invention.
  • the invention comprises non-splicing variants of gp350/220 DNA and/or homogeneous protein further modified by deletion of all or a portion of the DNA encoding and/or amino acid sequence comprising the transmembrane region of gp350/220 and even further modified by deletion of the remaining C-terminal DNA and/or amino acid sequences of gp350/220.
  • the invention comprises non-splicing variant DNA sequences encoding the homogeneous gp350 proteins of the invention.
  • DNA sequences comprise the DNA sequence of Figure 1 encoding amino acids 1 through 907 and further comprising the nucleotide substitutions taught herein to remove the donor and acceptor splice sites.
  • DNA sequences optionally comprise truncated DNA sequences in which the nucleotides encoding all or part of the transmembrane domain and C-terminus comprising amino acids 861 through 907 are deleted and deletion variants in which the nucleotides encoding all or part of the transmembrane domain comprising amino acids 861 through 881 are deleted.
  • the DNA sequences of the present invention encoding homogeneous gp350 proteins may also comprise DNA capable of hybridizing under appropriate stringency conditions, or which would be capable of hybridizing under such conditions but for the degeneracy of the genetic code, to an isolated DNA sequence of Figure 1. Accordingly, the DNA sequences of this invention may contain modifications in the non-coding sequences, signal sequences or coding sequences, based on allelic variation, species variation or deliberate modification. These non-splicing variant gp350/220 DNA sequences as disclosed herein can be constructed using methods well known in the art. The modified DNA sequences of this invention can be expressed recombinantly, likewise using known methods, to produce the homogeneous gp350 proteins of this invention. Such recombinant proteins can be purified and incorporated into pharmaceutical compositions for the prophylactic treatment and prevention of EBV related diseases.
  • the non-splicing variants of gp350 220 DNA of this invention can be expressed recombinantly in different types of cells using the appropriate expression control systems as is known in the art.
  • suitable cells known and available in the art include, but are not limited to, yeast cells such as Saccharomyces cerevisiae. bacterial cells such as R coli and Bacillus subtilis and mammalian cells such as GH3, CHO, NSO, MDCK and C-127 cells.
  • Vectors used with cell types are selected based on their compatibility with the cell type and expression control system used. Cells and vectors that allow for the expression of secreted products of the gp350/220 gene are preferred.
  • R coli is transformed using derivatives of pBR322 which have been modified using conventional techniques to contain the DNA sequences for expression of the desired protein, in this instance the non-splicing variant sequences of EBV gp350, with or without the sequences encoding the C-terminus and/or membrane spanning region.
  • pBR322 contains genes for ampicillin and tetracycline resistance, which can be used as markers. See, Bolivar, Gene 2:95(1977).
  • promoters for transcription initiation and optionally an operator or enhancer include the beta-lactamase and lac promoter systems (see Chang, Nature 198: 1056(1977)), the tryptophan promoter system (see Goeddel, Nucleic Acids Res. 8:4057(1980)) and the lambda-derived PL promoter and N-gene ribosome binding site (see Shimatake. Nature 292: 128(1981).
  • any available promoter system or expression control system that is compatible with prokaryotic host cells can be used.
  • Other exemplary host cells, plasmid and expression vehicles are disclosed in United States Patent Nos.
  • Insect cells may also be used as host cells employing insect cell expression.
  • the components of the expression system include a transfer vector, usually a bacterial plasmid, which contains both a fragment of the baculovirus genome, and a convenient restriction site for insertion of the heterologous gene or genes to be expressed; a wild type baculovirus with a sequence homologous to the baculovirus-specific fragment in the transfer vector (this allows for the homologous recombination of the heterologous gene in to the baculovirus genome); and appropriate insect host cells and growth media.
  • pAc373 the most commonly used transfer vector for introducing foreign genes into AcNPV.
  • Many other vectors known to those of skill in the art, have also been designed. These include, for example, pVL985 (which alters the polyhedrin start codon from ATG to ATT, and which introduces a BamHI cloning site 32 basepairs downstream from the ATT; see Luckow and Summers, Virology (1989) 17:31.
  • the plasmid usually also contains the polyhedrin polyadenylation signal (Miller et al. (1988) Ann. Rev. Microbiol.. 42:177) and a procaryotic ampicillin-resistance (amp) gene and origin of replication for selection and propagation in R coli.
  • Baculovirus transfer vectors usually contain a baculovirus promoter.
  • a baculovirus promoter is any DNA sequence capable of binding a baculovirus RNA polymerase and initiating the downstream (5' to 3') transcription of a coding sequence (e.g. structural gene) into mRNA.
  • a promoter will have a transcription initiation region which is usually placed proximal to the 5' end of the coding sequence.
  • This transcription initiation region typically includes an RNA polymerase binding site and a transcription initiation site.
  • a baculovirus transfer vector can also have a second domain called an enhancer, which, if present, is usually distal to the structural gene. Expression can be either regulated or constitutive. For insect cell expression technology, see EP patent publication 155 476. Yeast, for example Saccharomyces cervisiae. may also be used as a host cell.
  • plasmid vectors suitable for yeast expression are known, as are promoter and expression control systems. See for example, Myanohara, Proc. natl. Acad. Sci. 80:l(19831(PHO5 promoterl. EP Patent Publication 012 873 (leader sequences), Kurtz, Mol. Cell. Biol. 6:142(19861. Ito, Bacteriol. 153: 163(19831 and Hinnen, Proc. Natl. Acad. Sci. 75:1929(1979)(transformation procedures and suitable vectors). Eukaryotic cells from multicellular organisms may of course also be used as hosts cells for the expression of genes encoding proteins and polypeptides of interest.
  • Useful host cell lines include VERO and HeLa cells, and Chinese hamster ovary cells (CHO).
  • Expression vectors compatible with such cells are also available and typically include promoters and expression control sequences, such as for example, the early and late promoters from SV40 (see Fiers, Nature 273:113(1978)) and promoters from polyoma virus, adenovirus 2, bovine papilloma virus or avian sarcoma virus.
  • Exemplary host cells, promoters, selectable markers and techniques are also disclosed in United States Patent Nos.
  • Transformation of suitable host cells is accomplished using standard techniques appropriate to such cells, such as CaCl 2 treatment for prokaryotes as disclosed in Cohen Proc. Natl. Acad. Sci. 69:2110(19721 and CaPO 4 precipitation for mammalian cells as disclosed in Graham, Virology 52:546(19781.
  • Yeast transformation can be carried out as described in Hsiao, Proc. Natl. Acad. Sci. 76:3829(19791 or as described in Klebe, Gene 25:333(1983).
  • suitable vectors containing the non-splicing variant gp350 sequence is accomplished using conventional ligation and restriction techniques now well known in the art.
  • Site specific DNA cleavage is performed by treating with suitable restriction enzyme(s) under standard conditions, the particulars of which are typically specified by the restriction enzyme manufacturer.
  • Polyacrylamide gel or agarose gel electrophoresis may be performed to size separate the cleaved fragments using standard techniques and the fragments blunt ended by treatment with the Klenow fragment of R co ⁇ polymerase I in the presence of the four deoxynucleotide triphosphates.
  • Treatment with SI nuclease hydrolyzes any single-stranded portions.
  • Synthetic oligonucleotides can be made using for example, the diethylphosphoamidite method known in the art. See United States Patent No. 4,415,732 (1983). Ligations can be performed using T4 DNA ligase under standard conditions and temperatures and correct ligations confirmed by transforming R . cpJi or COS cells with the ligation mixture. Successful transformants are selected by ampicillin, tetracycline or other antibiotic resistance or using other markers as are known in the art.
  • the invention comprises vectors containing the non-splicing variants of gp350/220 DNA sequences and host cells and further comprises a method of making a non-splicing variant of gp350/220 protein by culturing said host cells containing a vector that is carrying a non-splicing variant of a gp350/220 DNA sequence operatively linked to an expression control sequence under culture conditions enabling expression of the homogeneous gp350 protein.
  • the expressed homogeneous gp350 is purified from cell and culture medium constituents using conventional glycoprotein purification techniques such as, but not limited to, ultrafiltration, free flow electrophoresis, gel filtration chromatography, affinity chromatography, SDS-PAGE, differential NH 4 SO 4 precipitation, lectin columns, ion exchange columns and hydrophobicity columns as is known in the art.
  • Small scale analytical preparations of gp350 are most readily purified using SDS-PAGE or lectin affinity columns and such small scale preparations for use in vaccination or immune response experiments are most readily purified using liquid chromatography.
  • gp350 For large scale production of commercially significant quantities of gp350 for use in vaccine compositions, a combination of ultrafiltration, gel filtration, ion exchange, and hydrophobic interaction chromatography are preferred.
  • the purified, homogeneous gp350 proteins of the present invention may be employed in therapeutic and/or immunogenic compositions for preventing and treating EBV related conditions and diseases such as infectitious mononucleosis, Burkitt' s lymphoma and nasopharyngeal carcinoma.
  • Such pharmaceutical compositions comprise an immunogenically-inducing effective amount of one or more of the homogeneous gp350 proteins of the present invention in admixture with a pharmaceutically acceptable carrier, for example an adjuvant/antigen presentation system such as alum.
  • a pharmaceutically acceptable carrier for example an adjuvant/antigen presentation system such as alum.
  • adjuvant/antigen presentation systems for instance, MF59 (Chiron Corp.), QS-21 (Cambridge Biotech Corp.), 3-DMPL (3-Deacyl-Monophosphoryl Lipid A) (RibilmmunoChem Research, Inc.), clinical grade incomplete Freund's adjuvant (IFA), fusogenic liposomes, water soluble polymers or Iscoms (Immune stimulating complexes) may also be used.
  • Other exemplary pharmaceutically acceptable carriers or solutions are aluminum hydroxide, saline and phosphate buffered saline.
  • the composition can be systemically administered, preferably subcutaneously or intramuscularly, in the form of an acceptable subcutaneous or intramuscular solution.
  • inoculation can be effected by surface scarification or by inoculation of a body cavity.
  • the preparation of such solutions, having due regard to pH, isotonicity, stability and the like is within the skill in the art.
  • the dosage regimen will be determined by the attending physician considering various factors known to modify the action of drugs such as for example, physical condition, body weight, sex, diet, severity of the condition, time of administration and other clinical factors. Exemplary dosage ranges comprise between about 1 ⁇ g to about 1000 ⁇ g of protein.
  • an immunologically- inducing effective amount of homogeneous gp350 protein is administered to a human patient in need of therapeutic or prophylactic treatment.
  • An immunologically inducing effective amount of a composition of this invention is contemplated to be in the range of about 1 microgram to about 1 milligram per dose administered. The number of doses administered may vary, depending on the above mentioned factors.
  • the invention is further described in the following examples, which are intended to illustrate the invention without limiting its scope.
  • the gp350/220 gene from the EBV B95-8 strain (Miller, et al , 1972), is available in a BamHI library as an open reading frame called BLLFl (Baer, Nature 310:207. 1984).
  • BLLFl BamHI library
  • the gp350/220 gene was cloned in two parts: 1) BLSH1, a 2.3 kb Hin ⁇ TH/Bfal 3' fragment and 2) BLSH2, a 337 b.p. Banl/Hindm 5' fragment ( Figure 2A).
  • the plasmid pDTM is comprised of a gp350/220 nucleic acid sequence that lacks a complete TM coding region. This construct was made using two staging vectors pSTGl and pSTG3. A 450 bp PCR product, SYCT, that introduced a Bfal site at the 3' end of the TM region was made using a BLLFl clone target sequence ( Figure 2).
  • PCR primers used are as follows:
  • Primer 1 GG ATC CTA GAC TGC GCC TTT AGG CGT A
  • BLLFl ... GAC TGC GCC TTT AGG CGT A.. A.A.: ... Asp Cys Ala Phe Arg Arg ...
  • Primer 2 GGA TCC TCT GTT CCT TCT GCT CCA GTG BLLFl: TCT GTT CCT TCT GCT CCA GTG
  • Primer 1 The Bfal site of Primer 1 was used to clone a Bfal/Xmal fragment of SCYT into pSTGl .
  • the remainder of Primer 1 corresponds to the amino acid sequence encoded by clone BLLFl .
  • Primer 2 corresponds to a region outside the gp350/220 open reading frame on the 3' side of the gene.
  • the SCYT PCR fragment was cut with Bfal and Xmal to produce a 136 base pair fragment which was cloned into a pMTll vector (Spaete and Mocarski, 1985) along with a second fragment, a BLSH1 Hindi ⁇ /Bfal fragment, to create pSTGl .
  • the plasmid pSTOP comprises a gp350/220 gene that lacks a TM region and the C-terminal cytoplasmic region adjacent the TM region.
  • a 16 base pair Bfal/EcoRI oligonucleotide linker was created with stop codons (underlined) in three frames following the Bfal sticky end as shown below:
  • the 5' overhang (TA) of the upper sequence is a sticky end for a Bfal restriction site and the 5' overhang (TTAA) of the lower sequence is an EcoRI sticky end.
  • This 16 base pair linker was used to clone a BLSH1 HindHI/Bfal fragment into pMTll, in order to create pSTG2.
  • a 2.3 kb pSTG2 Hindm/EcoRI fragment and the pSTG3 0.3 kb Xbal/Hindm fragment were cloned into ⁇ EE14 to create pSTOP.
  • oligonucleotide sequence and translated amino acid sequence of the wild type, pSTOP, and pDTM 3' ends of gp350 DNA and amino acid sequences are shown in Table 2 below. Arrows indicate the beginning and end of the wild-type transmembrane domain (TM). Only two amino acids from the transmembrane domain are retained in pDTM and pSTOP, Met 861 and Leu g62 (see also Figure 1). Note that a stop codon immediately follows Leu 862 in pSTOP. In pDTM the former location of the deleted transmembrane region is marked " ⁇ TM" . (In the Table, the native amino acids are indicated.)
  • the highly conserved and conserved bases of the gp350/220 gene splice site were changed. Four bases were changed in the donor splice site, including the highly conserved GT pair that occurs in 100% of all splice sites. Two conserved donor site bases, AA, were replaced with GT. The two highly conserved (invariant) donor splice site bases were changed from GT to CA. At the acceptor splice site, only one of the highly conserved acceptor splice site bases was altered to preserve the amino acid sequence. A second conserved acceptor splice site base was changed as indicated in Table 3. Table 3 summarizes the bases changed in the donor and acceptor splice sites of the gp350/220 gene.
  • Acceptor Splice site acceptor acceptor
  • the bases changed by oligonucleotide-based mutagenesis are marked with an asterisk in the mutant sequences.
  • the actual site of splicing is indicated by an arrow, and the encoded amino acids are shown. Note that the amino acid sequence does not change as a result of the nucleotide substitutions.
  • BamHI/XhoI fragments of the gp350/220 nucleotide sequence were cloned into the polylinker of plasmid M13TAC using Asp718 and BamHI restriction sites on the poly linker, combined with a 19 bp oligonucleotide linker containing Asp718 and Xhol sticky ends.
  • the plasmids M13DTM and M13STOP of Example 1 ( Figure 2B), were used for the mutagenesis.
  • PrDonorl and PrAcceptorl Two 42-mer oligonucleotides, PrDonorl and PrAcceptorl , were made for use in the mutagenesis. Each was designed to be complementary to gp350/220 gene sequences centering on either the donor or acceptor splice sites. The only region of the oligonucleotides that were not complementary to the gp350/220 gene were the bases representing the desired mutations. Mutagenesis oligonucleotides comprised the following: PrDonorl
  • PrAcceptorl Primer CTG TGT TAT ATT TTC ACC TC J C AGT TGG GTG AGC GGA GGT TAG
  • the sequence of the mutagenesis oligonucleotides are labelled "Primer,” while the DNA sequence spanning the gp350/220 gene splice sites are labelled "EBV.” Bases that were changed as a result of the mutagenesis are marked with an asterisk. The dashed line indicated the location of the splice.
  • the oligonucleotides PrDonorl and PrAcceptorl were hybridized to single- stranded clones of M13-DTM and M13-STOP. T4 DNA polymerase holoenzyme was used to produce double-stranded M13 DNA and E. coli was transformed with the double-stranded DNA.
  • any clone that contained the desired mutation could be identified by a color change from white to blue in the presence of X-gal and isothiopropylgalactate. Blue plaques were picked and grown up, and DNA sequencing across splice junctions was used for the final identification of mutant clones, labelled M13-MDTM and M13-MSTOP.
  • One method for producing high levels of homogeneous gp350 protein of the invention from mammalian cells involves the construction of cells containing multiple copies of the heterologous gp350 DNA sequence.
  • the heterologous DNA sequence is operatively linked to an amplifiable marker, in this example, the glutamine synthetase gene for which cells can be amplified using methionine sulphoximine.
  • the pMDTM and pMSTOP vectors made in Example 2 were transfected into CHO cells as discussed below, according to the procedures of Crockett,
  • CHO-K1 cells ATCC CCR61 were maintained in glutamine-free EMEM (Eagles Minimal Essential Medium) supplemented with 10% fetal bovine serum, 100 units/ml penicillin, 100 mg/ml streptomycin, MEM (Modified Eagle's
  • Plasmid DNA from the pMDTM, pMSTOP plasmids was applied by CaPO 4 precipitation using conventional techniques. 10 ⁇ gs of each plasmid DNA precipitate was incubated with the CHO-K1 cells plus 2 ml of serum-free media at
  • Clones were picked by scraping colonies off the dish with a sterile 200 ⁇ l pipetman tip and transferred to one well of a 96-well plate in media without methionine sulphoximine. 1-2 days later the media was replaced with media + 25 ⁇ M methionine sulphoximine. After 4 days the culture supernatants were harvested and assayed for protein products in an ELISA assay, as discussed below.
  • CHO cells were also transfected with the pEE14 control vector alone (which contains no EBV sequences) and 24 clones of CHO-pEE14 were also picked and transferred to plates to serve as controls. (The control clones were identified on the basis of survival in methionine sulphoximine.)
  • ELISA Assay Following transfection, 241 clones of CHO-pMDTM and 158 clones of CHO- pMSTOP were picked and grown up. Supernatants from these clones were tested for gp350 protein production. 96-well plates were coated with affinity-purified rabbit anti-gp350/220 antibody (antibody MDP1; gift of Andrew Morgan) diluted 1:2000 in 50mM sodium borate buffer, pH 9. The plates were incubated at 37 °C for 3-4 hours and washed 3 times with PBS + 0.05 % Tween 20 using a Nunc ImmunoWasher.
  • the plates were blocked by incubating with 2 % BSA in PBS + 0.01 % Thimerosal at 37°C for 0.5 hours and washed again. Supernatants from the transfected cells and control cells were added to the wells and incubated for 2 hours at 37 °C. The plates were then incubated with the primary detection antibody, a mouse monoclonal antibody against gp350/220 (antibody #C65221M; Biodesign
  • tissue culture supernatants from the pMDTM transfections were assayed for activity in a Western Blot.
  • CHO cell supernatants were purified on 5 % SDS-PAGE gels, transferred to nitrocellulose overnight, and probed with anti-gp350 antibodies. Seven pMDTM clones were found to be positive for gp350 in the Western blot analysis.
  • the pMDTM clones that were positive in the Western blot were further tested by radioimmunoprecipitation for the presence of gp220.
  • Selected transformed pMDTM cells, pEE14 control and GH3 ⁇ 19 control cells (described below) were grown overnight in six-well plates so that they were approximately three-quarters confluent on the day of the experiment. Each well contained approximately 5X10 6 cells.
  • the media was removed from each well and replaced with 0.7 ml of methionine-free MEM (10% fetal calf serum) + 100 ⁇ Ci 35 S -methionine. The cells were incubated 5.5 hours at 37°C and then microcentrifuged at 4000 rpm for
  • gp350 protein in the supernatant was immunoprecipitated by addition of 10 ⁇ l of Sepharose-Protein A (Sigma) in a 50% slurry and 20 ⁇ l monoclonal anti-gp350/220 (antibody #C65221M, lOOmg/ml; Biodesign International), with overnight rocking at 4°C.
  • the mixture was then pelleted at 2000 rpm, 2 minutes at room temperature in a microcentrifuge and washed four times with several volumes of phosphate-buffered saline. After the final wash, all liquid was removed from the pellet and replaced with 50 ⁇ l protein gel sample buffer.
  • CHO cells were transfected with the pEE14 vector alone and selected by methionine sulphoximine in parallel with the pMDTM transfection.
  • supernatants S
  • Ip immunoprecipitates
  • control lane 2 precipitation from the GH3 ⁇ 19 control cells results in two strong protein bands at approximately 220 and 350 kD demonstrating production of the truncated splice variant gp350 and gp220 proteins in about a 1 : 1 ratio.
  • This homogeneous gp350 protein expressed in CHO cell lines, or in other mammalian or non-mammalian cell lines, can be further scaled up and homogenous gp350 protein can be isolated and purified from conditioned medium from the cell line using methods familiar in the art, including techniques such as lectin-affinity chromatography, reverse phase HPLC, FPLC, gel filtration and the like. See David, J. Immunol. Methods 108:231(1988) and Madej, Vaccine 10:777(19921.
  • DNA probes complementary to gp350 were made from pDTM, see Example 1.
  • XP464 recognizes both gp350 and gp220.
  • pDTM was cut with Ncol and Ndel, two overlapping 580 b.p. fragments were isolated, and this mixture was cut with XmnI to eliminate one contaminating fragment and with AM to yield a 537 b.p. Alul/Ndel fragment internal to the gp350/220 splice sites.
  • AN537 is specific for the region spliced out of gp220 and is thus specific for gp350 message.
  • DNA probes were labelled by 32 P-dCTP nick translation (Amersham) using DNA fragments XP464 and AN537.
  • MDTM-1 and CHO-DTM-7 cells 90% confluent, was aspirated and the cells were lysed and scraped off in denaturing buffer (10 ml 4M Guanidine thiocyanate, 25mM sodium citrate, pH 7, 0.5 % sarkosyl, 100 mM 2-mercaptoethanol).
  • denaturing buffer 10 ml 4M Guanidine thiocyanate, 25mM sodium citrate, pH 7, 0.5 % sarkosyl, 100 mM 2-mercaptoethanol.
  • denaturing buffer 10 ml 4M Guanidine thiocyanate, 25mM sodium citrate, pH 7, 0.5 % sarkosyl, 100 mM 2-mercaptoethanol.
  • denaturing buffer 10 ml 4M Guanidine thiocyanate, 25mM sodium citrate, pH 7, 0.5 % sarkosyl, 100 mM 2-mercaptoethanol.
  • Each 10 ml lysate was supplemented with 1 m
  • RNA was precipitated from the aqueous phase by addition of one volume of isopropanol at 20 °C for 1 hour, pelleted at 4°C, resuspended in denaturing buffer and reprecipitated. The RNA pellet was washed IX in 70% ethanol, dried in a Speed- Vac and resuspended in DEPC -treated water.
  • DTM-7 and MDTM-1 whole cell RNA was denatured at 65 °C for 15 min in 15% formamide and 6% formaldehyde, run on 1 % agarose/6.6% formaldehyde gels and transferred to nitrocellulose by capillary action and probed with labeled XP464 and AN537.
  • the DNA probes were denatured by boiling 5 minutes, hybridized in 5X SSPE at 65 %C overnight and the nitrocellulose was washed at high stringency. Autoradiography was performed using a Bio-Rad phosphorimager. Northern blots of total cell RNA from CHO-MDTM cells show the effectiveness of splice mutations in preventing gp220-specific RNA production.
  • the reason for the difference in apparent mobility of the gp350 message in MDTM-1 versus DTM-7 lanes is not known.
  • the MDTM-1 species is overloaded compared with DTM-7, which may affect migration in the gel.
  • the gp350 message runs close to a large ribosomal RNA band on the gel, which may distort the apparent molecular weight. Either way, the presence of a single species complementary to gp350 DNA sequences suggests that this signal represents gp350 mRNA.
  • mutations in the donor and acceptor splice sites are effective at preventing gp220 message production, as judged by
  • the purified homogeneous gp350 proteins are incorporated into appropriate vehicles for administration and administered to mice as follows.
  • a 2x adjuvant- vehicle concentrate is prepared by mixing Pluronic L121 and squalane in 0.4% (v/v) Tween 80 in phosphate buffered saline with (Thr 1 ) MDP in accordance with the procedure of David, J. Immunol. Methods 108:231(19881 and Allison, J. Immunol. Methods 95: 157(19861.
  • composition for administration is prepared by addition of equal volumes of protein and adjuvant- vehicle on the day of administration.
  • the protein content should be with range of 5 micrograms to 50 micrograms per dose.
  • mice are immunized with three 0.1 ml intramuscular injections at 0, 21 and 42 days.
  • a pre-immunization bleed and successive bleeds taken 10 days after each injection are obtained from the retro-orbital sinus.
  • Serum antibody levels are determined by an ELISA according to the procedures described in Example 3.
  • EBV neutralizing antibodies in the sera are quantified by their ability to inhibit transformation of fetal cord blood lymphocytes by EBV in vitro according to the methods of Moss, J Gen. Virol. 17:233(19721 and
  • New Zealand white rabbits are inoculated by intramuscular administration of five doses of protein emulsified in the foregoing adjuvant at 0, 21 , 42, 63 and 84 days.
  • the dose should be in the range of about 5 ⁇ g to 50 ⁇ g per inoculation.
  • Sera is obtained two weeks following the last dose and tested for antibody titers to the antigen, for cross-reactive antibody to viral gp350/220 from B95-8 cells and for in vitro EBV-neutralizing activity following the methods of Emini, Virology 166:387(19881.
  • CTGTGTTATA TTTTCACCAC CTGTTGGGTG AGCGGAGGTT AG 42
  • GGC CAG CTG ACA CCC CAT ACG AAG GCT GTC TAC CAA CCT CGA GGT GCA 1256 Gly Gin Leu Thr Pro His Thr Lys Ala Val Tyr Gin Pro Arg Gly Ala 70 75 80
  • AAG GTT ATA TTC TCC AAG GCA CCC GAG AGC ACC ACC ACC TCC CCT ACC 2312 Lys Val He Phe Ser Lys Ala Pro Glu Ser Thr Thr Thr Ser Pro Thr 420 425 430
  • CAA CCA AAA AAT GCA ACC AGT GCT GTT ACC ACA GGC

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pulmonology (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Otolaryngology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Addition Polymer Or Copolymer, Post-Treatments, Or Chemical Modifications (AREA)
  • Treatment Of Liquids With Adsorbents In General (AREA)
  • Detergent Compositions (AREA)
  • Transition And Organic Metals Composition Catalysts For Addition Polymerization (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Polarising Elements (AREA)
  • Paper (AREA)
  • Fats And Perfumes (AREA)

Abstract

Composition comprising gp350 variant DNA and amino acid sequences are provided, as are vectors and host cells containing such sequences. Also provided is a process for producing homogeneous gp350 protein recombinantly and in the absence of production of gp220 protein, pharmaceutical compositions containing such protein and prophylactic uses.

Description

NON-SPLICING VARIANTS OF gp350/220
Epstein-Barr virus (EBV), a member of the herpesvirus group, causes infectious mononucleosis in humans. The disease affects more than 90% of the population. Health analysts estimate the cost of the disease in the United States is 100 million dollars per year. The virus is spread primarily by exchange of saliv: from individuals who shed the virus. Children infected with EBV are largely asym tomatic or have very mild symptoms, while adolescents and adults who become infected develop typical infectious mononucleosis, characterized by fever, pharyngitis, and adenopathy. People who have been infected maintain anti-EBV antibodies for the remainder of their lives, and are thus immune to further infection. Currently there is no commercially available EBV vaccine.
In addition to its infectious qualities, EBV has been shown to transform lymphocytes into rapidly dividing cells and has therefore been implicated in several different lymphomas, including Burkitt's lymphoma and oral hairy leukoplakia. EBV has also been detected in tissue' samples from nasopharyngeal tumors. Worldwide it is estimated that 80,000 cases of nasopharyngeal cancer occur and it is more prevalent in ethnic Chinese populations.
Development of a live, attenuated vaccine for EBV has been and still is problematic. Because of the potential oncogenic nature associated with EBV, researchers have been reluctant to use a live vaccine approach. This invention overcomes the problems associated with live vaccine development by creating methods and compositions for a subunit vaccine, that does not require the use of a potentially oncogenic live virus. A subunit vaccine uses one or more antigenic proteins from the virus that will elicit an immune response and confer immunity.
Two of the more important antigenic EBV proteins are glycoprotein(s) gp350/300 and gp220/200 that form part of the viral membrane envelope and allow virus particles to bind to and enter human target cells by interacting with the cellular membrane protein, CD21. See Nemerow. J. Virology 61:1416(1987 . They have long been singled out as subunit vaccine candidates but difficulties in obtaining antigenically active protein purified from native sources and low yields from recombinantly produced sources have hampered efforts of researcher and vaccine developers. In the literature these proteins are referred to using a variety of molecular weight ranges (350 or 300 kilodaltons (kD) for one of the proteins and 220 or 200 kDs for the other protein). The gp350 or 300 protein is herein referred to as gp350 protein and the gp220 or 200 protein is herein referred to as gp220 protein. Collectively, both proteins are herein referred to as gp350/220 protein(s).
An alternatively spliced, single gene encodes the gp350/220 proteins and results in the generation of gp350 and gp220 mRNA transcripts; no naturally occurring variations in the gp350/220 gene splice sites are known. The gene produces two expression products, the gp350 and gp220 proteins. The open reading frame for the gp350/220 DNA sequence is 2721 base pairs (bp). The entire reading frame encodes the 907 amino acids of gρ350. See U.S. Patent No. 4,707,358 issued to Kieff (1987). The spliced version of the reading frame covers 2130 bases and translates into gp220 protein, a 710 amino acid sequence. The theoretical molecular weights of gp350 protein and gp220 protein are 95 kD and 70 kD, respectively. The measured molecular weights of expressed gp350 protein and gp220 protein vary but are approximately 350 kilodaltons and 220 kilodaltons (kD), respectively. The extensive glycosylation of the proteins accounts for difference between the predicted and actual molecular weights. In any one cell, both gp350 and gp220 proteins are produced at a molar ratio ranging from about 6: 1 to 1:1. For example, in B95-8 cells, which are persistently infected with EBV, the ratio appears to vary but sometimes approaches the 6: 1 range. See. Miller. Proc. Natl. Acad. Sci. 69:383(19721.
Similarly, recombinant production of these glycoproteins has heretofore usually resulted in a mixture of gp350 and gp220 protein being produced. Heretodate, the gp350/220 proteins have been expressed in rat pituitary, Chinese hamster ovary VERO (African green monkey kidney) cells, as well as in yeast cells. See, Whang, J. Virol. 61 : 1796(1982), Motz, Gene 44:353(1986) and Emini, Virology 166:387(19881. A bovine papillomavirus virus expression system has also been used to make gp350/220 proteins in mouse fibroblast cells. See. Madei. Vaccine 10:777(19921. Laboratory and vaccine strains of Vaccinia virus have also been used to express gp 350/220 proteins. Modified recombinant versions of the EBV gp350/220 DNA and protein are known in the art. Specifically, recombinant truncated constructs of the gp350/220 gene lacking the membrane spanning sequence have been made. Such constructs still produce a mixture of the two gp 350 and gp220, but deletion of the membrane spanning region permits secretion of the proteins. See, Finerty, J. Gen. Virology 73:449(1992) and Madej, Vaccine 10:777(1992). Also, various recombinantly produced restriction fragments and fusion proteins comprising various gp350/220 sequences have also been made and expressed in E. coli. See EP Patent Publication 0 173 254 published July 24, 1991.
Accordingly, EBV research relating to gp350/220 heretodate has focused either on obtaining efficient expression of the native gp350/220 sequence or on a modified sequence lacking the transmembrane domain, resulting in a mixture of the two alternate spliced versions of the native or transmembrane lacking protein, or on production of epitopic fragment sequences in β .actosidase fusion proteins.
Partially purified preparations of gp350/220 are known. See, Finerty, J. Gen. Virology 73:449(1992) (recombinantly produced, partially purified). With respect to native gp350/220 protein, in most instances, the purification procedures resulted in inactivating the antigenicity of the protein, making it unacceptable for use in a subunit vaccine. However, highly purified preparations of antigenically active gp350 protein from native (i.e., non-recombinant) sources have been reported in the scientific literature. See, David, J. Immunol. Methods 108:231(19881. Additionally recombinant vaccine virus expressing gp350/220 protein was used to vaccinate cottontop tamarins against EBV-induced lymphoma. See, Morgan, J. Med. Virology 25:189(1988), Mackett, EMBO J. 4:3229(1985) and Mackett, VACCINES '86. pp293(Lerner RA, Chanock RM, Brown F Eds., 1986, Cold Spring Harbor Laboratory). However, the viral gp350/220 DNA sequence has not heretofore been engineered so as to enable expression solely of either one of the alternate spliced versions of the gene, thereby enabling and ensuring the production of pure gp350 or gp220 protein. Nor has a recombinant or mutant virus been made that expresses one or the other of the gp350 or gp220 proteins. Generally, splice sites facilitate the processing of pre-mRNA molecules into mRNA. In polyoma virus, splice sites are required for the efficient accumulation of late mRNA's. Alteration of the 3' and 5' splice sites in polyoma virus transcripts decreased or completely blocked mRNA accumulation. See, Treisman, Nature 292:595(1981). In SV40 virus, excisable intervening sequences facilitate mRNA transport out of the nucleus and mRNA stabilization in the nucleus and because these intron/exon junction sequences facilitate binding of small, nuclear, RNP particles, it is thought that prespliced mRNA's might fail to associate properly with processing pathways. It has been shown that point mutations at exon/intron splice sites reduce exon/intron cleavage and can disrupt pre-mRNA processing, nuclear transport and stability. See, Ryu, J. Virology 63:4386(1989) and Gross, Nature 286:634(19801.
Therefore, until the present invention, the effect of splice site modification on the functional expression and antigenic activity of the proteins encoded by the EBV gp350/220 sequence was at best unknown and unpredictable.
Additional background literature includes the following. EBV biology and disease is generally reviewed in Straus, Annal of Int. Med. 118:45(1993). A description of the EBV BLLFI open reading frame is found in Baer, Nature 310:207(1984). Descriptions of the Epstein-Barr virus gp350/220 DNA and amino acid sequences are found in articles by Beisel, J. Virology 54:665(19851 and Biggin, EMBO J. 3:1083(1984) and in United States Patent No. 4,707, 358 issued to Kieff, et al. (1987). A comparison of DNA sequences encoding gp350/220 in Epstein-Barr virus types A and B is disclosed in Lees, Virology 195:578(1993). Monoclonal antibodies that exhibit neutralizing activity against gp350/220 glycoprotein of EBV are disclosed in Thorley-Lawson, Proc. Natl. Acad. Sci. 77:5307(19801. Lastly, splice site consensus sequences for donor and acceptor splice sites are disclosed in Mount, Nucleic Acids Res.10:459(19821.
In one aspect this invention provides non- splicing variants of the EBV gp350/220 DNA sequence. The DNA sequences of the invention may include an isolated DNA sequence that encodes the expression of homogeneous gp350 protein. The DNA sequence coding for gp350 protein is characterized as comprising the same or substantially the same nucleotide sequence in Figure 1 wherein the native nucleotides at the donor and acceptor splice sites are replaced with non-native nucleotides, and fragments thereof. This DNA sequence may include 5' and 3' non-coding sequences flanking the coding sequence and further include an amino terminal signal sequence. Figure 1 illustrates the non-coding sequences and indicates the end of the putative signal sequence with an asterisk. It is understood, however, that the DNA sequences of this invention may exclude some or all of these flanking or signal sequences. The non-splicing variant DNA sequences of the invention are produced by introducing mutations into the Figure 1 DNA sequence in the donor and acceptor splice sites of the gene encoding gp350/220. This eliminates production of gp220 protein so t at only the gp350 protein is produced.
Accordingly, in another aspect the invention comprises homogeneous gp350 proteins, and methods of making the proteins by expression of the non-splicing variant of EBV gp350/220 DNA sequence in an appropriate prokaiyotic or eukaryotic h< t cell under the control of suitable expression control sequence. As the term is used here with respect to gp350 proteins, homogeneous means free or substantially free from gp220 protein. We note that homogeneous gp350 protein, recombinantly produced in mammalian or insect cells, has not to our knowledge ever been reported in the scientific literature heretofore. In yet another aspect, homogeneous gp350 proteins, additionally having deletions resulting in a secreted product are provided. Such deletions comprise either removal of the transmembrane region or removal of the transmembrane region and the remaining C-terminus of gp350. Such additionally modified DNA sequences and the proteins encoded thereby are yet another aspect of this invention. Also provided is a recombinant DNA molecule comprising vector DNA and a
DNA sequence encoding homogeneous gp350 protein. The DNA molecule provides the gp350 sequence in operative association with a suitable regulatory sequence capable of directing the replication and expression of homogeneous gp350 in a selected host cell. Host cells transformed with such DNA molecules for use in expressing recombinant homogeneous gp350 are also provided by this invention.
The DNA molecules and transformed host cells of the invention are employed in another aspect of the invention, a novel process for producing recombinant homogeneous gp350 protein or fragments thereof. In this process a cell line transformed with a DNA sequence encoding a homogeneous gp350 protein or fragment thereof (or a recombinant DNA molecule as described above) in operative association with a suitable regulatory or expression control sequence capable of controlling expression of the protein is cultured under appropriate conditions permitting expression of the recombinant DNA. The expressed protein is then harvested from the host cell or culture medium by suitable conventional means. The process may employ a number of known cells as host cells; presently preferred are mammalian cells and insect cells. The DNA sequences and proteins of the present invention are useful in the production of therapeutic and immunogenic compounds having EBV antigenic determinants. Such compounds find use in subunit vaccines for the prophylactic treatment and prevention of EBV related diseases, such as mononucleosis, Burkitt 's lymphoma and nasopharyngeal carcinoma. Accordingly, in yet another aspect the invention comprises such therapeutic and/or immunogenic pharmaceutical compositions for preventing and treating EBV related conditions and diseases in humans such as infectitious mononucleosis, Burkett's lymphoma and nasopharyngeal carcinoma. Such therapeutic and/or immunogenic pharmaceutical compositions comprise a immunogenically inducing effective amount of one or more of the homogeneous gp350 proteins of the present invention in admixture with a pharmaceutically acceptable carrier such as aluminum hydroxide, saline and phosphate buffered saline as are known in the art. By "immunogenically inducing" we mean an amount sufficient for stimulating in a mammal the production of antibodies to EBV. Alternatively, the active ingredient may be administered in the form of a liposome-containing aggregate. For prophylactic use, such pharmaceutical compositions may be formulated as subunit vaccines for administration in human patients. Patients may be vaccinated with a dose sufficient to stimulate antibody formation in the patient; and revaccinated after six months or one year.
A further aspect of the invention therefore is a method of treating EBV related diseases and conditions by administering to a patient, particularly to a human patient, an immunogenically inducing therapeutically effective amount of a homogeneous gp350 protein in a suitable pharmaceutical carrier. Still another aspect of the invention is a method of stimulating an immune response against EBV by administering to a patient an immunogenically inducing effective amount of a homogeneous gp350 protein in a suitable pharmaceutical vehicle. Figure 1 illustrates the DNA and amino acid sequence of gp350/220 (From
Beisel, J. Virology 54:665(1985)). The donor and acceptor splice sites are indicated. The transmembrane region is delineated with the horizontal arrows and an asterisk (*) marks the end of the putative signal sequence. Nucleotide numbering is shown at the left; amino acid numbering at the right.
Figure 2 illustrates construction of gp350 deletion and site directed mutants. The plasmid maps labelled pMDTM and pMSTOP exemplify the non-splicing gp350/220 variants of the invention. In section (A), a linear model of the gp350 protein is shown approximately to scale with the encoding clone, BLLF1, below. An N-terminal signal sequence (SS) and the transmembrane domains (TM) are indicated on the protein and important restriction sites are indicated on the gene diagram. The gp350 gene was cloned in two segments, the Hindiπ/Bfal BLSH1 fragment and the Banl/Hindm BLSH2 fragment. SC YT was created using the polymerase chain reaction from the region of BLLF1 indicated. In (B), the cloning scheme for pDTM, pSTOP, pMDTM, and pMSTOP is illustrated (plasmids not to scale). The details of the cloning are described in Examples 1 and 2. Plasmid maps are marked with the relevant restriction sites, the cloning vectors used and the gp350 gene fragments. Splice site mutations in pMDTM and pMSTOP are indicated by asterisks.
Figure 3 illustrates the results of immunoprecipitation of homogeneous gp350 protein from pMDTM clones as analyzed by SDS-PAGE. Positive control (GH3Δ19) cells secreting a truncated form of the gp350/220 proteins, negative control (pEE14) cells and several pMDTM clones were metabolically labeled with 35S-methionine for 5.5 hours; homogeneous gp350 protein was immunoprecipitated from the resulting tissue culture supernatants. For each cell type, samples of labeled tissue culture supernatants (S) and gp350/220 precipitations (Ip) were electrophoresed on 5 % SDS-PAGE (polyacrylamide gel electrophoresis). Location of molecular weight markers are indicated on the left side. Figure 4 illustrates the results of Northern blot analysis of protein from pMDTM clones expressed in CHO cells, as described in Example 3.4
Disclosed are compositions and methods comprising cloned EBV DNA sequences encoding non-splicing variants of gp350 protein. As noted, such non- splicing variants are referred to herein as homogeneous gp350 proteins. Normally, when the gp350/220 gene is expressed in mammalian cells two gene products are generated, gp350 and gp220, due to RNA splicing of the gene. The invention allows for only one gene product, gp350, to be produced. The invention involves removing some or all of the RNA splice site signals in the gp350 gene and expressing the gene in a suitable host cell. Mutations in the gp350/220 gene were introduced to prevent production of the 220 kD version of the protein when the gp350/220 gene is expressed in mammalian cells. As a result, mRNA transcripts encoding only gp350 are produced. The elimination of gp220 expression by using a gp350/220 gene non- splicing variant will result in increased production of gp350 relative to gp220. Production of gp220 is not essential for production of an effective anti-EBV vaccine because gp350 contains all the potential antigenic sites found on gp220.
Therefore, one aspect of this invention provides a DNA sequence encoding a polypeptide sequence substantially the same as gp350, except that the donor splice site codon encoding amino acid 501 and the acceptor splice site codon encoding amino acid 698 have been modified by replacement of native nucleotides with non-native nucleotides. Preferably the native nucleotides are replaced with non-native nucleotides such that the amino acid sequence remains the same. Specifically, in the example, native nucleotides AAGT at the donor splice site (nucleotides 1500 through 1504) and native nucleotides A and T flanking the GG acceptor splice site (nucleotides 2091 and 2094) were replaced with nucleotides GTCA and T and A, respectively. Consequently, the Glutamine at amino acid position 500 and the Serine at position 501 remained the same as a result of this substitution in the donor site. Likewise, the Threonine at amino acid position 697 and the Glycine at position 698 remained the same as a result of the modification in the acceptor site.
Analogously, substitutions other than those specifically exemplified could readily be performed by one skilled in the art as is more fully described below.
Therefore, in one aspect the invention comprises homogeneous gp350 proteins. The homogeneous gp350 proteins are further characterized by having an amino acid sequence substantially the same as that shown in Figure 1 from amino acids 1 through 907, from amino acids 1 through 862 or from amino acids 1 through 907 and excepting amino acids 863 through 881 , each with or without the N-terminal 18 amino acid signal sequence. In addition, analogs of homogeneous gp350 proteins are provided and include mutants in which there are variations in the amino acids sequence that retain antigenic activity and preferably have a homology of at least 80%, more preferably 90% , and most preferably 95%, with the corresponding region of the homogeneous gp350 proteins. Examples include proteins and polypeptides with minor amino acid variations from the amino acid sequence of Figure 1 ; in particular, conservative amino acids replacements. Conservative replacements are those that take place within a family of amino acids that are related in their side chains. Genetically encoded amino acids are generally divided into four families: (1) acidic = aspartate, glutamate; (2) basic = lysine, arginine, histidine; (3) non-polar = alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar = glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Phenylalanine, tryptophan and tyrosine are sometimes classified jointly as aromatic amino acids. For example, it is reasonable to expect that an isolated replacement of a leucine or a similar conservative replacement of an amino acid with a structurally related amino acid will not have a major effect on antigenic activity or functionality.
The invention offers the advantag *' simpler purification of gp350. Because gp350 and gp220 have s nilar biochemic properties, gp220 is often co-purified in preparations of gp350. Cells expressing only the non-splicing variant of the gp350/220 gene simplifies protein purification. This will reduce the costs of producing gp350. The invention also makes biochemical characterization of the starting material for gp350 purification easier. Because only one species is present, protein content analysis and amino acid sequence analysis may be performed without accounting for the presence of a second species.
The invention additionally offers the advantage of increased gp350 production. Prevention of gp350 gene splicing will shift the cell from dual production of gp350 and gp220 to the production of gp350 alone. In some cells, the concentrations of gp220 have been estimated to be 30% -100% of the gp350 concentration. With the gene splicing eliminated, gp350 production will be increased by the lack of gp220 production.
The DNA sequence of the gp350/220 gene is described by Beisel, J. Virology 54:665(1985) and Biggin, EMBO J. 3:1083(1984) and is illustrated in Figure 1. The gene is an open reading frame of 2721 bases, encoding 907 amino acids and specifying a primary translation product of about 95 kD. The difference between predicted and actual values represents extensive glycosylation of the protein. 591 bases (encoding 197 amino acids) are spliced out to produce gp220. The apparent molecular weight of gp350/220 gene products may also vary depending upon the type of measurement system used, glycosylation site utilization in different cell types, post-translational processing differences or selective gene mutation. Measured values vary for the products of different gp350/220 gene non-splice site variants but the term "homogeneous gp350 protein or proteins" encompasses gene products of the non- splicing variant, optionally having additional deletions or mutations such as the C- terminal deletions and/or transmembrane modifications also disclosed herein. The term "gp220 protein" refers to the alternatively spliced gp350/220 gene product with a molecular weight of approximately 220 kD. Splice-sites in the gp350/220 gene were identified by comparison of the gp350/220 gene with consensus donor and acceptor splice sequences based on other genes, predominantly from eukaryotic organisms. The consensus sequences developed by Mount, Nucleic Acids Res. 10:459(1982) from studying the splice sites in other genes are:
Donor: — AG/G 'T' — AGT C G
Acceptor: — Nn —A *G*/G
The bases asterisked above represent bases that appear in 100% of all splice sites (highly conserved). Positions with two bases or one base represent conserved positions (non highlighted positions). The slash indicates the actual site of splicing.
In the gp350/220 gene the donor splice site occurs after nucleotide 1501 and the acceptor splice occurs after nucleotide 2092, as shown by DNA sequencing (Biggin, EMBQ J. 3:1083(1984)) of the gρ350/220 gene. (The numbering used herein and in Figure 1 conforms to the numbering in Biggin). The splice site occurs in the corresponding gene region in the Type B strain of EBV (the donor splice site after A1J01 and the acceptor splice site after G20_o). The invention encompasses compositions made using either the A or B strain or another EBV strain's splice site to produce a single species of mRNA from the gp350/220 gene. The DNA sequence of the Type A form of the virus from strain B95-8 was used in the Examples although the DNA sequence of the Type B strain could equally have been used, because the translated gene products of Type A and B strains are 98% identical. The B strain lacks amino acids 507 through 520 and 570 through 576. The type A strain was used because it contains all the possible gp350 antigenic sites. Alternatively, EBV gp350/220 having strain-specific sequences could be used in accordance with the teachings herein to produce EBV strain-specific homogeneous gp350 proteins having immunogenic properties specific to a particular strain and t refore useful in immunogenic and/or therapeutic compositions for the prevention or treatment of strain specific EBV related diseases. Table 1 shows the wild type nucleotide and amino acid sequences of the donor and acceptor splice sites. To prevent RNA splicing of the gp350/220 gene, mutations were introduced into the gp350/220 gene nucleic acid sequence to replace the relevant base pairs of the RNA splice site. To render a splice-site nonfunctional, preferably at least one of the bases out of the two highly conserved bases framing the donor site or acceptor site should be replaced with nonconserved bases, more preferably at least two highly conserved bases should be mutated to nonconserved bases. Other conserved bases, more than two bases away from the splice site, can also be replaced with nonconserved splice site bases to further decrease recognition of the splice site. Both the donor and the acceptor site can be changed to impair splicing mechanisms. Preferably, both the donor and the acceptor contain at least one change each, in one of the four highly conserved splice site base positions, and more preferably at least two changes in two of the four highly conserved splice site base positions. If one splice site is not mutable due to a desire to maintain the wild-type amino acid sequence then it is preferable to introduce at least two mutations to the other splice site.
Mutation at the gp350/220 splice sites may introduce changes into the amino acid sequence of the subsequently expressed gp350 protein. Preferably such changes should be conservative amino acid substitutions. Conservative substitutions in the amino acid sequence, as opposed to nonconservative changes in the amino acid sequence, will help preserve antigenic sites. Conservative amino acid changes can be made as long as the base change (or base changes) result in a suitable change in the invariant donor/acceptor bases. For example, Gly could be substituted for Ser50ι at the donor splice site, using any Gly-specific codons other than GGU (use of GGU would preserve the G nucleotide and would not result in the desired GT replacement in the splice signal). Likewise, at the acceptor splice site, Gly698 to Ala would be a conservative change, but since all Ala codons start with the highly conserved G nucleotide, this would not result in the desired replacement. Although Proline also might be a conservative amino acid change, proline would not be used to replace a wild type amino acid because it would result in modification of the tertiary structure of the protein and thereby mask one or more gp350 antigenic sites. Table 1 shows the acceptable conservative amino acid replacements in the wild-type sequences. At the bottom of Table 1 is an example of a mutation with conservative amino acid changes.
TABLE 1
Figure imgf000014_0001
Although one aspect of the present invention comprises a non-splicing variant of gp350/220, additional mutations of the gp350/220 coding sequence may also be desirable. In order to produce soluble homogeneous gp350 proteins ("soluble proteins" are either free in solution or membrane associated but are not membrane integrated), for example, to avoid cell toxicity problems incurred by the expression of full length gp350 as an integral membrane protein, the membrane spanning region (also known as the transmembrane region) of gp350 is modified by deletion of all or part of its encoding DNA sequence. The membrane spanning region of gp350/220 comprises amino acids 861 (methionine) through 881 (alanine). See, Beisel, J. Virology 54:665(1985). Preferably, at least 8 amino acids of the transmembrane region are deleted, more preferably at least 12 amino acids are deleted and most preferably between 18 and 21 amino acids are deleted. Accordingly, in another aspect, the invention provides non-splicing variants of gp350/220 DNA and/or gp350 homogeneous protein additionally comprising at least one deletion in the transmembrane region of the gp350/220 DNA and/or gp350 homogeneous protein that results in the expression of soluble homogeneous gp350 protein.
In addition to deleting all or part of the transmembrane domain of the non- splicing gp350/220 variant, the C-terminal sequence following the transmembrane domain and comprising amino acids 881 through 907 may also be deleted in whole or in part, as described herein, in accordance with the invention. Thus, in another aspect the invention comprises non-splicing variants of gp350/220 DNA and/or homogeneous protein further modified by deletion of all or a portion of the DNA encoding and/or amino acid sequence comprising the transmembrane region of gp350/220 and even further modified by deletion of the remaining C-terminal DNA and/or amino acid sequences of gp350/220.
Accordingly, in another aspect the invention comprises non-splicing variant DNA sequences encoding the homogeneous gp350 proteins of the invention. Such DNA sequences comprise the DNA sequence of Figure 1 encoding amino acids 1 through 907 and further comprising the nucleotide substitutions taught herein to remove the donor and acceptor splice sites. Such DNA sequences optionally comprise truncated DNA sequences in which the nucleotides encoding all or part of the transmembrane domain and C-terminus comprising amino acids 861 through 907 are deleted and deletion variants in which the nucleotides encoding all or part of the transmembrane domain comprising amino acids 861 through 881 are deleted. The DNA sequences of the present invention encoding homogeneous gp350 proteins may also comprise DNA capable of hybridizing under appropriate stringency conditions, or which would be capable of hybridizing under such conditions but for the degeneracy of the genetic code, to an isolated DNA sequence of Figure 1. Accordingly, the DNA sequences of this invention may contain modifications in the non-coding sequences, signal sequences or coding sequences, based on allelic variation, species variation or deliberate modification. These non-splicing variant gp350/220 DNA sequences as disclosed herein can be constructed using methods well known in the art. The modified DNA sequences of this invention can be expressed recombinantly, likewise using known methods, to produce the homogeneous gp350 proteins of this invention. Such recombinant proteins can be purified and incorporated into pharmaceutical compositions for the prophylactic treatment and prevention of EBV related diseases.
The non-splicing variants of gp350 220 DNA of this invention can be expressed recombinantly in different types of cells using the appropriate expression control systems as is known in the art. Suitable cells known and available in the art include, but are not limited to, yeast cells such as Saccharomyces cerevisiae. bacterial cells such as R coli and Bacillus subtilis and mammalian cells such as GH3, CHO, NSO, MDCK and C-127 cells. Vectors used with cell types are selected based on their compatibility with the cell type and expression control system used. Cells and vectors that allow for the expression of secreted products of the gp350/220 gene are preferred. Typically for example, R coli is transformed using derivatives of pBR322 which have been modified using conventional techniques to contain the DNA sequences for expression of the desired protein, in this instance the non-splicing variant sequences of EBV gp350, with or without the sequences encoding the C-terminus and/or membrane spanning region. pBR322 contains genes for ampicillin and tetracycline resistance, which can be used as markers. See, Bolivar, Gene 2:95(1977). Commonly used expression control sequences, i.e., promoters for transcription initiation and optionally an operator or enhancer, include the beta-lactamase and lac promoter systems (see Chang, Nature 198: 1056(1977)), the tryptophan promoter system (see Goeddel, Nucleic Acids Res. 8:4057(1980)) and the lambda-derived PL promoter and N-gene ribosome binding site (see Shimatake. Nature 292: 128(1981). However, any available promoter system or expression control system that is compatible with prokaryotic host cells can be used. Other exemplary host cells, plasmid and expression vehicles are disclosed in United States Patent Nos. 4,356,270 issued to Itakura (1982), 4,431,739 issued to Riggs (1984) and 4,440,859 issued to Rutter (1984). Insect cells may also be used as host cells employing insect cell expression. In the case of expression in insect cells, generally the components of the expression system include a transfer vector, usually a bacterial plasmid, which contains both a fragment of the baculovirus genome, and a convenient restriction site for insertion of the heterologous gene or genes to be expressed; a wild type baculovirus with a sequence homologous to the baculovirus-specific fragment in the transfer vector (this allows for the homologous recombination of the heterologous gene in to the baculovirus genome); and appropriate insect host cells and growth media.
Currently, the most commonly used transfer vector for introducing foreign genes into AcNPV is pAc373. Many other vectors, known to those of skill in the art, have also been designed. These include, for example, pVL985 (which alters the polyhedrin start codon from ATG to ATT, and which introduces a BamHI cloning site 32 basepairs downstream from the ATT; see Luckow and Summers, Virology (1989) 17:31.
The plasmid usually also contains the polyhedrin polyadenylation signal (Miller et al. (1988) Ann. Rev. Microbiol.. 42:177) and a procaryotic ampicillin-resistance (amp) gene and origin of replication for selection and propagation in R coli. Baculovirus transfer vectors usually contain a baculovirus promoter. A baculovirus promoter is any DNA sequence capable of binding a baculovirus RNA polymerase and initiating the downstream (5' to 3') transcription of a coding sequence (e.g. structural gene) into mRNA. A promoter will have a transcription initiation region which is usually placed proximal to the 5' end of the coding sequence. This transcription initiation region typically includes an RNA polymerase binding site and a transcription initiation site. A baculovirus transfer vector can also have a second domain called an enhancer, which, if present, is usually distal to the structural gene. Expression can be either regulated or constitutive. For insect cell expression technology, see EP patent publication 155 476. Yeast, for example Saccharomyces cervisiae. may also be used as a host cell.
Various strains are available and may be used. Likewise, plasmid vectors suitable for yeast expression are known, as are promoter and expression control systems. See for example, Myanohara, Proc. natl. Acad. Sci. 80:l(19831(PHO5 promoterl. EP Patent Publication 012 873 (leader sequences), Kurtz, Mol. Cell. Biol. 6:142(19861. Ito, Bacteriol. 153: 163(19831 and Hinnen, Proc. Natl. Acad. Sci. 75:1929(1979)(transformation procedures and suitable vectors). Eukaryotic cells from multicellular organisms may of course also be used as hosts cells for the expression of genes encoding proteins and polypeptides of interest. Useful host cell lines include VERO and HeLa cells, and Chinese hamster ovary cells (CHO). Expression vectors compatible with such cells are also available and typically include promoters and expression control sequences, such as for example, the early and late promoters from SV40 (see Fiers, Nature 273:113(1978)) and promoters from polyoma virus, adenovirus 2, bovine papilloma virus or avian sarcoma virus. Exemplary host cells, promoters, selectable markers and techniques are also disclosed in United States Patent Nos. 5,122,469 issued to Mather (1992), 4,399,216 issued to Axel (1983), 4,634,665 issued to Axel (1987), 4,713,339 issued to Levinson (1987), 4,656, 134 issued to Ringold (1987), 4,822,736 issued to Kellems (1989) and 4,874,702 issued to Fiers (1989).
Transformation of suitable host cells is accomplished using standard techniques appropriate to such cells, such as CaCl2 treatment for prokaryotes as disclosed in Cohen Proc. Natl. Acad. Sci. 69:2110(19721 and CaPO4 precipitation for mammalian cells as disclosed in Graham, Virology 52:546(19781. Yeast transformation can be carried out as described in Hsiao, Proc. Natl. Acad. Sci. 76:3829(19791 or as described in Klebe, Gene 25:333(1983).
The construction of suitable vectors containing the non-splicing variant gp350 sequence (with or without the additional modifications disclosed here resulting in deletion of the C-terminus and/or the membrane spanning region) is accomplished using conventional ligation and restriction techniques now well known in the art. Site specific DNA cleavage is performed by treating with suitable restriction enzyme(s) under standard conditions, the particulars of which are typically specified by the restriction enzyme manufacturer. Polyacrylamide gel or agarose gel electrophoresis may be performed to size separate the cleaved fragments using standard techniques and the fragments blunt ended by treatment with the Klenow fragment of R coϋ polymerase I in the presence of the four deoxynucleotide triphosphates. Treatment with SI nuclease hydrolyzes any single-stranded portions. Synthetic oligonucleotides can be made using for example, the diethylphosphoamidite method known in the art. See United States Patent No. 4,415,732 (1983). Ligations can be performed using T4 DNA ligase under standard conditions and temperatures and correct ligations confirmed by transforming R. cpJi or COS cells with the ligation mixture. Successful transformants are selected by ampicillin, tetracycline or other antibiotic resistance or using other markers as are known in the art.
Such recombinant DNA techniques are fully explained in the literature. See, e.g. , Sambrook, MOLECULAR CLONING: A LABORATORY MANUAL, 2D ED. (1989); DNA CLONING, Vol. I and π (DN Glover e 1985); OLIGONUCLEOTTDE SYNTHESIS (MJ Gait ed V 84); NUCLEIC ACID HYBRIDIZATION (BD Hames ed 1984); TRANSCRIPTION AND TRANSLATION (BD Hames ed 1984); ANIMAL CELL CULTURE (RI Freshney ed 1986); B. Perbal, A PRACTICAL GUIDE TO MOLECULAR CLONING (1984); GENE TRANSFER VECTORS FOR MAMMALIAN CELLS (JH Miller ed 1987 Cold Spring Harbor Laboratory); Scopes, PROTEIN PURIFICATION: PRINCIPLES AND PRACTICE, 2nd ed, (1987 Springer- Verlag NY) and HANDBOOK OF EXPERIMENTAL IMMUNOLOGY Vols I - IV (DM Weired 1986). All such publications mentioned herein are incorporated by reference for the substance of what they disclose.
Accordingly in another aspect the invention comprises vectors containing the non-splicing variants of gp350/220 DNA sequences and host cells and further comprises a method of making a non-splicing variant of gp350/220 protein by culturing said host cells containing a vector that is carrying a non-splicing variant of a gp350/220 DNA sequence operatively linked to an expression control sequence under culture conditions enabling expression of the homogeneous gp350 protein.
The expressed homogeneous gp350 is purified from cell and culture medium constituents using conventional glycoprotein purification techniques such as, but not limited to, ultrafiltration, free flow electrophoresis, gel filtration chromatography, affinity chromatography, SDS-PAGE, differential NH4SO4 precipitation, lectin columns, ion exchange columns and hydrophobicity columns as is known in the art. Small scale analytical preparations of gp350 are most readily purified using SDS-PAGE or lectin affinity columns and such small scale preparations for use in vaccination or immune response experiments are most readily purified using liquid chromatography. For large scale production of commercially significant quantities of gp350 for use in vaccine compositions, a combination of ultrafiltration, gel filtration, ion exchange, and hydrophobic interaction chromatography are preferred. The purified, homogeneous gp350 proteins of the present invention may be employed in therapeutic and/or immunogenic compositions for preventing and treating EBV related conditions and diseases such as infectitious mononucleosis, Burkitt' s lymphoma and nasopharyngeal carcinoma. Such pharmaceutical compositions comprise an immunogenically-inducing effective amount of one or more of the homogeneous gp350 proteins of the present invention in admixture with a pharmaceutically acceptable carrier, for example an adjuvant/antigen presentation system such as alum. Other adjuvant/antigen presentation systems, for instance, MF59 (Chiron Corp.), QS-21 (Cambridge Biotech Corp.), 3-DMPL (3-Deacyl-Monophosphoryl Lipid A) (RibilmmunoChem Research, Inc.), clinical grade incomplete Freund's adjuvant (IFA), fusogenic liposomes, water soluble polymers or Iscoms (Immune stimulating complexes) may also be used. Other exemplary pharmaceutically acceptable carriers or solutions are aluminum hydroxide, saline and phosphate buffered saline. The composition can be systemically administered, preferably subcutaneously or intramuscularly, in the form of an acceptable subcutaneous or intramuscular solution. Also inoculation can be effected by surface scarification or by inoculation of a body cavity. The preparation of such solutions, having due regard to pH, isotonicity, stability and the like is within the skill in the art. The dosage regimen will be determined by the attending physician considering various factors known to modify the action of drugs such as for example, physical condition, body weight, sex, diet, severity of the condition, time of administration and other clinical factors. Exemplary dosage ranges comprise between about 1 μg to about 1000 μg of protein.
In practicing the method of treatment of this invention, an immunologically- inducing effective amount of homogeneous gp350 protein is administered to a human patient in need of therapeutic or prophylactic treatment. An immunologically inducing effective amount of a composition of this invention is contemplated to be in the range of about 1 microgram to about 1 milligram per dose administered. The number of doses administered may vary, depending on the above mentioned factors. The invention is further described in the following examples, which are intended to illustrate the invention without limiting its scope. EXAMPLE 1
Deletion of the gp350/220 Transmembrane Region and Transmembrane Region through C-terminus to create pDTM and pSTOP The gp350/220 gene from the EBV B95-8 strain (Miller, et al , 1972), is available in a BamHI library as an open reading frame called BLLFl (Baer, Nature 310:207. 1984). To create the desired constructs (shown diagrammatically in Figure 2B), the gp350/220 gene was cloned in two parts: 1) BLSH1, a 2.3 kb HinαTH/Bfal 3' fragment and 2) BLSH2, a 337 b.p. Banl/Hindm 5' fragment (Figure 2A). These fragments were cloned into staging vectors so that the deletions of the C-terminal cytoplasmic and/or transmembrane-encoding domains could be performed. Because the Bfal site occurs at the 5' end of the region encoding the gp350 transmembrane (TM) domain, it was used to construct the TM domain deletions and TM domain deletions with adjacent C-terminus deletions. Using Bfal, it was possible to create deletions retaining only two amino acids of the TM region (Table 2). 1. Construction of pDTM From pSTGl. and pSTG3
The plasmid pDTM is comprised of a gp350/220 nucleic acid sequence that lacks a complete TM coding region. This construct was made using two staging vectors pSTGl and pSTG3. A 450 bp PCR product, SYCT, that introduced a Bfal site at the 3' end of the TM region was made using a BLLFl clone target sequence (Figure 2).
The PCR primers used are as follows:
Bfal
Primer 1: GG ATC CTA GAC TGC GCC TTT AGG CGT A
BLLFl: ... GAC TGC GCC TTT AGG CGT A.. A.A.: ... Asp Cys Ala Phe Arg Arg ...
t End of TM Region
Primer 2: GGA TCC TCT GTT CCT TCT GCT CCA GTG BLLFl: TCT GTT CCT TCT GCT CCA GTG
The Bfal site of Primer 1 was used to clone a Bfal/Xmal fragment of SCYT into pSTGl . The remainder of Primer 1 corresponds to the amino acid sequence encoded by clone BLLFl . Primer 2 corresponds to a region outside the gp350/220 open reading frame on the 3' side of the gene. The SCYT PCR fragment was cut with Bfal and Xmal to produce a 136 base pair fragment which was cloned into a pMTll vector (Spaete and Mocarski, 1985) along with a second fragment, a BLSH1 Hindiπ/Bfal fragment, to create pSTGl . Sequencing across the Bfal site indicated that all of the TM amino acid coding region was deleted except for amino acids Met and Leu (see Table 2). A third BLLFl fragment, BLSH2, was cloned into pMTll to create pSTG3. A 16 base pair Banl/Xbal oligonucleotide linker outside of the gp350/220 gene coding sequence was used to clone the BLSH2 Banl/Hindm fragment into the pSTG3. A 2.4 Hindm/Xmal pSTGl fragment, was cloned into a pEE14 vector (Celltech, England) together with a 0.3 Xbal/HinαTH pSTG3 fragment to complete the pDTM construct.
2. Construction of DSTOP using vectors pSTG2 and pSTG3
The plasmid pSTOP comprises a gp350/220 gene that lacks a TM region and the C-terminal cytoplasmic region adjacent the TM region. To create this construct, a 16 base pair Bfal/EcoRI oligonucleotide linker was created with stop codons (underlined) in three frames following the Bfal sticky end as shown below:
TAT AGA CTA GTC TAG G
A TCT GAT CAG ATC CTT AA
The 5' overhang (TA) of the upper sequence is a sticky end for a Bfal restriction site and the 5' overhang (TTAA) of the lower sequence is an EcoRI sticky end. This 16 base pair linker was used to clone a BLSH1 HindHI/Bfal fragment into pMTll, in order to create pSTG2. A 2.3 kb pSTG2 Hindm/EcoRI fragment and the pSTG3 0.3 kb Xbal/Hindm fragment were cloned into ρEE14 to create pSTOP.
3. Comparison of the wild-tvpe. pDTM and pSTOP sequences at the TM region
The oligonucleotide sequence and translated amino acid sequence of the wild type, pSTOP, and pDTM 3' ends of gp350 DNA and amino acid sequences are shown in Table 2 below. Arrows indicate the beginning and end of the wild-type transmembrane domain (TM). Only two amino acids from the transmembrane domain are retained in pDTM and pSTOP, Met861 and Leug62 (see also Figure 1). Note that a stop codon immediately follows Leu862 in pSTOP. In pDTM the former location of the deleted transmembrane region is marked "ΔTM" . (In the Table, the native amino acids are indicated.)
TABLE 2 3' End of gp350 Wild-Type Sequence
...AAC CTC TCC ATG CTA GTA CTG GTC ATG GCG GAC TGC GCC
...Asn Leu Ser Met Leu862 Val Leu Val Met Ala Asp882 Cys Ala t t
TM start TM end 3' End of pSTOP
...AAC CTC TCC ATG CTA TAG ACT AGT TCT AGG ... ...Asn Leu Ser Met Leu862 STOP
3' End of pDTM ...AAC CTC TCC ATG CTA GAC TGC GCC...
...Asn Leu Ser Met Leu862 Asp882 Cys Ala...
ΔTM
EXAMPLE 2
Removal of the gp350/220 Gene Donor and Acceptor Splice Sites to create pMDTM and pMSTOP
In order to obtain homogeneous production of a gp350 protein the highly conserved and conserved bases of the gp350/220 gene splice site were changed. Four bases were changed in the donor splice site, including the highly conserved GT pair that occurs in 100% of all splice sites. Two conserved donor site bases, AA, were replaced with GT. The two highly conserved (invariant) donor splice site bases were changed from GT to CA. At the acceptor splice site, only one of the highly conserved acceptor splice site bases was altered to preserve the amino acid sequence. A second conserved acceptor splice site base was changed as indicated in Table 3. Table 3 summarizes the bases changed in the donor and acceptor splice sites of the gp350/220 gene.
TABLE 3: EBV gp350/220 Gene Splice Site Changes Donor Splice site:
donor donor
Wild-type: GAA AI GT mutant: GAG* T*C*A*
Glu Ser501 Glu Ser501
Acceptor Splice site: acceptor acceptor
Wild-type: ACA GI GT mutant: ACT* GGA* Thr Gly698 Thr Gly698
The bases changed by oligonucleotide-based mutagenesis are marked with an asterisk in the mutant sequences. The actual site of splicing is indicated by an arrow, and the encoded amino acids are shown. Note that the amino acid sequence does not change as a result of the nucleotide substitutions.
These nucleotide substitutions to the wild type gp350/220 donor splice site and accepter splice site DNA sequences were accomplished using oligonucleotide-mediated mutagenesis. A modified phage vector, M13TAC, was employed to produce mutations as described in Zoller, M.E. and Smith, M. (1983) Methods of Enzymol. 100:468. BamHI/XhoI fragments of the gp350/220 nucleotide sequence were cloned into the polylinker of plasmid M13TAC using Asp718 and BamHI restriction sites on the poly linker, combined with a 19 bp oligonucleotide linker containing Asp718 and Xhol sticky ends. The plasmids M13DTM and M13STOP of Example 1 (Figure 2B), were used for the mutagenesis.
Two 42-mer oligonucleotides, PrDonorl and PrAcceptorl , were made for use in the mutagenesis. Each was designed to be complementary to gp350/220 gene sequences centering on either the donor or acceptor splice sites. The only region of the oligonucleotides that were not complementary to the gp350/220 gene were the bases representing the desired mutations. Mutagenesis oligonucleotides comprised the following: PrDonorl
Primer: GGT CAT GTC GGG GGC CTT TG |A CTC TGT GCC GTT GTC CCA TGG ** I * *
EBV: GGT CAT GTC GGG GGC CTT AC |T TTC TGT GCC GTT GTC CCA TGG
PrAcceptorl Primer: CTG TGT TAT ATT TTC ACC TC J C AGT TGG GTG AGC GGA GGT TAG
EBV: CTG TGT TAT ATT TTC ACC AC |C TGT TGG GTG AGC GGA GGT TAG
The sequence of the mutagenesis oligonucleotides are labelled "Primer," while the DNA sequence spanning the gp350/220 gene splice sites are labelled "EBV." Bases that were changed as a result of the mutagenesis are marked with an asterisk. The dashed line indicated the location of the splice. The oligonucleotides PrDonorl and PrAcceptorl were hybridized to single- stranded clones of M13-DTM and M13-STOP. T4 DNA polymerase holoenzyme was used to produce double-stranded M13 DNA and E. coli was transformed with the double-stranded DNA. Using the vector M13TAC, any clone that contained the desired mutation could be identified by a color change from white to blue in the presence of X-gal and isothiopropylgalactate. Blue plaques were picked and grown up, and DNA sequencing across splice junctions was used for the final identification of mutant clones, labelled M13-MDTM and M13-MSTOP.
After identifying clones containing the desired mutations, BamHt/XhoI fragments were cut out of M13-MDTM and M13-MSTOP and ligated back into pDTM or pSTOP backbones to create the constructs pMDTM and pMSTOP, respectively. These constructs were transfected into CHO cells to express the non- splicing variant gp350/220 DNA sequences as described in Example 3. EXAMPLE 3 Expression of gp350 in CHO cells 1. Transfection of gp350/220 gene constructs
One method for producing high levels of homogeneous gp350 protein of the invention from mammalian cells involves the construction of cells containing multiple copies of the heterologous gp350 DNA sequence. The heterologous DNA sequence is operatively linked to an amplifiable marker, in this example, the glutamine synthetase gene for which cells can be amplified using methionine sulphoximine.
The pMDTM and pMSTOP vectors made in Example 2 were transfected into CHO cells as discussed below, according to the procedures of Crockett,
Bio/Technology 8:662(1990) and as described in the Celltech Instruction Manual for the glutamine synthetase gene amplification system (1992).
CHO-K1 cells (ATCC CCR61) were maintained in glutamine-free EMEM (Eagles Minimal Essential Medium) supplemented with 10% fetal bovine serum, 100 units/ml penicillin, 100 mg/ml streptomycin, MEM (Modified Eagle's
Medium) nonessential amino acids, and 1 mM sodium pyruvate (all obtained from JRH Biosciences). The media was also supplemented with 60 mg/ml glutamic acid, 60 mg/ml asparagine, 7 mg/ml adenosine, 7 mg/ml guanosine, 7 mg/ml cytidine, 7 mg/ml uridine, and 2.4 mg/ml thymidine (all from Sigma.) This media preparation was used throughout the transfection, with deviations from this recipe as noted.
One day prior to transfection 10-cm dishes were seeded with 3 x 106 CHO-K1 cells. On the day of transfection the cells were washed with 10 ml serum-free media per dish. Plasmid DNA (from the pMDTM, pMSTOP plasmids) was applied by CaPO4 precipitation using conventional techniques. 10 μgs of each plasmid DNA precipitate was incubated with the CHO-K1 cells plus 2 ml of serum-free media at
37°C for 4.5 hours. Three replicates of each of the four plasmid DNA transfections were made. The cells were then shocked for 1.5 minutes with 15% glycerol in HEPES-buffered saline. After rinsing with serum-free media, the cells were re-fed with serum-containing media and incubated for 24 hours. The following day the media was changed to include 10% dialyzed fetal bovine serum (JRH Biosciences) and amplified by the addition of 25 μM methionine sulphoximine (Sigma). Cells were re-fed with methionine sulphoximine-containing media every 3-5 days until the amplified clones were large enough for picking, approximately 13-14 days later. Clones were picked by scraping colonies off the dish with a sterile 200 μl pipetman tip and transferred to one well of a 96-well plate in media without methionine sulphoximine. 1-2 days later the media was replaced with media + 25 μM methionine sulphoximine. After 4 days the culture supernatants were harvested and assayed for protein products in an ELISA assay, as discussed below.
CHO cells were also transfected with the pEE14 control vector alone (which contains no EBV sequences) and 24 clones of CHO-pEE14 were also picked and transferred to plates to serve as controls. (The control clones were identified on the basis of survival in methionine sulphoximine.)
2. ELISA Assay Following transfection, 241 clones of CHO-pMDTM and 158 clones of CHO- pMSTOP were picked and grown up. Supernatants from these clones were tested for gp350 protein production. 96-well plates were coated with affinity-purified rabbit anti-gp350/220 antibody (antibody MDP1; gift of Andrew Morgan) diluted 1:2000 in 50mM sodium borate buffer, pH 9. The plates were incubated at 37 °C for 3-4 hours and washed 3 times with PBS + 0.05 % Tween 20 using a Nunc ImmunoWasher.
After blotting dry, the plates were blocked by incubating with 2 % BSA in PBS + 0.01 % Thimerosal at 37°C for 0.5 hours and washed again. Supernatants from the transfected cells and control cells were added to the wells and incubated for 2 hours at 37 °C. The plates were then incubated with the primary detection antibody, a mouse monoclonal antibody against gp350/220 (antibody #C65221M; Biodesign
International) at lmg/ml diluted in PBS wash buffer, 37°C for 1 hour. After washing, the plates were incubated with the secondary antibody, horseradish peroxidase-conjugated goat F(ab)2 fragments directed against mouse immunoglobulins (Human Ig adsorbed; Biosource International.), 0.7 μg/ml in PBS + 0.05 % BSA and 0.01 % Thimerosal, at 37°C for 1 hour. The plates were washed and developed using
ABTS (Pierce Chemicals) dissolved in Stable Peroxide Substrate Buffer (Pierce Chemicals) for 0.5 hours at room temperature. The reaction was stopped with 1 % SDS and the plates were read at 405 and 650 nm wavelengths using a Molecular Devices Vmax ELISA plate reader. 24 pMDTM and 18 pMSTOP clones tested positive for secreted gp350. The clones exhibiting the highest ELISA signal were transferred to 24-well plates for scale-up and further testing in a Western Blot and a radioimmunoprecipitation assay.
3. Western Blot and Radio Immunoprecipitation Assay
In an initial screening, tissue culture supernatants from the pMDTM transfections were assayed for activity in a Western Blot. CHO cell supernatants were purified on 5 % SDS-PAGE gels, transferred to nitrocellulose overnight, and probed with anti-gp350 antibodies. Seven pMDTM clones were found to be positive for gp350 in the Western blot analysis.
The pMDTM clones that were positive in the Western blot were further tested by radioimmunoprecipitation for the presence of gp220. Selected transformed pMDTM cells, pEE14 control and GH3Δ19 control cells (described below) were grown overnight in six-well plates so that they were approximately three-quarters confluent on the day of the experiment. Each well contained approximately 5X106 cells. For labelling, the media was removed from each well and replaced with 0.7 ml of methionine-free MEM (10% fetal calf serum) + 100 μCi 35S -methionine. The cells were incubated 5.5 hours at 37°C and then microcentrifuged at 4000 rpm for
5 minutes. Homogeneous gp350 protein in the supernatant was immunoprecipitated by addition of 10 μl of Sepharose-Protein A (Sigma) in a 50% slurry and 20 μl monoclonal anti-gp350/220 (antibody #C65221M, lOOmg/ml; Biodesign International), with overnight rocking at 4°C. The mixture was then pelleted at 2000 rpm, 2 minutes at room temperature in a microcentrifuge and washed four times with several volumes of phosphate-buffered saline. After the final wash, all liquid was removed from the pellet and replaced with 50 μl protein gel sample buffer. The samples containing the precipitated immuno-complex were boiled 5 minutes and run on a 5 % SDS-PAGE. Immunoprecipitates were compared to gel samples of tissue culture supernatants mixed 1 : 1 with protein sample buffer. The gel was dried and autoradiographed with Hyperfilm β-Max (Amersham). Figure 3 shows the autoradiographic results of SDS-PAGE analysis of the radioimmunoprecipitation. The cell line used as a positive control was GH3Δ19 (gift of Elliot Keiff; Whang et al. , 1987). GH3Δ19 cells secrete a truncated form of the gp350/220 protein lacking the transmembrane and C-terminal cytoplasmic domains. For use as a negative control, CHO cells were transfected with the pEE14 vector alone and selected by methionine sulphoximine in parallel with the pMDTM transfection. In Figure 3, supernatants ("S") are shown in odd numbered lanes, alternated with immunoprecipitates ("Ip") shown in even numbered lanes. In control lane 2, precipitation from the GH3Δ19 control cells results in two strong protein bands at approximately 220 and 350 kD demonstrating production of the truncated splice variant gp350 and gp220 proteins in about a 1 : 1 ratio. As expected, these immunoprecipitated bands are concentrated with respect to the radiolabelled tissue culture supernatant (non-immunoprecipitated sample) in lane 1. Also, as expected, no bands are shown in the negative control (lane 4), since the pEE14 vector does not contain any of the gp350/220 constructs.
SDS-PAGE analysis of the immunoprecipitation from supernatants of pMDTM clones in lanes 6, 8 and 10 results in a single strong band at approximately 350 kD, the same as the higher molecular weight species in the GH3Δ19 control lane 2. In contrast to the GH3Δ19 control lane however, an additional strong band at approximately 220 kD is absent from lanes 6, 8 and 10, although in lane 8 a very faint band migrating at a slightly lower molecular weight is revealed. This could represent a degradation product, a co-precipitated cellular product or a small amount of gp220 protein resulting from a mistranslation or a mutational event that returns the deleted donor and acceptor splice sites to the native nucleotide or amino acid sequences. Strong single bands at approximately 350 kD were found in five other
MTDM replicates tested (data not shown).
It is unlikely that the complete absence of the band at 220 kD in lanes 6 and 10 is due to inefficient precipitation from MDTM supernatants since in the 35S-labelled GH3Δ19 control lane (2), a band at 220 kD is easily visualized. Also, additional assays using the pDTM constructs of Example 1 that contain the wild type splice sites result in two strong bands at 350 and 220 kD. Therefore, these results demonstrate that deletion of the splice sites results in production of gp350 protein in the absence of production of gp220 protein.
This homogeneous gp350 protein, expressed in CHO cell lines, or in other mammalian or non-mammalian cell lines, can be further scaled up and homogenous gp350 protein can be isolated and purified from conditioned medium from the cell line using methods familiar in the art, including techniques such as lectin-affinity chromatography, reverse phase HPLC, FPLC, gel filtration and the like. See David, J. Immunol. Methods 108:231(1988) and Madej, Vaccine 10:777(19921.
4. Northern Blot Analysis of pMDTM Protein
In this experiment, we show by Northern blot that MDTM-1 cells are making gp350 RNA and not gp220 RNA, providing confirmation on another level that splice site mutations prevent gp220 production.
DNA probes complementary to gp350 were made from pDTM, see Example 1. A gp350/220 probe template, XP464, was isolated as a 464 b.p. XhoI/PstI pDTM fragment. XP464 recognizes both gp350 and gp220. To make the gp350-specific probe AN537, pDTM was cut with Ncol and Ndel, two overlapping 580 b.p. fragments were isolated, and this mixture was cut with XmnI to eliminate one contaminating fragment and with AM to yield a 537 b.p. Alul/Ndel fragment internal to the gp350/220 splice sites. AN537 is specific for the region spliced out of gp220 and is thus specific for gp350 message. DNA probes were labelled by 32P-dCTP nick translation (Amersham) using DNA fragments XP464 and AN537.
Whole cell RNA was prepared essentially according to the method of Chomczyunski and Sacchi, Anal. Biochem.. 162:156-59 (1987). Media from two T- 250 flasks each of CHO-pEE14 (a negative control celll line, see Example 1), CHO-
MDTM-1 and CHO-DTM-7 cells, 90% confluent, was aspirated and the cells were lysed and scraped off in denaturing buffer (10 ml 4M Guanidine thiocyanate, 25mM sodium citrate, pH 7, 0.5 % sarkosyl, 100 mM 2-mercaptoethanol). Each 10 ml lysate was supplemented with 1 ml 2M sodium acetate ,pH 4, 10 ml saturated phenol, pH 4.5, and 2 ml chloroform/isoamyl alcohol; the lysate was incubated on ice 15 minutes, spun 10000 x g 20 minutes at 4°C, and the upper aqueous phase was removed. RNA was precipitated from the aqueous phase by addition of one volume of isopropanol at 20 °C for 1 hour, pelleted at 4°C, resuspended in denaturing buffer and reprecipitated. The RNA pellet was washed IX in 70% ethanol, dried in a Speed- Vac and resuspended in DEPC -treated water.
DTM-7 and MDTM-1 whole cell RNA was denatured at 65 °C for 15 min in 15% formamide and 6% formaldehyde, run on 1 % agarose/6.6% formaldehyde gels and transferred to nitrocellulose by capillary action and probed with labeled XP464 and AN537. The DNA probes were denatured by boiling 5 minutes, hybridized in 5X SSPE at 65 %C overnight and the nitrocellulose was washed at high stringency. Autoradiography was performed using a Bio-Rad phosphorimager. Northern blots of total cell RNA from CHO-MDTM cells show the effectiveness of splice mutations in preventing gp220-specific RNA production. The gp350-specific probe, AN537, bound to only one species of RNA in MDTM-1 and DTM-7 cells (Fig. 4, lanes 1 and 2) as expected for a gp350-specific probe. The XP464 probe, which is specific for both gp350 and gp220 RNA. recognized two species in DTM-7 and a single higher molecular weight band in MDTM-1 (Fig 4., lanes 4 and 3), as expected if splice mutations are preventing gp220 message production in MDTM-1. Even though the MDTM-1 lanes are overloaded for gp350- specific RNA, no distinct gp220 RNA is visible. The reason for the difference in apparent mobility of the gp350 message in MDTM-1 versus DTM-7 lanes is not known. The MDTM-1 species is overloaded compared with DTM-7, which may affect migration in the gel. Also, the gp350 message runs close to a large ribosomal RNA band on the gel, which may distort the apparent molecular weight. Either way, the presence of a single species complementary to gp350 DNA sequences suggests that this signal represents gp350 mRNA. Thus, mutations in the donor and acceptor splice sites are effective at preventing gp220 message production, as judged by
Northern blots. This result is further confirmed by radioimmunoprecipitation using monoclonal antibodies specific for gp350/220 as well as by Western blots of MDTM- 1 and DTM-7 supernatants. EXAMPLE 4
Testing the Homogeneous gp350 Proteins for Immunogenic Activity The purified homogeneous gp350 proteins are incorporated into appropriate vehicles for administration and administered to mice as follows.
A 2x adjuvant- vehicle concentrate is prepared by mixing Pluronic L121 and squalane in 0.4% (v/v) Tween 80 in phosphate buffered saline with (Thr1) MDP in accordance with the procedure of David, J. Immunol. Methods 108:231(19881 and Allison, J. Immunol. Methods 95: 157(19861.
The composition for administration is prepared by addition of equal volumes of protein and adjuvant- vehicle on the day of administration. The protein content should be with range of 5 micrograms to 50 micrograms per dose.
BALB/c mice are immunized with three 0.1 ml intramuscular injections at 0, 21 and 42 days. A pre-immunization bleed and successive bleeds taken 10 days after each injection are obtained from the retro-orbital sinus.
Serum antibody levels are determined by an ELISA according to the procedures described in Example 3. EBV neutralizing antibodies in the sera are quantified by their ability to inhibit transformation of fetal cord blood lymphocytes by EBV in vitro according to the methods of Moss, J Gen. Virol. 17:233(19721 and
De Schryver, Int. J. Cancer 13:353(19741.
Alternatively, New Zealand white rabbits are inoculated by intramuscular administration of five doses of protein emulsified in the foregoing adjuvant at 0, 21 , 42, 63 and 84 days. The dose should be in the range of about 5 μg to 50 μg per inoculation. Sera is obtained two weeks following the last dose and tested for antibody titers to the antigen, for cross-reactive antibody to viral gp350/220 from B95-8 cells and for in vitro EBV-neutralizing activity following the methods of Emini, Virology 166:387(19881.
Because the ability of the EBV gp350/220 protein to induce protective immunity in an animal model of EBV infection has already been established, see
Epstein, Clin. Exp. Immunol 63:485(19861. similar positive results from administration of a homogeneous gp350 protein composition are expected. The disclosures of all publication identified herein are expressly incorporated herein by reference. The foregoing detailed description is given for clearness of understanding only and no unnecessary limitations are either understood or inferred therefrom, as modifications within the scope of the invention will be obvious to those skilled in the art.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Spaete, Richard and Jackman, Winthrop, T.
(ii) TITLE OF INVENTION: Non Splicing Variants of gp350/220
(iii) NUMBER OF SEQUENCES: 19
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Cooley Godward Castro Huddleson & Tatum
((BB)) SSTTRREEEETT:: 5 Palo Alto Square
(O CITY: Palo Alto
(D) STATE: California
(E) COUNTRY USA
(F) ZIP: 94306
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: Patent In Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: 08/229,291
(B) FILING DATE: April 18, 1994 (C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Luann Cεerr
(B) REGISTRATION NUMBER: 31,822 (C) REFERENCE/DOCKET NUMBER: AVIR-003/00US
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 415-843-5163
(B) TELEFAX: 415-857-0663 (C) TELEX: 380816 CooleyPA (2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
GGATCCTAGA CTGCGCCTTT AGGCGTA 27
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: :
GACTGCGCCT TTAGGCGTA 19
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 6 amino acids
(B) TYPE: amino acid (C) TOPOLOGY: linear
(ii) MOLECULE TYPE: Protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
Asp Cys Ala Phe Arg Arg 1 5 (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
GGATCCTCTG TTCCTTCTGC TCCAGTG 27
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:
TCTGTTCCTT CTGCTCCAGT G 21
(2) INFORMATION FOR SEQ ID NO: 6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:
TATAGACTAG TCTAGG 16 (2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
ATCTGATCAG ATCCTTAA 18
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 39 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:
AACCTCTCCA TGCTAGTACT GGTCATGGCG GACTGCGCC 39
(2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 13 amino acid
(B) TYPE: amino acid
(C) TOPOLOGY: linear
(ii) MOLECULE TYPE: Protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: Asn Leu Ser Met Leu Val Leu Val Met Ala Asp Cys Ala 1 5 10
(2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:
AACCTCTCCA TGCTATAGAC TAGTTCTAGG 30
(2) INFORMATION FOR SEQ ID NO:11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 5 amino acids
(B) TYPE: amino acid (C) TOPOLOGY: linear
(ii) MOLECULE TYPE: Protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:
Asn Leu Ser Met Leu
1 5
(2) INFORMATION FOR SEQ ID NO:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: Oligomer DNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:
AACCTCTCCA TGCTAGACTG CGCC 24
(2) INFORMATION FOR SEQ ID NO:13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids
(B) TYPE: amino acid (C) TOPOLOGY: linear
(ii) MOLECULE TYPE: Protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
Asn Leu Ser Met Leu86- Asp882Cys Ala
1 5
(2) INFORMATION FOR SEQ ID NO:14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:
GGTCATGTCG GGGGCCTTTG ACTCTGTGCC GTTGTCCCAT GG 42
(2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:15: GGTCATGTCG GGGGCCTTAC TTTCTGTGCC GTTGTCCCAT GG 42
(2) INFORMATION FOR SEQ ID NO:16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:
CTGTGTTATA TTTTCACCTC CAGTTGGGTG AGCGGAGGTT AG 42
(2) INFORMATION FOR SEQ ID NO:17:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 42 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: Oligomer DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:17:
CTGTGTTATA TTTTCACCAC CTGTTGGGTG AGCGGAGGTT AG 42
(2) INFORMATION FOR SEQ ID NO:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3833 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1014..3734 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
GAATTCCATA AATGAAACAC GCTGGTCAGG TGTTAAAACT TCCTCCCAGA TTTTCGTGAG 60
GCTCCTGTGT ATAGCCATAT AGTCAAAGAA AATACTGTAG CGGGGATTAC AGCTCTGTAC 120
AATGTTACCC ACGGAGCTCT GAACATACAA CCACTGGCGA TCCCCGGGGG TACATCGCGG 180
CAGCTTAAAG GTGCCGGCGG AAAAGGTCAC GTGACACCTA CGGCCACCTG TGCACCCAAG 240
TGTCGCCTGG AGATGTACGA ATGTGGGAGT CGTCTGGTGA TCGGTGTAGC TGTACATCCA 300
GCTGCTGTAT GCCTGGTAAC CCATAGGCCA TCCGGCGGCC AGGGTTTGCA GTCTCCATTT 360
GGCCTGATCT CTACGAGAAG CTGGATTTCT CCGACGATCT CTAATGGCCT GTCGAATGGC 420
CATGGCATAC ATTATGTACA TCTCGGTATT TGAAATCTGG ATCCGAAAAA CTGGTCTATG 480
GCTCGTGTGT CGATGCGCTG AAACCAACGG CAACAAATTA CTTACCTTGT TGTTGTGTGA 540
TGGGTAAAAA CACACATCAC ACACTTAGGC CATAGGGATG CTCACCGTAG CCGCGGCTCC 600
AATCGCTTGA AGAAGTGTTC TTAGATCTAG TGGAAACCTG CGGAGAATGG CTTCTCGCCC 660
AGGGAGATCC GGCTGGGGTG GGAGCATGGG TCGTGCTGGA GCTGACCCAC CGGCATCATG 720
ATCGACCCGC TTTCTCTTCG TACCCTTCTG GGCCGGCTCC AGGTGGGCAT CTTCTGCTTC 780
CTTTTCTGAG CTGCTATCTG ATAACTCTAT GAGGACATTT TCCCAATCTC CCGCCGATAC 840
CTGTTCCTGC ACAACCGAGG TAGATGGGAC TTCTTCTTCC ATGTTGTCAT CCAGGGCCGG 900
GGGACCCGGC CTGTCCTTGT CCATTTTGTC TGCAACAAAA GTGTGACTCA CCAACACCGC 960
ACCCCCCTTG TACCTATTAA AGAGGATGCT GCCTAGAAAT CGGTGCCGAG ACA ATG 1016
Met
1
GAG GCA GCC TTG CTT GTG TGT CAG TAC ACC ATC CAG AGC CTG ATC CAT 1064 Glu Ala Ala Leu Leu Val Cys Gin Tyr Thr lie Gin Ser Leu He His 5 10 15 CTC ACG GGT GAA GAT CCT GGT TTT TTC AAT GTT GAG ATT CCG GAA TTC 1112 Leu Thr Gly Glu Asp Pro Gly Phe Phe Asn Val Glu He Pro Glu Phe 20 25 30
CCA TTT TAC CCC ACA TGC AAT GTT TGC ACG GCA GAT GTC AAT GTA ACT 1160 Pro Phe Tyr Pro Thr Cys Asn Val Cys Thr Ala Asp Val Asn Val Thr 35 40 45
ATC AAT TTC GAT GTC GGG GGC AAA AAG CAT CAA CTT GAT CTT GAC TTT 1208 He Asn Phe Asp Val Gly Gly Lys Lys His Gin Leu Asp Leu Asp Phe 50 55 60 65
GGC CAG CTG ACA CCC CAT ACG AAG GCT GTC TAC CAA CCT CGA GGT GCA 1256 Gly Gin Leu Thr Pro His Thr Lys Ala Val Tyr Gin Pro Arg Gly Ala 70 75 80
TTT GGT GGC TCA GAA AAT GCC ACC AAT CTC TTT CTA CTG GAG CTC CTT 1304
Phe Gly Gly Ser Glu Asn Ala Thr Asn Leu Phe Leu Leu Glu Leu Leu
85 90 95
GGT GCA GGA GAA TTG GCT CTA ACT ATG CGG TCT AAG AAG CTT CCA ATT 1352
Gly Ala Gly Glu Leu Ala Leu Thr Met Arg Ser Lys Lys Leu Pro He 100 105 110
AAC GTC ACC ACC GGA GAG GAG CAA CAA GTA AGC CTG GAA TCT GTA GAT 1400 Asn Val Thr Thr Gly Glu Glu Gin Gin Val Ser Leu Glu Ser Val Asp 115 120 125
GTC TAC TTT CAA GAT GTG TTT GGA ACC ATG TGG TGC CAC CAT GCA GAA 1448 Val Tyr Phe Gin Asp Val Phe Gly Thr Met Trp Cys His His Ala Glu 130 135 140 145
ATG CAA AAC CCC GTG TAC CTG ATA CCA GAA ACA GTG CCA TAC ATA AAG 1496 Met Gin Asn Pro Val Tyr Leu He Pro Glu Thr Val Pro Tyr He Lys 150 155 160
TGG GAT AAC TGT AAT TCT ACC AAT ATA ACG GCA GTA GTG AGG GCA CAG 1544 Trp Asp Asn Cys Asn Ser Thr Asn He Thr Ala Val Val Arg Ala Gin 165 170 175
GGG CTG GAT GTC ACG CTA CCC TTA AGT TTG CCA ACG TCA GCT CAA GAC 1592 Gly Leu Asp Val Thr Leu Pro Leu Ser Leu Pro Thr Ser Ala Gin Asp 180 185 190 TCG AAT TTC AGC GTA AAA ACA GAA ATG CTC GGT AAT GAG ATA GAT ATT 1640 Ser Asn Phe Ser Val Lys Thr Glu Met Leu Gly Asn Glu He Asp He 195 200 205
GAG TGT ATT ATG GAG GAT GGC GAA ATT TCA CAA GTT CTG CCC GGA GAC 1688 Glu Cys He Met Glu Asp Gly Glu He Ser Gin Val Leu Pro Gly Asp 210 215 220 225
AAC AAA TTT AAC ATC ACC TGC AGT GGA TAC GAG AGC CAT GTT CCC AGC 1736 Asn Lys Phe Asn He Thr Cys Ser Gly Tyr Glu Ser His Val Pro Ser
230 235 240
GGC GGA ATT CTC ACA TCA ACG AGT CCC GTG GCC ACC CCA ATA CCT GGT 1784 Gly Gly He Leu Thr Ser Thr Ser Pro Val Ala Thr Pro He Pro Gly 245 250 255
ACA GGG TAT GCA TAC AGC CTG CGT CTG ACA CCA CGT CCA GTG TCA CGA 1832 Thr Gly Tyr Ala Tyr Ser Leu Arg Leu Thr Pro Arg Pro Val Ser Arg 260 265 270
TTT CTT GGC AAT AAC AGT ATC CTG TAC GTG TTT TAC TCT GGG AAT GGA 1880 Phe Leu Gly Asn Asn Ser He Leu Tyr Val Phe Tyr Ser Gly Asn Gly 275 280 285
CCG AAG GCG AGC GGG GGA GAT TAC TGC ATT CAG TCC AAC ATT GTG TTC 1928 Pro Lys Ala Ser Gly Gly Asp Tyr Cys He Gin Ser Asn He Val Phe 290 295 300 305
TCT GAT GAG ATT CCA GCT TCA CAG GAC ATG CCG ACA AAC ACC ACA GAC 1976 Ser Asp Glu He Pro Ala Ser Gin Asp Met Pro Thr Asn Thr Thr Asp
310 315 320
ATC ACA TAT GTG GGT GAC AAT GCT ACC TAT TCA GTG CCA ATG GTC ACT 2024 He Thr Tyr Val Gly Asp Asn Ala Thr Tyr Ser Val Pro Met Val Thr 325 330 335
TCT GAG GAC GCA AAC TCG CCA AAT GTT ACA GTG ACT GCC TTT TGG GCC 2072 Ser Glu Asp Ala Asn Ser Pro Asn Val Thr Val Thr Ala Phe Trp Ala 340 345 350
TGG CCA AAC AAC ACT GAA ACT GAC TTT AAG TGC AAA TGG ACT CTC ACC 21 0 Trp Pro Asn Asn Thr Glu Thr Asp Phe Lys Cys Lys Trp Thr Leu Thr 355 360 365 TCG GGG ACA CCT TCG GGT TGT GAA AAT ATT TCT GGT GCA TTT GCG AGC 2168 Ser Gly Thr Pro Ser Gly Cys Glu Asn He Ser Gly Ala Phe Ala Ser 370 375 380 385
AAT CGG ACA TTT GAC ATT ACT GTC TCG GGT CTT GGC ACG GCC CCC AAG 2216 Asn Arg Thr Phe Asp He Thr Val Ser Gly Leu Gly Thr Ala Pro Lys 390 395 400
ACA CTC ATT ATC ACA CGA ACG GCT ACC AAT GCC ACC ACA ACA ACC CAC 2264 Thr Leu He He Thr Arg Thr Ala Thr Asn Ala Thr Thr Thr Thr His 405 410 415
AAG GTT ATA TTC TCC AAG GCA CCC GAG AGC ACC ACC ACC TCC CCT ACC 2312 Lys Val He Phe Ser Lys Ala Pro Glu Ser Thr Thr Thr Ser Pro Thr 420 425 430
TTG AAT ACA ACT GGA TTT GCT GAT CCC AAT ACA ACG ACA GGT CTA CCC 2360
Leu Asn Thr Thr Gly Phe Ala Asp Pro Asn Thr Thr Thr Gly Leu Pro 435 440 445
AGC TCT ACT CAC GTG CCT ACC AAC CTC ACC GCA CCT GCA AGC ACA GGC 2408
Ser Ser Thr His Val Pro Thr Asn Leu Thr Ala Pro Ala Ser Thr Gly 450 455 460 465
CCC ACT GTA TCC ACC GCG GAT GTC ACC AGC CCA ACA CCA GCC GGC ACA 2456 Pro Thr Val Ser Thr Ala Asp Val Thr Ser Pro Thr Pro Ala Gly Thr 470 475 480
ACG TCA GGC GCA TCA CCG GTG ACA CCA AGT CCA TCT CCA TGG GAC AAC 2504 Thr Ser Gly Ala Ser Pro Val Thr Pro Ser Pro Ser Pro Trp Asp Asn 485 490 495
GGC ACA GAA AGT AAG GCC CCC GAC ATG ACC AGC TCC ACC TCA CCA GTG 2552 Gly Thr Glu Ser Lys Ala Pro Asp Met Thr Ser Ser Thr Ser Pro Val 500 505 510
ACT ACC CCA ACC CCA AAT GCC ACC AGC CCC ACC CCA GCA GTG ACT ACC 2600
Thr Thr Pro Thr Pro Asn Ala Thr Ser Pro Thr Pro Ala Val Thr Thr 515 520 525
CCA ACC CCA AAT GCC ACC AGC CCC ACC CCA GCA GTG ACT ACC CCA ACC 2648 Pro Thr Pro Asn Ala Thr Ser Pro Thr Pro Ala Val Thr Thr Pro Thr 530 535 540 545 CCA AAT GCC ACC AGC CCC ACC TTG GGA AAA ACA AGT CCT ACC TCA GCA 2696 Pro Asn Ala Thr Ser Pro Thr Leu Gly Lys Thr Ser Pro Thr Ser Ala 550 555 560
GTG ACT ACC CCA ACC CCA AAT GCC ACC AGC CCC ACC TTG GGA AAA ACA 2744 Val Thr Thr Pro Thr Pro Asn Ala Thr Ser Pro Thr Leu Gly Lys Thr 565 570 575
AGC CCC ACC TCA GCA GTG ACT ACC CCA ACC CCA AAT GCC ACC AGC CCC 2792 Ser Pro Thr Ser Ala Val Thr Thr Pro Thr Pro Asn Ala Thr Ser Pro 580 585 590
ACC TTG GGA AAA ACA AGC CCC ACC TCA GCA GTG ACT ACC CCA ACC CCA 2840 Thr Leu Gly Lys Thr Ser Pro Thr Ser Ala Val Thr Thr Pro Thr Pro 595 600 605
AAT GCC ACC GGC CCT ACT GTG GGA GAA ACA AGT CCA CAG GCA AAT GCC 2888 Asn Ala Thr Gly Pro Thr Val Gly Glu Thr Ser Pro Gin Ala Asn Ala 610 615 620 625
ACC AAC CAC ACC TTA GGA GGA ACA AGT CCC ACC CCA GTA GTT ACC AGC 2936 Thr Asn His Thr Leu Gly Gly Thr Ser Pro Thr Pro Val Val Thr Ser 630 635 640
CAA CCA AAA AAT GCA ACC AGT GCT GTT ACC ACA GGC CAA CAT AAC ATA 2984 Gin Pro Lys Asn Ala Thr Ser Ala Val Thr Thr Gly Gin His Asn He 645 650 655
ACT TCA AGT TCA ACC TCT TCC ATG TCA CTG AGA CCC AGT TCA AAC CCA 3032 Thr Ser Ser Ser Thr Ser Ser Met Ser Leu Arg Pro Ser Ser Asn Pro 660 665 670
GAG ACA CTC AGC CCC TCC ACC AGT GAC AAT TCA ACG TCA CAT ATG CCT 3080 Glu Thr Leu Ser Pro Ser Thr Ser Asp Asn Ser Thr Ser His Met Pro 675 680 685
TTA CTA ACC TCC GCT CAC CCA ACA GGT GGT GAA AAT ATA ACA CAG GTG 3128 Leu Leu Thr Ser Ala His Pro Thr Gly Gly Glu Asn He Thr Gin Val 690 695 700 705
ACA CCA GCC TCT ATC AGC ACA CAT CAT GTG TCC ACC AGT TCG CCA GAA 3176 Thr Pro Ala Ser He Ser Thr His His Val Ser Thr Ser Ser Pro Glu 710 715 720 CCC CGC CCA GGC ACC ACC AGC CAA GCG TCA GGC CCT GGA AAC AGT TCC 3224 Pro Arg Pro Gly Thr Thr Ser Gin Ala Ser Gly Pro Gly Asn Ser Ser 725 730 735
5 ACA TCC ACA AAA CCG GGG GAG GTT AAT GTC ACC AAA GGC ACG CCC CCC 3272 Thr Ser Thr Lys Pro Gly Glu Val Asn Val Thr Lys Gly Thr Pro Pro 740 745 750
CAA AAT GCA ACG TCG CCC CAG GCC CCC AGT GGC CAA AAG ACG GCG GTT 3320 10 Gin Asn Ala Thr Ser Pro Gin Ala Pro Ser Gly Gin Lys Thr Ala Val 755 760 765
CCC ACG GTC ACC TCA ACA GGT GGA AAG GCC AAT TCT ACC ACC GGT GGA 3368 Pro Thr Val Thr Ser Thr Gly Gly Lys Ala Asn Ser Thr Thr Gly Gly 15 770 775 780 785
AAG CAC ACC ACA GGA CAT GGA GCC CGG ACA AGT ACA GAG CCC ACC ACA 3416 Lys His Thr Thr Gly His Gly Ala Arg Thr Ser Thr Glu Pro Thr Thr 790 795 800
20
GAT TAC GGC GGT GAT TCA ACT ACG CCA AGA CCG AGA TAC AAT GCG ACC 3464 Asp Tyr Gly Gly Asp Ser Thr Thr Pro Arg Pro Arg Tyr Asn Ala Thr 805 810 815
25 ACC TAT CTA CCT CCC AGC ACT TCT AGC AAA CTG CGG CCC CGC TGG ACT 3512 Thr Tyr Leu Pro Pro Ser Thr Ser Ser Lys Leu Arg Pro Arg Trp Thr 820 825 830
TTT ACG AGC CCA CCG GTT ACC ACA GCC CAA GCC ACC GTG CCA GTC CCG 3560 30 Phe Thr Ser Pro Pro Val Thr Thr Ala Gin Ala Thr Val Pro Val Pro 835 840 845
CCA ACG TCC CAG CCC AGA TTC TCA AAC CTC TCC ATG CTA GTA CTG CAG 3608 Pro Thr Ser Gin Pro Arg Phe Ser Asn Leu Ser Met Leu Val Leu Gin 35 850 855 860 865
TGG GCC TCT CTG GCT GTG CTG ACC CTT CTG CTG CTG CTG GTC ATG GCG 3656 Trp Ala Ser Leu Ala Val Leu Thr Leu Leu Leu Leu Leu Val Met Ala 870 875 880
40
GAC TGC GCC TTT AGG CGT AAC TTG TCT ACA TCC CAT ACC TAC ACC ACC 3704 Asp Cys Ala Phe Arg Arg Asn Leu Ser Thr Ser His Thr Tyr Thr Thr 885 890 895 CCA CCA TAT GAT GAC GCC GAG ACC TAT GTA TAAAGTCAAT AAAAATTTAT 3754 Pro Pro Tyr Asp Asp Ala Glu Thr Tyr Val 900 905
TAATCAGAAA TTTGCACTTT CTTTGCTTCA CGTCCCCGGG AGCGGGAGCG GGCACGTCGG 3814
GTGGCGTTGG GGTCGTTTG 3833
(2) INFORMATION FOR SEQ ID NO:19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 907 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:19:
Met Glu Ala Ala Leu Leu Val Cys Gin Tyr Thr He Gin Ser Leu He 1 5 10 15
His Leu Thr Gly Glu Asp Pro Gly Phe Phe Asn Val Glu He Pro Glu 20 25 30
Phe Pro Phe Tyr Pro Thr Cys Asn Val Cys Thr Ala Asp Val Asn Val 35 40 45
Thr He Asn Phe Asp Val Gly Gly Lys Lys His Gin Leu Asp Leu Asp 50 55 60
Phe Gly Gin Leu Thr Pro His Thr Lys Ala Val Tyr Gin Pro Arg Gly 65 70 75 80
Ala Phe Gly Gly Ser Glu Asn Ala Thr Asn Leu Phe Leu Leu Glu Leu 85 90 95
Leu Gly Ala Gly Glu Leu Ala Leu Thr Met Arg Ser Lys Lys Leu Pro 100 105 110
He Asn Val Thr Thr Gly Glu Glu Gin Gin Val Ser Leu Glu Ser Val 115 120 125 46
Asp Val Tyr Phe Gin Asp Val Phe Gly Thr Met Trp Cys His His Ala 130 135 140
Glu Met Gin Asn Pro Val Tyr Leu He Pro Glu Thr Val Pro Tyr He 5 145 150 155 160
Lys Trp Asp Asn Cys Asn Ser Thr Asn He Thr Ala Val Val Arg Ala 165 170 175
10 Gin Gly Leu Asp Val Thr Leu Pro Leu Ser Leu Pro Thr Ser Ala Gin 180 185 190
Asp Ser Asn Phe Ser Val Lys Thr Glu Met Leu Gly Asn Glu He Asp 195 200 205
15
He Glu Cys He Met Glu Asp Gly Glu He Ser Gin Val Leu Pro Gly 210 215 220
Asp Asn Lys Phe Asn He Thr Cys Ser Gly Tyr Glu Ser His Val Pro 20 225 230 235 240
Ser Gly Gly He Leu Thr Ser Thr Ser Pro Val Ala Thr Pro He Pro 245 250 255
25 Gly Thr Gly Tyr Ala Tyr Ser Leu Arg Leu Thr Pro Arg Pro Val Ser 260 265 270
Arg Phe Leu Gly Asn Asn Ser He Leu Tyr Val Phe Tyr Ser Gly Asn 275 280 285
30
Gly Pro Lys Ala Ser Gly Gly Asp Tyr Cys He Gin Ser Asn He Val 290 295 300
Phe Ser Asp Glu He Pro Ala Ser Gin Asp Met Pro Thr Asn Thr Thr 35 305 310 315 320
Asp He Thr Tyr Val Gly Asp Asn Ala Thr Tyr Ser Val Pro Met Val 325 330 335
40 Thr Ser Glu Asp Ala Asn Ser Pro Asn Val Thr Val Thr Ala Phe Trp 340 345 350
Ala Trp Pro Asn Asn Thr Glu Thr Asp Phe Lys Cys Lys Trp Thr Leu 355 360 365
Thr Ser Gly Thr Pro Ser Gly Cys Glu Asn He Ser Gly Ala Phe Ala 370 375 380
Ser Asn Arg Thr Phe Asp He Thr Val Ser Gly Leu Gly Thr Ala Pro 385 390 395 400
Lys Thr Leu He He Thr Arg Thr Ala Thr Asn Ala Thr Thr Thr Thr 405 410 415
His Lys Val He Phe Ser Lys Ala Pro Glu Ser Thr Thr Thr Ser Pro 420 425 430
Thr Leu Asn Thr Thr Gly Phe Ala Asp Pro Asn Thr Thr Thr Gly Leu 435 440 445
Pro Ser Ser Thr His Val Pro Thr Asn Leu Thr Ala Pro Ala Ser Thr 450 455 460
Gly Pro Thr Val Ser Thr Ala Asp Val Thr Ser Pro Thr Pro Ala Gly 465 470 475 480
Thr Thr Ser Gly Ala Ser Pro Val Thr Pro Ser Pro Ser Pro Trp Asp 485 490 495
Asn Gly Thr Glu Ser Lys Ala Pro Asp Met Thr Ser Ser Thr Ser Pro 500 505 510
Val Thr Thr Pro Thr Pro Asn Ala Thr Ser Pro Thr Pro Ala Val Thr 515 520 525
Thr Pro Thr Pro Asn Ala Thr Ser Pro Thr Pro Ala Val Thr Thr Pro 530 535 540
Thr Pro Asn Ala Thr Ser Pro Thr Leu Gly Lys Thr Ser Pro Thr Ser 545 550 555 560
Ala Val Thr Thr Pro Thr Pro Asn Ala Thr Ser Pro Thr Leu Gly Lys 565 570 575
Thr Ser Pro Thr Ser Ala Val Thr Thr Pro Thr Pro Asn Ala Thr Ser 580 585 590 Pro Thr Leu Gly Lys Thr Ser Pro Thr Ser Ala Val Thr Thr Pro Thr 595 600 605
Pro Asn Ala Thr Gly Pro Thr Val Gly Glu Thr Ser Pro Gin Ala Asn 610 615 620
Ala Thr Asn His Thr Leu Gly Gly Thr Ser Pro Thr Pro Val Val Thr 625 630 635 640
Ser Gin Pro Lys Asn Ala Thr Ser Ala Val Thr Thr Gly Gin His Asn
645 650 655
He Thr Ser Ser Ser Thr Ser Ser Met Ser Leu Arg Pro Ser Ser Asn 660 665 670
Pro Glu Thr Leu Ser Pro Ser Thr Ser Asp Asn Ser Thr Ser His Met 675 680 685
Pro Leu Leu Thr Ser Ala His Pro Thr Gly Gly Glu Asn He Thr Gin 690 695 700
Val Thr Pro Ala Ser He Ser Thr His His Val Ser Thr Ser Ser Pro 705 710 715 720
Glu Pro Arg Pro Gly Thr Thr Ser Gin Ala Ser Gly Pro Gly Asn Ser
725 730 735
Ser Thr Ser Thr Lys Pro Gly Glu Val Asn Val Thr Lys Gly Thr Pro 740 745 750
Pro Gin Asn Ala Thr Ser Pro Gin Ala Pro Ser Gly Gin Lys Thr Ala 755 760 765
Val Pro Thr Val Thr Ser Thr Gly Gly Lys Ala Asn Ser Thr Thr Gly 770 775 780
Gly Lys His Thr Thr Gly His Gly Ala Arg Thr Ser Thr Glu Pro Thr 785 790 795 800
Thr Asp Tyr Gly Gly Asp Ser Thr Thr Pro Arg Pro Arg Tyr Asn Ala
805 810 815
Thr Thr Tyr Leu Pro Pro Ser Thr Ser Ser Lys Leu Arg Pro Arg Trp 820 825 830
Thr Phe Thr Ser Pro Pro Val Thr Thr Ala Gin Ala Thr Val Pro Val 835 840 845
Pro Pro Thr Ser Gin Pro Arg Phe Ser Asn Leu Ser Met Leu Val Leu 850 855 860
Gin Trp Ala Ser Leu Ala Val Leu Thr Leu Leu Leu Leu Leu Val Met 865 870 875 880
Ala Asp Cys Ala Phe Arg Arg Asn Leu Ser Thr Ser His Thr Tyr Thr 885 890 895
Thr Pro Pro Tyr Asp Asp Ala Glu Thr Tyr Val 900 905

Claims

CLAIMS We claim:
1. An isolated DNA sequence that codes on expression for an homogeneous gp350 protein.
2. A DNA sequence of Claim 1 further characterized by having a nucleotide sequence in which at least one native nucleotide encoding serine at position 501 of Fig. 1 is replaced with a non-native nucleotide and in which at least one native nucleotide encoding glycine at position 698 is replaced with a non-native nucleotide.
3. A DNA sequence of Claim 1 wherein said homogeneous gp350 protein is further characterized by encoding an amino acid sequence selected from the group consisting of
(a) amino acid 19 through amino acid 862 of Fig. 1. ( ) amino acid 1 through amino acid 862 of Fig. 1. (c) amino acid 19 through amino acid 862 and amino acid 882 through amino acid 907 of Fig. 1.
(d) amino acid 1 through amino acid 862 and amino acid 882 through amino acid 907 of Fig. 1.
(e) amino acid 1 through amino acid 907 wherein nucleotides encoding at least 8 amino acids in the transmembrane region are deleted.
4. A vector comprising a DNA sequence of Claim 1, Claim 2 or Claim 3.
5. A host cell transformed with a DNA sequence of Claim 1, Claim 2 or Claim 3 in operative association with an expression control sequence capable of directing replicated and expression of said DNA sequence.
6. A process for producing a homogenous gp350 protein comprising cultuiing a host cell of Claim 5 in a suitable culture medium and isolating said homogeneous gp350 protein from said cell.
7. A homogeneous gp350 protein, made according to the process of
Claim 6.
8. A homogeneous gp350 protein, wherein one or more of the native nucleotides encoding the donor splice site and acceptor splice site are replaced with substitute nucleotide(s) different from said native nucleotide(s) which preserve the amino acid sequence of said donor and acceptor sites.
9. A pharmaceutical composition comprising the homogeneous gp350 protein of Claim 7 or Claim 8 in admixture with a pharmaceutically acceptable carrier.
10. Use of a homogenous gp350 protein for the preparation of a pharmaceutic composition suitable for the prophylactic treatment of a EBV-related disease or condition.
PCT/US1995/004611 1994-04-18 1995-04-13 NON-SPLICING VARIANTS OF gp350/220 WO1995028488A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
EP95916984A EP0769056B1 (en) 1994-04-18 1995-04-13 NON-SPLICING VARIANTS OF gp350/220
CZ19963054A CZ292283B6 (en) 1994-04-18 1995-04-13 Isolated DNA code sequence, vector, host cell, protein, pharmaceutical composition and vaccine
PL95316941A PL181881B1 (en) 1994-04-18 1995-04-13 Variant ofgp 350/220 being not susceptible to composition
AT95916984T ATE210184T1 (en) 1994-04-18 1995-04-13 UNSPLICED VARIANTS OF THE GP350/220
HU9602894A HU221647B1 (en) 1994-04-18 1995-04-13 Dna-sequence encoding protein ebv gp350
JP52710995A JP3447743B2 (en) 1994-04-18 1995-04-13 Non-splicing variants of gp350 / 220
SK1343-96A SK283446B6 (en) 1994-04-18 1995-04-13 Isolated DNA variant, host cell, method for production of homogeneous gp350 protein, pharmaceutical composition containing it and use
AU23838/95A AU707837B2 (en) 1994-04-18 1995-04-13 Non-splicing variants of gp350/220
DE69524415T DE69524415T2 (en) 1994-04-18 1995-04-13 UNSPLEATED VARIANTS OF THE GP350 / 220
UA96114355A UA47403C2 (en) 1994-04-18 1995-04-13 Separated dna fragment coding protein gp350 ebv or truncated variant of gp350 ebv, vector, host cell transformed by vector, process for producing gp350 protein, homogenous gp350 ebv protein, pharmaceutical composition, method of prophylactic treatment of disease or state related to ebv (variants), composition containing homogenous gp350 ebv
BR9507473A BR9507473A (en) 1994-04-18 1995-04-13 DNA sequence isolated vector host cell process to produce a homogeneous gp350 protein homogeneous gp350 protein pharmaceutical composition and use in homogeneous gp350 protein
CA002187908A CA2187908C (en) 1994-04-18 1995-04-13 Non-splicing variants of gp350/220
SI9530569T SI0769056T1 (en) 1994-04-18 1995-04-13 NON-SPLICING VARIANTS OF gp350/220
DK95916984T DK0769056T3 (en) 1994-04-18 1995-04-13 Non-splicing variants of gp350 / 220
NO19964431A NO319382B1 (en) 1994-04-18 1996-10-18 Isolated DNA fragment encoding the EBV gp350 protein or an abbreviated version thereof, as well as vector, host cell and pharmaceutical composition containing the DNA fragment or protein and method of preparation and use thereof
FI964186A FI118224B (en) 1994-04-18 1996-10-18 non-looping variants of gp350 / 220
LVP-96-430A LV11803B (en) 1994-04-18 1996-11-12 Non-splicing variants of gp350/220
FI20075338A FI20075338A (en) 1994-04-18 2007-05-10 No looping changes to gb350 / 220

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US22929194A 1994-04-18 1994-04-18
US08/229,291 1994-04-18

Publications (1)

Publication Number Publication Date
WO1995028488A1 true WO1995028488A1 (en) 1995-10-26

Family

ID=22860582

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/004611 WO1995028488A1 (en) 1994-04-18 1995-04-13 NON-SPLICING VARIANTS OF gp350/220

Country Status (25)

Country Link
US (3) US6054130A (en)
EP (1) EP0769056B1 (en)
JP (4) JP3447743B2 (en)
KR (1) KR100380953B1 (en)
CN (2) CN100415895C (en)
AT (1) ATE210184T1 (en)
AU (1) AU707837B2 (en)
BR (1) BR9507473A (en)
CA (1) CA2187908C (en)
CZ (1) CZ292283B6 (en)
DE (1) DE69524415T2 (en)
DK (1) DK0769056T3 (en)
ES (1) ES2170144T3 (en)
FI (2) FI118224B (en)
HU (1) HU221647B1 (en)
LV (1) LV11803B (en)
MY (1) MY114769A (en)
NO (1) NO319382B1 (en)
PL (1) PL181881B1 (en)
PT (1) PT769056E (en)
RU (1) RU2178807C2 (en)
SK (1) SK283446B6 (en)
TW (1) TW496897B (en)
UA (1) UA47403C2 (en)
WO (1) WO1995028488A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999064603A2 (en) * 1998-06-12 1999-12-16 Henry M. Jackson Foundation For The Advancement Of Military Medicine ENHANCEMENT OF B CELL ACTIVATION AND IMMUNOGLOBULIN SECRETION BY CO-STIMULATION OF RECEPTORS FOR ANTIGEN AND EBV Gp350/220
US6268470B1 (en) * 1996-03-15 2001-07-31 Millennium Pharmaceuticals, Inc. Composition and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation
US6723695B1 (en) * 1997-07-10 2004-04-20 Council Of The Queensland Institute Of Medical Research CTL epitopes from EBV

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6692749B1 (en) * 1994-04-18 2004-02-17 Medimmune Vaccines, Inc. Non-splicing variants of gp350/220
MY114769A (en) 1994-04-18 2003-01-31 Aviron Inc Non-splicing variants of gp350/220
JP2002517180A (en) * 1997-12-05 2002-06-18 ジ・イミユーン・リスポンス・コーポレーシヨン Novel vectors and genes exhibiting increased expression
GB0210682D0 (en) * 2002-05-09 2002-06-19 Glaxosmithkline Biolog Sa Novel use
CA2799995A1 (en) * 2009-05-22 2010-11-25 Intelligent Medical Devices, Inc. Optimized probes and primers and methods of using same for the detection, screening, quantitation, isolation and sequencing of cytomegalovirus and epstein-barr virus
US10461635B1 (en) * 2018-05-15 2019-10-29 Analog Devices Global Unlimited Company Low VIN high efficiency chargepump

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3069310D1 (en) * 1980-11-03 1984-10-31 Itt Ind Gmbh Deutsche Binary mos ripple carry parallel adder/subtractor and appropriate adding/subtracting stage
US4707358A (en) * 1984-01-30 1987-11-17 The University Of Chicago Vaccine against Epstein-Barr Virus
US5244792A (en) * 1984-04-06 1993-09-14 Chiron Corporation Expression of recombinant glyoprotein B from herpes simplex virus
US5171568A (en) * 1984-04-06 1992-12-15 Chiron Corporation Recombinant herpes simplex gb-gd vaccine
EP0173254B1 (en) * 1984-08-23 1991-07-24 Hans Joachim Wolf Dna sequences of the ebv genome, recombinant dna molecules, processes for producing ebv-related antigens, diagnostic compositions and pharmaceutical compositions containing said antigens
EP0312164A1 (en) * 1987-10-16 1989-04-19 Merck & Co. Inc. Purification of recombinant epstein-barr virus antigens from vero cells, yeast cells or L cells
CA2090295A1 (en) * 1992-02-03 1993-08-04 Anthony B. Nesburn Process for the expression of herpes simplex virus type 1 glycoprotein e and methods of use
AU1648092A (en) * 1992-03-19 1993-10-21 Cancer Research Campaign Technology Limited Defective recombinant adenoviruses expressing characteristic epstein-barr virus proteins
US5474914A (en) * 1992-07-29 1995-12-12 Chiron Corporation Method of producing secreted CMV glycoprotein H
MY114769A (en) 1994-04-18 2003-01-31 Aviron Inc Non-splicing variants of gp350/220

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
EMBO JOURNAL, Volume 3, Number 5, issued 1984, BIGGEN et al., "Transcription and DNA Sequence of the BamHI L Fragment of B95-8 Epstein-Barr Virus", pages 1083-1090. *
EMBO JOURNAL, Volume 4, Number 12, issued 1985, MACKETT et al., "Recombinant Vaccinia Virus Induces Neutralizing Antibodies in Rabbits Against Epstein-Barr Virus Membrane Antigen gp340", pages 3229-3234. *
GENE, Volume 44, issued 1986, MOTZ et al., "Expression of the Epstein-Barr Virus Major Membrane Protein in Chinese Hamster Ovary Cells", pages 353-359. *
JOURNAL OF GENERAL VIROLOGY, Volume 73, issued 1992, FINERTY et al., "Protective Immunization Against Epstein-Barr Virus-Induced Disease in Cottontop Tamarins Using the Virus Envelope Glycoprotein gp340 Produced by a Bovine Pappillomavirus Expression Vector", pages 449-453. *
JOURNAL OF IMMUNOLOGICAL METHODS, Volume 108, issued 1988, DAVID et al., "Efficient Purification of Epstein-Barr Virus Membrane Antigen gp340 by Fast Protein Liquid Chromatography", pages 231-236. *
JOURNAL OF VIROLOGY, Volume 61, issued June 1987, WHANG et al., "Expression of the Epstein-Barr Virus gp350/220 Gene in Rodent and Primate Cells", pages 1796-1807. *
JOURNAL OF VIROLOGY, Volume 61, Number 5, issued May 1987, NEMEROW et al., "Identification of gp350 as the Viral Glycoprotein Mediating Attachment of Epstein-Bar Virus (EBV) to the EBV/C3d Receptor of B Cells; Sequence Homology or gp350 and C3 Complement Fragment C3d", pages 1416-1420. *
NUCLEIC ACIDS RESEARCH, Volume 10, Number 2, issued 1982, MOUNT, "A Catalog of Splice Junction Sequences", pages 459-472. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6268470B1 (en) * 1996-03-15 2001-07-31 Millennium Pharmaceuticals, Inc. Composition and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation
US6723695B1 (en) * 1997-07-10 2004-04-20 Council Of The Queensland Institute Of Medical Research CTL epitopes from EBV
WO1999064603A2 (en) * 1998-06-12 1999-12-16 Henry M. Jackson Foundation For The Advancement Of Military Medicine ENHANCEMENT OF B CELL ACTIVATION AND IMMUNOGLOBULIN SECRETION BY CO-STIMULATION OF RECEPTORS FOR ANTIGEN AND EBV Gp350/220
WO1999064603A3 (en) * 1998-06-12 2000-02-10 Jackson H M Found Military Med ENHANCEMENT OF B CELL ACTIVATION AND IMMUNOGLOBULIN SECRETION BY CO-STIMULATION OF RECEPTORS FOR ANTIGEN AND EBV Gp350/220
US6432679B1 (en) 1998-06-12 2002-08-13 Henry M. Jackson Foundation For The Advancement Of Military Medicine Enhancement of B cell activation and immunoglobulin secretion by co-stimulation of receptors for antigen and EBV Gp350/220

Also Published As

Publication number Publication date
CN100415895C (en) 2008-09-03
DE69524415D1 (en) 2002-01-17
CN1118573C (en) 2003-08-20
US5824508A (en) 1998-10-20
JP2004290199A (en) 2004-10-21
CN1152940A (en) 1997-06-25
DK0769056T3 (en) 2002-04-02
LV11803A (en) 1997-06-20
JP2005333990A (en) 2005-12-08
PT769056E (en) 2002-05-31
NO964431D0 (en) 1996-10-18
MY114769A (en) 2003-01-31
FI20075338A (en) 2007-05-10
NO319382B1 (en) 2005-07-25
RU2178807C2 (en) 2002-01-27
CZ292283B6 (en) 2003-08-13
AU2383895A (en) 1995-11-10
US6458364B1 (en) 2002-10-01
KR100380953B1 (en) 2003-10-10
HU9602894D0 (en) 1996-12-30
UA47403C2 (en) 2002-07-15
SK134396A3 (en) 1997-06-04
SK283446B6 (en) 2003-07-01
CA2187908A1 (en) 1995-10-26
EP0769056B1 (en) 2001-12-05
FI964186A0 (en) 1996-10-18
PL181881B1 (en) 2001-09-28
FI118224B (en) 2007-08-31
CA2187908C (en) 2002-09-17
PL316941A1 (en) 1997-02-17
LV11803B (en) 1998-01-20
JP4317786B2 (en) 2009-08-19
DE69524415T2 (en) 2002-08-01
HU221647B1 (en) 2002-12-28
CZ305496A3 (en) 1997-10-15
ATE210184T1 (en) 2001-12-15
EP0769056A1 (en) 1997-04-23
HUT75831A (en) 1997-05-28
NO964431L (en) 1996-12-11
US6054130A (en) 2000-04-25
JPH10501687A (en) 1998-02-17
ES2170144T3 (en) 2002-08-01
CN1495262A (en) 2004-05-12
BR9507473A (en) 1997-09-23
JP3447743B2 (en) 2003-09-16
JP2003230396A (en) 2003-08-19
TW496897B (en) 2002-08-01
EP0769056A4 (en) 1998-03-04
FI964186A (en) 1996-12-17
AU707837B2 (en) 1999-07-22

Similar Documents

Publication Publication Date Title
EP0139417B1 (en) Vaccines based on membrane bound proteins and process for making them
Robbins et al. Pseudorabies virus gene encoding glycoprotein gIII is not essential for growth in tissue culture
US6054131A (en) Vaccine composition for herpes simplex virus and method of using
JP2005333990A (en) Gp350/220 non-splicing variant
Bowles et al. The ICP0 protein of equine herpesvirus 1 is an early protein that independently transactivates expression of all classes of viral promoters
US20130064844A1 (en) Non-splicing variants of gp350/220
US5807557A (en) Soluble herpesvirus glycoprotein complex
US6713296B1 (en) VZV gene, mutant VZV and immunogenic compositions
WO1998015289A1 (en) Proteins of kaposi&#39;s sarcoma associated herpesvirus
WO1998015289A9 (en) Proteins of kaposi&#39;s sarcoma associated herpesvirus
AU6404594A (en) Recombinant epstein-barr virus protein and its use in vaccine

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 95193673.5

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TT UA UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2187908

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1019960705822

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: PV1996-3054

Country of ref document: CZ

Ref document number: 964186

Country of ref document: FI

Ref document number: 134396

Country of ref document: SK

WWE Wipo information: entry into national phase

Ref document number: 1199690348

Country of ref document: VN

WWE Wipo information: entry into national phase

Ref document number: 1995916984

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1995916984

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV1996-3054

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 1995916984

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: PV1996-3054

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 20075338

Country of ref document: FI