WO2001038350A2 - 85kDa NEISSERIAL ANTIGEN - Google Patents

85kDa NEISSERIAL ANTIGEN Download PDF

Info

Publication number
WO2001038350A2
WO2001038350A2 PCT/IB2000/001851 IB0001851W WO0138350A2 WO 2001038350 A2 WO2001038350 A2 WO 2001038350A2 IB 0001851 W IB0001851 W IB 0001851W WO 0138350 A2 WO0138350 A2 WO 0138350A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
protein
sequence
expression
antigen against
Prior art date
Application number
PCT/IB2000/001851
Other languages
French (fr)
Other versions
WO2001038350A3 (en
Inventor
Marzia Monica Giuliani
Mariagrazia Pizza
Rino Rappuoli
Johan Holst
Original Assignee
Chiron Spa
Statens Institutt For Folkehelse
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 claimed from GBGB9928197.4A external-priority patent/GB9928197D0/en
Priority claimed from GB0005698A external-priority patent/GB0005698D0/en
Application filed by Chiron Spa, Statens Institutt For Folkehelse filed Critical Chiron Spa
Priority to CA2390344A priority Critical patent/CA2390344C/en
Priority to JP2001540113A priority patent/JP4840956B2/en
Priority to BR0015961-1A priority patent/BR0015961A/en
Priority to EP00981520A priority patent/EP1234039A2/en
Priority to AU18753/01A priority patent/AU1875301A/en
Publication of WO2001038350A2 publication Critical patent/WO2001038350A2/en
Publication of WO2001038350A3 publication Critical patent/WO2001038350A3/en
Priority to US11/264,676 priority patent/US7700119B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/22Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Neisseriaceae (F)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to an antigen from the related bacteria Neisseria meningitidis and Neisseria gonorrhoeae.
  • Neisseria meningitidis is a non-motile, Gram-negative diplococcus human pathogen. It colonises the pharynx, causing meningitis and, occasionally, septicaemia in the absence of meningitis. It is closely related to N. gonorrhoeae, although one feature that clearly differentiates meningococcus from gonococcus is the presence of a polysaccharide capsule that is present in all pathogenic meningococci.
  • N. meningitidis causes both endemic and epidemic disease. In the United States the attack rate is 0.6- 1 per 100,000 persons per year, and it can be much greater during outbreaks [Lieber an et al. (1996) JAMA 275(19):1499-1503; Schuchat et al (1997) N Engl J Med 337(14):970-976]. In developing countries, endemic disease rates are much higher and during epidemics incidence rates can reach 500 cases per 100,000 persons per year. Mortality is extremely high, at 10-20% in the United States, and much higher in developing countries. Following the introduction of the conjugate vaccine against Haemophilus influenzae, N. meningitidis is the major cause of bacterial meningitis at all ages in the United States [Schuchat et al (1997) supra].
  • the meningococcal vaccine currently in use is a tetravalent polysaccharide vaccine composed of serogroups A, C, Y and W135. Following the success of the vaccination against H.influenzae, however, conjugate vaccines against serogroups A and C have been developed
  • Meningococcus B remains a problem, however. This serotype currently is responsible for approximately 50% of total meningitis in the United States, Europe, and South America.
  • the polysaccharide approach cannot be used because the menB capsular polysaccharide is a polymer of ⁇ (2-8)-Iinked N-acetyl neuraminic acid that is also present in mammalian tissue. This results in tolerance to the antigen; indeed, if a response were elicited, it would be anti-self, and therefore undesirable.
  • the capsular polysaccharide has, for instance, been chemically modified substituting the N-acetyl groups with N-propionyl groups, leaving the specific antigenicity unaltered [Romero & Outschoorn (1994) Clin Microbiol Rev 7(4):559-575].
  • OMPs outer membrane proteins
  • the invention provides a protein comprising one or more of the following amino acid sequences: SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 1 1, SEQ ID 12, SEQ ID 13.
  • a protein comprising a sequence having sequence identity to SEQ ID 1 , SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 11, SEQ ID 12, or SEQ ID 13.
  • the degree of sequence identity is preferably greater than 50% (e.g. 60%, 70%, 80%, 90%, 95%, 99% or more).
  • These homologous sequences include mutants and allelic variants.
  • 50% identity or more between two proteins is considered to be an indication of functional equivalence.
  • the invention further provides a protein comprising a fragment of SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 11, SEQ ID 12, or SEQ ID 13.
  • the fragment should comprise at least n consecutive amino acids from the sequence and, depending on the particular sequence, n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20 or more).
  • n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20 or more).
  • the fragment comprises an epitope from the sequence.
  • the proteins of the invention can, of course, be prepared by various means (e.g. recombinant expression, purification from cell culture, chemical synthesis etc.) and in various forms (e.g. native, fusions etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other Neisseria or host cell proteins).
  • the invention provides antibodies which bind to these proteins. These may be polyclonal or monoclonal and may be produced by any suitable means.
  • the invention provides nucleic acid comprising SEQ ID 2, SEQ ID 6, or SEQ ID 10.
  • the invention provides nucleic acid comprising sequences having sequence identity to SEQ ID 2, SEQ ID 6, or SEQ ID 10.
  • the degree of sequence identity is preferably greater than 50% (e.g. 60%, 70%, 80%, 90%, 95%, 99% or more).
  • nucleic acid which can hybridise to SEQ ID 2, SEQ ID 6, or SEQ ID 10, preferably under "high stringency” conditions (e.g. 65°C in a 0. IxSSC, 0.5% SDS solution).
  • Nucleic acid comprising fragments of SEQ ID 2, SEQ ID 6, or SEQ ID 10 are also provided. These should comprise at least n consecutive nucleotides from SEQ ID 2, SEQ ID 6, or SEQ ID 10 and, depending on the particular sequence, n is 10 or more (eg 12, 14, 15, 18, 20, 25, 30, 35, 40 or more).
  • the invention provides nucleic acid encoding the proteins and protein fragments of the invention.
  • nucleic acid comprising sequences complementary to those described above (e.g. for antisense or probing pu ⁇ oses).
  • Nucleic acid according to the invention can, of course, be prepared in many ways (e.g. by chemical synthesis, from genomic or cDNA libraries, from the organism itself etc.) and can take various forms (e.g. single stranded, double stranded, vectors, probes etc.).
  • nucleic acid includes DNA and RNA, and also their analogues, such as those containing modified backbones, and also peptide nucleic acids (PNA) etc.
  • the invention provides vectors comprising nucleotide sequences of the invention (e.g. expression vectors) and host cells transformed with such vectors.
  • vectors comprising nucleotide sequences of the invention (e.g. expression vectors) and host cells transformed with such vectors.
  • compositions comprising protein, antibody, and/or nucleic acid according to the invention.
  • These compositions may be suitable as vaccines, for instance, or as diagnostic reagents, or as immunogenic compositions.
  • compositions of the invention may further comprise immunogenic components selected from one or more of the following:
  • compositions further comprise immunogenic components selected from one or more of the following:
  • a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8,
  • a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, & 90, as disclosed in W099/36544 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
  • a protein encoded by one of the 21 60 genes N M B 0001 to N M B 2160 disclosed in Tettelin et al [Science (2000) 287: 1809-1815] or a protein com prising an im m unogenic fragm ent of one or m ore of these 2160 genes, or a protein com prising a sequence having sequence identity (preferably greater than 50% e.g. 60% , 70% , 80% , 90% , 95 % , 99% or m ore) to one of these 2 160 genes);
  • a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62,
  • a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1-8 disclosed in WO96/29412 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
  • a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1-23 disclosed in WO95/03413 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
  • a protein comprising an amino acid sequence consisting of SEQ ID 2 disclosed in WO99/31 132 (or a protein comprising an immunogenic fragment of SEQ ID 2, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to SEQ ID 2);
  • a polysaccharide antigen against Neisseria meningitidis serogroup C such as that described in Costantino et al. (1992) Vaccine 10:691 -698;
  • a polysaccharide antigen against pneumococcus • A protective antigen against diphtheria, consisting of a diphtheria toxoid, such as the
  • CRM197 mutant [e.g. Del Guidice et al. (1998) Molecular Aspects of Medicine 19: 1 -70].
  • a protective antigen against tetanus consisting of a tetanus toxoid [e.g. Wassilak & Orenstein, Chapter 4 of Vaccines (eds. Plotkin & Mortimer), 1988]
  • a protective antigen against whooping cough comprising pertussis holotoxin (PT) and filamentous haemagglutinin (FHA); optionally further comprising pertactin and/or agglutinogens 2 and 3 [e.g. Gustafsson et al. (1996) N. Engl. J. Med. 334:349-355; Rappuoli et al (1991) T1BTECH 9:232-238].
  • a protective antigen against H. pylori comprising one or more of CagA (e.g. WO93/18150), VacA (e.g. WO93/18150), NAP (e.g. WO99/53310), HopX (e.g. WO98/04702), HopY (e.g. WO98/04702), urease.
  • CagA e.g. WO93/18150
  • VacA e.g. WO93/18150
  • NAP e.g. WO99/53310
  • HopX e.g. WO98/04702
  • HopY e.g. WO98/04702
  • a protective antigen against hepatitis B virus consisting of a HBV surface antigen and/or a HBV core antigen.
  • the composition comprises an antigen against diphtheria, it preferably also comprises antigens against tetanus and polio.
  • the composition comprises an antigen against tetanus, it preferably also comprises antigens against diphtheria and polio.
  • the composition comprises an antigen against polio, it preferably also comprises antigens against diphtheria and tetanus.
  • Pertussis toxin is a toxic protein and, when present in the composition, it is preferably detoxified. Detoxification may be by chemical and/or genetic means.
  • a preferred detoxified mutant is the 9K/129G double mutant [e.g. Rappuoli (1997) Nature Medicine 3:374-376].
  • composition includes a protein that exists in different nascent and mature forms
  • the mature form of the protein is preferably used.
  • NspA is included, (WO96/29412; see also Martin et al. (1997) J. Exp. Med Xj_5 1 173-1183) the mature form of the protein lacking the signal peptide is preferably used.
  • composition includes a polysaccharide antigen
  • the polysaccharide is preferably conjugated to a carrier protein.
  • the proteins of the invention present in the compositions preferably interact synergistically with at least one of the protective antigens in the composition.
  • the invention also provides the compositions of the invention for use as medicaments (preferably as vaccines) or as diagnostic reagents. It also provides the use of a composition according to the invention in the manufacture of: (i) a medicament for treating or preventing infection due to Neisserial bacteria; (ii) a diagnostic reagent for detecting the presence of Neisserial bacteria or of antibodies raised against Neisserial bacteria; and/or (iii) a reagent which can raise antibodies against Neisserial bacteria.
  • Said Neisserial bacteria may be any species or strain (such as N. gonorrhoeae) but are preferably N. meningitidis, especially serogroup B.
  • the invention also provides a method of treating a patient, comprising administering to the patient a therapeutically effective amount of nucleic acid, protein, and/or antibody according to the invention.
  • the method is preferably immunisation.
  • Vaccines according to the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i.e. to treat disease after infection).
  • the invention provides various processes.
  • a process for producing proteins of the invention comprising the step of culturing a host cell according to the invention under conditions which induce protein expression.
  • a process for producing protein or nucleic acid of the invention wherein the protein or nucleic acid is synthesised in part or in whole using chemical means.
  • a process for detecting polynucleotides of the invention comprising the steps of: (a) contacting a nucleic probe according to the invention with a biological sample under hybridizing conditions to form duplexes; and (b) detecting said duplexes.
  • a process for detecting proteins of the invention comprising the steps of: (a) contacting an antibody according to the invention with a biological sample under conditions suitable for the formation of an antibody-antigen complexes; and (b) detecting said complexes.
  • a composition containing X is "substantially free of Y when at least 85% by weight of the total X+Y in the composition is X.
  • X comprises at least about 90% by weight of the total of X+Y in the composition, more preferably at least about 95% or even 99% by weight.
  • composition comprising X may consist exclusively of X or may include something additional to X, such as X+Y.
  • heterologous refers to two biological components that are not found together in nature.
  • the components may be host cells, genes, or regulatory regions, such as promoters.
  • heterologous components are not found together in nature, they can function together, as when a promoter heterologous to a gene is operably linked to the gene
  • a Neisserial sequence is heterologous to a mouse host cell.
  • a further examples would be two epitopes from the same or different proteins which have been assembled in a single protein in an arrangement not found in nature.
  • An "origin of replication” is a polynucleotide sequence that initiates and regulates replication of polynucleotides, such as an expression vector.
  • the origin of replication behaves as an autonomous unit of polynucleotide replication within a cell, capable of replication under its own control.
  • An origin of replication may be needed for a vector to replicate in a particular host cell. With certain origins of replication, an expression vector can be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells.
  • a "mutant" sequence is defined as DNA, RNA or amino acid sequence differing from but having sequence identity with the native or disclosed sequence.
  • the degree of sequence identity between the native or disclosed sequence and the mutant sequence is preferably greater than 50% (e g. 60%, 70%, 80%, 90%, 95%, 99% or more, calculated using the Smith-Waterman algorithm as descnbed above)
  • an "allelic variant" of a nucleic acid molecule, or region, for which nucleic acid sequence is provided herein is a nucleic acid molecule, or region, that occurs essentially at the same locus in the genome of another or second isolate, and that, due to natural variation caused by, for example, mutation or recombination, has a similar but not identical nucleic acid sequence.
  • allelic variant typically encodes a protein having similar activity to that of the protein encoded by the gene to which it is being compared.
  • allelic variant can also comprise an alteration in the 5' or 3' untranslated regions of the gene, such as in regulatory control regions (e.g. see US patent 5,753,235).
  • Neisse ⁇ al nucleotide sequences can be expressed in a variety of different expression systems; for example those used with mammalian cells, baculov_ruses, plants, bacteria, and yeast. l Mammalian Systems
  • a mammalian promoter is any DNA sequence capable of binding mammalian RNA polymerase and initiating the downstream (3') transcription of a coding sequence (e.g. structural gene) into mRNA.
  • a promoter will have a transcription initiating region, which is usually placed proximal to the 5' end of the coding sequence, and a TATA box, usually located 25-30 base pairs (bp) upstream of the transcription initiation site. The TATA box is thought to direct RNA polymerase II to begin RNA synthesis at the correct site.
  • a mammalian promoter will also contain an upstream promoter element, usually located within 100 to 200 bp upstream of the TATA box.
  • An upstream promoter element determines the rate at which transcription is initiated and can act in either orientation [Sambrook et al. (1989) "Expression of Cloned Genes in Mammalian Cells.” In Molecular Cloning: A Laboratory Manual, 2nd ed.].
  • Mammalian viral genes are often highly expressed and have a broad host range; therefore sequences encoding mammalian viral genes provide particularly useful promoter sequences. Examples include the SV40 early promoter, mouse mammary tumor virus LTR promoter, adenovirus major late promoter (Ad MLP), and he ⁇ es simplex virus promoter. In addition, sequences derived from non- viral genes, such as the murine metallotheionein gene, also provide useful promoter sequences. Expression may be either constitutive or regulated (inducible), depending on the promoter can be induced with glucocorticoid in hormone-responsive cells.
  • Enhancer is a regulatory DNA sequence that can stimulate transcription up to 1000-fold when linked to homologous or heterologous promoters, with synthesis beginning at the normal RNA start site. Enhancers are also active when they are placed upstream or downstream from the transcription initiation site, in either normal or flipped orientation, or at a distance of more than 1000 nucleotides from the promoter [Maniatis et al. (1987) Science 236:1237; Alberts et al. (1989) Molecular Biology of the Cell, 2nd ed.]. Enhancer elements derived from viruses may be particularly useful, because they usually have a broader host range.
  • Examples include the SV40 early gene enhancer [Dijkema et al (1985) EMBO J. 4:761] and the enhancer/promoters derived from the long terminal repeat (LTR) of the Rous Sarcoma Virus [Gorman et al. (1982b) Proc. Natl. Acad. Sci. 79:6777] and from human cytomegalovirus [Boshart et al. (1985) Cell 47:521]. Additionally, some enhancers are regulatable and become active only in the presence of an inducer, such as a hormone or metal ion [Sassone-Corsi and Borelli (1986) Trends Genet. 2:2X5; Maniatis et al.
  • an inducer such as a hormone or metal ion
  • a DNA molecule may be expressed intracellularly in mammalian cells.
  • a promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus of the recombinant protein will always be a methionine, which is encoded by the ATG start codon. If desired, the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide.
  • foreign proteins can also be secreted from the cell into the growth media by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provides for secretion of the foreign protein in mammalian cells.
  • the leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell.
  • the adenovirus triparite leader is an example of a leader sequence that provides for secretion of a foreign protein in mammalian cells.
  • transcription termination and polyadenylation sequences recognized by mammalian cells are regulatory regions located 3' to the translation stop codon and thus, together with the promoter elements, flank the coding sequence.
  • the 3' terminus of the mature mRNA is formed by site- specific post-transcriptional cleavage and polyadenylation [Birnstiel et al. (1985) Cell 41:349; Proudfoot and Whitelaw (1988) "Termination and 3' end processing of eukaryotic RNA. In Transcription and splicing (ed. B.D. Hames and D.M. Glover); Proudfoot (1989) Trends Biochem. Sci. 74:105].
  • transcription terminater/polyadenylation signals include those derived from SV40 [Sambrook et al (1989) "Expression of cloned genes in cultured mammalian cells.” In Molecular Cloning: A Laboratory Manual].
  • the above described components comprising a promoter, polyadenylation signal, and transcription termination sequence are put together into expression constructs.
  • Enhancers, introns with functional splice donor and acceptor sites, and leader sequences may also be included in an expression construct, if desired.
  • Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g. plasmids) capable of stable maintenance in a host, such as mammalian cells or bacteria.
  • Mammalian replication systems include those derived from animal viruses, which require trans-acting factors to replicate.
  • plasmids containing the replication systems of papova viruses such as SV40 [Gluzman (1981) Cell 23:175] or polyomavirus
  • SV40 Gluzman (1981) Cell 23:175
  • polyomavirus replicate to extremely high copy number in the presence of the appropriate viral T antigen.
  • mammalian replicons include those derived from bovine papillomavirus and Epstein-Barr virus.
  • the replicon may have two replicaton systems, thus allowing it to be maintained, for example, in mammalian cells for expression and in a prokaryotic host for cloning and amplification.
  • mammalian-bacteria shuttle vectors include pMT2 [Kaufman et al. (1989) Mol. Cell. Biol. 9:946] and pHEBO [Shimizu et al.
  • the transformation procedure used depends upon the host to be transformed.
  • Methods for introduction of heterologous polynucleotides into mammalian cells include dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
  • Mammalian cell lines available as hosts for expression are known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to, Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g. Hep G2), and a number of other cell lines.
  • ATCC American Type Culture Collection
  • CHO Chinese hamster ovary
  • HeLa cells HeLa cells
  • BHK baby hamster kidney cells
  • COS monkey kidney cells
  • human hepatocellular carcinoma cells e.g. Hep G2
  • the polynucleotide encoding the protein can also be inserted into a suitable insect expression vector, and is operably linked to the control elements within that vector.
  • Vector construction employs techniques w hich are known in the art.
  • the components of the expression system include a transfer vector, usually a bacterial plasm id, 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 hom ologous to the baculovirus-specific fragment in the transfer vector (this allows for the hom ologous recom bination of the heterologous gene in to the baculovirus genom e); and appropriate insect host cells and growth m edia.
  • the vector and the wild type viral genome are transfected into an insect host cell where the vector and viral genome are allowed to recombine.
  • the packaged recombinant virus is expressed and recombinant plaques are identified and purified.
  • Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Invitrogen, San Diego CA ("MaxBac” kit). These techniques are generally known to those skilled in the art and fully described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987) (hereinafter "Summers and Smith”).
  • the above described components comprising a prom oter, leader (if desired), coding sequence of interest, and transcription term ination sequence, are usually assembled into an interm ediate transplacement construct (transfer vector).
  • This construct m ay contain a single gene and operably linked regulatory elements; multiple genes, each with its owned set of operably linked regulatory elements; or multiple genes, regulated by the same set of regulatory elements.
  • Intermediate transplacem ent constructs are often m aintained in a replicon, such as an extrachromosomal element (e.g.
  • plasm ids capable of stable m aintenance in a host, such as a bacterium .
  • the replicon w ill have a replication system , thus allowing it to be m aintained in a suitable host for cloning and amplification.
  • 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) 77:31.
  • the plasmid usually also contains the polyhedrin polyadenylation signal (Miller et al. (1988) Ann. Rev. Microbiol, 42:177) and a prokaryotic ampicillin-resistance (amp) gene and origin of replication for selection and propagation in E. 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 usually includes an RNA polymerase binding site and a transcription initiation site.
  • a baculovirus transfer vector may also have a second domain called an enhancer, which, if present, is usually distal to the structural gene.
  • Expression may be either regulated or constitutive. Structural genes, abundantly transcribed at late times in a viral infection cycle, provide particularly useful promoter sequences. Examples include sequences derived from the gene encoding the viral polyhedron protein, Friesen et al., (1986) "The Regulation of Baculovirus Gene Expression," in: The Molecular Biology of Baculoviruses (ed. Walter Doerfler); EPO Publ. Nos.
  • DNA encoding suitable signal sequences can be derived from genes for secreted insect or baculovirus proteins, such as the baculovirus polyhedrin gene (Carbonell et al. (1988) Gene, 73:409).
  • the signals for mammalian cell posttranslational modifications (such as signal peptide cleavage, proteolytic cleavage, and phosphorylation) appear to be recognized by insect cells, and the signals required for secretion and nuclear accumulation also appear to be conserved between the invertebrate cells and vertebrate cells, leaders of non-insect origin, such as those derived from genes encoding human ⁇ -interferon, Maeda et al., (1985), Nature 315:592; human gastrin-releasing peptide, Lebacq-Verheyden et al., (1988), Molec. Cell. Biol. S:3129; human IL-2, Smith et al., (1985) Proc.
  • mammalian cell posttranslational modifications such as signal peptide cleavage, proteolytic cleavage, and phosphorylation
  • a recombinant polypeptide or polyprotein may be expressed intracellularly or, if it is expressed with the proper regulatory sequences, it can be secreted.
  • Good intracellular expression of nonfused foreign proteins usually requires heterologous genes that ideally have a short leader sequence containing suitable translation initiation signals preceding an ATG start signal. If desired, methionine at the N-terminus may be cleaved from the mature protein by in vitro incubation with cyanogen bromide.
  • recombinant polyproteins or proteins which are not naturally secreted can be secreted from the insect cell by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provides for secretion of the foreign protein in insects.
  • the leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the translocation of the protein into the endoplasmic reticulum.
  • an insect cell host is co-transformed with the heterologous DNA of the transfer vector and the genomic DNA of wild type baculovirus — usually by co-transfection.
  • the promoter and transcription termination sequence of the construct will usually comprise a 2-5kb section of the baculovirus genome.
  • the insertion can be into a gene such as the polyhedrin gene, by homologous double crossover recombination; insertion can also be into a restriction enzyme site engineered into the desired baculovirus gene. Miller et al., (1989), Bioessays 4:91.
  • the DNA sequence, when cloned in place of the polyhedrin gene in the expression vector, is flanked both 5' and 3' by polyhedrin-specific sequences and is positioned downstream of the polyhedrin promoter.
  • the newly formed baculovirus expression vector is subsequently packaged into an infectious recombinant baculovirus. Homologous recombination occurs at low frequency (between about 1% and about 5%); thus, the majority of the virus produced after cotransfection is still wild-type virus. Therefore, a method is necessary to identify recombinant viruses.
  • An advantage of the expression system is a visual screen allowing recombinant viruses to be distinguished.
  • the polyhedrin protein which is produced by the native virus, is produced at very high levels in the nuclei of infected cells at late times after viral infection. Accumulated polyhedrin protein forms occlusion bodies that also contain embedded particles.
  • occlusion bodies up to 15 ⁇ m in size, are highly retractile, giving them a bright shiny appearance that is readily visualized under the light microscope.
  • Cells infected with recombinant viruses lack occlusion bodies.
  • the transfection supernatant is plaqued onto a monolayer of insect cells by techniques known to those skilled in the art. Namely, the plaques are screened under the light microscope for the presence (indicative of wild-type virus) or absence (indicative of recombinant virus) of occlusion bodies.
  • Recombinant baculovirus expression vectors have been developed for infection into several insect cells.
  • recombinant baculoviruses have been developed for, inter alia: Aedes aegypti , Autographa californica, Bombyx mori, Drosophila melanogaster, Spodoptera frugiperda, and Trichoplusia ni (WO 89/046699; Carbonell et al., (1985) J. Virol. 5(5:153; Wright (1986) Nature 327:718; Smith et al., (1983) Mol. Cell. Biol. 3:2156; and see generally, Fraser, et al. (1989) In Vitro Cell. Dev. Biol. 25:225).
  • Cells and cell culture media are commercially available for both direct and fusion expression of heterologous polypeptides in a baculovirus/expression system; cell culture technology is generally known to those skilled in the art. See, e.g. Summers and Smith supra.
  • the modified insect cells may then be grown in an appropriate nutrient medium, which allows for stable maintenance of the plasmid(s) present in the modified insect host. Where the expression product gene is under inducible control, the host may be grown to high density, and expression induced. Alternatively, where expression is constitutive, the product will be continuously expressed into the medium and the nutrient medium must be continuously circulated, while removing the product of interest and augmenting depleted nutrients.
  • the product may be purified by such techniques as chromatography, e.g.
  • the product may be further purified, as required, so as to remove substantially any insect proteins which are also secreted in the medium or result from lysis of insect cells, so as to provide a product which is at least substantially free of host debris, e.g. proteins, lipids and polysaccharides.
  • recombinant host cells derived from the transformants are incubated under conditions which allow expression of the recombinant protein encoding sequence. These conditions will vary, dependent upon the host cell selected. However, the conditions are readily ascertainable to those of ordinary skill in the art, based upon what is known in the art. iii. Plant Systems
  • a desired polynucleotide sequence is inserted into an expression cassette comprising genetic regulatory elements designed for operation in plants.
  • the expression cassette is inserted into a desired expression vector with companion sequences upstream and downstream from the expression cassette suitable for expression in a plant host.
  • the companion sequences will be of plasmid or viral origin and provide necessary characteristics to the vector to permit the vectors to move DNA from an original cloning host, such as bacteria, to the desired plant host.
  • the basic bacterial/plant vector construct will preferably provide a broad host range prokaryote replication origin; a prokaryote selectable marker; and, for Agrobacterium transformations, T DNA sequences for Agrobacterium-mediated transfer to plant chromosomes.
  • the construct will preferably also have a selectable marker gene suitable for determining if a plant cell has been transformed.
  • a selectable marker gene suitable for determining if a plant cell has been transformed is found in Wilmink and Dons, 1993, Plant Mol. Biol. Reptr, 11 (2): 165-185.
  • Sequences suitable for permitting integration of the heterologous sequence into the plant genome are also recommended. These might include transposon sequences and the like for homologous recombination as well as Ti sequences which permit random insertion of a heterologous expression cassette into a plant genome. Suitable prokaryote selectable markers include resistance toward antibiotics such as ampicillin or tetracycline. Other DNA sequences encoding additional functions may also be present in the vector, as is known in the art.
  • the nucleic acid molecules of the subject invention may be included into an expression cassette for expression of the protein(s) of interest.
  • the recombinant expression cassette will contain in addition to the heterologous protein encoding sequence the following elements, a promoter region, plant 5' untranslated sequences, initiation codon depending upon whether or not the structural gene comes equipped with one, and a transcription and translation termination sequence.
  • Unique restriction enzyme sites at the 5' and 3' ends of the cassette allow for easy insertion into a pre-existing vector.
  • a heterologous coding sequence may be for any protein relating to the present invention.
  • the sequence encoding the protein of interest will encode a signal peptide which allows processing and translocation of the protein, as appropriate, and will usually lack any sequence which might result in the binding of the desired protein of the invention to a membrane. Since, for the most part, the transcriptional initiation region will be for a gene which is expressed and translocated during germination, by employing the signal peptide which provides for translocation, one may also provide for translocation of the protein of interest. In this way, the protein(s) of interest will be translocated from the cells in which they are expressed and may be efficiently harvested.
  • the ultimate expression of the desired gene product will be in a eucaryotic cell it is desirable to determine whether any portion of the cloned gene contains sequences which will be processed out as introns by the host's splicosome machinery. If so, site-directed mutagenesis of the "intron" region may be conducted to prevent losing a portion of the genetic message as a false intron code, Reed and Maniatis, Cell 41 :95-105, 1985.
  • the vector can be microinjected directly into plant cells by use of micropipettes to mechanically transfer the recombinant DNA. Crossway, Mol. Gen. Genet, 202:179-185, 1985.
  • the genetic material may also be transferred into the plant cell by using polyethylene glycol, Krens, et al., Nature, 296, 72-74, 1982.
  • Another method of introduction of nucleic acid segments is high velocity ballistic penetration by small particles with the nucleic acid either within the matrix of small beads or particles, or on the surface, Klein, et al., Nature, 327, 70-73, 1987 and Knudsen and Muller, 1991, Planta, 185:330-336 teaching particle bombardment of barley endosperm to create transgenic barley.
  • Yet another method of introduction would be fusion of protoplasts with other entities, either minicells, cells, lysosomes or other fusible lipid-surfaced bodies, Fraley, et al., Proc. Natl. Acad. Sci. USA, 79, 1859-1863, 1982.
  • the vector may also be introduced into the plant cells by electroporation. (Fromm et al., Proc. Natl Acad. Sci. USA 82:5824, 1985).
  • plant protoplasts are electroporated in the presence of plasmids containing the gene construct. Electrical impulses of high field strength reversibly permeabilize biomembranes allowing the introduction of the plasmids. Electroporated plant protoplasts reform the cell wall, divide, and form plant callus.
  • All plants from which protoplasts can be isolated and cultured to give whole regenerated plants can be transformed by the present invention so that whole plants are recovered which contain the transferred gene. It is known that practically all plants can be regenerated from cultured cells or tissues, including but not limited to all major species of sugarcane, sugar beet, cotton, fruit and other trees, legumes and vegetables.
  • Some suitable plants include, for example, species from the genera Fragaria, Lotus, Medicago, Onobrychis, Trifolium, Trigonella, Vigna, Citrus, Linum, Geranium, Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Datura, Hyoscyamus, Lycopersion, Nicotiana, Solanum, Petunia, Digitalis, Majorana, Cichorium, Helianthus, Lactuca, Bromus, Asparagus, Antirrhinum, Hererocallis, Nemesia, Pelargonium, Panicum, Pennisetum, Ranunculus, Senecio, Salpiglossis, Cucumis, Browaalia, Glycine, Lolium, Zea, Triticum, Sorghum, and Datura.
  • Means for regeneration vary from species to species of plants, but generally a suspension of transformed protoplasts containing copies of the heterologous gene is first provided. Callus tissue is formed and shoots may be induced from callus and subsequently rooted. Alternatively, embryo formation can be induced from the protoplast suspension. These embryos germinate as natural embryos to form plants.
  • the culture media will generally contain various amino acids and hormones, such as auxin and cytokinins. It is also advantageous to add glutamic acid and proline to the medium, especially for such species as corn and alfalfa. Shoots and roots normally develop simultaneously. Efficient regeneration will depend on the medium, on the genotype, and on the history of the culture. If these three variables are controlled, then regeneration is fully reproducible and repeatable.
  • the desired protein of the invention may be excreted or alternatively, the protein may be extracted from the whole plant. W ere the desired protein of the invention is secreted into the medium, it may be collected. Alternatively, the embryos and embryoless-half seeds or other plant tissue may be mechanically disrupted to release any secreted protein between cells and tissues. The mixture may be suspended in a buffer solution to retrieve soluble proteins. Conventional protein isolation and purification methods will be then used to purify the recombinant protein. Parameters of time, temperature pH, oxygen, and volumes will be adjusted through routine methods to optimize expression and recovery of heterologous protein. iv. Bacterial Systems
  • a bacterial promoter is any DNA sequence capable of binding bacterial RNA polymerase and initiating the downstream (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 usually includes an RNA polymerase binding site and a transcription initiation site.
  • a bacterial promoter may also have a second domain called an operator, that may overlap an adjacent RNA polymerase binding site at which RNA synthesis begins. The operator permits negative regulated (inducible) transcription, as a gene repressor protein may bind the operator and thereby inhibit transcription of a specific gene.
  • Constitutive expression may occur in the absence of negative regulatory elements, such as the operator.
  • positive regulation may be achieved by a gene activator protein binding sequence, which, if present is usually proximal (5') to the RNA polymerase binding sequence.
  • An example of a gene activator protein is the catabolite activator protein (CAP), which helps initiate transcription of the lac operon in Escherichia coli (E. coli) [Raibaud et al. (1984) Annu. Rev. Genet. 75:173].
  • Regulated expression may therefore be either positive or negative, thereby either enhancing or reducing transcription.
  • Sequences encoding metabolic pathway enzymes provide particularly useful promoter sequences. Examples include promoter sequences derived from sugar metabolizing enzymes, such as galactose, lactose (lac) [Chang et al. (1977) Nature 798:1056], and maltose. Additional examples include promoter sequences derived from biosynthetic enzymes such as tryptophan (trp) [Goeddel et al. (1980) Nuc. Acids Res. 8:4057; Yelverton et al. (1981) Nucl. Acids Res. 9:731 ; US patent 4,738,921; EP-A-0036776 and EP-A-0121775].
  • the tac promoter is a hybrid trp-lac promoter comprised of both trp promoter and lac operon sequences that is regulated by the lac repressor [Amann et al. (1983) Gene 25:167; de Boer et al. (1983) Proc. Natl. Acad. Sci. 80:21].
  • a bacterial promoter can include naturally occurring promoters of non-bacterial origin that have the ability to bind bacterial RNA polymerase and initiate transcription.
  • a naturally occurring promoter of non-bacterial origin can also be coupled with a compatible RNA polymerase to produce high levels of expression of some genes in prokaryotes.
  • bacteriophage T7 RNA polymerase/promoter system is an example of a coupled promoter system [Studier et al. (1986) J. Mol. Biol. 789:1 13; Tabor et al. (1985) Proc Natl. Acad. Sci. 82: 1074].
  • a hybrid promoter can also be comprised of a bacteriophage promoter and an E. coli operator region (EPO- A-0 267 851).
  • an efficient ribosome binding site is also useful for the expression of foreign genes in prokaryotes.
  • the ribosome binding site is called the Shine-Dalgarno (SD) sequence and includes an initiation codon (ATG) and a sequence 3-9 nucleotides in length located 3-1 1 nucleotides upstream of the initiation codon [Shine et al. (1975) Nature 254:34].
  • SD sequence is thought to promote binding of mRNA to the ribosome by the pairing of bases between the SD sequence and the 3' and of E. coli 16S rRNA [Steitz et al.
  • a DNA molecule may be expressed intracellularly.
  • a promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus will always be a methionine, which is encoded by the ATG start codon. If desired, methionine at the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide or by either in vivo on in vitro incubation with a bacterial methionine N-terminal peptidase (EPO-A-0219 237).
  • Fusion proteins provide an alternative to direct expression. Usually, a DNA sequence encoding the
  • N-terminal portion of an endogenous bacterial protein, or other stable protein is fused to the 5' end of heterologous coding sequences.
  • this construct will provide a fusion of the two amino acid sequences.
  • the bacteriophage lambda cell gene can be linked at the 5' terminus of a foreign gene and expressed in bacteria.
  • the resulting fusion protein preferably retains a site for a processing enzyme (factor Xa) to cleave the bacteriophage protein from the foreign gene
  • Fusion proteins can also be made with sequences from the lacZ [Jia et al (1987) Gene 60:197], trpE [Allen et al. (1987) J. Biotechnol. 5:93; Makoff et al.
  • the DNA sequence at the junction of the two amino acid sequences may or may not encode a cleavable site.
  • a ubiquitin fusion protein is made with the ubiquitin region that preferably retains a site for a processing enzyme (e.g. ubiquitin specific processing-protease) to cleave the ubiquitin from the foreign protein.
  • a processing enzyme e.g. ubiquitin specific processing-protease
  • foreign proteins can also be secreted from the cell by creating chimeric DNA molecules that encode a fusion protein comprised of a signal peptide sequence fragment that provides for secretion of the foreign protein in bacteria [US patent 4,336,336].
  • the signal sequence fragment usually encodes a signal peptide comprised of hydrophobic am ino acids w hich direct the secretion of the protein from the cell.
  • the protein is either secreted into the growth media (gram-positive bacteria) or into the periplasmic space, located between the inner and outer membrane of the cell (gram-negative bacteria).
  • w hich can be cleaved either in vivo or in vitro encoded between the signal peptide fragment and the foreign gene.
  • DNA encoding suitable signal sequences can be derived from genes for secreted bacterial proteins, such as the E. coli outer membrane protein gene (ompA) [Masui et al (1983), in: Experimental Manipulation of Gene Expression; Ghrayeb et al. (1984) EMBO J. 3:2437] and the E. coli alkaline phosphatase signal sequence (phoA) [Oka et al. (1985) Proc. Natl. Acad. Sci. 82:72X2].
  • the signal sequence of the alpha-amylase gene from various Bacillus strains can be used to secrete heterologous proteins from B. subtilis [Palva et al. (1982) Proc. Natl Acad. Sci. USA 79:5582; EP-A-0 244 042].
  • transcription termination sequences recognized by bacteria are regulatory regions located 3' to the translation stop codon, and thus together with the promoter flank the coding sequence. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Transcription termination sequences frequently include DNA sequences of about 50 nucleotides capable of forming stem loop structures that aid in terminating transcription. Examples include transcription termination sequences derived from genes with strong promoters, such as the trp gene in E. coli as well as other biosynthetic genes. Usually, the above described components, comprising a promoter, signal sequence (if desired), coding sequence of interest, and transcription termination sequence, are put together into expression constructs.
  • Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g. plasmids) capable of stable maintenance in a host, such as bacteria.
  • the replicon will have a replication system, thus allowing it to be maintained in a prokaryotic host either for expression or for cloning and amplification.
  • a replicon may be either a high or low copy number plasmid.
  • a high copy number plasmid will generally have a copy number ranging from about 5 to about 200, and usually about 10 to about 150.
  • a host containing a high copy number plasmid will preferably contain at least about 10, and more preferably at least about 20 plasmids. Either a high or low copy number vector may be selected, depending upon the effect of the vector and the foreign protein on the host.
  • the expression constructs can be integrated into the bacterial genome with an integrating vector.
  • Integrating vectors usually contain at least one sequence homologous to the bacterial chromosome that allows the vector to integrate. Integrations appear to result from recombinations between homologous DNA in the vector and the bacterial chromosome.
  • integrating vectors constructed with DNA from various Bacillus strains integrate into the Bacillus chromosome (EP-A- 0 127 328). Integrating vectors may also be comprised of bacteriophage or transposon sequences.
  • extrachromosomal and integrating expression constructs may contain selectable markers to allow for the selection of bacterial strains that have been transformed.
  • Selectable markers can be expressed in the bacterial host and may include genes which render bacteria resistant to drugs such as ampicillin, chloramphenicol, erythromycin, kanamycin (neomycin), and tetracycline [Davies et al. (1978) Annu. Rev. Microbiol. 32:469].
  • Selectable markers may also include biosynthetic genes, such as those in the histidine, tryptophan, and leucine biosynthetic pathways.
  • Transformation vectors are usually comprised of a selectable market that is either maintained in a replicon or developed into an integrating vector, as described above.
  • Expression and transformation vectors have been developed for transformation into many bacteria.
  • expression vectors have been developed for, inter alia, the following bacteria: Bacillus subtilis [Palva et al (1982) Proc. Natl. Acad. Sci. USA 79:5582; EP-A-0 036 259 and EP-A-0 063 953; WO 84/04541], Escherichia coli [Shimatake et al. (1981) Nature 292:128; A ann et al. (1985) Gene 40:183; Studier et al. (1986) J. Mol. Biol.
  • Methods of introducing exogenous DNA into bacterial hosts are well-known in the art, and usually include either the transformation of bacteria treated with CaCl 2 or other agents, such as divalent cations and DMSO. DNA can also be introduced into bacterial cells by electroporation.
  • Transformation procedures usually vary with the bacterial species to be transformed. See e.g.
  • a yeast promoter is any DNA sequence capable of binding yeast RNA polymerase and initiating the downstream (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 usually includes an RNA polymerase binding site (the "TATA Box") and a transcription initiation site.
  • a yeast promoter may also have a second domain called an upstream activator sequence (UAS), which, if present, is usually distal to the structural gene.
  • the UAS permits regulated (inducible) expression. Constitutive expression occurs in the absence of a UAS. Regulated expression may be either positive or negative, thereby either enhancing or reducing transcription.
  • Yeast is a fermenting organism with an active metabolic pathway, therefore sequences encoding enzymes in the metabolic pathway provide particularly useful promoter sequences. Examples include alcohol dehydrogenase (ADH) (EP-A-0 284 044), enolase, glucokinase, glucose-6- phosphate isomerase, glyceraldehyde-3-phosphate-dehydrogenase (GAP or GAPDH), hexokinase, phosphofructokinase, 3-phosphoglycerate mutase, and pyruvate kinase (PyK) (EPO-A-0 329 203).
  • the yeast PH05 gene encoding acid phosphatase, also provides useful promoter sequences [Myanohara et al. ( 1983) Proc. Natl. Acad. Sci. USA 80: 1 ] .
  • synthetic promoters which do not occur in nature also function as yeast promoters.
  • UAS sequences of one yeast promoter may be joined with the transcription activation region of another yeast promoter, creating a synthetic hybrid promoter.
  • hybrid promoters include the ADH regulatory sequence linked to the GAP transcription activation region (US Patent Nos. 4,876,197 and 4,880,734).
  • Other examples of hybrid promoters include promoters which consist of the regulatory sequences of either the ADH2, GAL4, GAL10, OR PH05 genes, combined with the transcriptional activation region of a glycolytic enzyme gene such as GAP or PyK (EP-A-0 164 556).
  • a yeast promoter can include naturally occurring promoters of non-yeast origin that have the ability to bind yeast RNA polymerase and initiate transcription. Examples of such promoters include, ter alia, [Cohen et al. (1980) Proc. Natl. Acad. Sci. USA 77: 1078; Henikoff et al. (1981) Nature 283:835; Hollenberg et al. (1981) Curr. Topics Microbiol. Immunol. 96:1 19; Hollenberg et al. (1979) "The Expression of Bacterial Antibiotic Resistance Genes in the Yeast Saccharomyces cerevisiae," in: Plasmids of Medical, Environmental and Commercial Importance (eds. K.N. Timmis and A. Puhler); Mercerau-Puigalon et al. (1980) Gene 77:163; Panthier et al. (1980) Curr. Genet. 2: 109;].
  • a DNA molecule may be expressed intracellularly in yeast.
  • a promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus of the recombinant protein will always be a methionine, which is encoded by the ATG start codon. If desired, methionine at the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide.
  • Fusion proteins provide an alternative for yeast expression systems, as well as in mammalian, baculovirus, and bacterial expression systems.
  • a DNA sequence encoding the N-terminal portion of an endogenous yeast protein, or other stable protein is fused to the 5' end of heterologous coding sequences.
  • this construct will provide a fusion of the two amino acid sequences.
  • the yeast or human superoxide dismutase (SOD) gene can be linked at the 5' terminus of a foreign gene and expressed in yeast.
  • the DNA sequence at the junction of the two amino acid sequences may or may not encode a cleavable site. See e.g. EP-A-0 196 056.
  • a ubiquitin fusion protein is made with the ubiquitin region that preferably retains a site for a processing enzyme (e.g. ubiquitin-specific processing protease) to cleave the ubiquitin from the foreign protein.
  • a processing enzyme e.g. ubiquitin-specific processing protease
  • native foreign protein can be isolated (e.g. WO88/024066).
  • foreign proteins can also be secreted from the cell into the growth media by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provide for secretion in yeast of the foreign protein.
  • a leader sequence fragment that provide for secretion in yeast of the foreign protein.
  • the leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell.
  • DNA encoding suitable signal sequences can be derived from genes for secreted yeast proteins, such as the yeast invertase gene (EP-A-0 012 873; JPO. 62,096,086) and the A-factor gene (US patent 4,588,684).
  • leaders of non-yeast origin such as an interferon leader, exist that also provide for secretion in yeast (EP-A-0060057).
  • a preferred class of secretion leaders are those that employ a fragment of the yeast alpha-factor gene, which contains both a "pre" signal sequence, and a "pro” region.
  • the types of alpha-factor fragments that can be employed include the full-length pre-pro alpha factor leader (about 83 amino acid residues) as well as truncated alpha-factor leaders (usually about 25 to about 50 amino acid residues) (US Patents 4,546,083 and 4,870,008; EP-A-0 324 274).
  • Additional leaders employing an alpha-factor leader fragment that provides for secretion include hybrid alpha-factor leaders made with a presequence of a first yeast, but a pro-region from a second yeast alphafactor. (e.g. see WO 89/02463.)
  • transcription termination sequences recognized by yeast are regulatory regions located 3' to the translation stop codon, and thus together with the promoter flank the coding sequence. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Examples of transcription terminator sequence and other yeast-recognized termination sequences, such as those coding for glycolytic enzymes.
  • Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g. plasmids) capable of stable maintenance in a host, such as yeast or bacteria.
  • the replicon may have two replication systems, thus allowing it to be maintained, for example, in yeast for expression and in a prokaryotic host for cloning and amplification.
  • yeast- bacteria shuttle vectors include YEp24 [Botstein et al. (1979) Gene 8:17-24], pCl/1 [Brake et al. (1984) Proc. Natl Acad.
  • a replicon may be either a high or low copy number plasmid.
  • a high copy number plasmid will generally have a copy number ranging from about 5 to about 200, and usually about 10 to about 150.
  • a host containing a high copy number plasmid will preferably have at least about 10, and more preferably at least about 20. Enter a high or low copy number vector may be selected, depending upon the effect of the vector and the foreign protein on the host. See e.g. Brake et al, supra.
  • the expression constructs can be integrated into the yeast genome with an integrating vector.
  • Integrating vectors usually contain at least one sequence homologous to a yeast chromosome that allows the vector to integrate, and preferably contain two homologous sequences flanking the expression construct. Integrations appear to result from recombinations between homologous DNA in the vector and the yeast chromosome [Orr- Weaver et al. (1983) Methods in Enzymol. 707:228- 245].
  • An integrating vector may be directed to a specific locus in yeast by selecting the appropriate homologous sequence for inclusion in the vector. See Orr- Weaver et al, supra.
  • One or more expression construct may integrate, possibly affecting levels of recombinant protein produced [Rine et al.
  • the chromosomal sequences included in the vector can occur either as a single segment in the vector, which results in the integration of the entire vector, or two segments homologous to adjacent segments in the chromosome and flanking the expression construct in the vector, which can result in the stable integration of only the expression construct.
  • extrachromosomal and integrating expression constructs may contain selectable markers to allow for the selection of yeast strains that have been transformed.
  • Selectable markers may include biosynthetic genes that can be expressed in the yeast host, such as ADE2, H1S4, LEU2, TRP1, and ALG7, and the G418 resistance gene, which confer resistance in yeast cells to tunicamycin and G418, respectively.
  • a suitable selectable marker may also provide yeast with the ability to grow in the presence of toxic compounds, such as metal.
  • the presence of CUP1 allows yeast to grow in the presence of copper ions [Butt et al. (1987) Microbiol, Rev. 57:351].
  • Transformation vectors are usually comprised of a selectable marker that is either maintained in a replicon or developed into an integrating vector, as described above.
  • Expression and transformation vectors have been developed for transformation into many yeasts.
  • expression vectors have been developed for, ter alia, the following yeasts:Candida albicans [Kurtz, et al. (1986) Mol Cell. Biol. 6: 142], Candida maltosa [Kunze, et al. (1985) J. Basic Microbiol. 25:141]. Hansenula polymo ⁇ ha [Gleeson, et al. (1986) J. Gen. Microbiol. 732:3459; Roggenkamp et al (1986) Mol. Gen. Genet. 202:302], Kluyveromyces fragilis [Das, et al.
  • Methods of introducing exogenous DNA into yeast hosts are well-known in the art, and usually include either the transformation of spheroplasts or of intact yeast cells treated with alkali cations. Transformation procedures usually vary with the yeast species to be transformed. See e.g. [Kurtz et al. (1986) Mol. Cell. Biol. 6:142; Kunze et al. (1985) J. Basic Microbiol 25:141; Candida]; [Gleeson et al. (1986) J. Gen. Microbiol. 732:3459; Roggenkamp et al. (1986) Mol. Gen. Genet. 202:302; Hansenula]; [Das et al. (1984) J. Bacteriol.
  • antibody refers to a polypeptide or group of polypeptides composed of at least one antibody combining site.
  • An “antibody combining site” is the three-dimensional binding space with an internal surface shape and charge distribution complementary to the features of an epitope of an antigen, which allows a binding of the antibody with the antigen.
  • Antibody includes, for example, vertebrate antibodies, hybrid antibodies, chimeric antibodies, humanised antibodies, altered antibodies, univalent antibodies, Fab proteins, and single domain antibodies. Antibodies against the proteins of the invention are useful for affinity chromatography, immunoassays, and distinguishing/identifying Neisserial proteins.
  • Antibodies to the proteins of the invention may be prepared by conventional methods.
  • the protein is first used to immunize a suitable animal, preferably a mouse, rat, rabbit or goat. Rabbits and goats are preferred for the preparation of polyclonal sera due to the volume of serum obtainable, and the availability of labeled anti-rabbit and anti-goat antibodies.
  • Immunization is generally performed by mixing or emulsifying the protein in saline, preferably in an adjuvant such as Freund's complete adjuvant, and injecting the mixture or emulsion parenterally (generally subcutaneously or intramuscularly). A dose of 50-200 ⁇ g injection is typically sufficient.
  • Immunization is generally boosted 2-6 weeks later with one or more injections of the protein in saline, preferably using Freund's incomplete adjuvant.
  • Polyclonal antisera is obtained by bleeding the immunized animal into a glass or plastic container, incubating the blood at 25°C for one hour, followed by incubating at 4°C for 2-18 hours. The serum is recovered by centrifugation (e.g. 1 ,000g for 10 minutes). About 20-50 ml per bleed may be obtained from rabbits.
  • Monoclonal antibodies are prepared using the standard method of Kohler & Milstein [Nature (1975) 256:495-96], or a modification thereof.
  • a mouse or rat is immunized as described above.
  • the spleen (and optionally several large lymph nodes) is removed and dissociated into single cells.
  • the spleen cells may be screened (after removal of nonspecifically adherent cells) by applying a cell suspension to a plate or well coated with the protein antigen.
  • B-cells expressing membrane-bound immunoglobulin specific for the antigen bind to the plate, and are not rinsed away with the rest of the suspension.
  • Resulting B-cells, or all dissociated spleen cells are then induced to fuse with myeloma cells to form hybridomas, and are cultured in a selective medium (e.g. hypoxanthine, aminopterin, thymidine medium, "HAT").
  • a selective medium e.g. hypoxanthine, aminopterin, thymidine medium, "HAT”
  • the resulting hybridomas are plated by limiting dilution, and are assayed for the production of antibodies which bind specifically to the immunizing antigen (and which do not bind to unrelated antigens).
  • the selected MAb-secreting hybridomas are then cultured either in vitro (e.g. in tissue culture bottles or hollow fiber reactors), or in vivo (as ascites in mice).
  • Suitable labels include fluorophores, chro ophores, radioactive atom s (particularly 32 P and 125 I), electron-dense reagents, enzym es, and ligands having specific binding partners.
  • Enzym es are typically detected by their activity. For example, horseradish peroxidase is usually detected by its ability to convert 3 ,3 ',5 ,5 '-tetram ethylbenzidine (TM B ) to a blue pigm ent, quantifiable w ith a spectrophotom eter.
  • S pecific binding partner refers to a protein capable of binding a ligand m olecule w ith high specificity, as for ex am ple in the case of an antigen and a m onoclonal antibody specific therefor.
  • Other specific binding partners include biotin and avidin or streptavidin, IgG and protein A , and the num erous receptor-ligand couples know n in the art. It should be understood that the above description is not m eant to categorize the various labels into distinct classes, as the sam e label m ay serve in several different m odes. For example, l 25 I m ay serve as a radioactive label or as an electron-den se reagent.
  • H RP m ay serve as enzym e or as antigen for a M A b. Further, one m ay com bine various labels for desired effect.
  • M A bs and avidin also require labels in the practice of this invention: thus, one m ight label a M A b w ith biotin, and detect its presence w ith avidin labeled w ith l 25 I, or w ith an anti-biotin M A b labeled w ith H RP. O ther perm utations and possibilities w ill be readily apparent to those of ordinary skill in the art, and are considered as equivalents w ithin the scope of the instant invention.
  • compositions can comprise either polypeptides, antibodies, or nucleic acid of the invention.
  • the pharmaceutical compositions will comprise a therapeutically effective amount of either polypeptides, antibodies, or polynucleotides of the claimed invention.
  • therapeutically effective amount refers to an amount of a therapeutic agent to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect.
  • the effect can be detected by, for example, chemical markers or antigen levels.
  • Therapeutic effects also include reduction in physical symptoms, such as decreased body temperature.
  • the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance. However, the effective amount for a given situation can be determined by routine experimentation and is within the judgement of the clinician.
  • an effective dose w ill be from about 0.01 m g/ kg to 50 m g/kg or 0.05 m g/kg to about 1 0 m g/kg of the D N A constructs in the individual to w hich it is adm inistered.
  • a pharm aceutical composition can also contain a pharm aceutically acceptable carrier.
  • the term "pharm aceutically acceptable carrier" refers to a carrier for adm inistration of a therapeutic agent, such as antibodies or a polypeptide, genes, and other therapeutic agents.
  • Suitable carriers m ay be large, slow ly m etabolized m acrom olecules such as proteins, polysaccharides, poly lactic acids, poly glycolic acids, polym eric am ino acids, am ino acid copolym ers, and inactive virus particles.
  • Such carriers are w ell know n to those of ordinary skill in the art.
  • Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • Pharm aceutically acceptable carriers in therapeutic compositions m ay contain liquids such as w ater, saline, glycerol and ethanol.
  • aux iliary substances such as wetting or em ulsifying agents, pH buffering substances, and the like, m ay be present in such vehicles.
  • the therapeutic com positions are prepared as injectables, either as liqu id solutions or suspensions; solid form s suitable for solution in, or suspension in, liquid vehicles prior to injection m ay also be prepared.
  • Liposomes are included w ithin the definition of a pharm aceutically acceptable carrier. Delivery Methods
  • compositions of the invention can be administered directly to the subject.
  • the subjects to be treated can be animals; in particular, human subjects can be treated.
  • Direct delivery of the compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue.
  • the compositions can also be administered into a lesion.
  • Other modes of administration include oral and pulmonary administration, suppositories, and transdermal or transcutaneous applications (e.g. see WO98/20734), needles, and gene guns or hyposprays.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • Vaccines V accines com prise im m u nising antigen(s), im m unogen(s), polypeptide(s), protein(s) or nucleic acid, usually in com bination w ith “pharm aceutically acceptable carriers," w hich include any carrier that does not itself induce the production of antibodies harm ful to the individual receiving the composition .
  • Suitable carriers are typically large, slow ly m etabolized m acrom olecu les such as proteins, polysaccharides, poly lactic acids, polyglycoiic acids, polym eric am ino acids, am ino acid copolym ers, lipid aggregates (such as oil droplets or liposom es), and inactive virus particles.
  • Such carriers are w ell know n to those of ordinary skill in the art. A dditionally, these carriers m ay function as im m unostimu lating agents ("adju vants").
  • the antigen or im m unogen m ay be conjugated to a bacterial toxoid, such as a toxoid from diphtheria, tetanus, cholera, H. pylori, etc. pathogens.
  • a bacterial toxoid such as a toxoid from diphtheria, tetanus, cholera, H. pylori, etc. pathogens.
  • Preferred adjuvants to enhance effectiveness of the composition include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc; (2) oil-in-water emulsion formulations (with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial cell wall components), such as for example (a) MF59TM (WO 90/14837; Chapter 10 in Vaccine design: the subunit and adjuvant approach, eds.
  • aluminum salts alum
  • oil-in-water emulsion formulations with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial cell wall components
  • MF59TM WO 90/14837
  • Span 85 containing various amounts of MTP-PE (see below), although not required) formulated into submicron particles using a microfluidizer such as Model HOY microfluidizer (Microfluidics, Newton, MA), (b) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic- blocked polymer L121, and thr-MDP (see below) either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RibiTM adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose di ycolate (TDM), and cell wall skeleton (MPL), trehalose di ycolate (TDM), and cell wall skeleton (MPL), trehalose di
  • interferons e.g. gamma interferon
  • M-CSF macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • other substances that act as immunostimulating agents to enhance the effectiveness of the composition.
  • Alum and MF59TM are preferred.
  • muramyl peptides include, but are not limited to, N-acetyl -muramyl -L- threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N- acetylmuramyI- -alanyl-D-isoglutaminyl-L-aIanine-2-(r-2'-dipalmitoyl-.sn-glycero-3- hydroxyphosphoryloxy)-ethylamine (MTP-PE), etc.
  • thr-MDP N-acetyl -muramyl -L- threonyl-D-isoglutamine
  • nor-MDP N-acetyl-normuramyl-L-alanyl-D-isoglutamine
  • MTP-PE N-acetylmuramyI
  • the immunogenic compositions typically will contain diluents, such as water, saline, glycerol, ethanol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • diluents such as water, saline, glycerol, ethanol, etc.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • the im m unogenic com positions are prepared as injectables, either as liquid solutions or suspensions; solid form s suitable for solution in, or suspension in, liquid vehicles prior to injection m ay also be prepared.
  • the preparation also m ay be em ulsified or encapsulated in liposom es for enhanced adjuvant effect, as discussed above under pharm aceutically acceptable carriers.
  • Immunogenic compositions used as vaccines comprise an immunologically effective amount of the antigenic or immunogenic polypeptides, as well as any other of the above-mentioned components, as needed.
  • immunologically effective amount it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention. This amount varies depending upon the health and physical condition of the individual to be treated, the taxonomic group of individual to be treated (e.g. nonhuman primate, primate, etc.), the capacity of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
  • the immunogenic compositions are conventionally administered parenterally, e.g. by injection, either subcutaneously, intramuscularly, or transdermally/transcutaneously (e.g. WO98/20734). Additional formulations suitable for other modes of administration include oral and pulmonary formulations, suppositories, and transdermal applications. Dosage treatment may be a single dose schedule or a multiple dose schedule. The vaccine may be administered in conjunction with other immunoregulatory agents.
  • DNA vaccination may be employed [e.g. Robinson & Torres (1997) Seminars in Immunology 9:271-283; Donnelly et al. (1997) Annu Rev Immunol 15:617-648; see later herein].
  • Polynucleotide and polypeptide pharmaceutical compositions may be employed [e.g. Robinson & Torres (1997) Seminars in Immunology 9:271-283; Donnelly et al. (1997) Annu Rev Immunol 15:617-648; see later herein].
  • polypeptides which include, without limitation: asioloorosomucoid (ASOR); transferrin; asialoglycoproteins; antibodies; antibody fragments; ferritin; interleukins; interferons, granulocyte, macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), macrophage colony stimulating factor (M-CSF), stem cell factor and erythropoietin.
  • Viral antigens such as envelope proteins, can also be used.
  • proteins from other invasive organisms such as the 17 amino acid peptide from the circumsporozoite protein of plasmodium falciparum known as RII.
  • hormones for example: hormones, steroids, androgens, estrogens, thyroid hormone, or vitamins, folic acid.
  • polyalkylene glycol can be included with the desired polynucleotides/polypeptides.
  • the polyalkylene glycol is polyethlylene glycol.
  • mono-, di-, or polysaccharides can be included.
  • the polysaccharide is dextran or DEAE-dextran.
  • the desired polynucleotide/polypeptide can also be encapsulated in lipids or packaged in liposomes prior to delivery to the subject or to cells derived therefrom.
  • Lipid encapsulation is generally accomplished using liposomes which are able to stably bind or entrap and retain nucleic acid.
  • the ratio of condensed polynucleotide to lipid preparation can vary but will generally be around 1 : 1 (mg DNA:micromoles lipid), or more of lipid.
  • Liposomal preparations for use in the present invention include cationic (positively charged), anionic (negatively charged) and neutral preparations.
  • Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Feigner (1987) Proc. Natl. Acad. Sci. USA 84:7413-7416); mRNA (Malone (1989) Proc. Natl. Acad. Sci. USA 86:6077-6081); and purified transcription factors (Debs (1990) J. Biol. Chem. 265: 10189-10192), in functional form.
  • Cationic liposomes are readily available.
  • N[l-2,3-dioleyloxy)propyl]-N,N,N- triethylammonium (DOTMA) liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, NY. (See, also, Feigner supra).
  • Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger).
  • Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. Szoka (1978) Proc. Natl. Acad. Sci. USA 75:4194-4198; WO90/1 1092 for a description of the synthesis of DOTAP (l ,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes.
  • anionic and neutral liposom es are readily available, such as from Avanti Polar Lipids (B irm ingham , AL), or can be easily prepared using readily available m aterials.
  • Such m aterials include phosphatidyl choline, cholesterol, phosphatidyl ethanolam ine, dioleoylphosphatidyl choline (D0PC), dioleoylphosphatidyl glycerol (D 0 PG ), dioleoylphoshatidyl ethanolam ine (DOPE), am ong others.
  • m aterials can also be m ixed w ith the DOTM A and D OTA P starting m aterials in appropriate ratios.
  • M ethods for m aking liposom es using these m aterials are well known in the art.
  • the liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUNs), or large unilamellar vesicles (LUVs).
  • MLVs multilammelar vesicles
  • SUNs small unilamellar vesicles
  • LUVs large unilamellar vesicles
  • the various liposome-nucleic acid complexes are prepared using methods known in the art. See e.g. Straubinger (1983) Meth. Immunol 101:512-527; Szoka (1978)
  • lipoproteins can be included w ith the polynucleotide/polypeptide to be delivered.
  • lipoproteins to be utilized include: chylom icrons, HDL, ID L, LDL, and VLDL. M utants, fragm ents, or fusions of these proteins can also be used. A lso, m odifications of naturally occurring lipoproteins can be used, such as acetylated LDL.
  • These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors.
  • lipoproteins are including w ith the polynucleotide to be delivered, no other targeting ligand is included in the composition.
  • Naturally occurring lipoproteins comprise a lipid and a protein portion.
  • the protein portion are known as apoproteins.
  • apoproteins A, B, C, D, and E have been isolated and identified. At least two of these contain several proteins, designated by Roman numerals, Al, All, AIV; Cl, CII, Cffl.
  • a lipoprotein can comprise more than one apoprotein.
  • naturally occurring chylomicrons comprises of A, B, C, and E, over time these lipoproteins lose A and acquire C and E apoproteins.
  • VLDL comprises A, B, C, and E apoproteins
  • LDL comprises apoprotein B; and HDL comprises apoproteins A, C, and E.
  • Lipoproteins contain a variety of lipids including, triglycerides, cholesterol (free and esters), and phospholipids. The composition of the lipids varies in naturally occurring lipoproteins. For example, chylomicrons comprise mainly triglycerides.
  • the composition of the lipids are chosen to aid in conformation of the apoprotein for receptor binding activity.
  • the composition of lipids can also be chosen to facilitate hydrophobic interaction and association with the polynucleotide binding molecule.
  • Naturally occurring lipoproteins can be isolated from serum by ultracentrifugation, for instance. Such methods are described in Meth. Enzymol. (supra); Pitas (1980) J. Biochem. 255:5454-5460 and Mahey (1979) J Clin. Invest 64:743-750. Lipoproteins can also be produced by in vitro or recombinant methods by expression of the apoprotein genes in a desired host cell. See, for example, Atkinson (1986) Annu Rev Biophys Chem 15:403 and Radding (1958) Biochim Biophys Acta 30: 443. Lipoproteins can also be purchased from commercial suppliers, such as Biomedical Techniologies, Inc., Stoughton, Massachusetts, USA. Further description of lipoproteins can be found in Zuckermann et al. PCT/US97/ 14465. F.Polvcationic Agents
  • Polycationic agents can be included, with or without lipoprotein, in a composition with the desired polynucleotide/polypeptide to be delivered.
  • Polycationic agents typically, exhibit a net positive charge at physiological relevant pH and are capable of neutralizing the electrical charge of nucleic acids to facilitate delivery to a desired location. These agents have both in vitro, ex vivo, and in vivo applications. Polycationic agents can be used to deliver nucleic acids to a living subject either intram uscularly, subcutaneously, etc.
  • useful polypeptides as polycationic agents: polylysine, polyarginine, polyornithine, and protamine.
  • transcriptional factors also contain domains that bind DNA and therefore may be useful as nucleic aid condensing agents. Briefly, transcriptional factors such as C/CEBP, c-jun, c-fos, AP-1, AP-2, AP-3, CPF, Prot-1, Sp-1, Oct-1, Oct-2, CREP, and TFIID contain basic domains that bind DNA sequences.
  • Organic polycationic agents include: spermine, spermidine, and purtrescine.
  • polycationic agent The dimensions and of the physical properties of a polycationic agent can be extrapolated from the list above, to construct other polypeptide polycationic agents or to produce synthetic polycationic agents.
  • Synthetic polycationic agents which are useful include, for example, DEAE-dextran, polybrene.
  • LipofectinTM, and lipofect AMINETM are monomers that form polycationic complexes when combined with polynucleotides/polypeptides. Nucleic Acid Hybridisation
  • Hybridization refers to the association of two nucleic acid sequences to one another by hydrogen bonding. Typically, one sequence w ill be fixed to a solid support and the other will be free in solution. Then, the two sequences will be placed in contact with one another under conditions that favor hydrogen bonding. Factors that affect this bonding include: the type and volume of solvent; reaction tem perature; time of hybridization; agitation; agents to block the non-specific attachment of the liquid phase sequence to the solid support (Denhardt's reagent or B LOTTO); concentration of the sequences; use of compounds to increase the rate of association of sequences (dextran sulfate or polyethylene glycol); and the stringency of the washing conditions following hybridization.
  • “Stringency” refers to conditions in a hybridization reaction that favor association of very similar sequences over sequences that differ.
  • the combination of temperature and salt concentration should be chosen that is approximately 120 to 200°C below the calculated Tm of the hybrid under study.
  • the temperature and salt conditions can often be determined empirically in preliminary experiments in which samples of genomic DNA immobilized on filters are hybridized to the sequence of interest and then washed under conditions of different stringencies. See Sambrook et al. at page 9.50.
  • Variables to consider when performing, for example, a Southern blot are (1) the complexity of the DNA being blotted and (2) the homology between the probe and the sequences being detected.
  • the total amount of the fragment(s) to be studied can vary a magnitude of 10, from 0.1 to l ⁇ g for a plasmid or phage digest to 10 '9 to 10 "8 g for a single copy gene in a highly complex eukaryotic genome.
  • substantially shorter blotting, hybridization, and exposure times a smaller amount of starting polynucleotides, and lower specific activity of probes can be used.
  • a single-copy yeast gene can be detected with an exposure time of only 1 hour starting with 1 ⁇ g of yeast DNA, blotting for two hours, and hybridizing for 4-8 hours with a probe of 10 8 cpm/ ⁇ g.
  • a conservative approach would start with 10 ⁇ g of DNA, blot overnight, and hybridize overnight in the presence of 10% dextran sulfate using a probe of greater than 10 8 cpm/ ⁇ g, resulting in an exposure time of -24 hours.
  • Tm melting temperature
  • Tm 81 + 16.6(log 10 Ci) + 0.4[%(G + C)]-0.6(%formamide) - 600// ⁇ -1.5(%mismatch).
  • Ci is the salt concentration (monovalent ions)
  • n is the length of the hybrid in base pairs (slightly modified from Meinkoth & Wahl (1984) Anal. Biochem. 138: 267-284).
  • the temperature of the hybridization and washes and the salt concentration during the washes are the simplest to adjust. As the temperature of the hybridization increases (i.e.
  • formamide content should be lowered and temperature adjusted accordingly, using the equation above. If the homology between the probe and the target fragment are not known, the simplest approach is to start with both hybridization and wash conditions which are nonstringent. If non-specific bands or high background are observed after autoradiography, the filter can be washed at high stringency and reexposed. If the time required for exposure makes this approach impractical, several hybridization and/or washing stringencies should be tested in parallel.
  • N. meningitidis serogroup B contained a component of approximately 80-85kDa. This protein was purified from SDS-PAGE gels and N-terminal sequenced (SEQ ID 1).
  • Antibodies raised against the SDS-PAGE purified protein cross-reacted with equivalent proteins in more than 50 N. meningitidis strains of diverse serogroups and serotypes. Cross-reactivity with N. gonorrhoeae, N.polysaccharia and N. lactamica was also observed. Post-immune sera from vaccinated patients also reacted with the protein.
  • the complete gene was cloned from serogroup B N meningitidis (SEQ ID 2) and the encoded protein was inferred (SEQ ID 3).
  • SEQ ID 4 The complete gene was cloned from serogroup B N meningitidis (SEQ ID 2) and the encoded protein was inferred (SEQ ID 3).
  • SEQ ID 4 The complete gene was cloned from serogroup B N meningitidis (SEQ ID 2) and the encoded protein was inferred (SEQ ID 3).
  • SEQ ID 4 signal peptide
  • SEQ ED 5 a mature sequence
  • SEQ ID 6 The complete gene from serogroup A N.meningitidis is shown in SEQ ID 6, with the encoded protein in SEQ ID 7.
  • the signal peptide and mature sequence are SEQ IDs 8 and 9.
  • the complete gene from N.gonorrhoeae is shown in SEQ ID 10, with the encoded protein in SEQ ID 11.
  • the signal peptide and mature sequence are SEQ IDs 12 and 13.
  • the protein sequences were compared and are highly homologous.
  • the N.meningitidis serogroup B sequence and the N.gonorrhoeae sequence show 95.4% identity in 797 aa overlap:
  • pep LPGSK QYYSATHNQTWFFP SKTFTLM GGEVGIAGGYGRTKEIPFFENFYGGGLGSVR orf21ng .pep LPGSK QYYSATHNQTWFFP ⁇ KTFT M GGEVGIAGGYGRTKEIPFFE FYGGGLGSVR
  • N.meningitidis serogroup A and B sequences show 99.9% identity in 797 aa overlap:
  • pep KPEDEIQRFQFQLGTTF orf 2 la pep KPEDEIQRFQFQ GTTFX
  • N.meningitidis strains 2996 and MC58 were grown to exponential phase in 100ml of GC medium, harvested by centrifugation, and resuspended in 5ml buffer (20% w/v sucrose, 50mM Tris-HCl, 50mM EDTA, pH8). After 10 minutes incubation on ice, the bacteria were lysed by adding 10ml of lysis solution (50mM NaCl, 1% Na-Sarkosyl, 50 ⁇ g/ml Proteinase K), and the suspension incubated at 37°C for 2 hours. Two phenol extractions (equilibrated to pH 8) and one CHCl 3 /isoamylalcohol (24: 1) extraction were performed.
  • DNA was precipitated by addition of 0.3M sodium acetate and 2 volumes of ethanol, and collected by centrifugation. The pellet was washed once with 70%(v/v) ethanol and redissolved in 4.0ml TE buffer (lOmM Tris- HCl, ImM EDTA, pH 8.0). The DNA concentration was measured by reading OD 26 o-
  • ORF21 without the sequence encoding the leader peptide, was amplified by PCR using the following oligonucleotides: orf21 for ⁇ SEQ ID 105> (BamHI-Ndel) orf21 rev ⁇ SEQ ID 106> (Xhol)
  • the 5' primer included two restriction sites (BamHI-Ndel), the 3' primer included a Xhol restriction site, in order to direct the cloning of the amplification product (corresponding to ORF21) into the two expression vectors pGEX-KG (using BamHI-XhoT) to have a N-terminal GST-fusion, and pET21b+ (using Ndel-Xhol), to have a C-terminal His-fusion.
  • the standard PCR protocol was as follows: 200ng of genomic DNA from 2996 or MC58 strains or lOng of plasmid DNA preparation of recombinant clones were used as template in the presence of 40 ⁇ M of each oligonucletide primer, 400-800 ⁇ M dNTPs solution, lx PCR buffer (including 1.5mM MgCl 2 ), 2.5 units Taql DNA polymerase (using Perkin-Elmer AmpliTaQ, Boerhingher Mannheim ExpandTM Long Template).
  • each sample underwent a two-step amplification: the first 5 cycles were performed using the hybridisation temperature that excluded the restriction enzyme tail of the primer (T m ⁇ ). This was followed by 30 cycles according to the hybridisation temperature calculated for the whole length oligos (T m2 ). Elongation times, performed at 68°C or 72°C, varied according to the length of the Orf to be amplified. In the case of Orf 1 the elongation time, starting from 3 minutes, was increased by 15 seconds each cycle. The cycles were completed with a 10 minute extension step at 72°C.
  • the amplified DNA was either loaded directly on a 1% agarose gel.
  • the DNA fragment corresponding to the band of correct size was purified from the gel using the Qiagen Gel Extraction Kit, following the manufacturer's protocol.
  • the purified DNA corresponding to the amplified fragment was digested with the appropriate restriction enzymes for cloning into pET-21b+ or pET22b+.
  • Digested fragments were purified using the QIAquick PCR purification kit (following the manufacturer's instructions) and eluted with either H O or lOmM Tris, pH 8.5.
  • Plasmid vectors were digested with the appropriate restriction enzymes, loaded onto a 1.0% agarose gel and the band corresponding to the digested vector purified using the Qiagen QIAquick Gel Extraction Kit.
  • Recombinant plasmid was transformed into competent E.coli DH5 or HB101 by incubating the ligase reaction solution and bacteria for 40 minutes on ice, then at 37°C for 3 minutes. This was followed by the addition of 800 ⁇ l LB broth and incubation at 37°C for 20 minutes. The cells were centrifuged at maximum speed in an Eppendorf microfuge, resuspended in approximately 200 ⁇ l of the supernatant and plated onto LB ampicillin (lOOmg/ml ) agar.
  • Screening for recombinant clones was performed by growing randomly selected colonies overnight at 37°C in 4.0ml of LB broth + lOO ⁇ g/ml ampicillin. Cells were pelleted and plasmid DNA extracted using the Qiagen QIAprep Spin Miniprep Kit, following the manufacturer's instructions. Approximately l ⁇ g of each individual miniprep was digested with the appropriate restriction enzymes and the digest loaded onto a 1-1.5% agarose gel (depending on the expected insert size), in parallel with the molecular weight marker (lkb DNA Ladder, GIBCO). Positive clones were selected on the basis of the size of insert.
  • recombinant plasmids were transformed into E.coli strains suitable for expression of the recombinant protein, l ⁇ l of each construct was used to transform E.coli BL21-DE3 as described above.
  • Single recombinant colonies were inoculated into 2ml LB+Amp (lOO ⁇ g/ml), incubated at 37°C overnight, then diluted 1:30 in 20ml of LB+Amp (lOO ⁇ g/ml) in 100ml flasks, to give an OD 600 between 0.1 and 0.2.
  • OD 6 oo indicated exponential growth suitable for induction of expression (0.4-0.8 OD).
  • Protein expression was induced by addition of LOmM IPTG. After 3 hours incubation at 30°C or 37°C the OD 6 oo was measured and expression examined. 1.0ml of each sample was centrifuged in a microfuge, the pellet resuspended in PBS and analysed by SDS-PAGE and Coomassie Blue staining.
  • the GST fusion protein was expressed, but it was found to be insoluble (i.e. unpurifiable).
  • the His-fusion was expressed as insoluble protein.
  • a single colony was streaked and grown overnight at 37°C on a LB/Amp (100 ⁇ g/ml) agar plate.
  • An isolated colony from this plate was inoculated into 20ml of LB/Amp (100 ⁇ g/ml) liquid medium and grown overnight at 37°C with shaking.
  • the overnight culture was diluted 1:30 into 1.0 L LB/Amp (100 ⁇ g/ml) liquid medium and allowed to grow at the optimal temperature (30 or 37°C) until the OD5 5 0 reached 0.6-0.8.
  • Expression of recombinant protein was induced by addition of IPTG (final concentration LOmM) and the culture incubated for a further 3 hours.
  • Bacteria were harvested by centrifugation at 8000g for 15 min at 4°C. The bacterial pellet was resuspended in 7.5 ml of buffer B (8M urea, lOmM Tris-HCl, 100 mM phosphate buffer, pH 8.8). Cells were disrupted by sonication on ice four times for 30 sec at 40W using a Branson sonifier 450 and centrifuged at 13 OOOxg for 30 min at 4°C. Pellets were resuspended in 2.0 ml buffer C (6 M guanidine hydrochloride, 100 mM phosphate buffer, 10 mM Tris- HC1, pH 7.5 and treated with 10 passes of a Dounce homogenizer.
  • buffer B 8M urea, lOmM Tris-HCl, 100 mM phosphate buffer, pH 8.8
  • Cells were disrupted by sonication on ice four times for 30 sec at 40W using a Branson sonifier 450 and centrif
  • the homogenate was centrifuged at 13000g for 30 min and the supernatant retained. Supernatants were mixed with 150 ⁇ l Ni + -resin (previously equilibrated with buffer B) and incubated at room temperature with gentle agitation for 30 min.
  • the resin was Chelating Sepharose Fast Flow (Pharmacia), prepared according to the manufacturer's protocol. The batch-wise preparation was centrifuged at 700 x g for 5 min at 4°C and the supernatant discarded. The resin was washed twice (batch-wise) with 10ml buffer B for 10 min, resuspended in 1.0 ml buffer B and loaded onto a disposable column.
  • the resin continued to be washed with buffer B at room temperature, until the OD 280 of the flow-through reached 0.02- 0.01.
  • the resin was further washed with buffer D (8M urea, lOmM Tris-HCl, lOOmM phosphate buffer, pH 6.3) until OD 280 of the flow-through reached 0.02-0.01.
  • the His-fusion protein was eluted by addition of 700 ⁇ l of elution buffer B (8 M urea, 10 mM Tris-HCl, 100 mM phosphate buffer, pH 4.5) and fractions collected until the OD 8 o indicated all the recombinant protein was obtained. 20 ⁇ l aliquots of each elution fraction were analysed by SDS-PAGE. Protein concentrations were estimated using the Bradford assay.
  • glycerol was added to the denatured fractions obtained above to give a final concentration of 10% v/v.
  • the proteins were diluted to 200 ⁇ g/ml using dialysis buffer I (10% v/v glycerol, 0.5M arginine, 50 mM phosphate buffer, 5.0 mM reduced glutathione, 0.5 mM oxidised glutathione, 2.0 M urea, pH 8.8) and dialysed against the same buffer for 12-14 hours at 4°C.
  • mice were immunized with antigens on days 0, 21 and 35 and sera analyzed at day 49.
  • 0RF21 gave a positive results on the following ELISA assay: acapsulated M enB M 7 and the capsulated strains were plated on chocolate agar plates and incubated overnight at 37°C with 5% C0 2 .
  • B acterial colonies were collected from the agar plates using a sterile dracon swab and inoculated into M ueller-Hinton Broth (Difco) containing 0.25% glucose. B acterial growth was m onitored every 30 m inutes by following OU 2 o. The bacteria were let to grow until the 0D reached the value of 0.4-0.5. The culture was centrifuged for 10 minutes at 4000rpm .
  • the membrane was washed twice with washing buffer (3% skimmed milk, 0.1% Triton X100 in PBS) and incubated for 2 hours at 37°C with mice sera diluted 1:200 in washing buffer. The membrane was washed twice and incubated for 90 minutes with a 1:2000 dilution of horseradish peroxidase labelled anti-mouse Ig. The membrane was washed twice with 0.1% Triton X100 in PBS and developed with the Opti-4CN Substrate Kit (Bio-Rad). The reaction was stopped by adding water.
  • the OMVs were prepared as follows: N. meningitidis strain 2996 was grown overnight at 37 degrees with 5% CO 2 on 5 GC plates, harvested with a loop and resuspended in 10 ml of 20mM
  • Tris-HCl pH 7.5, 2 mM EDTA Tris-HCl pH 7.5, 2 mM EDTA. Heat inactivation was performed at 56°C for 45 minutes and the bacteria disrupted by sonication for 5 minutes on ice (50% duty cycle, 50% output , Branson sonifier 3 mm microtip). Unbroken cells were removed by centrifugation at 5000g for 10 minutes, the supernatant containing the total cell envelope fraction recovered and further centrifuged overnight at 50000g at the temperature of 4°C . The pellet containing the membranes was resuspended in 2% sarkosyl, 20mM Tris-HCl pH 7.5, 2 mM EDTA and incubated at room temperature for 20 minutes to solubilyze the inner membranes.
  • the suspension was centrifuged at lOOOOg for 10 minutes to remove aggregates, the supernatant was further centrifuged at 50000g for 3 hours.
  • the pellet, containing the outer membranes was washed in PBS and resuspended in the same buffer.
  • the protein concentration was measured by the D.C. Bio-Rad Protein assay (Modified Lowry method), using BSA as a standard.
  • Total cell extracts were prepared as follows: N. meningitidis strain 2996 was grown overnight on a GC plate, harvested with a loop and resuspended in 1ml of 20mM Tris-HCl. Heat inactivation was performed at 56°C for 30 minutes. ELISA, FACS and Western analysis all show that ORF21 is surface-exposed.
  • B acteria were diluted in 50m M Phosphate buffer pH 7.2 containing l Om M M gC l 2 , l Om M CaCl 2 and 0.5 % (w/v) B SA (assay buffer) at the w orking dilution of 10 5 CFU/m l.
  • Controls included bacteria incubated with complement serum , im m une sera incubated with bacteria and with complem ent inactivated by heating at 56°C for 30'.
  • l O ⁇ l of the controls were plated on M ueller-Hinton agar plates using the tilt method (tim e 0).
  • the 96-wells plate was incubated for 1 hour at 37°C with rotation.
  • 7 ⁇ l of each sample were plated on M ueller-Hinton agar plates as spots, whereas l O ⁇ l of the controls were plated using the tilt method (time 1 ).
  • a gar plates were incubated for 18 hours at 37 degrees and the colonies corresponding to time 0 and time 1 were counted.
  • Computer analysis Figure 1 shows the sequence of ORF21, together with computer analysis data.
  • ORF21 has the following AMPHI regions [Gao et al. (1989) J. Immunol. 143:3007; Roberts et al (1996) AIDS Res Hum Retrovir 12:593; Quakyi et al (1992) Scand J Immunol suppl.l 1:9]: SEQ IDs 14 to 32.
  • the antigenic index algorithm identified the following regions: SEQ IDs 33 to 70.
  • Hopp & Woods analysis reveals the following hydrophilic regions: SEQ IDs 71 to 104.
  • SEQ IDs 14-104 may be used as these oligopeptides, or as part of longer polypeptides.
  • the three proteins identified above are expressed and used for immunisation. Good immune responses are observed against the proteins.
  • the proteins are each combined with antigens against other pathogenic organisms (e.g. the Chiron polysaccharide vaccine against serogroup C meningitis), and used for immunisation. Good immune responses are observed.
  • pathogenic organisms e.g. the Chiron polysaccharide vaccine against serogroup C meningitis

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

An 85kDa antigen from Neisseria meningitidis and Neisseria gonorrhoeae has been cloned, sequenced and expressed. The antigen is common to diverse strains, serogroups and serotypes of N.meningitidis, and also to N.gonorrhoeae, N.polysaccharia and N.lactamica. The protein sequences of N.meningitids (serogroups A and B) and N.gonorrhoeae are highly homologous.

Description

85kDa NEISSERIAL ANTIGEN
All documents cited herein are hereby incoφorated by reference in their entirety.
TECHNICAL FIELD
This invention relates to an antigen from the related bacteria Neisseria meningitidis and Neisseria gonorrhoeae.
BACKGROUND ART
Neisseria meningitidis is a non-motile, Gram-negative diplococcus human pathogen. It colonises the pharynx, causing meningitis and, occasionally, septicaemia in the absence of meningitis. It is closely related to N. gonorrhoeae, although one feature that clearly differentiates meningococcus from gonococcus is the presence of a polysaccharide capsule that is present in all pathogenic meningococci.
N. meningitidis causes both endemic and epidemic disease. In the United States the attack rate is 0.6- 1 per 100,000 persons per year, and it can be much greater during outbreaks [Lieber an et al. (1996) JAMA 275(19):1499-1503; Schuchat et al (1997) N Engl J Med 337(14):970-976]. In developing countries, endemic disease rates are much higher and during epidemics incidence rates can reach 500 cases per 100,000 persons per year. Mortality is extremely high, at 10-20% in the United States, and much higher in developing countries. Following the introduction of the conjugate vaccine against Haemophilus influenzae, N. meningitidis is the major cause of bacterial meningitis at all ages in the United States [Schuchat et al (1997) supra].
Based on the organism's capsular polysaccharide, 12 serogroups of N. meningitidis have been identified. The meningococcal vaccine currently in use is a tetravalent polysaccharide vaccine composed of serogroups A, C, Y and W135. Following the success of the vaccination against H.influenzae, however, conjugate vaccines against serogroups A and C have been developed
Meningococcus B remains a problem, however. This serotype currently is responsible for approximately 50% of total meningitis in the United States, Europe, and South America. The polysaccharide approach cannot be used because the menB capsular polysaccharide is a polymer of α(2-8)-Iinked N-acetyl neuraminic acid that is also present in mammalian tissue. This results in tolerance to the antigen; indeed, if a response were elicited, it would be anti-self, and therefore undesirable. In order to avoid induction of autoimmunity and to induce a protective immune response, the capsular polysaccharide has, for instance, been chemically modified substituting the N-acetyl groups with N-propionyl groups, leaving the specific antigenicity unaltered [Romero & Outschoorn (1994) Clin Microbiol Rev 7(4):559-575]. Altemative approaches to menB vaccines have used complex mixtures of outer membrane proteins (OMPs), containing either the OMPs alone, or OMPs enriched in porins, or deleted of the class 4 OMPs that are believed to induce antibodies that block bactericidal activity. These vaccines are poorly characterised and are only effective against homologous strains. To overcome antigenic variability, multivalent vaccines containing up to nine different porins have been constructed [e.g. Poolman JT (1992) Infect. Agents Dis. 4: 13-28]. Additional proteins to be used in outer membrane vaccines have been the opa and ope proteins, but none of these approaches have been able to overcome the antigenic variability [e.g. Ala'Aldeen & Borriello (1996) Vaccine 14(l):49-53]. The Norwegian National Institute of Public Health vaccine, however, is safe, elicits strain-specific immunity in children and adults, and is efficacious in preventing disease in adolescents [Fredriksen et al. (X99X) NIPH Ann 14(2):67-80 & 107-123].
These vaccines are, however, poorly characterised, and the molecular basis for their efficacy and protection has not been dissected. It is an object of the present invention to define antigenic components of vaccines, to characterise them in order to allow better defined vaccines to be produced (e.g. acellular sub-unit vaccines), and to broaden the Neisseria strains against which immunity is elicited.
DISCLOSURE OF THE INVENTION
Proteins
The invention provides a protein comprising one or more of the following amino acid sequences: SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 1 1, SEQ ID 12, SEQ ID 13.
It also provides a protein comprising a sequence having sequence identity to SEQ ID 1 , SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 11, SEQ ID 12, or SEQ ID 13. Depending on the particular SEQ ID, the degree of sequence identity is preferably greater than 50% (e.g. 60%, 70%, 80%, 90%, 95%, 99% or more). These homologous sequences include mutants and allelic variants. Typically, 50% identity or more between two proteins is considered to be an indication of functional equivalence. Identity between proteins is preferably determined by the Smith-Waterman homology search algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affine gap search with parameters gap open penalty=12 and gap extension penalty -1.
The invention further provides a protein comprising a fragment of SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 11, SEQ ID 12, or SEQ ID 13. The fragment should comprise at least n consecutive amino acids from the sequence and, depending on the particular sequence, n is 7 or more (e.g. 8, 10, 12, 14, 16, 18, 20 or more). Preferably the fragment comprises an epitope from the sequence.
The proteins of the invention can, of course, be prepared by various means (e.g. recombinant expression, purification from cell culture, chemical synthesis etc.) and in various forms (e.g. native, fusions etc.). They are preferably prepared in substantially pure form (i.e. substantially free from other Neisseria or host cell proteins).
Antibodies
According to a further aspect, the invention provides antibodies which bind to these proteins. These may be polyclonal or monoclonal and may be produced by any suitable means.
Nucleic acids
According to a further aspect, the invention provides nucleic acid comprising SEQ ID 2, SEQ ID 6, or SEQ ID 10. In addition, the invention provides nucleic acid comprising sequences having sequence identity to SEQ ID 2, SEQ ID 6, or SEQ ID 10. Depending on the particular SEQ ID, the degree of sequence identity is preferably greater than 50% (e.g. 60%, 70%, 80%, 90%, 95%, 99% or more). Sequence identity is preferably determined by the Smith-Waterman ho ology search algorithm as implemented in the MPSRCH program (Oxford Molecular), using an affine gap search with parameters gap open penalty-12 and gap extension penalty=l .
Furthermore, the invention provides nucleic acid which can hybridise to SEQ ID 2, SEQ ID 6, or SEQ ID 10, preferably under "high stringency" conditions (e.g. 65°C in a 0. IxSSC, 0.5% SDS solution).
Nucleic acid comprising fragments of SEQ ID 2, SEQ ID 6, or SEQ ID 10 are also provided. These should comprise at least n consecutive nucleotides from SEQ ID 2, SEQ ID 6, or SEQ ID 10 and, depending on the particular sequence, n is 10 or more (eg 12, 14, 15, 18, 20, 25, 30, 35, 40 or more).
According to a further aspect, the invention provides nucleic acid encoding the proteins and protein fragments of the invention.
It should also be appreciated that the invention provides nucleic acid comprising sequences complementary to those described above (e.g. for antisense or probing puφoses). Nucleic acid according to the invention can, of course, be prepared in many ways (e.g. by chemical synthesis, from genomic or cDNA libraries, from the organism itself etc.) and can take various forms (e.g. single stranded, double stranded, vectors, probes etc.).
In addition, the term "nucleic acid" includes DNA and RNA, and also their analogues, such as those containing modified backbones, and also peptide nucleic acids (PNA) etc.
According to a further aspect, the invention provides vectors comprising nucleotide sequences of the invention (e.g. expression vectors) and host cells transformed with such vectors.
Compositions
According to a further aspect, the invention provides compositions comprising protein, antibody, and/or nucleic acid according to the invention. These compositions may be suitable as vaccines, for instance, or as diagnostic reagents, or as immunogenic compositions.
The compositions of the invention may further comprise immunogenic components selected from one or more of the following:
• the proteins disclosed in WO99/57280 or immunogenic fragments thereof; • the proteins disclosed in WO99/36544 or immunogenic fragments thereof;
• the proteins disclosed in WO99/24578 or immunogenic fragments thereof;
• the proteins disclosed in Tettelin et al. [Science (2000) 287:1809-1815; NMB0001 to NMB2160] or immunogenic fragments thereof;
• the proteins disclosed in WO97/28273 or immunogenic fragments thereof; • the proteins disclosed in WO96/29412 or immunogenic fragments thereof;
• the proteins disclosed in WO95/03413 or immunogenic fragments thereof;
• the proteins disclosed in WO99/31 132 or immunogenic fragments thereof;
• a protective antigen against Neisseria meningitidis serogroup A;
• a protective antigen against Neisseria meningitidis serogroup C; • a protective antigen against Neisseria meningitidis serogroup Y;
• a protective antigen against Neisseria meningitidis serogroup W;
• a protective antigen against Haemophilus influenzae;
• a protective antigen against pneumococcus;
• a protective antigen against diphtheria; • a protective antigen against tetanus;
• a protective antigen against whooping cough; • a protective antigen against Helicobacter pylori;
• a protective antigen against polio; and/or
• a protective antigen against hepatitis B virus.
Preferably, the compositions further comprise immunogenic components selected from one or more of the following:
• a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8,
10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64,66,68,70,72,74,76,78,80,82,84,86,88,90,92,94,96,98, 100, 102, 104, 106, 108, 110, 112, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192,
194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 256, 258, 260, 262, 264, 266, 268, 270, 272, 274, 276, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296, 298, 300, 302, 304, 306, 308, 310, 312, 314, 316, 318, 320, 322, 324, 326, 328, 330, 332, 334, 336, 338, 340, 342, 344, 346, 348, 350, 352, 354, 356, 358, 360, 362, 364, 366, 368, 370, 372, 374, 376, 378, 380, 382, 384, 386, 388, 390, 392,
394, 396, 398, 400, 402, 404, 406, 408, 410, 412, 414, 416, 418, 420, 422, 424, 426, 428, 430, 432, 434, 436, 438, 440, 442, 444, 446, 448, 450, 452, 454, 456, 458, 460, 462, 464, 466, 468, 470, 472, 474, 476, 478, 480, 482, 484, 486, 488, 490, 492, 494, 496, 498, 500, 502, 504, 506, 508, 510, 512, 514, 516, 518, 520, 522, 524, 526, 528, 530, 532, 534, 536, 538, 540, 542, 544, 546, 548, 550, 552, 554, 556, 558, 560, 562, 564, 566, 568, 570, 572, 574, 576, 578, 580, 582, 584, 586, 588, 590, 592,
594, 596, 598, 600, 602, 604, 606, 608, 610, 612, 614, 616, 618, 620, 622, 624, 626, 628, 630, 632, 634, 636, 638, 640, 642, 644, 646, 648, 650, 652, 654, 656, 658, 660, 662, 664, 666, 668, 670, 672, 674, 676, 678, 680, 682, 684, 686, 688, 690, 692, 694, 696, 698, 700, 702, 704, 706, 708, 710, 712, 714, 716, 718, 720, 722, 724, 726, 728, 730, 732, 734, 736, 738, 740, 742, 744, 746, 748, 750, 752, 754, 756, 758, 760, 762, 764, 766, 768, 770, 772, 774, 776, 778, 780, 782, 784, 786, 788, 790, 792,
794, 796, 798, 800, 802, 804, 806, 808, 810, 812, 814, 816, 818, 820, 822, 824, 826, 828, 830, 832, 834, 836, 838, 840, 842, 844, 846, 848, 850, 852, 854, 856, 858, 860, 862, 864, 866, 868, 870, 872, 874, 876, 878, 880, 882, 884, 886, 888, 890, & 892, as disclosed in W099/24578 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%,
70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
• a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, & 90, as disclosed in W099/36544 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
• a protein encoded by one of the 21 60 genes N M B 0001 to N M B 2160 disclosed in Tettelin et al [Science (2000) 287: 1809-1815] (or a protein com prising an im m unogenic fragm ent of one or m ore of these 2160 genes, or a protein com prising a sequence having sequence identity (preferably greater than 50% e.g. 60% , 70% , 80% , 90% , 95 % , 99% or m ore) to one of these 2 160 genes);
• a protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62,
64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 1 10, 1 12,
1 14, 1 16, 1 1 8, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152,
154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192,
194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 21 8, 220, 222, 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 256, 258, 260, 262, 264, 266, 268, 270, 272,
274, 276, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296, 298, 300, 302, 304, 306, 308, 310, 312,
314, 316, 318, 320, 322, 324, 326, 328, 330, 332, 334, 336, 338, 340, 342, 344, 346, 348, 350, 352,
354, 356, 358, 360, 362, 364, 366, 368, 370, 372, 374, 376, 378, 380, 382, 384, 386, 388, 390, 392,
394, 396, 398, 400, 402, 404, 406, 408, 410, 412, 414, 416, 41 8, 420, 422, 424, 426, 428, 430, 432, 434, 436, 438, 440, 442, 444, 446, 448, 450, 452, 454, 456, 458, 460, 462, 464, 466, 468, 470, 472,
474, 476, 478, 480, 482, 484, 486, 488, 490, 492, 494, 496, 498, 500, 502, 504, 506, 508, 510, 512,
514, 516, 5 18, 520, 522, 524, 526, 528, 530, 532, 534, 536, 538, 540, 542, 544, 546, 548, 550, 552,
554, 556, 558, 560, 562, 564, 566, 568, 570, 572, 574, 576, 578, 580, 582, 584, 586, 588, 590, 592,
594, 596, 598, 600, 602, 604, 606, 608, 610, 612, 614, 616, 618, 620, 622, 624, 626, 628, 630, 632, 634, 636, 638, 640, 642, 644, 646, 648, 650, 652, 654, 656, 658, 660, 662, 664, 666, 668, 670, 672,
674, 676, 678, 680, 682, 684, 686, 688, 690, 692, 694, 696, 698, 700, 702, 704, 706, 708, 710, 712,
714, 716, 718, 720, 722, 724, 726, 728, 730, 732, 734, 736, 738, 740, 742, 744, 746, 748, 750, 752,
754, 756, 758, 760, 762, 764, 766, 768, 770, 772, 774, 776, 778, 780, 782, 784, 786, 788, 790, 792,
794, 796, 798, 800, 802, 804, 806, 808, 810, 812, 814, 816, 818, 820, 822, 824, 826, 828, 830, 832, 834, 836, 838, 840, 842, 844, 846, 848, 850, 852, 854, 856, 858, 860, 862, 864, 866, 868, 870, 872,
874, 876, 878, 880, 882, 884, 886, 888, 890, 892, 894, 896, 898, 900, 902, 904, 906, 908, 910, 912,
914, 916, 918, 920, 922, 924, 926, 928, 930, 932, 934, 936, 938, 940, 942, 944, 946, 948, 950, 952,
954, 956, 958, 960, 962, 964, 966, 968, 970, 972, 974, 976, 978, 980, 982, 984, 986, 988, 990, 992,
994, 996, 998, 1000, 1002, 1004, 1006, 1008, 1010, 1012, 1014, 1016, 1018, 1020, 1022, 1024, 1026, 1028, 1030, 1032, 1034, 1036, 1038, 1040, 1042, 1044, 1046, 1048, 1050, 1052, 1054, 1056, 1058, 1060, 1062, 1064, 1066, 1068, 1070, 1072, 1074, 1076, 1078, 1080, 1082, 1084, 1086, 1088, 1090, 1092, 1094, 1096, 1098, 1100, 1102, 1104, 1106, 1108, 1110, 1112, 1114, 1116, 1118, 1120, 1122, 1124, 1126, 1128, 1130, 1132, 1134, 1136, 1138, 1140, 1142, 1144, 1146, 1148, 1150, 1152, 1154, 1156, 1158, 1160, 1162, 1164, 1166, 1168, 1170, 1172, 1174, 1176, 1178, 1180, 1182, 1184, 1186, 1188, 1190, 1192, 1194, 1196, 1198, 1200, 1202, 1204, 1206, 1208, 1210, 1212, 1214, 1216, 1218, 1220, 1222, 1224, 1226, 1228, 1230, 1232, 1234, 1236, 1238, 1240, 1242, 1244, 1246, 1248, 1250, 1252, 1254, 1256, 1258, 1260, 1262, 1264, 1266, 1268, 1270, 1272, 1274, 1276, 1278, 1280, 1282, 1284, 1286, 1288, 1290, 1292, 1294, 1296, 1298, 1300, 1302, 1304, 1306, 1308, 1310, 1312, 1314, 1316, 1318, 1320, 1322, 1324, 1326, 1328, 1330, 1332, 1334, 1336, 1338, 1340, 1342, 1344, 1346, 1348, 1350, 1352, 1354, 1356, 1358, 1360, 1362, 1364, 1366, 1368, 1370, 1372, 1374, 1376, 1378, 1380, 1382, 1384, 1386, 1388, 1390, 1392, 1394, 1396, 1398, 1400, 1402, 1404, 1406, 1408, 1410, 1412, 1414, 1416, 1418, 1420, 1422, 1424, 1426, 1428, 1430, 1432, 1434, 1436, 1438, 1440, 1442, 1444, 1446, 1448, 1450, 1452, 1454, 1456, 1458, 1460, 1462, 1464, 1466, 1468, 1470, 1472, 1474, 1476, 1478, 1480, 1482, 1484, 1486, 1488, 1490, 1492, 1494, 1496, 1498, 1500, 1502, 1504, 1506, 1508, 1510, 1512, 1514, 1516, 1518, 1520, 1522, 1524, 1526, 1528, 1530, 1532, 1534, 1536, 1538, 1540, 1542, 1544, 1546, 1548, 1550, 1552, 1554, 1556, 1558, 1560, 1562, 1564, 1566, 1568, 1570, 1572, 1574, 1576, 1578, 1580, 1582, 1584, 1586, 1588, 1590, 1592, 1594, 1596, 1598, 1600, 1602, 1604, 1606, 1608, 1610, 1612, 1614, 1616, 1618, 1620, 1622, 1624, 1626, 1628, 1630, 1632, 1634, 1636, 1638, 1640, 1642, 1644, 1646, 1648, 1650, 1652, 1654, 1656, 1658, 1660, 1662, 1664, 1666, 1668, 1670, 1672, 1674, 1676, 1678, 1680, 1682, 1684, 1686.1688, 1690, 1692, 1694, 1696, 1698, 1700, 1702, 1704, 1706, 1708, 1710, 1712, 1714, 1716, 1718, 1720, 1722, 1724, 1726, 1728, 1730, 1732, 1734, 1736, 1738, 1740, 1742, 1744, 1746, 1748, 1750, 1752, 1754, 1756, 1758, 1760, 1762, 1764, 1766, 1768, 1770, 1772, 1774, 1776, 1778, 1780, 1782, 1784, 1786, 1788, 1790, 1792, 1794, 1796, 1798, 1800, 1802, 1804, 1806, 1808, 1810, 1812, 1814, 1816, 1818, 1820, 1822, 1824, 1826, 1828, 1830, 1832, 1834, 1836, 1838, 1840, 1842, 1844, 1846, 1848, 1850, 1852, 1854, 1856, 1858, 1860, 1862, 1864, 1866, 1868, 1870, 1872, 1874, 1876, 1878, 1880, 1882, 1884, 1886, 1888, 1890, 1892, 1894, 1896, 1898, 1900, 1902, 1904, 1906, 1908, 1910, 1912, 1914, 1916, 1918, 1920, 1922, 1924, 1926, 1928, 1930, 1932, 1934, 1936, 1938, 1940, 1942, 1944, 1946, 1948, 1950, 1952, 1954, 1956, 1958, 1960, 1962, 1964, 1966, 1968, 1970, 1972, 1974, 1976, 1978, 1980, 1982, 1984, 1986, 1988, 1990, 1992, 1994, 1996, 1998, 2000, 2002, 2004, 2006, 2008, 2010, 2012, 2014, 2016, 2018, 2020, 2022, 2024, 2026, 2028, 2030, 2032, 2034, 2036, 2038, 2040, 2042, 2044, 2046, 2048, 2050, 2052, 2054, 2056, 2058, 2060, 2062, 2064, 2066, 2068, 2070, 2072, 2074, 2076, 2078, 2080, 2082, 2084, 2086, 2088, 2090, 2092, 2094, 2096, 2098, 2100, 2102, 2104, 2106, 2108, 2110, 2112, 2114, 2116, 2118, 2120, 2122, 2124, 2126, 2128, 2130, 2132, 2134, 2136, 2138, 2140, 2142, 2144, 2146, 2148, 2150, 2152, 2154, 2156, 2158, 2160, 2162, 2164, 2166, 2168, 2170, 2172, 2174, 2176, 2178, 2180, 2182, 2184, 2186, 2188, 2190, 2192, 2194, 2196, 2198, 2200, 2202, 2204, 2206, 2208, 2210, 2212, 2214, 2216, 2218, 2220, 2222, 2224, 2226, 2228, 2230, 2232, 2234, 2236, 2238, 2240, 2242, 2244, 2246, 2248, 2250, 2252, 2254, 2256, 2258, 2260, 2262, 2264, 2266, 2268, 2270, 2272, 2274, 2276, 2278, 2280, 2282, 2284, 2286, 2288, 2290, 2292, 2294, 2296, 2298, 2300, 2302, 2304, 2306, 2308, 2310, 2312, 2314, 2316, 2318, 2320, 2322, 2324, 2326, 2328, 2330, 2332, 2334, 2336, 2338, 2340, 2342, 2344, 2346, 2348, 2350, 2352, 2354, 2356, 2358, 2360, 2362, 2364, 2366, 2368, 2370, 2372, 2374, 2376, 2378, 2380, 2382, 2384, 2386, 2388, 2390, 2392, 2394, 2396, 2398, 2400, 2402, 2404, 2406, 2408, 2410, 2412, 2414, 2416, 2418, 2420, 2422, 2424, 2426, 2428, 2430, 2432, 2434, 2436, 2438, 2440, 2442, 2444, 2446, 2448, 2450, 2452, 2454, 2456, 2458, 2460, 2462, 2464, 2466, 2468, 2470, 2472, 2474, 2476, 2478, 2480, 2482, 2484, 2486, 2488, 2490, 2492, 2494, 2496, 2498, 2500, 2502, 2504, 2506, 2508, 2510, 2512, 2514, 2516, 2518, 2520, 2522, 2524, 2526, 2528, 2530, 2532, 2534, 2536, 2538, 2540, 2542, 2544, 2546, 2548, 2550, 2552, 2554, 2556, 2558, 2560, 2562, 2564, 2566, 2568, 2570, 2572, 2574, 2576, 2578, 2580, 2582, 2584, 2586, 2588, 2590, 2592, 2594, 2596, 2598, 2600, 2602, 2604, 2606, 2608, 2610, 2612, 2614, 2616, 2618, 2620, 2622, 2624, 2626, 2628, 2630, 2632, 2634, 2636, 2638, 2640, 2642, 2644, 2646, 2648, 2650, 2652, 2654, 2656, 2658, 2660, 2662, 2664, 2666, 2668, 2670, 2672, 2674, 2676, 2678, 2680, 2682, 2684, 2686, 2688, 2690, 2692, 2694, 2696, 2698, 2700, 2702, 2704, 2706, 2708, 2710, 2712, 2714, 2716, 2718, 2720, 2722, 2724, 2726, 2728, 2730, 2732, 2734, 2736, 2738, 2740, 2742, 2744, 2746, 2748, 2750, 2752, 2754, 2756, 2758, 2760, 2762, 2764, 2766, 2768, 2770, 2772, 2774, 2776, 2778, 2780, 2782, 2784, 2786, 2788, 2790, 2792, 2794, 2796, 2798, 2800, 2802, 2804, 2806, 2808, 2810, 2812, 2814, 2816, 2818, 2820, 2822, 2824, 2826, 2828, 2830, 2832, 2834, 2836, 2838, 2840, 2842, 2844, 2846, 2848, 2850, 2852, 2854, 2856, 2858, 2860, 2862, 2864, 2866, 2868, 2870, 2872, 2874, 2876, 2878, 2880, 2882, 2884, 2886, 2888, 2890, 2892, 2894, 2896, 2898, 2900, 2902, 2904, 2906, 2908, 2910, 2912, 2914, 2916, 2918, 2920, 2922, 2924, 2926, 2928, 2930, 2932, 2934, 2936, 2938, 2940, 2942, 2944, 2946, 2948, 2950, 2952, 2954, 2956, 2958, 2960, 2962, 2964, 2966, 2968, 2970, 2972, 2974, 2976, 2978, 2980, 2982, 2984, 2986, 2988, 2990, 2992, 2994, 2996, 2998, 3000, 3002, 3004, 3006, 3008, 3010, 3012, 3014, 3016, 3018 & 3020, as disclosed in WO99/57280 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g.60%, 70%, 80%, 90%, 95%, 99% or more) to one ofthese SEQ IDs);
• The protein disclosed in Figure 4 or Figure 13 ofW097/28273;
• A protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1-8 disclosed in WO96/29412 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
• A protein comprising an amino acid sequence selected from the group consisting of SEQ IDs 1-23 disclosed in WO95/03413 (or a protein comprising an immunogenic fragment of one or more of these SEQ IDs, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to one of these SEQ IDs);
• A protein comprising an amino acid sequence consisting of SEQ ID 2 disclosed in WO99/31 132 (or a protein comprising an immunogenic fragment of SEQ ID 2, or a protein comprising a sequence having sequence identity (preferably greater than 50% e.g. 60%, 70%, 80%, 90%, 95%, 99% or more) to SEQ ID 2);
• A polysaccharide antigen against Neisseria meningitidis serogroup A;
• A polysaccharide antigen against Neisseria meningitidis serogroup C, such as that described in Costantino et al. (1992) Vaccine 10:691 -698;
• A polysaccharide antigen against Neisseria meningitidis serogroup Y;
• A polysaccharide antigen against Neisseria meningitidis serogroup W;
• A polysaccharide antigen against Haemophilus influenzae;
• A polysaccharide antigen against pneumococcus; • A protective antigen against diphtheria, consisting of a diphtheria toxoid, such as the
CRM197 mutant [e.g. Del Guidice et al. (1998) Molecular Aspects of Medicine 19: 1 -70].
• A protective antigen against tetanus, consisting of a tetanus toxoid [e.g. Wassilak & Orenstein, Chapter 4 of Vaccines (eds. Plotkin & Mortimer), 1988]
• A protective antigen against whooping cough, comprising pertussis holotoxin (PT) and filamentous haemagglutinin (FHA); optionally further comprising pertactin and/or agglutinogens 2 and 3 [e.g. Gustafsson et al. (1996) N. Engl. J. Med. 334:349-355; Rappuoli et al (1991) T1BTECH 9:232-238].
• A protective antigen against H. pylori, comprising one or more of CagA (e.g. WO93/18150), VacA (e.g. WO93/18150), NAP (e.g. WO99/53310), HopX (e.g. WO98/04702), HopY (e.g. WO98/04702), urease.
• A protective antigen against hepatitis B virus, consisting of a HBV surface antigen and/or a HBV core antigen. Where the composition comprises an antigen against diphtheria, it preferably also comprises antigens against tetanus and polio. Where the composition comprises an antigen against tetanus, it preferably also comprises antigens against diphtheria and polio. Where the composition comprises an antigen against polio, it preferably also comprises antigens against diphtheria and tetanus.
Pertussis toxin is a toxic protein and, when present in the composition, it is preferably detoxified. Detoxification may be by chemical and/or genetic means. A preferred detoxified mutant is the 9K/129G double mutant [e.g. Rappuoli (1997) Nature Medicine 3:374-376].
Where the composition includes a protein that exists in different nascent and mature forms, the mature form of the protein is preferably used. For example, where NspA is included, (WO96/29412; see also Martin et al. (1997) J. Exp. Med Xj_5 1 173-1183) the mature form of the protein lacking the signal peptide is preferably used.
Where the composition includes a polysaccharide antigen, the polysaccharide is preferably conjugated to a carrier protein.
The proteins of the invention present in the compositions preferably interact synergistically with at least one of the protective antigens in the composition.
Therapy, prophylaxis, diagnosis
The invention also provides the compositions of the invention for use as medicaments (preferably as vaccines) or as diagnostic reagents. It also provides the use of a composition according to the invention in the manufacture of: (i) a medicament for treating or preventing infection due to Neisserial bacteria; (ii) a diagnostic reagent for detecting the presence of Neisserial bacteria or of antibodies raised against Neisserial bacteria; and/or (iii) a reagent which can raise antibodies against Neisserial bacteria. Said Neisserial bacteria may be any species or strain (such as N. gonorrhoeae) but are preferably N. meningitidis, especially serogroup B.
The invention also provides a method of treating a patient, comprising administering to the patient a therapeutically effective amount of nucleic acid, protein, and/or antibody according to the invention. The method is preferably immunisation.
Vaccines according to the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i.e. to treat disease after infection).
Processes According to further aspects, the invention provides various processes. A process for producing proteins of the invention is provided, comprising the step of culturing a host cell according to the invention under conditions which induce protein expression.
A process for producing protein or nucleic acid of the invention is provided, wherein the protein or nucleic acid is synthesised in part or in whole using chemical means.
A process for detecting polynucleotides of the invention is provided, comprising the steps of: (a) contacting a nucleic probe according to the invention with a biological sample under hybridizing conditions to form duplexes; and (b) detecting said duplexes.
A process for detecting proteins of the invention is provided, comprising the steps of: (a) contacting an antibody according to the invention with a biological sample under conditions suitable for the formation of an antibody-antigen complexes; and (b) detecting said complexes.
MODES FOR CARRYING OUT THE INVENTION
A summary of standard techniques and procedures which may be employed in order to perform the invention (e.g. to utilise the disclosed sequences for vaccination or diagnostic puφoses) follows. This summary is not a limitation on the invention but, rather, gives examples that may be used, but are not required.
General
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature e.g. Sambrook Molecular Cloning; A Laboratory Manual, Second Edition (1989); DNA Cloning, Volumes I and ii (D.N Glover ed. 1985); Oligonucleotide Synthesis (M.J. Gait ed, 1984); Nucleic Acid Hybridization (B.D. Hames & S.J. Higgins eds. 1984); Transcription and Translation (B.D. Hames & S.J. Higgins eds. 1984); Animal Cell Culture (R.I. Freshney ed. 1986); Immobilized Cells and Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide to Molecular Cloning (1984); the Methods in Enzymology series (Academic Press, Inc.), especially volumes 154 & 155; Gene Transfer Vectors for Mammalian Cells (J.H. Miller and M.P. Calos eds. 1987, Cold Spring Harbor Laboratory); Mayer and Walker, eds. (1987), Immunochemical Methods in Cell and Molecular Biology (Academic Press, London); Scopes, (1987) Protein Purification: Principles and Practice, Second Edition (Springer- Verlag, N.Y.), and Handbook of Experimental Immunology, Volumes I-IV (D.M. Weir and C. C. Blackwell eds 1986).
Standard abbreviations for nucleotides and amino acids are used in this specification. A composition containing X is "substantially free of Y when at least 85% by weight of the total X+Y in the composition is X. Preferably, X comprises at least about 90% by weight of the total of X+Y in the composition, more preferably at least about 95% or even 99% by weight.
The term "comprising" means "including" as well as "consisting" e g a composition "comprising" X may consist exclusively of X or may include something additional to X, such as X+Y.
The term "heterologous" refers to two biological components that are not found together in nature. The components may be host cells, genes, or regulatory regions, such as promoters. Although the heterologous components are not found together in nature, they can function together, as when a promoter heterologous to a gene is operably linked to the gene Another example is where a Neisserial sequence is heterologous to a mouse host cell. A further examples would be two epitopes from the same or different proteins which have been assembled in a single protein in an arrangement not found in nature.
An "origin of replication" is a polynucleotide sequence that initiates and regulates replication of polynucleotides, such as an expression vector. The origin of replication behaves as an autonomous unit of polynucleotide replication within a cell, capable of replication under its own control. An origin of replication may be needed for a vector to replicate in a particular host cell. With certain origins of replication, an expression vector can be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells. A "mutant" sequence is defined as DNA, RNA or amino acid sequence differing from but having sequence identity with the native or disclosed sequence. Depending on the particular sequence, the degree of sequence identity between the native or disclosed sequence and the mutant sequence is preferably greater than 50% (e g. 60%, 70%, 80%, 90%, 95%, 99% or more, calculated using the Smith-Waterman algorithm as descnbed above) As used herein, an "allelic variant" of a nucleic acid molecule, or region, for which nucleic acid sequence is provided herein is a nucleic acid molecule, or region, that occurs essentially at the same locus in the genome of another or second isolate, and that, due to natural variation caused by, for example, mutation or recombination, has a similar but not identical nucleic acid sequence. A coding region allelic variant typically encodes a protein having similar activity to that of the protein encoded by the gene to which it is being compared. An allelic variant can also comprise an alteration in the 5' or 3' untranslated regions of the gene, such as in regulatory control regions (e.g. see US patent 5,753,235).
Expression systems
The Neisseπal nucleotide sequences can be expressed in a variety of different expression systems; for example those used with mammalian cells, baculov_ruses, plants, bacteria, and yeast. l Mammalian Systems
Mammalian expression systems are known in the art. A mammalian promoter is any DNA sequence capable of binding mammalian RNA polymerase and initiating the downstream (3') transcription of a coding sequence (e.g. structural gene) into mRNA. A promoter will have a transcription initiating region, which is usually placed proximal to the 5' end of the coding sequence, and a TATA box, usually located 25-30 base pairs (bp) upstream of the transcription initiation site. The TATA box is thought to direct RNA polymerase II to begin RNA synthesis at the correct site. A mammalian promoter will also contain an upstream promoter element, usually located within 100 to 200 bp upstream of the TATA box. An upstream promoter element determines the rate at which transcription is initiated and can act in either orientation [Sambrook et al. (1989) "Expression of Cloned Genes in Mammalian Cells." In Molecular Cloning: A Laboratory Manual, 2nd ed.].
Mammalian viral genes are often highly expressed and have a broad host range; therefore sequences encoding mammalian viral genes provide particularly useful promoter sequences. Examples include the SV40 early promoter, mouse mammary tumor virus LTR promoter, adenovirus major late promoter (Ad MLP), and heφes simplex virus promoter. In addition, sequences derived from non- viral genes, such as the murine metallotheionein gene, also provide useful promoter sequences. Expression may be either constitutive or regulated (inducible), depending on the promoter can be induced with glucocorticoid in hormone-responsive cells.
The presence of an enhancer element (enhancer), combined with the promoter elements described above, will usually increase expression levels. An enhancer is a regulatory DNA sequence that can stimulate transcription up to 1000-fold when linked to homologous or heterologous promoters, with synthesis beginning at the normal RNA start site. Enhancers are also active when they are placed upstream or downstream from the transcription initiation site, in either normal or flipped orientation, or at a distance of more than 1000 nucleotides from the promoter [Maniatis et al. (1987) Science 236:1237; Alberts et al. (1989) Molecular Biology of the Cell, 2nd ed.]. Enhancer elements derived from viruses may be particularly useful, because they usually have a broader host range. Examples include the SV40 early gene enhancer [Dijkema et al (1985) EMBO J. 4:761] and the enhancer/promoters derived from the long terminal repeat (LTR) of the Rous Sarcoma Virus [Gorman et al. (1982b) Proc. Natl. Acad. Sci. 79:6777] and from human cytomegalovirus [Boshart et al. (1985) Cell 47:521]. Additionally, some enhancers are regulatable and become active only in the presence of an inducer, such as a hormone or metal ion [Sassone-Corsi and Borelli (1986) Trends Genet. 2:2X5; Maniatis et al. (1987) Science 236:1237]. A DNA molecule may be expressed intracellularly in mammalian cells. A promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus of the recombinant protein will always be a methionine, which is encoded by the ATG start codon. If desired, the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide. Alternatively, foreign proteins can also be secreted from the cell into the growth media by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provides for secretion of the foreign protein in mammalian cells. Preferably, there are processing sites encoded between the leader fragment and the foreign gene that can be cleaved either in vivo or in vitro. The leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell. The adenovirus triparite leader is an example of a leader sequence that provides for secretion of a foreign protein in mammalian cells.
Usually, transcription termination and polyadenylation sequences recognized by mammalian cells are regulatory regions located 3' to the translation stop codon and thus, together with the promoter elements, flank the coding sequence. The 3' terminus of the mature mRNA is formed by site- specific post-transcriptional cleavage and polyadenylation [Birnstiel et al. (1985) Cell 41:349; Proudfoot and Whitelaw (1988) "Termination and 3' end processing of eukaryotic RNA. In Transcription and splicing (ed. B.D. Hames and D.M. Glover); Proudfoot (1989) Trends Biochem. Sci. 74:105]. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Examples of transcription terminater/polyadenylation signals include those derived from SV40 [Sambrook et al (1989) "Expression of cloned genes in cultured mammalian cells." In Molecular Cloning: A Laboratory Manual].
Usually, the above described components, comprising a promoter, polyadenylation signal, and transcription termination sequence are put together into expression constructs. Enhancers, introns with functional splice donor and acceptor sites, and leader sequences may also be included in an expression construct, if desired. Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g. plasmids) capable of stable maintenance in a host, such as mammalian cells or bacteria. Mammalian replication systems include those derived from animal viruses, which require trans-acting factors to replicate. For example, plasmids containing the replication systems of papova viruses, such as SV40 [Gluzman (1981) Cell 23:175] or polyomavirus, replicate to extremely high copy number in the presence of the appropriate viral T antigen. Additional examples of mammalian replicons include those derived from bovine papillomavirus and Epstein-Barr virus. Additionally, the replicon may have two replicaton systems, thus allowing it to be maintained, for example, in mammalian cells for expression and in a prokaryotic host for cloning and amplification. Examples of such mammalian-bacteria shuttle vectors include pMT2 [Kaufman et al. (1989) Mol. Cell. Biol. 9:946] and pHEBO [Shimizu et al.
(1986) Mol. Cell. Biol 6:1074].
The transformation procedure used depends upon the host to be transformed. Methods for introduction of heterologous polynucleotides into mammalian cells are known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
Mammalian cell lines available as hosts for expression are known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to, Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g. Hep G2), and a number of other cell lines. ii. Baculovirus Systems
The polynucleotide encoding the protein can also be inserted into a suitable insect expression vector, and is operably linked to the control elements within that vector. Vector construction employs techniques w hich are known in the art. Generally, the components of the expression system include a transfer vector, usually a bacterial plasm id, 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 hom ologous to the baculovirus-specific fragment in the transfer vector (this allows for the hom ologous recom bination of the heterologous gene in to the baculovirus genom e); and appropriate insect host cells and growth m edia. After inserting the DNA sequence encoding the protein into the transfer vector, the vector and the wild type viral genome are transfected into an insect host cell where the vector and viral genome are allowed to recombine. The packaged recombinant virus is expressed and recombinant plaques are identified and purified. Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Invitrogen, San Diego CA ("MaxBac" kit). These techniques are generally known to those skilled in the art and fully described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987) (hereinafter "Summers and Smith").
Prior to inserting the DN A sequence encoding the protein into the baculovirus genom e, the above described components, comprising a prom oter, leader (if desired), coding sequence of interest, and transcription term ination sequence, are usually assembled into an interm ediate transplacement construct (transfer vector). This construct m ay contain a single gene and operably linked regulatory elements; multiple genes, each with its owned set of operably linked regulatory elements; or multiple genes, regulated by the same set of regulatory elements. Intermediate transplacem ent constructs are often m aintained in a replicon, such as an extrachromosomal element (e.g. plasm ids) capable of stable m aintenance in a host, such as a bacterium . The replicon w ill have a replication system , thus allowing it to be m aintained in a suitable host for cloning and amplification.
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) 77:31.
The plasmid usually also contains the polyhedrin polyadenylation signal (Miller et al. (1988) Ann. Rev. Microbiol, 42:177) and a prokaryotic ampicillin-resistance (amp) gene and origin of replication for selection and propagation in E. 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 usually includes an RNA polymerase binding site and a transcription initiation site. A baculovirus transfer vector may also have a second domain called an enhancer, which, if present, is usually distal to the structural gene. Expression may be either regulated or constitutive. Structural genes, abundantly transcribed at late times in a viral infection cycle, provide particularly useful promoter sequences. Examples include sequences derived from the gene encoding the viral polyhedron protein, Friesen et al., (1986) "The Regulation of Baculovirus Gene Expression," in: The Molecular Biology of Baculoviruses (ed. Walter Doerfler); EPO Publ. Nos. 127 839 and 155 476; and the gene encoding the plO protein, Vlak et al., (1988), J. Gen. Virol. 69:765. DNA encoding suitable signal sequences can be derived from genes for secreted insect or baculovirus proteins, such as the baculovirus polyhedrin gene (Carbonell et al. (1988) Gene, 73:409). Alternatively, since the signals for mammalian cell posttranslational modifications (such as signal peptide cleavage, proteolytic cleavage, and phosphorylation) appear to be recognized by insect cells, and the signals required for secretion and nuclear accumulation also appear to be conserved between the invertebrate cells and vertebrate cells, leaders of non-insect origin, such as those derived from genes encoding human α-interferon, Maeda et al., (1985), Nature 315:592; human gastrin-releasing peptide, Lebacq-Verheyden et al., (1988), Molec. Cell. Biol. S:3129; human IL-2, Smith et al., (1985) Proc. Nat'l Acad. Sci. USA, §2:8404; mouse IL-3, (Miyajima et al., (1987) Gene 58:273; and human glucocerebrosidase, Martin et al. (1988) DNA, 7:99, can also be used to provide for secretion in insects.
A recombinant polypeptide or polyprotein may be expressed intracellularly or, if it is expressed with the proper regulatory sequences, it can be secreted. Good intracellular expression of nonfused foreign proteins usually requires heterologous genes that ideally have a short leader sequence containing suitable translation initiation signals preceding an ATG start signal. If desired, methionine at the N-terminus may be cleaved from the mature protein by in vitro incubation with cyanogen bromide.
Alternatively, recombinant polyproteins or proteins which are not naturally secreted can be secreted from the insect cell by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provides for secretion of the foreign protein in insects. The leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the translocation of the protein into the endoplasmic reticulum.
After insertion of the DNA sequence and/or the gene encoding the expression product precursor of the protein, an insect cell host is co-transformed with the heterologous DNA of the transfer vector and the genomic DNA of wild type baculovirus — usually by co-transfection. The promoter and transcription termination sequence of the construct will usually comprise a 2-5kb section of the baculovirus genome. Methods for introducing heterologous DNA into the desired site in the baculovirus virus are known in the art. (See Summers and Smith supra; Ju et al. (1987); Smith et al., Mol. Cell. Biol. (1983) 3:2156; and Luckow and Summers (1989)). For example, the insertion can be into a gene such as the polyhedrin gene, by homologous double crossover recombination; insertion can also be into a restriction enzyme site engineered into the desired baculovirus gene. Miller et al., (1989), Bioessays 4:91. The DNA sequence, when cloned in place of the polyhedrin gene in the expression vector, is flanked both 5' and 3' by polyhedrin-specific sequences and is positioned downstream of the polyhedrin promoter.
The newly formed baculovirus expression vector is subsequently packaged into an infectious recombinant baculovirus. Homologous recombination occurs at low frequency (between about 1% and about 5%); thus, the majority of the virus produced after cotransfection is still wild-type virus. Therefore, a method is necessary to identify recombinant viruses. An advantage of the expression system is a visual screen allowing recombinant viruses to be distinguished. The polyhedrin protein, which is produced by the native virus, is produced at very high levels in the nuclei of infected cells at late times after viral infection. Accumulated polyhedrin protein forms occlusion bodies that also contain embedded particles. These occlusion bodies, up to 15 μm in size, are highly retractile, giving them a bright shiny appearance that is readily visualized under the light microscope. Cells infected with recombinant viruses lack occlusion bodies. To distinguish recombinant virus from wild-type virus, the transfection supernatant is plaqued onto a monolayer of insect cells by techniques known to those skilled in the art. Namely, the plaques are screened under the light microscope for the presence (indicative of wild-type virus) or absence (indicative of recombinant virus) of occlusion bodies. "Current Protocols in Microbiology" Vol. 2 (Ausubel et al. eds) at 16.8 (Supp. 10, 1990); Summers and Smith, supra; Miller et al. (1989).
Recombinant baculovirus expression vectors have been developed for infection into several insect cells. For example, recombinant baculoviruses have been developed for, inter alia: Aedes aegypti , Autographa californica, Bombyx mori, Drosophila melanogaster, Spodoptera frugiperda, and Trichoplusia ni (WO 89/046699; Carbonell et al., (1985) J. Virol. 5(5:153; Wright (1986) Nature 327:718; Smith et al., (1983) Mol. Cell. Biol. 3:2156; and see generally, Fraser, et al. (1989) In Vitro Cell. Dev. Biol. 25:225).
Cells and cell culture media are commercially available for both direct and fusion expression of heterologous polypeptides in a baculovirus/expression system; cell culture technology is generally known to those skilled in the art. See, e.g. Summers and Smith supra. The modified insect cells may then be grown in an appropriate nutrient medium, which allows for stable maintenance of the plasmid(s) present in the modified insect host. Where the expression product gene is under inducible control, the host may be grown to high density, and expression induced. Alternatively, where expression is constitutive, the product will be continuously expressed into the medium and the nutrient medium must be continuously circulated, while removing the product of interest and augmenting depleted nutrients. The product may be purified by such techniques as chromatography, e.g. HPLC, affinity chromatography, ion exchange chromatography, etc.; electrophoresis; density gradient centrifugation; solvent extraction, or the like. As appropriate, the product may be further purified, as required, so as to remove substantially any insect proteins which are also secreted in the medium or result from lysis of insect cells, so as to provide a product which is at least substantially free of host debris, e.g. proteins, lipids and polysaccharides.
In order to obtain protein expression, recombinant host cells derived from the transformants are incubated under conditions which allow expression of the recombinant protein encoding sequence. These conditions will vary, dependent upon the host cell selected. However, the conditions are readily ascertainable to those of ordinary skill in the art, based upon what is known in the art. iii. Plant Systems
There are many plant cell culture and whole plant genetic expression systems known in the art. Exemplary plant cellular genetic expression systems include those described in patents, such as: US 5,693,506; US 5,659,122; and US 5,608,143. Additional examples of genetic expression in plant cell culture has been described by Zenk, Phytochemistry 30:3861-3863 (1991). Descriptions of plant protein signal peptides may be found in addition to the references described above in Vaulco be et al., Mol. Gen. Genet. 209:33-40 (1987); Chandler et al., Plant Molecular Biology 3:407-418 (1984); Rogers, J. Biol. Chem. 260:3731-3738 (1985); Rothstein et al., Gene 55:353-356 (1987); Whittier et al., Nucleic Acids Research 15:2515-2535 (1987); Wirsel et al., Molecular Microbiology 3:3-14 (1989); Yu et al., Gene 122:247-253 (1992). A description of the regulation of plant gene expression by the phytohormone, gibberellic acid and secreted enzymes induced by gibberellic acid can be found in R.L. Jones and J. MacMillin, Gibberellins: in: Advanced Plant Physiology,. Malcolm B. Wilkins, ed., 1984 Pitman Publishing Limited, London, pp. 21-52. References that describe other metabolically-regulated genes: Sheen, Plant Cell, 2:1027- 1038(1990); Maas et al., EMBO J. 9:3447-3452 (1990); Benkel and Hickey, Proc. Nail. Acad. Sci. 84:1337-1339 (1987)
Typically, using techniques known in the art, a desired polynucleotide sequence is inserted into an expression cassette comprising genetic regulatory elements designed for operation in plants. The expression cassette is inserted into a desired expression vector with companion sequences upstream and downstream from the expression cassette suitable for expression in a plant host. The companion sequences will be of plasmid or viral origin and provide necessary characteristics to the vector to permit the vectors to move DNA from an original cloning host, such as bacteria, to the desired plant host. The basic bacterial/plant vector construct will preferably provide a broad host range prokaryote replication origin; a prokaryote selectable marker; and, for Agrobacterium transformations, T DNA sequences for Agrobacterium-mediated transfer to plant chromosomes. Where the heterologous gene is not readily amenable to detection, the construct will preferably also have a selectable marker gene suitable for determining if a plant cell has been transformed. A general review of suitable markers, for example for the members of the grass family, is found in Wilmink and Dons, 1993, Plant Mol. Biol. Reptr, 11 (2): 165-185.
Sequences suitable for permitting integration of the heterologous sequence into the plant genome are also recommended. These might include transposon sequences and the like for homologous recombination as well as Ti sequences which permit random insertion of a heterologous expression cassette into a plant genome. Suitable prokaryote selectable markers include resistance toward antibiotics such as ampicillin or tetracycline. Other DNA sequences encoding additional functions may also be present in the vector, as is known in the art.
The nucleic acid molecules of the subject invention may be included into an expression cassette for expression of the protein(s) of interest. Usually, there will be only one expression cassette, although two or more are feasible. The recombinant expression cassette will contain in addition to the heterologous protein encoding sequence the following elements, a promoter region, plant 5' untranslated sequences, initiation codon depending upon whether or not the structural gene comes equipped with one, and a transcription and translation termination sequence. Unique restriction enzyme sites at the 5' and 3' ends of the cassette allow for easy insertion into a pre-existing vector.
A heterologous coding sequence may be for any protein relating to the present invention. The sequence encoding the protein of interest will encode a signal peptide which allows processing and translocation of the protein, as appropriate, and will usually lack any sequence which might result in the binding of the desired protein of the invention to a membrane. Since, for the most part, the transcriptional initiation region will be for a gene which is expressed and translocated during germination, by employing the signal peptide which provides for translocation, one may also provide for translocation of the protein of interest. In this way, the protein(s) of interest will be translocated from the cells in which they are expressed and may be efficiently harvested. Typically secretion in seeds are across the aleurone or scutellar epithelium layer into the endosperm of the seed. While it is not required that the protein be secreted from the cells in which the protein is produced, this facilitates the isolation and purification of the recombinant protein.
Since the ultimate expression of the desired gene product will be in a eucaryotic cell it is desirable to determine whether any portion of the cloned gene contains sequences which will be processed out as introns by the host's splicosome machinery. If so, site-directed mutagenesis of the "intron" region may be conducted to prevent losing a portion of the genetic message as a false intron code, Reed and Maniatis, Cell 41 :95-105, 1985.
The vector can be microinjected directly into plant cells by use of micropipettes to mechanically transfer the recombinant DNA. Crossway, Mol. Gen. Genet, 202:179-185, 1985. The genetic material may also be transferred into the plant cell by using polyethylene glycol, Krens, et al., Nature, 296, 72-74, 1982. Another method of introduction of nucleic acid segments is high velocity ballistic penetration by small particles with the nucleic acid either within the matrix of small beads or particles, or on the surface, Klein, et al., Nature, 327, 70-73, 1987 and Knudsen and Muller, 1991, Planta, 185:330-336 teaching particle bombardment of barley endosperm to create transgenic barley. Yet another method of introduction would be fusion of protoplasts with other entities, either minicells, cells, lysosomes or other fusible lipid-surfaced bodies, Fraley, et al., Proc. Natl. Acad. Sci. USA, 79, 1859-1863, 1982.
The vector may also be introduced into the plant cells by electroporation. (Fromm et al., Proc. Natl Acad. Sci. USA 82:5824, 1985). In this technique, plant protoplasts are electroporated in the presence of plasmids containing the gene construct. Electrical impulses of high field strength reversibly permeabilize biomembranes allowing the introduction of the plasmids. Electroporated plant protoplasts reform the cell wall, divide, and form plant callus.
All plants from which protoplasts can be isolated and cultured to give whole regenerated plants can be transformed by the present invention so that whole plants are recovered which contain the transferred gene. It is known that practically all plants can be regenerated from cultured cells or tissues, including but not limited to all major species of sugarcane, sugar beet, cotton, fruit and other trees, legumes and vegetables. Some suitable plants include, for example, species from the genera Fragaria, Lotus, Medicago, Onobrychis, Trifolium, Trigonella, Vigna, Citrus, Linum, Geranium, Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Datura, Hyoscyamus, Lycopersion, Nicotiana, Solanum, Petunia, Digitalis, Majorana, Cichorium, Helianthus, Lactuca, Bromus, Asparagus, Antirrhinum, Hererocallis, Nemesia, Pelargonium, Panicum, Pennisetum, Ranunculus, Senecio, Salpiglossis, Cucumis, Browaalia, Glycine, Lolium, Zea, Triticum, Sorghum, and Datura. Means for regeneration vary from species to species of plants, but generally a suspension of transformed protoplasts containing copies of the heterologous gene is first provided. Callus tissue is formed and shoots may be induced from callus and subsequently rooted. Alternatively, embryo formation can be induced from the protoplast suspension. These embryos germinate as natural embryos to form plants. The culture media will generally contain various amino acids and hormones, such as auxin and cytokinins. It is also advantageous to add glutamic acid and proline to the medium, especially for such species as corn and alfalfa. Shoots and roots normally develop simultaneously. Efficient regeneration will depend on the medium, on the genotype, and on the history of the culture. If these three variables are controlled, then regeneration is fully reproducible and repeatable. In some plant cell culture systems, the desired protein of the invention may be excreted or alternatively, the protein may be extracted from the whole plant. W ere the desired protein of the invention is secreted into the medium, it may be collected. Alternatively, the embryos and embryoless-half seeds or other plant tissue may be mechanically disrupted to release any secreted protein between cells and tissues. The mixture may be suspended in a buffer solution to retrieve soluble proteins. Conventional protein isolation and purification methods will be then used to purify the recombinant protein. Parameters of time, temperature pH, oxygen, and volumes will be adjusted through routine methods to optimize expression and recovery of heterologous protein. iv. Bacterial Systems
Bacterial expression techniques are known in the art. A bacterial promoter is any DNA sequence capable of binding bacterial RNA polymerase and initiating the downstream (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 usually includes an RNA polymerase binding site and a transcription initiation site. A bacterial promoter may also have a second domain called an operator, that may overlap an adjacent RNA polymerase binding site at which RNA synthesis begins. The operator permits negative regulated (inducible) transcription, as a gene repressor protein may bind the operator and thereby inhibit transcription of a specific gene. Constitutive expression may occur in the absence of negative regulatory elements, such as the operator. In addition, positive regulation may be achieved by a gene activator protein binding sequence, which, if present is usually proximal (5') to the RNA polymerase binding sequence. An example of a gene activator protein is the catabolite activator protein (CAP), which helps initiate transcription of the lac operon in Escherichia coli (E. coli) [Raibaud et al. (1984) Annu. Rev. Genet. 75:173]. Regulated expression may therefore be either positive or negative, thereby either enhancing or reducing transcription.
Sequences encoding metabolic pathway enzymes provide particularly useful promoter sequences. Examples include promoter sequences derived from sugar metabolizing enzymes, such as galactose, lactose (lac) [Chang et al. (1977) Nature 798:1056], and maltose. Additional examples include promoter sequences derived from biosynthetic enzymes such as tryptophan (trp) [Goeddel et al. (1980) Nuc. Acids Res. 8:4057; Yelverton et al. (1981) Nucl. Acids Res. 9:731 ; US patent 4,738,921; EP-A-0036776 and EP-A-0121775]. The g-laotamase (bid) promoter system [Weissmann (1981) "The cloning of interferon and other mistakes." In Interferon 3 (ed. I. Gresser)], bacteriophage lambda PL [Shimatake et al. (1981) Nature 292:128] and T5 [US patent 4,689,406] promoter systems also provide useful promoter sequences. In addition, synthetic promoters which do not occur in nature also function as bacterial promoters. For example, transcription activation sequences of one bacterial or bacteriophage promoter may be joined with the operon sequences of another bacterial or bacteriophage promoter, creating a synthetic hybrid promoter [US patent 4,551,433]. For example, the tac promoter is a hybrid trp-lac promoter comprised of both trp promoter and lac operon sequences that is regulated by the lac repressor [Amann et al. (1983) Gene 25:167; de Boer et al. (1983) Proc. Natl. Acad. Sci. 80:21]. Furthermore, a bacterial promoter can include naturally occurring promoters of non-bacterial origin that have the ability to bind bacterial RNA polymerase and initiate transcription. A naturally occurring promoter of non-bacterial origin can also be coupled with a compatible RNA polymerase to produce high levels of expression of some genes in prokaryotes. The bacteriophage T7 RNA polymerase/promoter system is an example of a coupled promoter system [Studier et al. (1986) J. Mol. Biol. 789:1 13; Tabor et al. (1985) Proc Natl. Acad. Sci. 82: 1074]. In addition, a hybrid promoter can also be comprised of a bacteriophage promoter and an E. coli operator region (EPO- A-0 267 851).
In addition to a functioning promoter sequence, an efficient ribosome binding site is also useful for the expression of foreign genes in prokaryotes. In E. coli, the ribosome binding site is called the Shine-Dalgarno (SD) sequence and includes an initiation codon (ATG) and a sequence 3-9 nucleotides in length located 3-1 1 nucleotides upstream of the initiation codon [Shine et al. (1975) Nature 254:34]. The SD sequence is thought to promote binding of mRNA to the ribosome by the pairing of bases between the SD sequence and the 3' and of E. coli 16S rRNA [Steitz et al. (1979) "Genetic signals and nucleotide sequences in messenger RNA." In Biological Regulation and Development: Gene Expression (ed. R.F. Goldberger)]. To express eukaryotic genes and prokaryotic genes with weak ribosome-binding site [Sambrook et al. (1989) "Expression of cloned genes in Escherichia coli." In Molecular Cloning: A Laboratory Manual].
A DNA molecule may be expressed intracellularly. A promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus will always be a methionine, which is encoded by the ATG start codon. If desired, methionine at the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide or by either in vivo on in vitro incubation with a bacterial methionine N-terminal peptidase (EPO-A-0219 237).
Fusion proteins provide an alternative to direct expression. Usually, a DNA sequence encoding the
N-terminal portion of an endogenous bacterial protein, or other stable protein, is fused to the 5' end of heterologous coding sequences. Upon expression, this construct will provide a fusion of the two amino acid sequences. For example, the bacteriophage lambda cell gene can be linked at the 5' terminus of a foreign gene and expressed in bacteria. The resulting fusion protein preferably retains a site for a processing enzyme (factor Xa) to cleave the bacteriophage protein from the foreign gene
[Nagai et al. (1984) Nature 309:810]. Fusion proteins can also be made with sequences from the lacZ [Jia et al (1987) Gene 60:197], trpE [Allen et al. (1987) J. Biotechnol. 5:93; Makoff et al.
(1989) J. Gen. Microbiol. 735:1 1], and Chey [EP-A-0 324 647] genes. The DNA sequence at the junction of the two amino acid sequences may or may not encode a cleavable site. Another example is a ubiquitin fusion protein. Such a fusion protein is made with the ubiquitin region that preferably retains a site for a processing enzyme (e.g. ubiquitin specific processing-protease) to cleave the ubiquitin from the foreign protein. Through this method, native foreign protein can be isolated
[Miller et al. (1989) Bio/Technology 7:698]. Alternatively, foreign proteins can also be secreted from the cell by creating chimeric DNA molecules that encode a fusion protein comprised of a signal peptide sequence fragment that provides for secretion of the foreign protein in bacteria [US patent 4,336,336]. The signal sequence fragment usually encodes a signal peptide comprised of hydrophobic am ino acids w hich direct the secretion of the protein from the cell. The protein is either secreted into the growth media (gram-positive bacteria) or into the periplasmic space, located between the inner and outer membrane of the cell (gram-negative bacteria). Preferably there are processing sites, w hich can be cleaved either in vivo or in vitro encoded between the signal peptide fragment and the foreign gene.
DNA encoding suitable signal sequences can be derived from genes for secreted bacterial proteins, such as the E. coli outer membrane protein gene (ompA) [Masui et al (1983), in: Experimental Manipulation of Gene Expression; Ghrayeb et al. (1984) EMBO J. 3:2437] and the E. coli alkaline phosphatase signal sequence (phoA) [Oka et al. (1985) Proc. Natl. Acad. Sci. 82:72X2]. As an additional example, the signal sequence of the alpha-amylase gene from various Bacillus strains can be used to secrete heterologous proteins from B. subtilis [Palva et al. (1982) Proc. Natl Acad. Sci. USA 79:5582; EP-A-0 244 042].
Usually, transcription termination sequences recognized by bacteria are regulatory regions located 3' to the translation stop codon, and thus together with the promoter flank the coding sequence. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Transcription termination sequences frequently include DNA sequences of about 50 nucleotides capable of forming stem loop structures that aid in terminating transcription. Examples include transcription termination sequences derived from genes with strong promoters, such as the trp gene in E. coli as well as other biosynthetic genes. Usually, the above described components, comprising a promoter, signal sequence (if desired), coding sequence of interest, and transcription termination sequence, are put together into expression constructs. Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g. plasmids) capable of stable maintenance in a host, such as bacteria. The replicon will have a replication system, thus allowing it to be maintained in a prokaryotic host either for expression or for cloning and amplification. In addition, a replicon may be either a high or low copy number plasmid. A high copy number plasmid will generally have a copy number ranging from about 5 to about 200, and usually about 10 to about 150. A host containing a high copy number plasmid will preferably contain at least about 10, and more preferably at least about 20 plasmids. Either a high or low copy number vector may be selected, depending upon the effect of the vector and the foreign protein on the host.
Alternatively, the expression constructs can be integrated into the bacterial genome with an integrating vector. Integrating vectors usually contain at least one sequence homologous to the bacterial chromosome that allows the vector to integrate. Integrations appear to result from recombinations between homologous DNA in the vector and the bacterial chromosome. For example, integrating vectors constructed with DNA from various Bacillus strains integrate into the Bacillus chromosome (EP-A- 0 127 328). Integrating vectors may also be comprised of bacteriophage or transposon sequences.
Usually, extrachromosomal and integrating expression constructs may contain selectable markers to allow for the selection of bacterial strains that have been transformed. Selectable markers can be expressed in the bacterial host and may include genes which render bacteria resistant to drugs such as ampicillin, chloramphenicol, erythromycin, kanamycin (neomycin), and tetracycline [Davies et al. (1978) Annu. Rev. Microbiol. 32:469]. Selectable markers may also include biosynthetic genes, such as those in the histidine, tryptophan, and leucine biosynthetic pathways.
Alternatively, some of the above described components can be put together in transformation vectors. Transformation vectors are usually comprised of a selectable market that is either maintained in a replicon or developed into an integrating vector, as described above.
Expression and transformation vectors, either extra-chromosomal replicons or integrating vectors, have been developed for transformation into many bacteria. For example, expression vectors have been developed for, inter alia, the following bacteria: Bacillus subtilis [Palva et al (1982) Proc. Natl. Acad. Sci. USA 79:5582; EP-A-0 036 259 and EP-A-0 063 953; WO 84/04541], Escherichia coli [Shimatake et al. (1981) Nature 292:128; A ann et al. (1985) Gene 40:183; Studier et al. (1986) J. Mol. Biol. 789:113; EP-A-0 036 776,EP-A-0 136 829 and EP-A-0 136 907], Streptococcus cremoris [Powell et al. (1988) Appl. Environ. Microbiol. 54:655]; Streptococcus lividans [Powell et al. (1988) Appl. Environ. Microbiol. 54:655], Streptomyces lividans [US patent 4,745,056].
Methods of introducing exogenous DNA into bacterial hosts are well-known in the art, and usually include either the transformation of bacteria treated with CaCl2 or other agents, such as divalent cations and DMSO. DNA can also be introduced into bacterial cells by electroporation.
Transformation procedures usually vary with the bacterial species to be transformed. See e.g.
[Masson et al. (1989) FEMS Microbiol. Lett. 60:273; Palva et al. (1982) Proc. Natl. Acad. Sci. USA
79:5582; EP-A-0 036 259 and EP-A-0 063 953; WO 84/04541, Bacillus], [Miller et al. (1988) Proc.
Natl. Acad. Sci. 85:856; Wang et al. (1990) J. Bacteriol. 772:949, Campylobacter], [Cohen et al. (1973) Proc. Natl. Acad. Sci. 69:2110; Dower et al. (1988) Nucleic Acids Res. 76:6127; Kushner
(1978) "An improved method for transformation of Escherichia coli with ColEl-derived plasmids.
In Genetic Engineering: Proceedings of the International Symposium on Genetic Engineering (eds.
H.W. Boyer and S. Nicosia); Mandel et al (1970) J. Mol. Biol. 53:159; Taketo (1988) Biochim.
Biophys. Acta 949:318; Escherichia], [Chassy et al (1987) FEMS Microbiol. Lett. 44:173 Lactobacillus]; [Fiedler et al. (1988) Anal. Biochem 770:38, Pseudomonas]; [Augustin et al. (1990)
FEMS Microbiol Lett. 66:203, Staphylococcus], [Barany et al. (1980) J. Bacteriol. 744:698;
Harlander (1987) "Transformation of Streptococcus lactis by electroporation, in: Streptococcal
Genetics (ed. J. Ferretti and R. Curtiss III); Perry et al. (1981 ) Infect. Immun. 32: 1295; Powell et al.
(1988) Appl. Environ. Microbiol. 54:655; Somkuti et al. (1987) Proc. 4th Evr. Cong. Biotechnology 7:412, Streptococcus] . v. Yeast Expression
Yeast expression systems are also known to one of ordinary skill in the art. A yeast promoter is any DNA sequence capable of binding yeast RNA polymerase and initiating the downstream (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 usually includes an RNA polymerase binding site (the "TATA Box") and a transcription initiation site. A yeast promoter may also have a second domain called an upstream activator sequence (UAS), which, if present, is usually distal to the structural gene. The UAS permits regulated (inducible) expression. Constitutive expression occurs in the absence of a UAS. Regulated expression may be either positive or negative, thereby either enhancing or reducing transcription.
Yeast is a fermenting organism with an active metabolic pathway, therefore sequences encoding enzymes in the metabolic pathway provide particularly useful promoter sequences. Examples include alcohol dehydrogenase (ADH) (EP-A-0 284 044), enolase, glucokinase, glucose-6- phosphate isomerase, glyceraldehyde-3-phosphate-dehydrogenase (GAP or GAPDH), hexokinase, phosphofructokinase, 3-phosphoglycerate mutase, and pyruvate kinase (PyK) (EPO-A-0 329 203). The yeast PH05 gene, encoding acid phosphatase, also provides useful promoter sequences [Myanohara et al. ( 1983) Proc. Natl. Acad. Sci. USA 80: 1 ] .
In addition, synthetic promoters which do not occur in nature also function as yeast promoters. For example, UAS sequences of one yeast promoter may be joined with the transcription activation region of another yeast promoter, creating a synthetic hybrid promoter. Examples of such hybrid promoters include the ADH regulatory sequence linked to the GAP transcription activation region (US Patent Nos. 4,876,197 and 4,880,734). Other examples of hybrid promoters include promoters which consist of the regulatory sequences of either the ADH2, GAL4, GAL10, OR PH05 genes, combined with the transcriptional activation region of a glycolytic enzyme gene such as GAP or PyK (EP-A-0 164 556). Furthermore, a yeast promoter can include naturally occurring promoters of non-yeast origin that have the ability to bind yeast RNA polymerase and initiate transcription. Examples of such promoters include, ter alia, [Cohen et al. (1980) Proc. Natl. Acad. Sci. USA 77: 1078; Henikoff et al. (1981) Nature 283:835; Hollenberg et al. (1981) Curr. Topics Microbiol. Immunol. 96:1 19; Hollenberg et al. (1979) "The Expression of Bacterial Antibiotic Resistance Genes in the Yeast Saccharomyces cerevisiae," in: Plasmids of Medical, Environmental and Commercial Importance (eds. K.N. Timmis and A. Puhler); Mercerau-Puigalon et al. (1980) Gene 77:163; Panthier et al. (1980) Curr. Genet. 2: 109;].
A DNA molecule may be expressed intracellularly in yeast. A promoter sequence may be directly linked with the DNA molecule, in which case the first amino acid at the N-terminus of the recombinant protein will always be a methionine, which is encoded by the ATG start codon. If desired, methionine at the N-terminus may be cleaved from the protein by in vitro incubation with cyanogen bromide.
Fusion proteins provide an alternative for yeast expression systems, as well as in mammalian, baculovirus, and bacterial expression systems. Usually, a DNA sequence encoding the N-terminal portion of an endogenous yeast protein, or other stable protein, is fused to the 5' end of heterologous coding sequences. Upon expression, this construct will provide a fusion of the two amino acid sequences. For example, the yeast or human superoxide dismutase (SOD) gene, can be linked at the 5' terminus of a foreign gene and expressed in yeast. The DNA sequence at the junction of the two amino acid sequences may or may not encode a cleavable site. See e.g. EP-A-0 196 056. Another example is a ubiquitin fusion protein. Such a fusion protein is made with the ubiquitin region that preferably retains a site for a processing enzyme (e.g. ubiquitin-specific processing protease) to cleave the ubiquitin from the foreign protein. Through this method, therefore, native foreign protein can be isolated (e.g. WO88/024066).
Alternatively, foreign proteins can also be secreted from the cell into the growth media by creating chimeric DNA molecules that encode a fusion protein comprised of a leader sequence fragment that provide for secretion in yeast of the foreign protein. Preferably, there are processing sites encoded between the leader fragment and the foreign gene that can be cleaved either in vivo or in vitro. The leader sequence fragment usually encodes a signal peptide comprised of hydrophobic amino acids which direct the secretion of the protein from the cell. DNA encoding suitable signal sequences can be derived from genes for secreted yeast proteins, such as the yeast invertase gene (EP-A-0 012 873; JPO. 62,096,086) and the A-factor gene (US patent 4,588,684). Alternatively, leaders of non-yeast origin, such as an interferon leader, exist that also provide for secretion in yeast (EP-A-0060057).
A preferred class of secretion leaders are those that employ a fragment of the yeast alpha-factor gene, which contains both a "pre" signal sequence, and a "pro" region. The types of alpha-factor fragments that can be employed include the full-length pre-pro alpha factor leader (about 83 amino acid residues) as well as truncated alpha-factor leaders (usually about 25 to about 50 amino acid residues) (US Patents 4,546,083 and 4,870,008; EP-A-0 324 274). Additional leaders employing an alpha-factor leader fragment that provides for secretion include hybrid alpha-factor leaders made with a presequence of a first yeast, but a pro-region from a second yeast alphafactor. (e.g. see WO 89/02463.)
Usually, transcription termination sequences recognized by yeast are regulatory regions located 3' to the translation stop codon, and thus together with the promoter flank the coding sequence. These sequences direct the transcription of an mRNA which can be translated into the polypeptide encoded by the DNA. Examples of transcription terminator sequence and other yeast-recognized termination sequences, such as those coding for glycolytic enzymes.
Usually, the above described components, comprising a promoter, leader (if desired), coding sequence of interest, and transcription termination sequence, are put together into expression constructs. Expression constructs are often maintained in a replicon, such as an extrachromosomal element (e.g. plasmids) capable of stable maintenance in a host, such as yeast or bacteria. The replicon may have two replication systems, thus allowing it to be maintained, for example, in yeast for expression and in a prokaryotic host for cloning and amplification. Examples of such yeast- bacteria shuttle vectors include YEp24 [Botstein et al. (1979) Gene 8:17-24], pCl/1 [Brake et al. (1984) Proc. Natl Acad. Sci USA 87:4642-4646], and YRpl7 [Stinchcomb et al. (1982) J. Mol. Biol. 758:157]. In addition, a replicon may be either a high or low copy number plasmid. A high copy number plasmid will generally have a copy number ranging from about 5 to about 200, and usually about 10 to about 150. A host containing a high copy number plasmid will preferably have at least about 10, and more preferably at least about 20. Enter a high or low copy number vector may be selected, depending upon the effect of the vector and the foreign protein on the host. See e.g. Brake et al, supra.
Alternatively, the expression constructs can be integrated into the yeast genome with an integrating vector. Integrating vectors usually contain at least one sequence homologous to a yeast chromosome that allows the vector to integrate, and preferably contain two homologous sequences flanking the expression construct. Integrations appear to result from recombinations between homologous DNA in the vector and the yeast chromosome [Orr- Weaver et al. (1983) Methods in Enzymol. 707:228- 245]. An integrating vector may be directed to a specific locus in yeast by selecting the appropriate homologous sequence for inclusion in the vector. See Orr- Weaver et al, supra. One or more expression construct may integrate, possibly affecting levels of recombinant protein produced [Rine et al. (1983) Proc. Natl. Acad. Sci. USA 80:6750]. The chromosomal sequences included in the vector can occur either as a single segment in the vector, which results in the integration of the entire vector, or two segments homologous to adjacent segments in the chromosome and flanking the expression construct in the vector, which can result in the stable integration of only the expression construct.
Usually, extrachromosomal and integrating expression constructs may contain selectable markers to allow for the selection of yeast strains that have been transformed. Selectable markers may include biosynthetic genes that can be expressed in the yeast host, such as ADE2, H1S4, LEU2, TRP1, and ALG7, and the G418 resistance gene, which confer resistance in yeast cells to tunicamycin and G418, respectively. In addition, a suitable selectable marker may also provide yeast with the ability to grow in the presence of toxic compounds, such as metal. For example, the presence of CUP1 allows yeast to grow in the presence of copper ions [Butt et al. (1987) Microbiol, Rev. 57:351].
Alternatively, some of the above described components can be put together into transformation vectors. Transformation vectors are usually comprised of a selectable marker that is either maintained in a replicon or developed into an integrating vector, as described above.
Expression and transformation vectors, either extrachromosomal replicons or integrating vectors, have been developed for transformation into many yeasts. For example, expression vectors have been developed for, ter alia, the following yeasts:Candida albicans [Kurtz, et al. (1986) Mol Cell. Biol. 6: 142], Candida maltosa [Kunze, et al. (1985) J. Basic Microbiol. 25:141]. Hansenula polymoφha [Gleeson, et al. (1986) J. Gen. Microbiol. 732:3459; Roggenkamp et al (1986) Mol. Gen. Genet. 202:302], Kluyveromyces fragilis [Das, et al. (1984) /. Bacteriol. 758:1165], Kluyveromyces lactis [De Louvencourt et al. (1983) J. Bacteriol. 754:737; Van den Berg et al. (1990) Bio/Technology 8:135], Pichia guillerimondii [Kunze et al. (1985) J. Basic Microbiol. 25:141], Pichia pastoris [Cregg, et al. (1985) Mol. Cell. Biol. 5:3376; US Patent Nos. 4,837,148 and 4,929,555], Saccharomyces cerevisiae [Hinnen et al. (1978) Proc. Natl. Acad. Sci. USA 75:1929; Ito et al. (1983) J. Bacteriol 753:163], Schizosaccharomyces pombe [Beach and Nurse (1981) Nature 300:706], and Yarrowia lipolytica [Davidow, et al. (1985) Curr. Genet. 70:380471 Gaillardin, et al. (1985) Curr. Genet. 70:49].
Methods of introducing exogenous DNA into yeast hosts are well-known in the art, and usually include either the transformation of spheroplasts or of intact yeast cells treated with alkali cations. Transformation procedures usually vary with the yeast species to be transformed. See e.g. [Kurtz et al. (1986) Mol. Cell. Biol. 6:142; Kunze et al. (1985) J. Basic Microbiol 25:141; Candida]; [Gleeson et al. (1986) J. Gen. Microbiol. 732:3459; Roggenkamp et al. (1986) Mol. Gen. Genet. 202:302; Hansenula]; [Das et al. (1984) J. Bacteriol. 758: 1165; De Louvencourt et al. (1983) J. Bacteriol. 154;X 165; Van den Berg et al. (1990) Bio/Technology 8:135; Kluyveromyces]; [Cregg et al. (1985) Mol. Cell. Biol. 5:3376; Kunze et al. (1985) J. Basic Microbiol. 25:141; US Patent Nos. 4,837,148 and 4,929,555; Pichia]; [Hinnen et al. (1978) Proc. Natl. Acad. Sci. USA 75; 1929; Ito et al. (1983) J. Bacteriol. 753:163 Saccharomyces]; [Beach and Nurse (1981) Nature 300:706; Schizosaccharomyces]; [Davidow et al. (1985) Curr. Genet. 70:39; Gaillardin et al. (1985) Curr. Genet. 70:49; Yarrowia].
Antibodies
As used herein, the term "antibody" refers to a polypeptide or group of polypeptides composed of at least one antibody combining site. An "antibody combining site" is the three-dimensional binding space with an internal surface shape and charge distribution complementary to the features of an epitope of an antigen, which allows a binding of the antibody with the antigen. "Antibody" includes, for example, vertebrate antibodies, hybrid antibodies, chimeric antibodies, humanised antibodies, altered antibodies, univalent antibodies, Fab proteins, and single domain antibodies. Antibodies against the proteins of the invention are useful for affinity chromatography, immunoassays, and distinguishing/identifying Neisserial proteins.
Antibodies to the proteins of the invention, both polyclonal and monoclonal, may be prepared by conventional methods. In general, the protein is first used to immunize a suitable animal, preferably a mouse, rat, rabbit or goat. Rabbits and goats are preferred for the preparation of polyclonal sera due to the volume of serum obtainable, and the availability of labeled anti-rabbit and anti-goat antibodies. Immunization is generally performed by mixing or emulsifying the protein in saline, preferably in an adjuvant such as Freund's complete adjuvant, and injecting the mixture or emulsion parenterally (generally subcutaneously or intramuscularly). A dose of 50-200 μg injection is typically sufficient. Immunization is generally boosted 2-6 weeks later with one or more injections of the protein in saline, preferably using Freund's incomplete adjuvant. One may alternatively generate antibodies by in vitro immunization using methods known in the art, which for the puφoses of this invention is considered equivalent to in vivo immunization. Polyclonal antisera is obtained by bleeding the immunized animal into a glass or plastic container, incubating the blood at 25°C for one hour, followed by incubating at 4°C for 2-18 hours. The serum is recovered by centrifugation (e.g. 1 ,000g for 10 minutes). About 20-50 ml per bleed may be obtained from rabbits.
Monoclonal antibodies are prepared using the standard method of Kohler & Milstein [Nature (1975) 256:495-96], or a modification thereof. Typically, a mouse or rat is immunized as described above. However, rather than bleeding the animal to extract serum, the spleen (and optionally several large lymph nodes) is removed and dissociated into single cells. If desired, the spleen cells may be screened (after removal of nonspecifically adherent cells) by applying a cell suspension to a plate or well coated with the protein antigen. B-cells expressing membrane-bound immunoglobulin specific for the antigen bind to the plate, and are not rinsed away with the rest of the suspension. Resulting B-cells, or all dissociated spleen cells, are then induced to fuse with myeloma cells to form hybridomas, and are cultured in a selective medium (e.g. hypoxanthine, aminopterin, thymidine medium, "HAT"). The resulting hybridomas are plated by limiting dilution, and are assayed for the production of antibodies which bind specifically to the immunizing antigen (and which do not bind to unrelated antigens). The selected MAb-secreting hybridomas are then cultured either in vitro (e.g. in tissue culture bottles or hollow fiber reactors), or in vivo (as ascites in mice).
If desired, the an tibodies (w hether polyclonal or m onoclonal) m ay be labeled using conventional techniques. Suitable labels include fluorophores, chro ophores, radioactive atom s (particularly 32P and 125I), electron-dense reagents, enzym es, and ligands having specific binding partners. Enzym es are typically detected by their activity. For example, horseradish peroxidase is usually detected by its ability to convert 3 ,3 ',5 ,5 '-tetram ethylbenzidine (TM B ) to a blue pigm ent, quantifiable w ith a spectrophotom eter. "S pecific binding partner" refers to a protein capable of binding a ligand m olecule w ith high specificity, as for ex am ple in the case of an antigen and a m onoclonal antibody specific therefor. Other specific binding partners include biotin and avidin or streptavidin, IgG and protein A , and the num erous receptor-ligand couples know n in the art. It should be understood that the above description is not m eant to categorize the various labels into distinct classes, as the sam e label m ay serve in several different m odes. For example, l 25I m ay serve as a radioactive label or as an electron-den se reagent. H RP m ay serve as enzym e or as antigen for a M A b. Further, one m ay com bine various labels for desired effect. For exam ple, M A bs and avidin also require labels in the practice of this invention: thus, one m ight label a M A b w ith biotin, and detect its presence w ith avidin labeled w ith l 25I, or w ith an anti-biotin M A b labeled w ith H RP. O ther perm utations and possibilities w ill be readily apparent to those of ordinary skill in the art, and are considered as equivalents w ithin the scope of the instant invention.
Pharmaceutical Compositions
Pharmaceutical compositions can comprise either polypeptides, antibodies, or nucleic acid of the invention. The pharmaceutical compositions will comprise a therapeutically effective amount of either polypeptides, antibodies, or polynucleotides of the claimed invention.
The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect. The effect can be detected by, for example, chemical markers or antigen levels. Therapeutic effects also include reduction in physical symptoms, such as decreased body temperature. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance. However, the effective amount for a given situation can be determined by routine experimentation and is within the judgement of the clinician.
For purposes of the present in vention, an effective dose w ill be from about 0.01 m g/ kg to 50 m g/kg or 0.05 m g/kg to about 1 0 m g/kg of the D N A constructs in the individual to w hich it is adm inistered. A pharm aceutical composition can also contain a pharm aceutically acceptable carrier. The term "pharm aceutically acceptable carrier" refers to a carrier for adm inistration of a therapeutic agent, such as antibodies or a polypeptide, genes, and other therapeutic agents. The term refers to any pharm aceutical carrier that does not itself induce the production of antibodies harm ful to the individual receiving the com position, and w hich m ay be adm inistered w ithout undue toxicity. Suitable carriers m ay be large, slow ly m etabolized m acrom olecules such as proteins, polysaccharides, poly lactic acids, poly glycolic acids, polym eric am ino acids, am ino acid copolym ers, and inactive virus particles. Such carriers are w ell know n to those of ordinary skill in the art.
Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of pharmaceutically acceptable excipients is available in Remington's Pharmaceutical Sciences (Mack Pub. Co., NJ. 1991).
Pharm aceutically acceptable carriers in therapeutic compositions m ay contain liquids such as w ater, saline, glycerol and ethanol. A dditionally, aux iliary substances, such as wetting or em ulsifying agents, pH buffering substances, and the like, m ay be present in such vehicles. Typically, the therapeutic com positions are prepared as injectables, either as liqu id solutions or suspensions; solid form s suitable for solution in, or suspension in, liquid vehicles prior to injection m ay also be prepared. Liposomes are included w ithin the definition of a pharm aceutically acceptable carrier. Delivery Methods
Once formulated, the compositions of the invention can be administered directly to the subject. The subjects to be treated can be animals; in particular, human subjects can be treated.
Direct delivery of the compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue. The compositions can also be administered into a lesion. Other modes of administration include oral and pulmonary administration, suppositories, and transdermal or transcutaneous applications (e.g. see WO98/20734), needles, and gene guns or hyposprays. Dosage treatment may be a single dose schedule or a multiple dose schedule.
Vaccines V accines com prise im m u nising antigen(s), im m unogen(s), polypeptide(s), protein(s) or nucleic acid, usually in com bination w ith "pharm aceutically acceptable carriers," w hich include any carrier that does not itself induce the production of antibodies harm ful to the individual receiving the composition . Suitable carriers are typically large, slow ly m etabolized m acrom olecu les such as proteins, polysaccharides, poly lactic acids, polyglycoiic acids, polym eric am ino acids, am ino acid copolym ers, lipid aggregates (such as oil droplets or liposom es), and inactive virus particles. Such carriers are w ell know n to those of ordinary skill in the art. A dditionally, these carriers m ay function as im m unostimu lating agents ("adju vants"). Furtherm ore, the antigen or im m unogen m ay be conjugated to a bacterial toxoid, such as a toxoid from diphtheria, tetanus, cholera, H. pylori, etc. pathogens. Preferred adjuvants to enhance effectiveness of the composition include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc; (2) oil-in-water emulsion formulations (with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial cell wall components), such as for example (a) MF59™ (WO 90/14837; Chapter 10 in Vaccine design: the subunit and adjuvant approach, eds. Powell & Newman, Plenum Press 1995), containing 5% Squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE (see below), although not required) formulated into submicron particles using a microfluidizer such as Model HOY microfluidizer (Microfluidics, Newton, MA), (b) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic- blocked polymer L121, and thr-MDP (see below) either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) Ribi™ adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose di ycolate (TDM), and cell wall skeleton (CWS), preferably MPL + CWS (Detox™); (3) saponin adjuvants, such as Stimulon™ (Cambridge Bioscience, Worcester, MA) may be used or particles generated therefrom such as ISCOMs (immunostimulating complexes); (4) Complete Freund's Adjuvant (CFA) and Incomplete Freund's Adjuvant (IFA); (5) cytokines, such as interleukins (e.g. IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g. gamma interferon), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), etc; and (6) other substances that act as immunostimulating agents to enhance the effectiveness of the composition. Alum and MF59™ are preferred.
As mentioned above, muramyl peptides include, but are not limited to, N-acetyl -muramyl -L- threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N- acetylmuramyI- -alanyl-D-isoglutaminyl-L-aIanine-2-(r-2'-dipalmitoyl-.sn-glycero-3- hydroxyphosphoryloxy)-ethylamine (MTP-PE), etc.
The immunogenic compositions (e.g. the immunising antigen/immunogen/polypeptide/protein/ nucleic acid, pharmaceutically acceptable carrier, and adjuvant) typically will contain diluents, such as water, saline, glycerol, ethanol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles. Typically, the im m unogenic com positions are prepared as injectables, either as liquid solutions or suspensions; solid form s suitable for solution in, or suspension in, liquid vehicles prior to injection m ay also be prepared. The preparation also m ay be em ulsified or encapsulated in liposom es for enhanced adjuvant effect, as discussed above under pharm aceutically acceptable carriers.
Immunogenic compositions used as vaccines comprise an immunologically effective amount of the antigenic or immunogenic polypeptides, as well as any other of the above-mentioned components, as needed. By "immunologically effective amount", it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention. This amount varies depending upon the health and physical condition of the individual to be treated, the taxonomic group of individual to be treated (e.g. nonhuman primate, primate, etc.), the capacity of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.
The immunogenic compositions are conventionally administered parenterally, e.g. by injection, either subcutaneously, intramuscularly, or transdermally/transcutaneously (e.g. WO98/20734). Additional formulations suitable for other modes of administration include oral and pulmonary formulations, suppositories, and transdermal applications. Dosage treatment may be a single dose schedule or a multiple dose schedule. The vaccine may be administered in conjunction with other immunoregulatory agents.
As an alternative to protein-based vaccines, DNA vaccination may be employed [e.g. Robinson & Torres (1997) Seminars in Immunology 9:271-283; Donnelly et al. (1997) Annu Rev Immunol 15:617-648; see later herein]. Polynucleotide and polypeptide pharmaceutical compositions
In addition to the pharmaceutically acceptable carriers and salts described above, the following additional agents can be used with polynucleotide and/or polypeptide compositions.
A.Pol ypeptides
One example are polypeptides which include, without limitation: asioloorosomucoid (ASOR); transferrin; asialoglycoproteins; antibodies; antibody fragments; ferritin; interleukins; interferons, granulocyte, macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), macrophage colony stimulating factor (M-CSF), stem cell factor and erythropoietin. Viral antigens, such as envelope proteins, can also be used. Also, proteins from other invasive organisms, such as the 17 amino acid peptide from the circumsporozoite protein of plasmodium falciparum known as RII.
B.Hormones. Vitamins, etc.
Other groups that can be included are, for example: hormones, steroids, androgens, estrogens, thyroid hormone, or vitamins, folic acid.
C.Polvalkylenes, Polysaccharides. etc. Also, polyalkylene glycol can be included with the desired polynucleotides/polypeptides. In a preferred embodiment, the polyalkylene glycol is polyethlylene glycol. In addition, mono-, di-, or polysaccharides can be included. In a preferred embodiment of this aspect, the polysaccharide is dextran or DEAE-dextran. Also, chitosan and poly(lactide-co-glycolide)
D.Lipids. and Liposomes The desired polynucleotide/polypeptide can also be encapsulated in lipids or packaged in liposomes prior to delivery to the subject or to cells derived therefrom. Lipid encapsulation is generally accomplished using liposomes which are able to stably bind or entrap and retain nucleic acid. The ratio of condensed polynucleotide to lipid preparation can vary but will generally be around 1 : 1 (mg DNA:micromoles lipid), or more of lipid. For a review of the use of liposomes as carriers for delivery of nucleic acids, see, Hug and Sleight (1991) Biochim. Biophys. Acta. 1097: 1-17; Straubinger (1983) Meth. Enzymol. 101 :512-527.
Liposomal preparations for use in the present invention include cationic (positively charged), anionic (negatively charged) and neutral preparations. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Feigner (1987) Proc. Natl. Acad. Sci. USA 84:7413-7416); mRNA (Malone (1989) Proc. Natl. Acad. Sci. USA 86:6077-6081); and purified transcription factors (Debs (1990) J. Biol. Chem. 265: 10189-10192), in functional form.
Cationic liposomes are readily available. For example, N[l-2,3-dioleyloxy)propyl]-N,N,N- triethylammonium (DOTMA) liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, NY. (See, also, Feigner supra). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger). Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. Szoka (1978) Proc. Natl. Acad. Sci. USA 75:4194-4198; WO90/1 1092 for a description of the synthesis of DOTAP (l ,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes.
Sim ilarly, anionic and neutral liposom es are readily available, such as from Avanti Polar Lipids (B irm ingham , AL), or can be easily prepared using readily available m aterials. Such m aterials include phosphatidyl choline, cholesterol, phosphatidyl ethanolam ine, dioleoylphosphatidyl choline (D0PC), dioleoylphosphatidyl glycerol (D 0 PG ), dioleoylphoshatidyl ethanolam ine (DOPE), am ong others. These m aterials can also be m ixed w ith the DOTM A and D OTA P starting m aterials in appropriate ratios. M ethods for m aking liposom es using these m aterials are well known in the art.
The liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUNs), or large unilamellar vesicles (LUVs). The various liposome-nucleic acid complexes are prepared using methods known in the art. See e.g. Straubinger (1983) Meth. Immunol 101:512-527; Szoka (1978)
Proc. Natl. Acad. Sci. USA 75:4194-4198; Papahadjopoulos (1975) Biochim. Biophys. Acta
394:483; Wilson (1979) Cell 17:77); Deamer & Bangham (1976) Biochim. Biophys. Acta 443:629;
Ostro (1977) Biochem. Biophys. Res. Commun. 76:836; Fraley (1979) Proc. Natl. Acad. Sci. USA 76:3348); Enoch & Strittmatter (1979) Proc. Natl. Acad. Sci. USA 76:145; Fraley (1980) J. Biol.
Chem. (1980) 255:10431 ; Szoka & Papahadjopoulos (1978) Proc. Natl. Acad. Sci. USA 75:145; and
Schaefer-Ridder (1982) Science 215 : 166.
E.Lipoproteins
In addition, lipoproteins can be included w ith the polynucleotide/polypeptide to be delivered. Examples of lipoproteins to be utilized include: chylom icrons, HDL, ID L, LDL, and VLDL. M utants, fragm ents, or fusions of these proteins can also be used. A lso, m odifications of naturally occurring lipoproteins can be used, such as acetylated LDL. These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors. Preferably, if lipoproteins are including w ith the polynucleotide to be delivered, no other targeting ligand is included in the composition.
Naturally occurring lipoproteins comprise a lipid and a protein portion. The protein portion are known as apoproteins. At the present, apoproteins A, B, C, D, and E have been isolated and identified. At least two of these contain several proteins, designated by Roman numerals, Al, All, AIV; Cl, CII, Cffl.
A lipoprotein can comprise more than one apoprotein. For example, naturally occurring chylomicrons comprises of A, B, C, and E, over time these lipoproteins lose A and acquire C and E apoproteins. VLDL comprises A, B, C, and E apoproteins, LDL comprises apoprotein B; and HDL comprises apoproteins A, C, and E.
The amino acid of these apoproteins are known and are described in, for example, Breslow (1985) Annu Rev. Biochem 54:699; Law (1986) Adv. Exp Med. Biol. 151 : 162; Chen (1986) J Biol Chem 261:12918; Kane (1980) Proc Natl Acad Sci USA 77:2465; and Utermann (1984) Hum Genet 65:232. Lipoproteins contain a variety of lipids including, triglycerides, cholesterol (free and esters), and phospholipids. The composition of the lipids varies in naturally occurring lipoproteins. For example, chylomicrons comprise mainly triglycerides. A more detailed description of the lipid content of naturally occurring lipoproteins can be found, for example, in Meth. Enzymol. 128 (1986). The composition of the lipids are chosen to aid in conformation of the apoprotein for receptor binding activity. The composition of lipids can also be chosen to facilitate hydrophobic interaction and association with the polynucleotide binding molecule.
Naturally occurring lipoproteins can be isolated from serum by ultracentrifugation, for instance. Such methods are described in Meth. Enzymol. (supra); Pitas (1980) J. Biochem. 255:5454-5460 and Mahey (1979) J Clin. Invest 64:743-750. Lipoproteins can also be produced by in vitro or recombinant methods by expression of the apoprotein genes in a desired host cell. See, for example, Atkinson (1986) Annu Rev Biophys Chem 15:403 and Radding (1958) Biochim Biophys Acta 30: 443. Lipoproteins can also be purchased from commercial suppliers, such as Biomedical Techniologies, Inc., Stoughton, Massachusetts, USA. Further description of lipoproteins can be found in Zuckermann et al. PCT/US97/ 14465. F.Polvcationic Agents
Polycationic agents can be included, with or without lipoprotein, in a composition with the desired polynucleotide/polypeptide to be delivered.
Polycationic agents, typically, exhibit a net positive charge at physiological relevant pH and are capable of neutralizing the electrical charge of nucleic acids to facilitate delivery to a desired location. These agents have both in vitro, ex vivo, and in vivo applications. Polycationic agents can be used to deliver nucleic acids to a living subject either intram uscularly, subcutaneously, etc. The following are examples of useful polypeptides as polycationic agents: polylysine, polyarginine, polyornithine, and protamine. Other examples include histones, protamines, human serum albumin, DNA binding proteins, non-histone chromosomal proteins, coat proteins from DNA viruses, such as (XI 74, transcriptional factors also contain domains that bind DNA and therefore may be useful as nucleic aid condensing agents. Briefly, transcriptional factors such as C/CEBP, c-jun, c-fos, AP-1, AP-2, AP-3, CPF, Prot-1, Sp-1, Oct-1, Oct-2, CREP, and TFIID contain basic domains that bind DNA sequences.
Organic polycationic agents include: spermine, spermidine, and purtrescine.
The dimensions and of the physical properties of a polycationic agent can be extrapolated from the list above, to construct other polypeptide polycationic agents or to produce synthetic polycationic agents.
Synthetic polycationic agents which are useful include, for example, DEAE-dextran, polybrene. Lipofectin™, and lipofect AMINE™ are monomers that form polycationic complexes when combined with polynucleotides/polypeptides. Nucleic Acid Hybridisation
"Hybridization" refers to the association of two nucleic acid sequences to one another by hydrogen bonding. Typically, one sequence w ill be fixed to a solid support and the other will be free in solution. Then, the two sequences will be placed in contact with one another under conditions that favor hydrogen bonding. Factors that affect this bonding include: the type and volume of solvent; reaction tem perature; time of hybridization; agitation; agents to block the non-specific attachment of the liquid phase sequence to the solid support (Denhardt's reagent or B LOTTO); concentration of the sequences; use of compounds to increase the rate of association of sequences (dextran sulfate or polyethylene glycol); and the stringency of the washing conditions following hybridization. See Sam brook et al. [supra] Volum e 2, chapter 9, pages 9.47 to 9.57. "Stringency" refers to conditions in a hybridization reaction that favor association of very similar sequences over sequences that differ. For example, the combination of temperature and salt concentration should be chosen that is approximately 120 to 200°C below the calculated Tm of the hybrid under study. The temperature and salt conditions can often be determined empirically in preliminary experiments in which samples of genomic DNA immobilized on filters are hybridized to the sequence of interest and then washed under conditions of different stringencies. See Sambrook et al. at page 9.50.
Variables to consider when performing, for example, a Southern blot are (1) the complexity of the DNA being blotted and (2) the homology between the probe and the sequences being detected. The total amount of the fragment(s) to be studied can vary a magnitude of 10, from 0.1 to lμg for a plasmid or phage digest to 10'9 to 10"8 g for a single copy gene in a highly complex eukaryotic genome. For lower complexity polynucleotides, substantially shorter blotting, hybridization, and exposure times, a smaller amount of starting polynucleotides, and lower specific activity of probes can be used. For example, a single-copy yeast gene can be detected with an exposure time of only 1 hour starting with 1 μg of yeast DNA, blotting for two hours, and hybridizing for 4-8 hours with a probe of 108 cpm/μg. For a single-copy mammalian gene a conservative approach would start with 10 μg of DNA, blot overnight, and hybridize overnight in the presence of 10% dextran sulfate using a probe of greater than 108 cpm/μg, resulting in an exposure time of -24 hours.
Several factors can affect the melting temperature (Tm) of a DNA-DNA hybrid between the probe and the fragment of interest, and consequently, the appropriate conditions for hybridization and washing. In many cases the probe is not 100% homologous to the fragment. Other commonly encountered variables include the length and total G+C content of the hybridizing sequences and the ionic strength and formamide content of the hybridization buffer. The effects of all of these factors can be approximated by a single equation:
Tm= 81 + 16.6(log10Ci) + 0.4[%(G + C)]-0.6(%formamide) - 600//ι-1.5(%mismatch). where Ci is the salt concentration (monovalent ions) and n is the length of the hybrid in base pairs (slightly modified from Meinkoth & Wahl (1984) Anal. Biochem. 138: 267-284). In designing a hybridization experiment, some factors affecting nucleic acid hybridization can be conveniently altered. The temperature of the hybridization and washes and the salt concentration during the washes are the simplest to adjust. As the temperature of the hybridization increases (i.e. stringency), it becomes less likely for hybridization to occur between strands that are nonhomologous, and as a result, background decreases. If the radiolabeled probe is not completely homologous with the immobilized fragment (as is frequently the case in gene family and interspecies hybridization experiments), the hybridization temperature must be reduced, and background will increase. The temperature of the washes affects the intensity of the hybridizing band and the degree of background in a similar manner. The stringency of the washes is also increased with decreasing salt concentrations. In general, convenient hybridization temperatures in the presence of 50% formamide are 42°C for a probe with is 95% to 100% homologous to the target fragment, 37°C for 90% to 95% homology, and 32°C for 85% to 90% homology. For lower homologies, formamide content should be lowered and temperature adjusted accordingly, using the equation above. If the homology between the probe and the target fragment are not known, the simplest approach is to start with both hybridization and wash conditions which are nonstringent. If non-specific bands or high background are observed after autoradiography, the filter can be washed at high stringency and reexposed. If the time required for exposure makes this approach impractical, several hybridization and/or washing stringencies should be tested in parallel. MODES FOR CARRYING OUT THE INVENTION
Identification of the meningococcal 80-85kDa protein
It was observed that various outer membrane vesicle preparations from N. meningitidis serogroup B contained a component of approximately 80-85kDa. This protein was purified from SDS-PAGE gels and N-terminal sequenced (SEQ ID 1).
Antibodies raised against the SDS-PAGE purified protein cross-reacted with equivalent proteins in more than 50 N. meningitidis strains of diverse serogroups and serotypes. Cross-reactivity with N. gonorrhoeae, N.polysaccharia and N. lactamica was also observed. Post-immune sera from vaccinated patients also reacted with the protein.
The complete gene was cloned from serogroup B N meningitidis (SEQ ID 2) and the encoded protein was inferred (SEQ ID 3). By comparison with the N-terminal sequencing described above, a signal peptide (SEQ ID 4) and a mature sequence (SEQ ED 5) are inferred.
Identification of corresponding genes in N.meningitidis serogroup A and N.gonorrhoeae
On the basis of the serogroup B N.meningitidis sequence, the corresponding genes from N.meningitidis serogroup A and N.gonorrhoeae were cloned and sequenced (called 'ORF2T).
The complete gene from serogroup A N.meningitidis is shown in SEQ ID 6, with the encoded protein in SEQ ID 7. The signal peptide and mature sequence are SEQ IDs 8 and 9.
The complete gene from N.gonorrhoeae is shown in SEQ ID 10, with the encoded protein in SEQ ID 11. The signal peptide and mature sequence are SEQ IDs 12 and 13.
Sequence comparisons
The protein sequences were compared and are highly homologous.
The N.meningitidis serogroup B sequence and the N.gonorrhoeae sequence show 95.4% identity in 797 aa overlap:
10 20 30 40 50 60 orf21.pep I^LKQIAS-ZUJMMLGISP ALADFTIQDIRVEG QRTEPSTVFNYLPVKVGDTYNDTHGSA orf2lng. ep MKLKQIASALM LGISP AFADFTIQDIRVEGLQRTEPSTVFNY PVKVGDTYNDTHGSA
10 20 30 40 50 60
70 80 90 100 110 120 orf21.pep IIKS YATGFFDDVRVETADGQLLTVIERPTIGSLNITGAKMLQNDAIKKNLESFGAQ orf2lng.pep IIKS YATGFFDDVRVETADGQL TVIERPTIGSLNITGAKMLQNDAIKKN ESFGLAQ 70 80 90 100 110 120 130 140 150 160 170 180 orf21.pep SQYFNQATLKQAVAGLKEEYLGRGKLNIQITPKVTK ARNRVDIDITIDEGKSAKITDIE
! 111 M 11 ! 11111 ' 11 M 11111 M I ! ! M 11 M 11 ! : I ! I M 11 II II (111 orf21ng .pep SQYFNQAT NQAVAGLKEEYLGRGK NIQITPKVTKLARNRVDIDITIDEGKSAKITDIE
130 140 150 160 170 180
190 200 210 220 230 240 orf21.pep FEGNQVYSDRKLMRQMSLTEGGIWTWLTRSNQFNEQKFAQDMEKVTDFYQNNGYFDFRIL orf21ng .pep FEGNQVYSDRK MRQMSLTEGGIWTWLTRSDRFDRQKFAQDMEKVTDFYQNNGYFDFRIL
190 200 210 220 230 240
250 260 270 280 290 300 o f21.pep DTDIQTNEDKTKQTIKITVHEGGRFR GKVSIEGDTNEVPKAE EKL TMKPGK YERQQ orf2lng.pep DTDIQTNEDKTRQTIKITVHEGGRFR GKVSIEGDTNEVPKAE EKLLTMKPGK YERQQ
250 260 270 280 290 300 310 320 330 340 350 360 orf21.pep MTAVLGEIQNRMGSAGYAYSEISVQP PNAETKTVDFVLHIEPGRKIYV EIHITGNKT or f 2 lng . pep IX1TAVLGEIQNRMGSAGYAYΞEISVQP PNAGTKTVDFVLHIEPGRKIYVNEIHITG NKT
310 320 330 340 350 360
370 380 390 400 410 420 or f 21. pep RDEWRRE RQMESAPYDTSKLQRSKERVE LGYFD VQFDAVP AGTPDKVD MS TE or f 2 lng . pep RDEWRRELRQMESAPYDTSKLQRSKERVE GYFDNVQFDAVP AGTPDKVDLMMSLTE 370 380 390 400 410 420
430 440 450 460 470 480 or f 21. pep RSTGSLDLSAGWVQDTG VMSAGVSQDN FGTGKSAA RASRSKTT NGSLSFTDPYFTA orf2lng .pep RSTGSLDLSAG VQDTG VMSAGVSQDN FGTGKSAA RASRSKTTLNGSLSFTDPYFTA
430 440 450 460 470 480
490 500 510 520 530 540 orf21.pep DGVSLGYDVYGKAFDPRKASTSIKQYKTTTAGAGIRMSVPVTEYDRVNFGLVAEHLTV T M I I I I I I : I I I I I || || || I I : I I I I I I I I I : I : I I : : I II M I I I I I I I : II I I I I I I orf21ng .pep DGVSLGYDIYGKAFDPRKASTSVKQYKTTTAGGGVRMGIPVTEYDRVFGLAAEHLTV T
490 500 510 520 530 540
550 560 570 580 590 600 orf21.pep YNKAPKHYADFIKKYGKTDGTDGSFKGWLYKGTVG GRNKTDSAL PTRGY TGVAEIA
IIIIHIIIMI IIIIMIIM 1111111111111111 111111111111111 orf2lng.pep YNKAPKRYADFIRKYGKTDGADGSFKG YKGTVGWGR KTDSAS PTRGYLTGVNAEIA
550 560 570 580 590 600 610 620 630 640 650 660 orf21.pep LPGSK QYYSATHNQTWFFP SKTFTLM GGEVGIAGGYGRTKEIPFFENFYGGGLGSVR orf21ng .pep LPGSK QYYSATHNQTWFFP ΞKTFT M GGEVGIAGGYGRTKEIPFFE FYGGGLGSVR
610 620 630 640 650 660
670 680 690 700 710 720 orf21.pep GYESGTLGPKVYDEYGEKISYGGNKKA VSAE LFPMPGAKDARTVRLSLFADAGSV DG orf21ng.pep GYESGTLGPKVYDEYGEKISYGGNKKA VSAELLFPMPGAKDARTVRLSLFADAGSV DG 670 680 690 700 710 720
730 740 750 760 770 780 orf21.pep KTYDDNSSSATGGRVQNIYGAGNTHKSTFT E RYSAGGAVT SPLGPMKFSYAYPLKK
HI :H = 1 ■ '■ '■ \■ ' I = 1111111111 II 111111111111111111111111 orf21ng .pep RTY TAAENGN KSVYSE-NAHKSTFTNELRYSAGGAVTVTC_SPLGPMKFSYAYP KK
730 740 750 760 770
790 orf21.pep KPEDEIQRFQFQLGTTF orf21ng.pep KPEDEIQRFQFQ GTTFX 780 790
The N.meningitidis serogroup A and B sequences show 99.9% identity in 797 aa overlap:
10 20 30 40 50 60 o f21.pep MK KQIASALMMLGISPLA ADFTIQDIRVEGLQRTEPSTVFNYLPVKVGDTYNDTHGSA orf2la.pep MK KQIASAIJWLGISPLALADFTIQDIRVEGLQRTEPSTVFNYLPVKVGDTYNDTHGSA
10 20 30 40 50 60
70 80 90 100 110 120 orf21.pep IIKSLYATGFFDDVRVETADGQLLLTVIERPTIGSLNITGAKMLQNDAIKK LESFG AQ orf2la.pep IIKSLYATGFFDDVRVETADGQL LTVIERPTIGSLNITGAKMLQ DAIKKNLESFG AQ
70 80 90 100 110 120
130 140 150 160 170 180 orf21.pep SQYFNQATLNQAVAG KEEYLGRGK NIQITPKVTKLARNRVDIDITIDEGKSAKITDIE orf2la.pep SQYFNQATLNQAVAGLKEEYLGRGKLNIQITPKVTKLARNRVDIDITIDEGKSAKITDIE
130 140 150 160 170 180 190 200 210 220 230 240 orf21.pep FEGNQVYSDRK MRQMSLTEGGI TWLTRSNQFNEQKFAQDMEKVTDFYQNNGYFDFRIL orf2la .pep FEGNQVYSDRK MRQMS TEGGI TWLTRSNQFNEQKFAQDMEKVTDFYQNNGYFDFR1L
190 200 210 220 230 240
250 260 270 280 290 300 orf21.pep DTDIQT EDKTKQTIKITVHEGGRFRWGKVSIEGDTNEVPKAELEKLLTMKPGKWYERQQ orf2la .pep DTDIQTNEDKTKQTIKITVHEGGRFRWGKVSIEGDTNEVPKAELEK TMKPGK YERQQ 250 260 270 280 290 300
310 320 330 340 350 360 orf21.pep MAVLGEIQNRMGSAGYAYSEISVQPLPNAETKTVDFV HIEPGRKIYVNEIHITGNNKT
MIIIIIMMIMIIIIIIIMIIIMIIIIIIIIIIIIIIIIIIMIMIIIIIIIII orf21a.pep MTAVLGEIQNRMGSAGYAYSEISVQP PNAETKTVDFVLHIEPGRKIYV EIHITGNNKT
310 320 330 340 350 360
370 380 390 400 410 420 orf21.pep RDEWRRELRQMESAPYDTSKLQRSKERVEL GYFDNVQFDAVP AGTPDKVDL MS TE or f 2 l . pep RDEWRRELRQMESAPYDTSKLQRSKERVE GYFDNVQFDAVPLAGTPDKVDLNMSLTE
370 380 390 400 410 420
430 440 450 460 470 480 orf 21.pep RSTGSLDLSAG VQDTG VMSAGVSQDNLFGTGKSAALRASRSKTTLNGSLSFTDPYFTA orf2la.pep RSTGSLD SAGWVQDTGLVMSAGVSQDNLFGTGKSAALRASRSKTTLNGSLSFTDPYFTA
430 440 450 460 470 480 490 500 510 520 530 540 orf21.pep DGVSLGYDVYGKAFDPRKASTSIKQYKTTTAGAGIRMSVPVTEYDRVFGLVAEHLTVN orf2la.pep DGVS GYDVYGKAFDPRKASTSIKQYKTTTAGAGIR SVPVTEYDRVNFGLVAEHLTV T
490 500 510 520 530 540
550 560 570 580 590 600 orf21.pep YNKAPKHYADFIKKYGKTDGTDGSFKG LYKG VG GR KTDSA PTRGY TGVAEIA orf2la.pep YNKAPKHYADFIKKYGKTDGTDGSFKG LYKGTVGWGRNKTDSA PTRGYTGVNAEIA 550 560 570 580 590 600
610 620 630 640 650 660 or f 21 . pep LPGSK QYYSATH QT FFPLSKTFT MLGGEVGIAGGYGRTKEIPFFENFYGGGLGSVR orf2la .pep LPGSKLQYYSATHNQT FFP SKTFT MLGGEVGIAGGYGRTKEIPFFENFYGGG GSVR
610 620 630 640 650 660
670 680 690 700 710 720 orf21. pep GYESGTLGPKVYDEYGEKISYGG KKANVSAELLFPMPGAKDARTVRLSLFADAGSVWDG orf2la .pep GYESGTLGPKVYDEYGEKISYGGNKKANVSAELLFPMPGAKDARTVRLSLFADAGSVDG 670 680 690 700 710 720
730 740 750 760 770 780 orf21.pep KTYDDNSSSATGGRVQNIYGAGNTHKSTFTNELRYSAGGAVT LSPLGPMKFSYAYPLKK orf2la .pep KTYDDNSSSATGGRVQNIYGAGNTHKSTFTNELRYSAGGAVT LSPLGPMKFSYAYPLKK
730 740 750 760 770 780
790 orf21.pep KPEDEIQRFQFQLGTTF orf 2 la . pep KPEDEIQRFQFQ GTTFX
790 The high degree of conservation suggests that a single protein may be able to induce immune responses against a variety of Neisseriae species.
Cloning, expression and purification
N.meningitidis strains 2996 and MC58 were grown to exponential phase in 100ml of GC medium, harvested by centrifugation, and resuspended in 5ml buffer (20% w/v sucrose, 50mM Tris-HCl, 50mM EDTA, pH8). After 10 minutes incubation on ice, the bacteria were lysed by adding 10ml of lysis solution (50mM NaCl, 1% Na-Sarkosyl, 50μg/ml Proteinase K), and the suspension incubated at 37°C for 2 hours. Two phenol extractions (equilibrated to pH 8) and one CHCl3/isoamylalcohol (24: 1) extraction were performed. DNA was precipitated by addition of 0.3M sodium acetate and 2 volumes of ethanol, and collected by centrifugation. The pellet was washed once with 70%(v/v) ethanol and redissolved in 4.0ml TE buffer (lOmM Tris- HCl, ImM EDTA, pH 8.0). The DNA concentration was measured by reading OD26o-
ORF21, without the sequence encoding the leader peptide, was amplified by PCR using the following oligonucleotides: orf21 for <SEQ ID 105> (BamHI-Ndel) orf21 rev <SEQ ID 106> (Xhol)
The 5' primer included two restriction sites (BamHI-Ndel), the 3' primer included a Xhol restriction site, in order to direct the cloning of the amplification product (corresponding to ORF21) into the two expression vectors pGEX-KG (using BamHI-XhoT) to have a N-terminal GST-fusion, and pET21b+ (using Ndel-Xhol), to have a C-terminal His-fusion. The standard PCR protocol was as follows: 200ng of genomic DNA from 2996 or MC58 strains or lOng of plasmid DNA preparation of recombinant clones were used as template in the presence of 40μM of each oligonucletide primer, 400-800 μM dNTPs solution, lx PCR buffer (including 1.5mM MgCl2), 2.5 units Taql DNA polymerase (using Perkin-Elmer AmpliTaQ, Boerhingher Mannheim Expand™ Long Template).
After a preliminary 3 minute incubation of the whole mix at 95°C, each sample underwent a two-step amplification: the first 5 cycles were performed using the hybridisation temperature that excluded the restriction enzyme tail of the primer (Tmι). This was followed by 30 cycles according to the hybridisation temperature calculated for the whole length oligos (Tm2). Elongation times, performed at 68°C or 72°C, varied according to the length of the Orf to be amplified. In the case of Orf 1 the elongation time, starting from 3 minutes, was increased by 15 seconds each cycle. The cycles were completed with a 10 minute extension step at 72°C.
The amplified DNA was either loaded directly on a 1% agarose gel. The DNA fragment corresponding to the band of correct size was purified from the gel using the Qiagen Gel Extraction Kit, following the manufacturer's protocol.
The purified DNA corresponding to the amplified fragment was digested with the appropriate restriction enzymes for cloning into pET-21b+ or pET22b+. Digested fragments were purified using the QIAquick PCR purification kit (following the manufacturer's instructions) and eluted with either H O or lOmM Tris, pH 8.5. Plasmid vectors were digested with the appropriate restriction enzymes, loaded onto a 1.0% agarose gel and the band corresponding to the digested vector purified using the Qiagen QIAquick Gel Extraction Kit.
The fragments corresponding to each gene, previously digested and purified, were ligated into pET21b+ or into pET22b+. A molar ratio of 3: 1 fragment/vector was used with T4 DNA ligase in the ligation buffer supplied by the manufacturer.
Recombinant plasmid was transformed into competent E.coli DH5 or HB101 by incubating the ligase reaction solution and bacteria for 40 minutes on ice, then at 37°C for 3 minutes. This was followed by the addition of 800μl LB broth and incubation at 37°C for 20 minutes. The cells were centrifuged at maximum speed in an Eppendorf microfuge, resuspended in approximately 200μl of the supernatant and plated onto LB ampicillin (lOOmg/ml ) agar.
Screening for recombinant clones was performed by growing randomly selected colonies overnight at 37°C in 4.0ml of LB broth + lOOμg/ml ampicillin. Cells were pelleted and plasmid DNA extracted using the Qiagen QIAprep Spin Miniprep Kit, following the manufacturer's instructions. Approximately lμg of each individual miniprep was digested with the appropriate restriction enzymes and the digest loaded onto a 1-1.5% agarose gel (depending on the expected insert size), in parallel with the molecular weight marker (lkb DNA Ladder, GIBCO). Positive clones were selected on the basis of the size of insert.
After cloning each gene into the expression vector, recombinant plasmids were transformed into E.coli strains suitable for expression of the recombinant protein, lμl of each construct was used to transform E.coli BL21-DE3 as described above. Single recombinant colonies were inoculated into 2ml LB+Amp (lOOμg/ml), incubated at 37°C overnight, then diluted 1:30 in 20ml of LB+Amp (lOOμg/ml) in 100ml flasks, to give an OD600 between 0.1 and 0.2. The flasks were incubated at 30°C or at 37°C in a gyratory water bath shaker until OD6oo indicated exponential growth suitable for induction of expression (0.4-0.8 OD). Protein expression was induced by addition of LOmM IPTG. After 3 hours incubation at 30°C or 37°C the OD6oo was measured and expression examined. 1.0ml of each sample was centrifuged in a microfuge, the pellet resuspended in PBS and analysed by SDS-PAGE and Coomassie Blue staining.
The GST fusion protein was expressed, but it was found to be insoluble (i.e. unpurifiable). The His-fusion was expressed as insoluble protein.
For each clone purified as a His-fusion, a single colony was streaked and grown overnight at 37°C on a LB/Amp (100 μg/ml) agar plate. An isolated colony from this plate was inoculated into 20ml of LB/Amp (100 μg/ml) liquid medium and grown overnight at 37°C with shaking. The overnight culture was diluted 1:30 into 1.0 L LB/Amp (100 μg/ml) liquid medium and allowed to grow at the optimal temperature (30 or 37°C) until the OD550 reached 0.6-0.8. Expression of recombinant protein was induced by addition of IPTG (final concentration LOmM) and the culture incubated for a further 3 hours. Bacteria were harvested by centrifugation at 8000g for 15 min at 4°C. The bacterial pellet was resuspended in 7.5 ml of buffer B (8M urea, lOmM Tris-HCl, 100 mM phosphate buffer, pH 8.8). Cells were disrupted by sonication on ice four times for 30 sec at 40W using a Branson sonifier 450 and centrifuged at 13 OOOxg for 30 min at 4°C. Pellets were resuspended in 2.0 ml buffer C (6 M guanidine hydrochloride, 100 mM phosphate buffer, 10 mM Tris- HC1, pH 7.5 and treated with 10 passes of a Dounce homogenizer. The homogenate was centrifuged at 13000g for 30 min and the supernatant retained. Supernatants were mixed with 150μl Ni +-resin (previously equilibrated with buffer B) and incubated at room temperature with gentle agitation for 30 min. The resin was Chelating Sepharose Fast Flow (Pharmacia), prepared according to the manufacturer's protocol. The batch-wise preparation was centrifuged at 700 x g for 5 min at 4°C and the supernatant discarded. The resin was washed twice (batch-wise) with 10ml buffer B for 10 min, resuspended in 1.0 ml buffer B and loaded onto a disposable column. The resin continued to be washed with buffer B at room temperature, until the OD280 of the flow-through reached 0.02- 0.01. The resin was further washed with buffer D (8M urea, lOmM Tris-HCl, lOOmM phosphate buffer, pH 6.3) until OD280 of the flow-through reached 0.02-0.01. The His-fusion protein was eluted by addition of 700μl of elution buffer B (8 M urea, 10 mM Tris-HCl, 100 mM phosphate buffer, pH 4.5) and fractions collected until the OD 8o indicated all the recombinant protein was obtained. 20μl aliquots of each elution fraction were analysed by SDS-PAGE. Protein concentrations were estimated using the Bradford assay.
To renature the protein, glycerol was added to the denatured fractions obtained above to give a final concentration of 10% v/v. The proteins were diluted to 200 μg/ml using dialysis buffer I (10% v/v glycerol, 0.5M arginine, 50 mM phosphate buffer, 5.0 mM reduced glutathione, 0.5 mM oxidised glutathione, 2.0 M urea, pH 8.8) and dialysed against the same buffer for 12-14 hours at 4°C. Further dialysis was performed with buffer II (10% v/v glycerol, 0.5M arginine, 50mM phosphate buffer, 5.0mM reduced glutathione, 0.5mM oxidised glutathione, pH 8.8) for 12-14 hours at 4°C. Protein concentration was estimated using the formula:
Protein (mg/ml) = (7.55 x OD2So) - (0.76 x OD26o)
For immunological characterisation of ORF21, Balb/C mice were immunized with antigens on days 0, 21 and 35 and sera analyzed at day 49.
0RF21 gave a positive results on the following ELISA assay: acapsulated M enB M 7 and the capsulated strains were plated on chocolate agar plates and incubated overnight at 37°C with 5% C02. B acterial colonies were collected from the agar plates using a sterile dracon swab and inoculated into M ueller-Hinton Broth (Difco) containing 0.25% glucose. B acterial growth was m onitored every 30 m inutes by following OU 2o. The bacteria were let to grow until the 0D reached the value of 0.4-0.5. The culture was centrifuged for 10 minutes at 4000rpm . The supernatant was discarded and bacteria were w ashed twice with PB S , resuspended in PB S containing 0.025% formaldehyde, and incubated for 1 hour at 37°C and then overnight at 4°C with stirring. l OOμ l bacterial cells were added to each well of a 96 well Greiner plate and incubated overnight at 4°C . The wells were then washed three tim es with PBT washing buffer (0.1 % Tween-20 in PB S ). 200μ l of saturation buffer (2.7% polyvinylpyrrolidone 10 in water) was added to each well and the plates incubated for 2 hours at 37°C . W ells were washed three times with PBT. 200μ l of diluted sera (Dilution buffer: 1 % BSA, 0.1 % Tween-20, 0.1 % NaNj in PB S) were added to each well and the plates incubated for 2 hours at 37°C . W ells were washed three times with PBT. l OOμ l of HRP-conjugated rabbit anti-mouse (Dako) serum diluted 1 :2000 in dilution buffer w ere added to each well and the plates were incubated for 90 minutes at 37°C . Wells were washed three times with PBT buffer. l OOμ l of substrate buffer for HRP (25ml of citrate buffer pH5 , l Omg of O-phenildiamine and l Oμ l of H2O2) w ere added to each well and the plates were left at room temperature for 20 minutes. l OOμ l 12.5% H2SO4 was added to each well and OD490 was followed. The ELISA titers were calculated abitrarely as the dilution of sera which gave an OD490 value of 0.4 above the level of preim une sera. The ELISA was considered positive when the dilution of sera with OD490 of 0.4 was higher than 1 :400.
To assess cellular location of ORF21, the following FACS assay was used: acapsulated MenB M7 strain was plated on chocolate agar plates and incubated overnight at 37°C with 5% CO2. Bacterial colonies were collected from the agar plates using a sterile dracon swab and inoculated into 4 tubes containing 8ml each Mueller-Hinton Broth (Difco) containing 0.25% glucose. Bacterial growth was monitored every 30 minutes by following OD62o. The bacteria were let to grow until the OD reached the value of 0.35-0.5. The culture was centrifuged for 10 minutes at 4000rpm. The supernatant was discarded and the pellet was resuspended in blocking buffer (1% BSA in PBS, 0.4% NaN3) and centrifuged for 5 minutes at 4000rpm. Cells were resuspended in blocking buffer to reach OD62o of 0.05. lOOμl bacterial cells were added to each well of a Costar 96 well plate. lOOμl of diluted (1: 100, 1 :200, 1:400) sera (in blocking buffer) were added to each well and plates incubated for 2 hours at 4°C. Cells were centrifuged for 5 minutes at 4000rpm, the supernatant aspirated and cells washed by addition of 200μl/well of blocking buffer in each well. lOOμl of R-Phicoerytrin conjugated F(ab)2 goat anti-mouse, diluted 1:100, was added to each well and plates incubated for 1 hour at 4°C. Cells were spun down by centrifugation at 4000rpm for 5 minutes and washed by addition of 200μl/well of blocking buffer. The supernatant was aspirated and cells resuspended in 2O0μl/well of PBS, 0.25% formaldehyde. Samples were transferred to FACScan tubes and read. The condition for FACScan (Laser Power 15mW) setting were: FL2 on; FSC-H threshold:92; FSC PMT Voltage: E 01; SSC PMT: 474; Amp. Gains 6.1; FL-2 PMT: 586; compensation values: 0.
As well as FACS, Western analysis was used to assess cellular location: purified proteins (500ng/lane), outer membrane vesicles (5μg) and total cell extracts (25μg) derived from MenB strain 2996 were loaded onto a 12% SDS-polyacrylamide gel and transferred to a nitrocellulose membrane. The transfer was performed for 2 hours at 150mA at 4°C, using transfer buffer (0.3% Tris base, 1.44% glycine, 20% (v/v) methanol). The membrane was saturated by overnight incubation at 4°C in saturation buffer (10% skimmed milk, 0.1% Triton X100 in PBS). The membrane was washed twice with washing buffer (3% skimmed milk, 0.1% Triton X100 in PBS) and incubated for 2 hours at 37°C with mice sera diluted 1:200 in washing buffer. The membrane was washed twice and incubated for 90 minutes with a 1:2000 dilution of horseradish peroxidase labelled anti-mouse Ig. The membrane was washed twice with 0.1% Triton X100 in PBS and developed with the Opti-4CN Substrate Kit (Bio-Rad). The reaction was stopped by adding water.
The OMVs were prepared as follows: N. meningitidis strain 2996 was grown overnight at 37 degrees with 5% CO2 on 5 GC plates, harvested with a loop and resuspended in 10 ml of 20mM
Tris-HCl pH 7.5, 2 mM EDTA. Heat inactivation was performed at 56°C for 45 minutes and the bacteria disrupted by sonication for 5 minutes on ice (50% duty cycle, 50% output , Branson sonifier 3 mm microtip). Unbroken cells were removed by centrifugation at 5000g for 10 minutes, the supernatant containing the total cell envelope fraction recovered and further centrifuged overnight at 50000g at the temperature of 4°C . The pellet containing the membranes was resuspended in 2% sarkosyl, 20mM Tris-HCl pH 7.5, 2 mM EDTA and incubated at room temperature for 20 minutes to solubilyze the inner membranes. The suspension was centrifuged at lOOOOg for 10 minutes to remove aggregates, the supernatant was further centrifuged at 50000g for 3 hours. The pellet, containing the outer membranes was washed in PBS and resuspended in the same buffer. The protein concentration was measured by the D.C. Bio-Rad Protein assay (Modified Lowry method), using BSA as a standard.
Total cell extracts were prepared as follows: N. meningitidis strain 2996 was grown overnight on a GC plate, harvested with a loop and resuspended in 1ml of 20mM Tris-HCl. Heat inactivation was performed at 56°C for 30 minutes. ELISA, FACS and Western analysis all show that ORF21 is surface-exposed.
0 RF21 gave good results using the follow ing bactericidal assay: N.meningitidis strain 2996 w as grown overnight at 37°C on chocolate agar plates (starting from a frozen stock) with 5% CO2. Colonies were collected and used to inoculate 7m l M ueller-Hinton broth, containing 0.25 % glucose to reach an OD620 of 0.05-0.08. The culture was incubated for approxim ately 1 .5 hours at 37 degrees with shacking until the OD620 reached the value of 0.23-0.24. B acteria were diluted in 50m M Phosphate buffer pH 7.2 containing l Om M M gC l2, l Om M CaCl2 and 0.5 % (w/v) B SA (assay buffer) at the w orking dilution of 105 CFU/m l. The total volume of the final reaction m ixture w as 50 μ l with 25 μ l of serial tw o fold dilution of test serum , 12.5 μ l of bacteria at the working dilution, 12.5 μ l of baby rabbit complem ent (final concentration 25% ). Controls included bacteria incubated with complement serum , im m une sera incubated with bacteria and with complem ent inactivated by heating at 56°C for 30'. Immediately after the addition of the baby rabbit com plement, l Oμ l of the controls were plated on M ueller-Hinton agar plates using the tilt method (tim e 0). The 96-wells plate was incubated for 1 hour at 37°C with rotation. 7μ l of each sample were plated on M ueller-Hinton agar plates as spots, whereas l Oμ l of the controls were plated using the tilt method (time 1 ). A gar plates were incubated for 18 hours at 37 degrees and the colonies corresponding to time 0 and time 1 were counted.
Computer analysis Figure 1 shows the sequence of ORF21, together with computer analysis data.
ORF21 has the following AMPHI regions [Gao et al. (1989) J. Immunol. 143:3007; Roberts et al (1996) AIDS Res Hum Retrovir 12:593; Quakyi et al (1992) Scand J Immunol suppl.l 1:9]: SEQ IDs 14 to 32.
The antigenic index algorithm identified the following regions: SEQ IDs 33 to 70.
Hopp & Woods analysis reveals the following hydrophilic regions: SEQ IDs 71 to 104.
SEQ IDs 14-104 may be used as these oligopeptides, or as part of longer polypeptides.
An alignment of ORF21 sequences from MenA, MenB and gonococcus is shown in Figure 2.
Vaccines
The three proteins identified above are expressed and used for immunisation. Good immune responses are observed against the proteins.
Combination vaccines
In addition, the proteins are each combined with antigens against other pathogenic organisms (e.g. the Chiron polysaccharide vaccine against serogroup C meningitis), and used for immunisation. Good immune responses are observed.
It will be understood that the invention has been described by way of example only and modifications may be made whilst remaining within the scope and spirit of the invention.

Claims

1. A protein comprising one or more of the following amino acid sequences: SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 11, SEQ ID 12, SEQ ID 13, and SEQ IDs 14 to 104.
2. A protein comprising a sequence having greater than 50% sequence identity to SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 11, SEQ ID 12, or SEQ ID 13.
3. A protein comprising a fragment of at least 7 consecutive amino acids from SEQ ID 1, SEQ ID 3, SEQ ID 4, SEQ ID 5, SEQ ID 7, SEQ ID 8, SEQ ID 9, SEQ ID 1 1, SEQ ID 12, or SEQ ID 13.
4. An antibody which bind to a protein according to any preceding claim.
5. Nucleic acid encoding a protein according to any one of claims 1 to 3.
6. Nucleic acid comprising SEQ ID 2, SEQ ID 6, or SEQ ID 10.
7. Nucleic acid comprising a sequence having greater than 50% sequence identity to SEQ ID 2, SEQ ID 6, or SEQ ID 10.
8. Nucleic acid comprising a fragment of at least 10 consecutive nucleotides from SEQ ID 2, SEQ ID 6, or SEQ ID 10.
9. A composition comprising a protein according to any one of claims 1 to 3, an antibody according to claim 4, and/or nucleic acid according to any one of claims 5 to 8.
10. The composition of claim 9, further comprising immunogenic components selected from one or more of the following:
• the proteins disclosed in WO99/57280 or immunogenic fragments thereof;
• the proteins disclosed in WO99/36544 or immunogenic fragments thereof;
• the proteins disclosed in WO99/24578 or immunogenic fragments thereof; • the proteins disclosed in WO97/28273 or immunogenic fragments thereof;
• the proteins disclosed in WO96/29412 or immunogenic fragments thereof;
• the proteins disclosed in WO95/03413 or immunogenic fragments thereof;
• the proteins disclosed in WO99/31132 or immunogenic fragments thereof;
• a protective antigen against Neisseria meningitidis serogroup A; • a protective antigen against Neisseria meningitidis serogroup C;
• a protective antigen against Neisseria meningitidis serogroup Y;
• a protective antigen against Neisseria meningitidis serogroup W;
• a protective antigen against Haemophilus influenzae; • a protective antigen against pneumococcus;
• a protective antigen against diphtheria;
• a protective antigen against tetanus;
• a protective antigen against whooping cough;
• a protective antigen against Helicobacter pylori; • a protective antigen against polio; and/or
• a protective antigen against hepatitis B virus.
11. The composition of claim 9 or claim 10 for use as a medicament.
12. The composition of claim 9 or claim 10 for use as a vaccine.
13. The use of the composition of claim 9 or claim 10 in the manufacture of a medicament for treating or preventing infection due to Neisserial bacteria.
14. A method of treating a patient, comprising administering to the patient a therapeutically effective amount of the composition of claim 9 or claim 10.
PCT/IB2000/001851 1999-11-29 2000-11-28 85kDa NEISSERIAL ANTIGEN WO2001038350A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CA2390344A CA2390344C (en) 1999-11-29 2000-11-28 85kda neisserial antigen
JP2001540113A JP4840956B2 (en) 1999-11-29 2000-11-28 85kDa Neisseria antigen
BR0015961-1A BR0015961A (en) 1999-11-29 2000-11-28 85kda neisserial antigen
EP00981520A EP1234039A2 (en) 1999-11-29 2000-11-28 85kDa NEISSERIAL ANTIGEN
AU18753/01A AU1875301A (en) 1999-11-29 2000-11-28 85kda neisserial antigen
US11/264,676 US7700119B2 (en) 1999-11-29 2005-11-01 85kDa neisserial antigen

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB9928197.4A GB9928197D0 (en) 1999-11-29 1999-11-29 85kDA antigen
GB9928197.4 1999-11-29
GB0005698.6 2000-03-09
GB0005698A GB0005698D0 (en) 2000-03-09 2000-03-09 85kDa Antigen

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10148534 A-371-Of-International 2000-11-28
US11/264,676 Division US7700119B2 (en) 1999-11-29 2005-11-01 85kDa neisserial antigen

Publications (2)

Publication Number Publication Date
WO2001038350A2 true WO2001038350A2 (en) 2001-05-31
WO2001038350A3 WO2001038350A3 (en) 2001-11-15

Family

ID=26243826

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2000/001851 WO2001038350A2 (en) 1999-11-29 2000-11-28 85kDa NEISSERIAL ANTIGEN

Country Status (13)

Country Link
US (1) US7700119B2 (en)
EP (2) EP1234039A2 (en)
JP (2) JP4840956B2 (en)
CN (1) CN1433471A (en)
AT (1) ATE460484T1 (en)
AU (1) AU1875301A (en)
BR (1) BR0015961A (en)
CA (1) CA2390344C (en)
CY (1) CY1110540T1 (en)
DE (1) DE60044005D1 (en)
DK (1) DK1741784T3 (en)
RU (1) RU2002117308A (en)
WO (1) WO2001038350A2 (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010070453A2 (en) 2008-12-17 2010-06-24 Novartis Ag Meningococcal vaccines including hemoglobin receptor
US7763589B2 (en) 1996-09-17 2010-07-27 Novartis Vaccines And Diagnostics, Inc. Compositions and methods for treating intracellular diseases
WO2010109323A1 (en) 2009-03-24 2010-09-30 Novartis Ag Adjuvanting meningococcal factor h binding protein
EP2248822A2 (en) 2001-07-27 2010-11-10 Novartis Vaccines and Diagnostics S.r.l. Meningococcus adhesins
EP2255826A2 (en) 2002-08-02 2010-12-01 GlaxoSmithKline Biologicals S.A. Neisserial vaccine compositions comprising a combination of antigens
EP2279746A2 (en) 2002-11-15 2011-02-02 Novartis Vaccines and Diagnostics S.r.l. Surface proteins in neisseria meningitidis
WO2011024072A2 (en) 2009-08-27 2011-03-03 Novartis Ag Hybrid polypeptides including meningococcal fhbp sequences
WO2011024071A1 (en) 2009-08-27 2011-03-03 Novartis Ag Adjuvant comprising aluminium, oligonucleotide and polycation
WO2011039631A2 (en) 2009-09-30 2011-04-07 Novartis Ag Expression of meningococcal fhbp polypeptides
WO2011051893A1 (en) 2009-10-27 2011-05-05 Novartis Ag Modified meningococcal fhbp polypeptides
WO2011161653A1 (en) 2010-06-25 2011-12-29 Novartis Ag Combinations of meningococcal factor h binding proteins
WO2012020326A1 (en) 2010-03-18 2012-02-16 Novartis Ag Adjuvanted vaccines for serogroup b meningococcus
WO2012032498A2 (en) 2010-09-10 2012-03-15 Novartis Ag Developments in meningococcal outer membrane vesicles
WO2012153302A1 (en) 2011-05-12 2012-11-15 Novartis Ag Antipyretics to enhance tolerability of vesicle-based vaccines
WO2013098589A1 (en) 2011-12-29 2013-07-04 Novartis Ag Adjuvanted combinations of meningococcal factor h binding proteins
WO2013113917A1 (en) 2012-02-02 2013-08-08 Novartis Ag Promoters for increased protein expression in meningococcus
WO2013132040A2 (en) 2012-03-08 2013-09-12 Novartis Ag In vitro potency assay for protein-based meningococcal vaccines
US8563007B1 (en) 2001-10-11 2013-10-22 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
WO2014037472A1 (en) 2012-09-06 2014-03-13 Novartis Ag Combination vaccines with serogroup b meningococcus and d/t/p
US8986710B2 (en) 2012-03-09 2015-03-24 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
EP3017826A1 (en) 2009-03-24 2016-05-11 Novartis AG Combinations of meningococcal factor h binding protein and pneumococcal saccharide conjugates
US9556240B2 (en) 2010-08-23 2017-01-31 Wyeth Llc Stable formulations of Neisseria meningitidis rLP2086 antigens
US9757443B2 (en) 2010-09-10 2017-09-12 Wyeth Llc Non-lipidated variants of Neisseria meningitidis ORF2086 antigens
US9802987B2 (en) 2013-03-08 2017-10-31 Pfizer Inc. Immunogenic fusion polypeptides
US9822150B2 (en) 2013-09-08 2017-11-21 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10183070B2 (en) 2017-01-31 2019-01-22 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10196429B2 (en) 2012-03-09 2019-02-05 Pfizer Inc. Neisseria meningitidis composition and methods thereof
US10888611B2 (en) 2015-02-19 2021-01-12 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
EP3782643A1 (en) 2014-02-28 2021-02-24 GlaxoSmithKline Biologicals SA Modified meningococcal fhbp polypeptides
WO2023067031A2 (en) 2021-10-21 2023-04-27 Glaxosmithkline Biologicals Sa Assay

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2270030E (en) * 2000-02-28 2012-07-24 Novartis Vaccines & Diagnostic Heterologous expression of neisserial proteins
AU2003274511B2 (en) * 2002-10-11 2009-06-04 Glaxosmithkline Biologicals S.A. Polypeptide-vaccines for broad protection against hypervirulent meningococcal lineages
GB0408977D0 (en) * 2004-04-22 2004-05-26 Chiron Srl Immunising against meningococcal serogroup Y using proteins
US9148335B2 (en) * 2008-09-30 2015-09-29 Qualcomm Incorporated Third party validation of internet protocol addresses

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990006696A2 (en) * 1988-12-19 1990-06-28 Praxis Biologics, Inc. Meningococcal class 1 outer-membrane protein vaccine
WO1997028273A1 (en) * 1996-02-01 1997-08-07 North American Vaccine, Inc. Expression of group b neisseria meningitidis outer membrane (mb3) protein from yeast and vaccines

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9313045D0 (en) 1993-06-24 1993-08-11 Source Vision Technology The L Printing and laminating
US5439808A (en) 1993-07-23 1995-08-08 North American Vaccine, Inc. Method for the high level expression, purification and refolding of the outer membrane group B porin proteins from Neisseria meningitidis
IL117483A (en) 1995-03-17 2008-03-20 Bernard Brodeur Proteinase k resistant surface protein of neisseria meningitidis
EP1900818A3 (en) 1997-11-06 2008-06-11 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigens
GB9726398D0 (en) 1997-12-12 1998-02-11 Isis Innovation Polypeptide and coding sequences
CA2317815A1 (en) 1998-01-14 1999-07-22 Chiron S.P.A. Neisseria meningitidis antigens
EP2261343A3 (en) 1998-05-01 2012-01-18 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
JP2004511201A (en) * 1998-10-09 2004-04-15 カイロン コーポレイション Neisseria genome sequences and methods of using them
US6610306B2 (en) * 1998-10-22 2003-08-26 The University Of Montana OMP85 protein of neisseria meningitidis, compositions containing the same and methods of use thereof
EP1123403A1 (en) * 1998-10-22 2001-08-16 The University Of Montana OMP85 PROTEINS OF $i(NEISSERIA GONORRHOEAE) AND $i(NEISSERIA MENINGITIDIS), COMPOSITIONS CONTAINING SAME AND METHODS OF USE THEREOF
DE60015084T2 (en) * 1999-02-26 2006-02-16 Chiron S.R.L. IMPROVEMENT OF BACTERIC ACIDITY OF NEISSERIA ANTIGENES CG-CONTAINING OLIGONUCLEOTIDES

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990006696A2 (en) * 1988-12-19 1990-06-28 Praxis Biologics, Inc. Meningococcal class 1 outer-membrane protein vaccine
WO1997028273A1 (en) * 1996-02-01 1997-08-07 North American Vaccine, Inc. Expression of group b neisseria meningitidis outer membrane (mb3) protein from yeast and vaccines

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MANNING D S ET AL: "OMP85 PROTEINS OF NEISSERIA GONORRHOEAE AND NEISSERIA MENINGITIDIS ARE SIMILAR TO HAEMOPHILUS INFLUENZAE D-15-AG AND PASTEURELLA MULTOCIDA OMA87" MICROBIAL PATHOGENESIS,US,ACADEMIC PRESS LIMITED, NEW YORK, NY, vol. 25, no. 1, 1998, pages 11-21, XP000857391 ISSN: 0882-4010 *

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7763589B2 (en) 1996-09-17 2010-07-27 Novartis Vaccines And Diagnostics, Inc. Compositions and methods for treating intracellular diseases
EP2248822A2 (en) 2001-07-27 2010-11-10 Novartis Vaccines and Diagnostics S.r.l. Meningococcus adhesins
US9168293B2 (en) 2001-10-11 2015-10-27 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
US9757444B2 (en) 2001-10-11 2017-09-12 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
US9623101B2 (en) 2001-10-11 2017-04-18 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
US9132182B2 (en) 2001-10-11 2015-09-15 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
US9107873B2 (en) 2001-10-11 2015-08-18 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
US10300122B2 (en) 2001-10-11 2019-05-28 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
US8563006B2 (en) 2001-10-11 2013-10-22 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
US8563007B1 (en) 2001-10-11 2013-10-22 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
US11116829B2 (en) 2001-10-11 2021-09-14 Wyeth Holdings Llc Immunogenic compositions for the prevention and treatment of meningococcal disease
EP2255826A2 (en) 2002-08-02 2010-12-01 GlaxoSmithKline Biologicals S.A. Neisserial vaccine compositions comprising a combination of antigens
EP2279746A2 (en) 2002-11-15 2011-02-02 Novartis Vaccines and Diagnostics S.r.l. Surface proteins in neisseria meningitidis
WO2010070453A2 (en) 2008-12-17 2010-06-24 Novartis Ag Meningococcal vaccines including hemoglobin receptor
WO2010109323A1 (en) 2009-03-24 2010-09-30 Novartis Ag Adjuvanting meningococcal factor h binding protein
EP3017826A1 (en) 2009-03-24 2016-05-11 Novartis AG Combinations of meningococcal factor h binding protein and pneumococcal saccharide conjugates
EP3017828A1 (en) 2009-08-27 2016-05-11 GlaxoSmithKline Biologicals SA Hybrid polypeptides including meningococcal fhbp sequences
WO2011024072A2 (en) 2009-08-27 2011-03-03 Novartis Ag Hybrid polypeptides including meningococcal fhbp sequences
WO2011024071A1 (en) 2009-08-27 2011-03-03 Novartis Ag Adjuvant comprising aluminium, oligonucleotide and polycation
WO2011039631A2 (en) 2009-09-30 2011-04-07 Novartis Ag Expression of meningococcal fhbp polypeptides
WO2011051893A1 (en) 2009-10-27 2011-05-05 Novartis Ag Modified meningococcal fhbp polypeptides
WO2012020326A1 (en) 2010-03-18 2012-02-16 Novartis Ag Adjuvanted vaccines for serogroup b meningococcus
WO2011161653A1 (en) 2010-06-25 2011-12-29 Novartis Ag Combinations of meningococcal factor h binding proteins
US9556240B2 (en) 2010-08-23 2017-01-31 Wyeth Llc Stable formulations of Neisseria meningitidis rLP2086 antigens
US10512681B2 (en) 2010-09-10 2019-12-24 Wyeth Llc Non-lipidated variants of Neisseria meningitidis ORF2086 antigens
US11077180B2 (en) 2010-09-10 2021-08-03 Wyeth Llc Non-lipidated variants of Neisseria meningitidis ORF2086 antigens
WO2012032498A2 (en) 2010-09-10 2012-03-15 Novartis Ag Developments in meningococcal outer membrane vesicles
US9757443B2 (en) 2010-09-10 2017-09-12 Wyeth Llc Non-lipidated variants of Neisseria meningitidis ORF2086 antigens
WO2012153302A1 (en) 2011-05-12 2012-11-15 Novartis Ag Antipyretics to enhance tolerability of vesicle-based vaccines
WO2013098589A1 (en) 2011-12-29 2013-07-04 Novartis Ag Adjuvanted combinations of meningococcal factor h binding proteins
US10596246B2 (en) 2011-12-29 2020-03-24 Glaxosmithkline Biological Sa Adjuvanted combinations of meningococcal factor H binding proteins
US9657297B2 (en) 2012-02-02 2017-05-23 Glaxosmithkline Biologicals Sa Promoters for increased protein expression in meningococcus
WO2013113917A1 (en) 2012-02-02 2013-08-08 Novartis Ag Promoters for increased protein expression in meningococcus
WO2013132040A2 (en) 2012-03-08 2013-09-12 Novartis Ag In vitro potency assay for protein-based meningococcal vaccines
US11209436B2 (en) 2012-03-08 2021-12-28 Glaxosmithkline Biologicals Sa Vitro potency assay for protein-based meningococcal vaccines
US10598666B2 (en) 2012-03-08 2020-03-24 Glaxosmithkline Biologicals Sa In vitro potency assay for protein-based meningococcal vaccines
US8986710B2 (en) 2012-03-09 2015-03-24 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10196429B2 (en) 2012-03-09 2019-02-05 Pfizer Inc. Neisseria meningitidis composition and methods thereof
US11472850B2 (en) 2012-03-09 2022-10-18 Pfizer Inc. Neisseria meningitidis composition and methods thereof
US10550159B2 (en) 2012-03-09 2020-02-04 Pfizer Inc. Neisseria meningitidis composition and methods thereof
US10829521B2 (en) 2012-03-09 2020-11-10 Pfizer Inc. Neisseria meningitidis composition and methods thereof
US9561269B2 (en) 2012-03-09 2017-02-07 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US9724402B2 (en) 2012-03-09 2017-08-08 Pfizer Inc. Neisseria meningitidis composition and methods thereof
WO2014037472A1 (en) 2012-09-06 2014-03-13 Novartis Ag Combination vaccines with serogroup b meningococcus and d/t/p
US9526776B2 (en) 2012-09-06 2016-12-27 Glaxosmithkline Biologicals Sa Combination vaccines with serogroup B meningococcus and D/T/P
US9802987B2 (en) 2013-03-08 2017-10-31 Pfizer Inc. Immunogenic fusion polypeptides
US11680087B2 (en) 2013-09-08 2023-06-20 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US9822150B2 (en) 2013-09-08 2017-11-21 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10899802B2 (en) 2013-09-08 2021-01-26 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
EP3782643A1 (en) 2014-02-28 2021-02-24 GlaxoSmithKline Biologicals SA Modified meningococcal fhbp polypeptides
US10888611B2 (en) 2015-02-19 2021-01-12 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10813989B2 (en) 2017-01-31 2020-10-27 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10543267B2 (en) 2017-01-31 2020-01-28 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US10183070B2 (en) 2017-01-31 2019-01-22 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US11730800B2 (en) 2017-01-31 2023-08-22 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
WO2023067031A2 (en) 2021-10-21 2023-04-27 Glaxosmithkline Biologicals Sa Assay

Also Published As

Publication number Publication date
BR0015961A (en) 2003-06-10
JP2011055838A (en) 2011-03-24
JP4840956B2 (en) 2011-12-21
US20060233827A1 (en) 2006-10-19
DK1741784T3 (en) 2010-05-25
CN1433471A (en) 2003-07-30
CA2390344A1 (en) 2001-05-31
RU2002117308A (en) 2004-03-10
EP1741784B1 (en) 2010-03-10
US7700119B2 (en) 2010-04-20
DE60044005D1 (en) 2010-04-22
CY1110540T1 (en) 2015-04-29
JP2003514575A (en) 2003-04-22
EP1234039A2 (en) 2002-08-28
ATE460484T1 (en) 2010-03-15
WO2001038350A3 (en) 2001-11-15
EP1741784A1 (en) 2007-01-10
CA2390344C (en) 2014-08-26
AU1875301A (en) 2001-06-04

Similar Documents

Publication Publication Date Title
US7700119B2 (en) 85kDa neisserial antigen
EP2275554B1 (en) Neisserial antigenic peptides
CA2871789C (en) Outer membrane vesicle (omv) vaccine comprising n. meningitidis serogroup b outer membrane proteins
EP1196587B1 (en) Antigenic meningococcal peptides
AU2006200732B2 (en) Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW 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)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2390344

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2001 540113

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/A/2002/005325

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2000981520

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 18753/01

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 519716

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2002 2002117308

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 008185719

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2000981520

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2000981520

Country of ref document: EP