WO2002026209A2 - Microparticles for delivery of the heterologous nucleic acids - Google Patents

Microparticles for delivery of the heterologous nucleic acids Download PDF

Info

Publication number
WO2002026209A2
WO2002026209A2 PCT/US2001/030540 US0130540W WO0226209A2 WO 2002026209 A2 WO2002026209 A2 WO 2002026209A2 US 0130540 W US0130540 W US 0130540W WO 0226209 A2 WO0226209 A2 WO 0226209A2
Authority
WO
WIPO (PCT)
Prior art keywords
microparticle
antigen
seq
microparticles
vector construct
Prior art date
Application number
PCT/US2001/030540
Other languages
French (fr)
Other versions
WO2002026209A3 (en
Inventor
Derek O'hagan
Gillis Otten
John James Donnelly
John M. Polo
Susan Barnett
Manmohan Singh
Jeffrey Ulmer
Thomas W. Dubensky, Jr.
Original Assignee
Chiron Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chiron Corporation filed Critical Chiron Corporation
Priority to BR0114305-0A priority Critical patent/BR0114305A/en
Priority to EP01975584A priority patent/EP1322287B1/en
Priority to CN018164595A priority patent/CN1468089B/en
Priority to AU9489701A priority patent/AU9489701A/en
Priority to DE60118228T priority patent/DE60118228T2/en
Priority to CA002421683A priority patent/CA2421683C/en
Priority to JP2002530039A priority patent/JP2004518631A/en
Priority to MXPA03002640A priority patent/MXPA03002640A/en
Publication of WO2002026209A2 publication Critical patent/WO2002026209A2/en
Publication of WO2002026209A3 publication Critical patent/WO2002026209A3/en
Priority to ZA2003/01744A priority patent/ZA200301744B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • A61K9/1647Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/167Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6093Synthetic polymers, e.g. polyethyleneglycol [PEG], Polymers or copolymers of (D) glutamate and (D) lysine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36141Use of virus, viral particle or viral elements as a vector
    • C12N2770/36143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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

  • Adjuvants are compounds which are capable of potentiating an irnmune response to antigens. Adjuvants can potentiate both humoral and cellular immunity. However, it is preferable for certain pathogens to stimulate cellular immunity and, particularly, Thl cells. In many instances, presently used adjuvants do not adequately induce Thl cell responses, and/or have deleterious side effects.
  • cationic Upid-based emulsions may be used as gene carriers. See, e.g., Yi et al., Proc. IntJ Symp. Control. Rel. Bioact. Mater., 24:653-654 (1997); Kim et al., Proc. IntJ Symp. Control. Rel. Bioact. Mater., 25:344-345 (1998); Kim et al., Proc. IntJ Symp. Control. Rel. Bioact. Mater., 26, #5438 (1999).
  • Cationic submicron emulsions a somewhat recent approach to pharmaceutical deUvery, were first shown to have carrying capacity for small molecule drugs (Elbaz et al 1993 Int. J.
  • U.S. Patents 5,814,482 and 6,015,686 disclose Eukaryotic Layered Vector Initiation Systems (ELVIS vectors), particularly those derived and constructed from alphavirus genomes (such as Sindbis virus), for use in stimulating an immune response to an antigen, in methods of inhibiting pathogenic agents, and in delivery of heterologous nucleotide sequences to eukaryotic cells and animals, among others.
  • ELVIS vectors Eukaryotic Layered Vector Initiation Systems
  • the antigenic polypeptide is from a virus such as, for example, hepatitis C virus (HCV), hepatitis B virus (HBV), herpes simplex virus (HSV), human irnmunodeficiency virus (HIV), cytomegalovirus (CMV), influenza virus (flu), and rabies virus.
  • HCV hepatitis C virus
  • HBV hepatitis B virus
  • HSV herpes simplex virus
  • HV human irnmunodeficiency virus
  • CMV cytomegalovirus
  • influenza virus flu
  • rabies virus hepatitis C virus
  • the antigenic polypeptide is selected from the group consisting of HSV glycoproteingD, HIV glycoprotein gpl20, HTV glycoprotein gpl40, HIV p55 gag, and polypeptides from the pol and tat regions.
  • the oligonucleotide comprises a CpG motif flanked by two purines immediately 5' to the motif and two pyrimidines immediately 3' to the motif
  • the oligonucleotide comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 19-28. Most preferred is SEQ ID NO:28.
  • the adjuvant composition further comprises a separate immunostimulating agent which is preferably selected from the group consisting of alum, a bacterial ceU wall component, and muramyl peptide.
  • the adjuvant composition itself may be in the form of a second microparticle.
  • microparticle refers to one or more microparticles, and the Uke.
  • Polymer microparticles for use herein are formed from materials that are ster ⁇ izable, non- toxic and biodegradable. Such materials include, without Umitation, poly( ⁇ -hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate.
  • Cationic detergents include, but are not limited to, cetrimide (cetyl trimethyl ammonium bromide, or "CTAB”), benzalkonium chloride, DDA (dimethyl dioctodecyl ammonium bromide), DOTAP(dioleoyl-3-trimethylammonium-propane), and the like.
  • Nonionic detergents include, but are not limited to, PVA, povidone (also known as polyvinylpyrroUdone or PVP), sorbitan esters, polysorbates, polyoxyethylated glycol monoethers, polyoxyethylated alkyl phenols, poloxamers, and the like.
  • macromolecule refers to, without Umitation, a pharmaceutical, a polynucleotide, a polypeptide, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof.
  • a pharmaceutical a polynucleotide, a polypeptide, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof.
  • conformational epitopes are readily identified by detern ⁇ ig spatial conformation of amino acids such as by, e.g., x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols, supra.
  • Such enhanced immunogenicity can be determined by aiiministering the microparticle/antigen composition, and antigen controls to animals and comparing antibody liters against the two using standard assays such as radioimmuno assay and ELISAs, weU known in the art.
  • vertebrate subject any member of the subphylum cordata, including, without Umitation, nDimimals such as cattle, sheep, pigs, goats, horses, and humans; domestic animals such as dogs and cats; and birds, including domestic, wild and game birds such as cocks and hens including chickens, turkeys and other gallinaceous birds.
  • the term does not denote a particular age. Thus, both adult and newborn animals are intended to be covered.
  • pharmaceutically acceptable or “pharmacologicaUy acceptable” is meant a material which is not biologicaUy or otherwise undesirable, i.e., the material may be administered to an individual along with the microparticle formulation without causing any undesirable biological effects in the individual or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • An alphavirus-derived RNA vector repUcon should contain the foUowing ordered elements: 5' viral sequences required in cis for repUcation (also referred to as 5' CSE), sequences which, when expressed, code for biologicaUy active alphavirus nonstructural proteins (e.g., nsPl, nsP2, nsP3, nsP4), 3' viral sequences required in cis for repUcation (also referred to as 3' CSE), and a polyadenylate tract.
  • 5' viral sequences required in cis for repUcation also referred to as 5' CSE
  • sequences which, when expressed, code for biologicaUy active alphavirus nonstructural proteins e.g., nsPl, nsP2, nsP3, nsP4
  • 3' viral sequences required in cis for repUcation also referred to as 3' CSE
  • a polyadenylate tract also referred to as 3' CSE
  • An alphavirus-derived RNA vector repUcon also may contain a viral subgenomic "junction region" promoter, sequences from one or more structural protein genes or portions thereof, extraneous nucleic acid molecule(s) which are of a size sufficient to aUow production of viable virus, as weU as heterologous sequence(s) to be expressed.
  • the vector construct may include a viral subgenomic "junction region" promoter, sequences from one or more structural protein genes or portions thereof, extraneous nucleic acid molecule(s) which are of a size sufficient to aUow production of viable virus, a 5' promoter which is capable of initiating the synthesis of viral RNA from cDNA in vitro or in vivo, a heterologous sequence to be expressed, and one or more restriction sites for insertion of heterologous sequences.
  • a viral subgenomic "junction region" promoter sequences from one or more structural protein genes or portions thereof, extraneous nucleic acid molecule(s) which are of a size sufficient to aUow production of viable virus, a 5' promoter which is capable of initiating the synthesis of viral RNA from cDNA in vitro or in vivo, a heterologous sequence to be expressed, and one or more restriction sites for insertion of heterologous sequences.
  • the vector construct may also optionaUy include a signal which directs pblyadenylation, a selectable marker, as weU as one or more restriction sites and a translation termination sequence.
  • the vector construct may include a packaging signal, long terminal repeats (LTRs), and positive and negative strand primer binding sites appropriate to the retrovirus used (if these are not already present).
  • LTRs long terminal repeats
  • vector constructs include ELVIS vectors, which comprise the cDNA complement of RNA vector constructs, RNA vector constructs themselves, alphavirus vector constructs, CMV vector constructs and the like.
  • a wide variety of macromolecules can be delivered in association with the microparticles including, but not limited to, pharmaceuticals such as antibiotics and antiviral agents, nonsteroidal antiinflammatory drugs, analgesics, vasodilators, cardiovascular drugs, psychotiopics, neuroleptics, antidepressants, antiparkinson drugs, beta blockers, calcium channel blockers, bradykinin inhibitors, ACE-inbibitors, vasodilators, prolactin inhibitors, steroids, hormone antagonists, antihistamines, serotonin antagonists, heparin, chemotherapeutic agents, antineoplastics and growth factors, including but not limited to PDGF, EGF, KGF, IGF-1 and IGF-2, FGF, polynucleotides which encode therapeutic or immunogenic proteins, immunogenic proteins and epitopes thereof for use in vaccines, hormones including peptide hormones such as insulin, promsulin, growth hormone, GHRH, LHRH, EGF, somatostatin
  • the invention is equaUy appUcable to other immunogenic proteins derived from any of the various HIV isolates, including any of the various envelope proteins such as gpl60 and gp41, gag antigens such as p24gag and p55gag, as weU as proteins derived from the pol and tat regions. Any of these proteins and antigens may also be modified for use in the present invention.
  • Figures 1, 2, 5, and 6 provide DNA sequences encoding modified gag antigens (SEQ ID NOs: 63, 64, 67, and 68), and
  • Figures 3 and 4 provide DNA sequences encoding modified envelope antigens (SEQ ID NOs: 65 and 66).
  • antigens from Neisseria meningitides B are disclosed in the foUowing co-owned patent appUcations: PCT/US99/09346; PCT IB98/01665; and PCT IB99/00103.
  • parasitic antigens include those derived from organisms causing malaria and Lyme disease.
  • a saccharide antigen fromN meningitidis serogroup A, C, W135 and/or Y such as the oUgosaccharide disclosed in Ref 12 below from serogroup C (see also Ref. 13).
  • microparticles manufactured in the presence of charged detergents may yield microparticles with a surface having a net negative or a net positive charge, which can adsorb a wide variety of molecules.
  • microparticles manufactured with anionic detergents such as sodium dodecyl sulfate (SDS), i.e. SDS-PLG microparticles, adsorb positively charged antigens, such as proteins.
  • microparticles manufactured with cationic detergents such as hexadecyltrimethylammonium bromide (CTAB), i.e. CTAB-PLG microparticles, adsorb negatively charged macromolecules, such as DNA. Where the macromolecules to be adsorbed have regions of positive and negative charge, cationic, anionic, nonionic or zwitterioinic detergents may be appropriate.
  • PLG copolymers with varying lactide:glycoUde ratios and molecular weights are readUy available commerciaUy from a number of sources including from Boehringer Ingelheim, Germany and Birmingham Polymers, Inc., Birmingham, AL. These polymers can also be synthesized by simple polycondensation of the lactic acid component using techniques weU known in the art, such as described in Tabata et al., J. Biomed. Mater. Res. (1988) 22:837-858.
  • the mixture is then homogenized to produce a stable w/o/w double emulsion.
  • Organic solvents are then evaporated.
  • the formulation parameters can be manipulated to allow the preparation of smaU microparticles on the order of 0.05 ⁇ m (50 nm) to larger microparticles 50 ⁇ m or even larger. See, e.g., jeffery et al., Pharm. Res. (1993) 10:362-368; McGee et al., J. Microencap. (1996).
  • reduced agitation results in larger microparticles, as does an increase in internal phase volume.
  • SmaU particles are produced by low aqueous phase volumes with high concentrations of emulsion stabilizers.
  • oU droplet emulsions include, for example, SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and Ribi® adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial ceU wall components from the group consisting of monophosphoryUpid A (MPL), trehalose dirnycolate (TDM), and ceU waU skeleton (CWS), preferably MPL + CWS (DetoxJ) (for a further discussion of suitable submicron ou-in- water emulsions for use herein, see commonly owned, patent appUcation no. 09/015,736, filed on January 29, 1998).
  • RFS Ribi® adjuvant system
  • MPL mono
  • micromolecules such as polypeptides and vector constructs may be adsorbed thereto as previously discussed.
  • the submicron emulsion microparticles of the present invention may also have macromolecules entrapped or encapsulated within them, as well as having macromolecules adsorbed thereon.
  • one of skiU in the art may prepare in accordance with the invention microparticles having encapsulated adjuvants with ELVIS vector adsorbed thereon,. or microparticles having encapsulated antigen with RNA vector construct adsorbed thereon.
  • the ELVIS vector capitalizes on the mechanism of RNA virus repUcation to achieve deUvery of a heterologous nucleotide sequence of interest by using a double-layered approach (for example, based on the above-described alphavirus vector construct).
  • an ELVIS vector provides a layered expression system capable of ampUfying the amount of RNA encoding the gene product of interest because the first layer initiates transcription of a second layer.
  • An alphavirus-based ELVIS vector may be constructed by first preparing a cDNA complementary to an alphavirus genome.
  • the cDNA corresponding to the genomic RNA is then deleted of sequences encoding one or more viral structural proteins which then may be replaced with heterologous DNA encoding the gene-product of interest, thereby preventing packaging of mature virus and enabling amplification of the heterologous sequence.
  • the modified cDNA containing the heterologous sequence is then inserted within the first layer of the ELVIS vector.
  • RNA is obtained from an RNA virus, then cDNA is synthesized by PCR ampUfication using appropriate primers for particular genes or portions of the RNA virus, which primers may also contain additional restriction sites as necessary.
  • the cDNA fragments are then cloned into a plasmid and transformed into an appropriate host such as E. coli. Positive colonies are grown for plasmid purification, and then plasmids are assembled into the desired ELVIS vector with a portion having heterologous DNA such as a reporter gene (e.g., GFP) or a desired gene coding for an antigen.
  • pSINCP a particular preferred ELVIS vector used in accordance with the instant invention.
  • W099/52549 (10) a polyoxyethylene sorbitan ester surfactant in combination with an octoxynol (WOO 1/21207) or a polyoxyethylene alkyl ether or ester surfactant in combination with at least one additional non-ionic surfactant such as an octoxynol (WO01/21152); (11) a saponin and an iiimiunostimulatory oUgonucleotide (e.g., a CpG oligonucleotide) (WO00/62800); (12 ) an immunostimulant and a particle of metal salt e.g.
  • Muramyl peptides include, but are not limited to, N-acetyl-muramyl-L-threonyl-D- isoglutamine (thr-MDP), N-acteyl-normuramyl-L-alanyl-D-isogluatme (nor-MDP), N- acetylmuramyl-L-alanyl-D-isogluatmmy huydroxyphosphoryloxy)-ethylamine (MTP-PE), etc.
  • thr-MDP N-acetyl-muramyl-L-threonyl-D- isoglutamine
  • nor-MDP N-acteyl-normuramyl-L-alanyl-D-isogluatme
  • MTP-PE N-acetylmuramyl-L-alanyl-D-isogluatmmy huydroxyphosphoryloxy
  • an optional additional component of the compositions of the present invention preferably is an adjuvant such as aluminum salts or an oUgonucleotide which comprises at least one CpG motif.
  • an adjuvant such as aluminum salts or an oUgonucleotide which comprises at least one CpG motif.
  • CpG motif' refers to a dinucleotide portion of an oligonucleotide which comprises a cytosine nucleotide followed by a guanosine nucleotide.
  • Such oUgonucleotides can be prepared using conventional oUgonucleotide synthesis weU known to the slatted artisan.
  • the oUgonucleotide preferably comprises between about 6 and about 100 nucleotides, more preferably between about 8 and about 50 nucleotides, most preferably between about 10 and about 40 nucleotides.
  • the oUgonucleotides of the invention can comprise substitutions of the sugar moieties and nitrogenous base moieties.
  • Preferred oUgonucleotides are disclosed in, for example, Krieg et al, Proc. Natl. Acad. Sci. USA, 1998, 95, 12631-12636, Klinman et al, Proc. Natl. Acad. Sci. USA, 1996, 93, 2879-2883, Weiner et al, Proc. Natl. Acad. Sci.
  • An epitope is within the scope of this definition of antigen.
  • An epitope is that portion of an antigenic molecule or antigenic complex that determines its immunological specificity. Commonly, an epitope is a peptide or polysaccharide in naturally occurring antigens. In artificial antigens it can be a low molecular weight substance such as an arsaniUc acid derivative.
  • An epitope will react specifically in vivo or in vitro with homologous antibodies or T lymphocytes.
  • Alternative descriptors are antigenic determinant, antigenic structural grouping and haptenic grouping.
  • a composition comprising an antigen wiU be used to boost the unmunological response of a previously ac ⁇ ministered vector construct, which preferably comprises a heterologous nucleic acid sequence that encodes the antigen. More preferably, the antigen is associated with (e.g., adsorbed to) the microparticles described herein and/or the antigen is coadministered with an adjuvant.
  • wfll provide specific guidance for each and every antigen which can be employed in this invention.
  • the effective amount of antigen wiU be a function of its inherent activity and purity and is empirically determined by those of ordinary skiU in the art via routine experimentation. It is contemplated that the adjuvant compositions of this invention can be used in conjunction with whole cell or viral immunogenic compositions as weU as with purified antigens or protein subunit or peptide immunogenic compositions prepared by recombinant DNA techniques or synthesis.
  • the immunogenic compositions contain about 500 micrograms to about 1 miUigram of nucleic acid.
  • One skiUed in the art can readily formulate an immunogenic composition comprising any desired amount of nucleic acid.
  • the immunogenic compositions according to the present invention are provided sterile and pyrogen free.
  • the immunogenic compositions can be conveniently administered in unit dosage form and can be prepared by any of the methods weU known in the pharmaceutical art, for example, as described in Remington 's Pharmaceutical Sciences (Mack Pub. Co., Easton, PA, 1980), the disclosure of which is incorporated herein by reference in its entirety.
  • the present invention is also directed to methods of stimulating an immune response in a host animal comprising administering to the animal one or more immunogenic compositions described above in an amount effective to induce an immune response.
  • the host animal is preferably a mammal, more preferably a human.
  • Preferred routes of administration include, but are not limited to, intramuscular, intraperitoneal, intradermal, subcutaneous, intravenous, intraarterial, intraoccular and oral as weU as transdermal or by inhalation or suppository. Most preferred routes of administration include intramuscular, intraperitoneal, intradermal and subcutaneous injection.
  • the immunogenic compositions are administered to a host animal using a needleless injection device, which are wett known and widely available.
  • a needleless injection device which are wett known and widely available.
  • One having ordinary skill in the art can, foUowing the teachings herein, use needleless injection devices to dehver immunogenic compositions to cells of an individual. Additionally, the embodiments of the invention may be deUvered together with
  • Sindbis vector construct which contains sequences derived from the SINDCchiron virus strain and also encodes a GFP reporter, was designated SINCR-GFP (also known as DCSP6SINgfp).
  • SINCR-GFP also known as DCSP6SINgfp
  • Preparation of replicon RNA from this reporter construct, as weU as Sindbis vector constructs expressing various other heterologous sequences (e.g., antigens, described in the specification and below) was performed by linearization of the DNA using Pmel, foUowed by in vitro transcription using bacteriophage SP6 polymerase as described previously (Polo et al., ibid; Dubensky et al., ibid).
  • the plasmid backbone first was modified by substituting the plasmid backbone with that frompCMVLink (zur Megede et al., J. Virol. 74:2628-2635, 2000) using two S ⁇ cl sites found in each plasmid, to generate the intermediate construct known as ELVIS1.5CB.
  • Group 5 used pSINCP- gpl40 and pSINCP - p55 gag adsorbed on PLG/CTAB microparticles.
  • PBMC peripheral blood mononuclear ceUs
  • PBMC peripheral blood mononuclear ceUs
  • RNA vector constructs include, but are not Umited to, alphavirus RNA vectors (see for example, US Patent 5843723, PCT pubUcation WO 99/18226, and Polo et al., 1999, PNAS 96:4598-4603), picornavirus RNA vectors (see for example, US Patent 6156538, and Vignuzzi et al., 2001, J Gen Virol. 82:1737-47), flavivirus RNA vectors (see for example, Vamavski et al. 1999, Virology 255:366-75), and rabivirus RNA vectors (see for example, Pugachev et al., 2000, J Virol.
  • RNA vector constructs for use in the present invention generaUy may be obtained from plasmid cDNA constructs as a source of starting material, by the standard process of in vitro transcription (see references above). Similarly to plasmid DNA, these RNA vector constructs then may be adsorbed to microparticles of the invention as described elsewhere in the examples . For example, the RNA vector constructs are adsorbed onto the microparticles by incubating 100 mg of cationic microparticles in a 1 mg/ml solution of DNA at 4°C for 6 hr. The microparticles are then separated by centrifugation, the peUet washed with TE buffer, and the microparticles freeze-dried.
  • Example 6 Adjuvants in Mice An experiment with mice was performed to analyze the effect of the adjuvant aluminum phosphate (alum) in mice. Polymer microparticles were prepared as above described with or without pCMV-p55gag adsorbed thereon. 10 micrograms of DNA, whether naked or adsorbed to the PLG microparticles was injected in groups of 6 CB6 FI mice on weeks 0 and 6, without or without alum The results, as geometric mean titers of antibody, are shown below in Table 9.
  • Example 8 Induction of Immune Response in Rhesus with Prime and Boost Immunizations An experiment was performed to determine the effect of priming with DNA and boosting with protein adsorbed to PLG microparticles. Particularly, PLG-SDS microparticles were prepared as ' described above and in commonly owned International patent appUcation PCT/US99/17308, and purified recombinant p55gag protein was adsorbed thereto. A group of animals was immunized with 1 mg pCMV-p55gag. The animals were immunized again at 4 weeks, then again at 8 weeks. The animals were boosted with p55gag protein adsorbed to PLG microparticles at 41 weeks.
  • results are shown in Table 10 below, which shows antibody titer for responders, induction of helper T ceU lymphoproUferation (mean stimulation index of responders), and induction of CTL (number of responders, based on greater than 10% lysis at two or more consecutive E:T ratios). Results were measured at 14 weeks post 3 rd prime for the prime columns, and 2 weeks post boost for the boost columns. Numbers in parentheses indicate the number of responders out of a total of 4 animals.
  • Plasmids encoding HIV-1 p55gag and gpl40env driven by the human cytomegalovirus (CMV) promoter were grown in Escherichia coli strain DH5 ⁇ , purified using a Qiagen Endofree Plasmid Giga kit (Qiagen, Inc.), and resuspended in 0.9% sodium chloride (Abbott Laboratories, North Chicago, IU.).
  • the pCMV vector used contains the immediate-early enhancer/promoter of CMV and a bovine growth hormone terminator and is described in detail elsewhere (Chapman, B. S., et al. 1991.
  • the HIV gag plasmid DNA vaccine contains a synthetically constructed p55gag gene, with codons reflecting mammaUan usage, derived from the HTV-1 SF2 strain as previously described (zur Megede, J., et al. 2000. "Increased expression and immunogenicity of sequence-modified human immunodeficiency virus type 1 gag gene.” J Virol. 74:2628-35).
  • the HIV env plasmid DNA vaccine (pCMVgpl40) consisted of a human tissue plasminogen activator (tPA) signal sequence and the gpl40 from HIV-1 SF162 strain, codon optimized for high level expression in mammaUan cells (Barnett, S. W., et. 2001. "The abiUty of an oUgomeric human immunodeficiency virus type 1 (HIV-1) envelope antigen to elicit neutralizing antibodies against primary HIV-1 isolates is improved foUowing partial deletion of the second hypervariable region.” J Virol. 75:5526-40).
  • the SINCP plasmid vector with either HIV-1 p55gag or gpl40env has been described in Example 3 above.
  • Example 11 Preparation of Proteins
  • the protein and cDNA sequences for the gpl60env.SF162 have been pubUshed in Cheng- Mayer, C, M. Quiroga, J. W. Tung, D. Dina, and J. A. Levy. 1990. 'Niral determinants of human immunodeficiency virus type 1 T-cell or macrophage tropism, cytopathogenicity, and CD4 antigen modulation.” J Virol. 64:4390-8. These sequences can be found under Genbank accession number M65024. Recombinant HTV-1 gl40.SF162(dV2) protein was expressed in Chinese hamster ovary cells and purified as previously described (Barnett, S. W., ET. 2001.
  • a pool of 51 synthetic peptides 20 amino acids (aa) long, overlapping by 10 aa, and spanning p55 gag, and a pool of 66 synthetic peptides 20 aa long, overlapping by 10 aa, and spanning gpl40 were prepared.
  • Rhesus macaque peripheral blood mononuclear ceUs (PBMC) were separated from heparinized blood by centrifugation on FicoU-Paque (Pharmacia Biotech, Piscataway, NJ.) gradients.
  • Rhesus PBMC (1 x 10 6 per weU) were cultured overnight in the presence of Brefeldin A (Pharmingen, San Diego, CA) and anti-CD28 monoclonal antibody (mAb) (Pharmingen) and in the presence or absence of the gag or env peptide pools.
  • DupUcate weUs were prepared for each condition of stimulation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Plant Pathology (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Microparticles with adsorbent surfaces, methods of making such microparticles, and uses thereof, are disclosed. The microparticles comprise a polymer, such as a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and the like, and are formed using cationic, anionic, or nonionic detergents. Also provided are microparticles in the form of submicron emulsions of an oil droplet emulsion having a metabolizable oil and an emulsifying agent. The surface of the microparticles efficiently adsorb polypeptides, such as antigens, and nucleic acids, such as ELVIS vectors and other vector constructs, containing heterologous nucleotide sequences encoding biologically active macromolecules, such as polypeptides, antigens, and adjuvants. Methods of stimulating an immune response, methods of immunizing a host animal against a viral, bacterial, or parasitic infection, and uses of the microparticle compositions for vaccines are also provided.

Description

MICROPARTICLES FOR DELIVERY OF HETEROLOGOUS NUCLEIC ACIDS
Technical Field
The present invention relates generally to pharmaceutical compositions. In particular, the invention relates to microparticles of polymers or submicron emulsions having adsorbent surfaces wherein biologically active agents, particularly nucleic acids, such as plasmid DNA, Eukaryotic Layered Vector Initiation Systems (ELVIS vectors) or RNA vector constructs, are adsorbed thereto, methods for preparing such microparticles and submicron emulsions, and uses thereof, including induction of immune responses, vaccines, and delivery of heterologous nucleotide sequences to eukaryotic cells and animals.
Background
Particulate carriers have been used in order to achieve controlled, parenteral delivery of therapeutic compounds. Such carriers are designed to maintain the active agent in the delivery system for an extended period of time. Examples of particulate carriers include those derived from polymethyl methacrylate polymers, as well as microparticles derived from poly(lactides) (see, e.g., U.S. Patent No. 3,773,919), poly(lactide-co-glycolides), known as PLG (see, e.g., U.S. Patent No. 4,767,628) and polyethylene glycol, known as PEG (see, e.g., U.S. Patent No. 5,648,095). Polymethyl methacrylate polymers are nondegradable while PLG particles biodegrade by random nonenaymatic hydrolysis of ester bonds to lactic and glycolic acids which are excreted along normal metabolic pathways.
For example, U.S. Patent No. 5,648,095 describes the use of microspheres with encapsulated pharmaceuticals as drug delivery systems for nasal, oral, pulmonary and oral delivery. Slow-release formulations containing various polypeptide growth factors have also been described. See, e.g., International Publication No. WO 94/12158, U.S. Patent No. 5,134,122 and International Publication No. WO 96/37216. Fattal et al., Journal of Controlled Release 53:137-143 (1998) describes nanoparticles prepared from polyalkylcyanoacrylates (PACA) having adsorbed oligonucleotides.
Particulate carriers, such as microparticles, have also been used with adsorbed or entrapped antigens in attempts to elicit adequate immune responses. Such carriers present multiple copies of a selected antigen to the immune system and promote trapping and retention of antigens in local lymph nodes. The particles can be phagocytosed by macrophages and can enhance antigen presentation through cytokine release. For example, commonly owned, co-pending Application No. 09/015,652, filed January 29, 1998, describes the use of antigen-adsorbed and antigen- encapsulated microparticles to stimulate cell-mediated immunological responses, as well as methods of making the microparticles.
In commonly owned Patent Application Serial No. 09/015,652, for example, a method of forming microparticles is disclosed which comprises combining a polymer with an organic solvent, then adding an emulsion stabilizer, such as polyvinyl alcohol (PVA), then evaporating the organic solvent, thereby forming microparticles. The surface of the microparticles comprises the polymer and the stabilizer. Macromolecules such as ELVIS vectors, other nucleotides (DNA or RNA), polypeptides, and antigens may then be adsorbed on those surfaces.
Adjuvants are compounds which are capable of potentiating an irnmune response to antigens. Adjuvants can potentiate both humoral and cellular immunity. However, it is preferable for certain pathogens to stimulate cellular immunity and, particularly, Thl cells. In many instances, presently used adjuvants do not adequately induce Thl cell responses, and/or have deleterious side effects.
Currently, the only adjuvants approved for human use in the United States are aluminum salts (alum). These adjuvants have been useful for some vaccines including hepatitis B, diphtheria, polio, rabies, and influenza, but may not be useful for others. For example, reports indicate that alum failed to improve the effectiveness of whooping cough and typhoid vaccines and provided only a slight effect with adenovirus vaccines. Additionally, problems such as, induction of granulomas at the injection site and lot-to-lot variation of alum preparations have been experienced.
Microparticles prepared from biodegradable and biocompatible polymers, known as the poly(lactide-co-glycolides) (PLG), have been demonstrated to be effective vehicles for a number of antigens. In addition, PLG microparticles can control the rate of release of entrapped antigens and, thus, offer potential for single-dose vaccines. Moreover, administration of biodegradable polymers with entrapped antigens has been demonstrated in a range of animal models to induce potent immune responses. O'Hagan et al, Advanced Drug Deliv. Rev., 1998, 32, 225-246 and Singh et al, Advanced Drug Deliv. Rev., 1998, 34, 285-304, the disclosures of which are incorporated herein by reference in their entirety.
An emulsion comprising squalene, sorbitan trioleate (Span85™), and polysorbate 80 (Tween 80™) microfluidized to provide uriiforrnly sized microdroplets, i.e. MF59, has also been shown to induce potent immune responses. MF59 formulations have been shown to induce antibody titers from 5 to >100 times greater than those obtained with aluminum salt adjuvants. MF59 has been demonstrated to enhance the immune response to antigens from numerous sources including, for example, herpes simplex virus (HSV), human immunodeficiency virus (HIV), influenza virus, hepatitis C virus (HCV), cytomegalovirus (CMV), hepatitis B virus (HBV), human papillomavirus (HPV), and malaria. Ott etal, Vaccine Design: The Subunit And Adjuvant Approach, 1995, M.F. Powell and MJ. Newman, Eds., Plenum Press, New York, p. 277-296; Singh et al, Vaccine, 1998, 16, 1822-1827; Ott et al, Vaccine, 1995, 13, 1557-1562; O'Hagan et al., Mol. Medicine Today, 1991, February, 69-75; and Traquina et al. , J. Infect. Dis., 1996, 174, 1168-75, the disclosures of which are incorporated herein by reference in their entirety. MF59 adjuvant improves the immunogenicity of subunit antigens while maintaining the safety and tolerability profile of alum adjuvant. Van Nest etal, Vaccines 92, 1992, Cold Spring Harbor Laboratory Press, 57-62 and Valensi et al., J. Immunol, 1994, 153, 4029-39, the disclosures of which are incorporated herein by reference in their entirety. MF59 is further described in co- pending U.S. application Serial No. 08/434,512, filed May 4, 1995, which is assigned to the assignee of the present invention, the disclosure of which is incorporated herein by reference in its entirety. In animal studies, MF59 has not been found to be genotoxic, teratogeniα, nor does it cause sensitization. The mechanism of action of MF59 appears to be dependent upon the generation of a strong CD4+ T cell, i.e., a Th2 cell response. MF59 adjuvants, however, elicit little, if any, Thl responses, or cytotoxic T lymphocyte (CTL) responses.
Oligonucleotides comprising CpG motifs mixed with antigens have been demonstrated to induce strong Thl immune responses. Roman et al, Nat. Med., 1991, 3, 849-854; Weiner et al, Proc. Natl. Acad. Sci. USA, 1991, 94, 10833-10837; Davis et al, J. Immunol, 1998, 160, 870- 876; Chu et al. , J. Exp. Med, 1991, 186, 1623-1631; Lipford et al, Eur. J. Immunol, 1991, 27, 2340-2344; and Moldoveanu et al, Vaccine, 1988, 16, 1216-1224, the disclosures of which are incorporated herein by reference in their entirety. Unmethylated CpG dinucleotides are relatively common in bacterial DNA, but are underrepresented and methylated in vertebrate DNA. Bird, Trends Genet, 1987, 3, 342-347. Bacterial DNA or synthetic ohgonucleotides containing unmethylated CpG motifs are also known to induce immune responses including, for example, B cell proliferation, interleukin-6 and immunoglobulin secretion, and apoptosis resistance. Krieg et al, Nature, 1995, 374, 546-549; Hinman etal, Proc. Natl. Acad. Sci. USA, 1996, 93, 2879-2883; Ballas etal, J. Immunol, 1996, 157, 1840-1845; Cowdery et al, J. Immunol, 1996, 156, 4570- 4575; Halpern et al, Cell. Immunol, 1996, 167, 72-78; Yamamoto et al, Jpn. J. Cancer Res., 1988, 79, 866-873; Stacey et al, J. Immunol, 1996, 157, 2116-2122; Messina et al, J. Immunol, 1991, 147, 17 '59-17 '64; Yi et al, J. Immunol, 1996, 157, 4918-4925; Yi etal., J. Immunol, 1996, 157, 5394-5402; Yi et al, J. Immunol, 1998, 160, 4755-4761; and Yi etal, J. Immunol, 1998, 160, 5898-5906; PCT PubUcation WO 96/02555; PCT PubUcation WO 98/16247; PCT PubUcation WO 98/18810; PCT PubUcation WO 98/40100; PCT PubUcation WO 98/55495; PCT PubUcation WO 98/37919; and PCT Publication WO 98/52581, the disclosures of which are incorporated herein by reference in their entirety.
It has also been shown that cationic Upid-based emulsions may be used as gene carriers. See, e.g., Yi et al., Proc. IntJ Symp. Control. Rel. Bioact. Mater., 24:653-654 (1997); Kim et al., Proc. IntJ Symp. Control. Rel. Bioact. Mater., 25:344-345 (1998); Kim et al., Proc. IntJ Symp. Control. Rel. Bioact. Mater., 26, #5438 (1999). Cationic submicron emulsions, a somewhat recent approach to pharmaceutical deUvery, were first shown to have carrying capacity for small molecule drugs (Elbaz et al 1993 Int. J. Pharm. 96 R1-R6). Use of the charged surface to stably bind and protect ohgonucleotides in serum has been demonstrated for both small oUgomers (Teixera et al (1999) Pharm Res 16 30-36) and plasmid DNA (Yi et al (2000) Pharm Res 17 314- 320.) DOTAP-based emulsions have been shown to enhance transfection in vitro and in vivo. (Kim et εϋ, supra).
An adjuvant which results in the increase of a Thl ceU response which can be used for prophylactic and therapeutic treatment is thus desirable. Such a response would be helpful in treatment of, for example, viral infections as well as for immunizing individuals susceptible to viral infections.
U.S. Patents 5,814,482 and 6,015,686 disclose Eukaryotic Layered Vector Initiation Systems (ELVIS vectors), particularly those derived and constructed from alphavirus genomes (such as Sindbis virus), for use in stimulating an immune response to an antigen, in methods of inhibiting pathogenic agents, and in delivery of heterologous nucleotide sequences to eukaryotic cells and animals, among others.
Commonly owned International patent appUcation PCT/US99/17308 and U.S. patent appUcation 09/715,902 disclose methods of making microparticles having adsorbed macromolecules, such as a pharmaceutical, a polynucleoti.de, a polypeptide, a protein, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metabolic pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof, and the Uke.
Commonly owned International patent appUcation PCT/USOO/03331 discloses methods of making submicron emulsions having adsorbed macromolecules, such as a pharmaceutical, a polynucleotide, a polypeptide, a protein, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof, and the like.
Summary of the invention
The inventors herein have discovered that the effectiveness of the various uses of nucleic acids, particularly vector constructs capable of expressing a nucleic acid sequence, and more particularly vector constructs comprising a heterologous nucleic acid sequence encoding an antigen, such as such pCMV vectors, ELVIS vectors or RNA vector constructs may be enhanced by adsorbing the vector constructs to polymer microparticles or submicron emulsions with adsorbent surfaces, which facilitates introduction of the vector constructs, and of heterologous nucleic acid sequences comprised in the vector constructs, into the cells of an animal.
As disclosed in above described International Patent AppUcation PCT/US99/17308, a method of forming microparticles with adsorbent surfaces capable of adsorbing a wide variety of macromolecules has been invented. In one embodiment, the microparticles are comprised of both a polymer and a detergent. The microparticles of the present invention adsorb such macromolecules more efficiently than other microparticles currently available.
Several embodiments of the present invention utilize microparticles that are derived from a polymer, such as a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, a polyalkylcyanoacrylate, a polycyanoacrylate, and the like, and are formed with detergents, such as cationic, anionic, or nonionic detergents, which detergents may be used in combination. The present inventors have discovered that these microparticles yield improved adsorption of vector constructs (e.g., ELVIS vectors, RNA vector constructs), as well as viral antigens, and provide for superior immune responses.
As disclosed in above-described International Patent AppUcation PCT/USOO/03331, a microparticle preparation comprising oil droplet submicron emulsions with ionic surfactants has been invented. Such compositions readily adsorb macromolecules such as DNA, protein, and other antigenic molecules. Several embodiments of the present invention utilize microparticles that are derived from an oil droplet emulsion that preferably comprises a metaboUzable oil and an emulsifying agent which are preferably present in the form of an oil-in-water emulsion having oil droplets substantiaUy aU of which are less than 1 micron in diameter, preferably smaller than 250 nm Preferably, the composition exists in the absence of any polyoxypropylene-polyoxyethylene block copolymer. The oil is preferably an animal oil, an unsaturated hydrocarbon, a terpenoid such as, for example, squalene, or a vegetable oil. The composition preferably comprises 0.5 to 20 % by volume of the oil in an aqueous medium The emulsifying agent preferably comprises a nonionic detergent such as a polyoxyethylene sorbitan mono-, di-, or triester or a sorbitan mono-, di-, or triether. Preferably, the composition comprises about 0.01 to about 5 % by weight of the emulsifying agent.
Hence, in some embodiments, the particulate portion of the invention's composition is a microparticle with an adsorbent surface, wherein the microparticle comprises a polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate.
In another embodiments, the particulate portion of the invention's composition is a submicron emulsion which comprise an oil droplet emulsion formulated with an ionic detergent. In other embodiments, the microparticle further comprises vector constructs capable of expressing a nucleic acid sequence, such as a selected ELVIS vector or RNA vector construct adsorbed on the microparticle's surface, with the vector construct comprising a heterologous nucleotide sequence encoding a polypeptide, a protein, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof and the Uke.
In other embodiments, the invention is directed to a microparticle composition comprising a nucleic acid, preferably vector constructs capable of expressing a nucleic acid sequence, such as a selected pCMV vector, ELVIS vector or RNA vector construct, adsorbed to a microparticle of the invention, and a pharmaceuticaUy acceptable excipient.
In other embodiments, the invention is directed to a method of producing a microparticle with an adsorbed nucleic acid, preferably vector constructs capable of expressing a nucleic acid sequence, such as an ELVIS vector or RNA vector construct, the method comprising:
(a) combining a polymer solution comprising a polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate, wherein the polymer is present at a concentration of about 1% to about 30% in an organic solvent; and an anionic, cationic, or nonionic detergent to the polymer solution, wherein the detergent is present at a ratio of 0.001 to 10 (w/w) detergent to polymer, to form a polymer/detergent mixture;
(b) dispersing the polymer/detergent mixture;
(c) removing the organic solvent;
(d) recovering the microparticle; and
(f) adsorbing an ELVIS vector or RNA vector construct to the surface of the microparticle, wherein the ELVIS vector or RNA vector construct comprises a heterologous nucleotide sequence encoding a polypeptide, a protein, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof, and the like. Preferably, the polymer/detergent mixture is emulsified to form an emulsion prior to removing the organic solvent. In other embodiments, the invention is directed to a microparticle produced by the above- described methods. More preferably, a microparticle composition is produced, which also comprises a pharmaceutically acceptable excipient.
In still other embodiments, the invention is directed to a method of deUvering a heterologous nucleotide sequence to a vertebrate subject, which comprises administering to a vertebrate subject any of the compositions described above.
In additional embodiments, the invention is directed to a method for eUciting a ceUular immune response in a vertebrate subject comprising administering to the vertebrate subject a therapeutically effective amount of a selected heterologous nucleotide sequence adsorbed to a microparticle of the invention.
In other embodiments, the invention is directed to a method of immunization which comprises administering to a vertebrate subject a therapeuticaUy effective amount of any of the microparticle compositions above. The composition may optionally contain unbound macromolecules, and also may optionally contain adjuvants, including aluminum salts such as aluminum phosphate, or an oUgonucleotide comprising at least one CpG motif.
In several preferred embodiments, the microparticles are formed from a poly(α-hydroxy acid); more preferably, a poly(D,L-lactide-co-glycoUde); and most preferably, a poly(D,L-lactide- co-glycoUde).
Each of the nonexhaustive previously described adsorbent microparticles may optionally also have macromolecules entrapped within them, or in free solution. Thus, the invention encompasses a variety of combinations wherein nucleic acid molecules are adsorbed on microparticles and other nucleic acid molecules are entrapped or adsorbed. Moreover, the microparticles of the invention may have more than one species of nucleic acid adsorbed thereon, as well as other antigenic macromolecules adsorbed thereon. Additionally, the microparticles may have several species of nucleic acid and/or other antigenic macromolecules entrapped within.
In other preferred embodiments, the microparticles are prepared in the form of submicron emulsions as described above.
The present invention is also directed to immunogenic compositions comprising an immunostimulating amount of a nucleic acid (e.g., a vector construct capable of expressing a nucleic acid sequence, such as a selected ELVIS vector or RNA vector construct, where the heterologous nucleotide sequence portion of the ELVIS vector or RNA vector construct may encode an antigen), and an immunostimulating amount of an adjuvant composition described herein. In some embodiments of the invention, the immunogenic composition comprises a CpG oUgonucleotide in combination with the nucleic acid- adsorbed microparticles. The adsorbed macromolecule itself is preferably an ELVIS vector or RNA vector construct encoding an antigenic polypeptide.
In some preferred embodiments of the invention, the antigenic polypeptide is from a virus such as, for example, hepatitis C virus (HCV), hepatitis B virus (HBV), herpes simplex virus (HSV), human irnmunodeficiency virus (HIV), cytomegalovirus (CMV), influenza virus (flu), and rabies virus. Preferably, the antigenic polypeptide is selected from the group consisting of HSV glycoproteingD, HIV glycoprotein gpl20, HTV glycoprotein gpl40, HIV p55 gag, and polypeptides from the pol and tat regions. In other preferred embodiments of the invention, the antigenic polypeptide is from a bacterium such as, for example, cholera, diphtheria, tetanus, streptococcus (e.g., streptococcus B), pertussis, Neisseria meningitidis (e.g., meningitis B), Neisseria gonorrhoeae, Helicobacter pylori, and Haemophilus influenza. In stiU other preferred embodiments of the invention, the antigenic polypeptide is from a parasite such as, for example, a malaria parasite.
Adjuvant compositions may comprise, for example, aluminum salts. Alternatively, adjuvant compositions may comprise an oUgonucleotide comprising at least one CpG motif. The adjuvant composition can also comprise an optional component which results in a positively charged emulsion. The oUgonucleotide preferably comprises at least one phosphorothioate bond or peptide nucleic acid bond. In preferred embodiments of the invention, the oUgonucleotide comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 1-28. In other preferred embodiments of the invention, the oligonucleotide comprises a CpG motif flanked by two purines immediately 5' to the motif and two pyrimidines immediately 3' to the motif In other preferred embodiments of the invention, the oligonucleotide comprises a nucleotide sequence selected from the group consisting of SEQ ID NOs: 19-28. Most preferred is SEQ ID NO:28. In some preferred embodiments of the invention, the adjuvant composition further comprises a separate immunostimulating agent which is preferably selected from the group consisting of alum, a bacterial ceU wall component, and muramyl peptide. The adjuvant composition itself may be in the form of a second microparticle. The second microparticle may have adsorbed and/or entrapped within a variety of nucleic acids and/or antigenic polypeptides, or other antigenic macromolecules. Additionally, the immunogenic compositions may include the presence of free nucleic acid in solution.
The present invention is also directed to methods of stimulating an immune response in a host animal comprising administering to the animal an immunogenic composition described herein in an amount effective to induce an immune response. The host animal is preferably a mammal, more preferably a Rhesus macaque, and stiU more preferably a human.
The present invention is also directed to methods of immunizing a host animal against a viral, bacterial, or parasitic infection comprising administering to the animal an immunogenic composition described herein in an amount effective to induce a protective response. The host animal is preferably a mammal, more preferably a Rhesus macaque, and still more preferably a human.
The present invention is also directed to methods of increasing a Thl immune response, or a CTL response, or lyphoproliferation, or cytokine production in a host animal comprising administering to the animal an immunogenic composition described herein in an amount effective to induce the Thl immune response, or the CTL response, or lyphoproliferation, or cytokine production. The host animal is preferably a mammal, more preferably a Rhesus macaque, and still more preferably a human.
The present invention is also directed to methods of raising an immune response in a host animal in which a microparticle-adsorbed macromolecule comprising a heterologous nucleic acid sequence encoding a first antigen (e.g., plasmid DNA, such as pCMV or an ELVIS vector, or an RNA vector construct) is first administered to the animal in an amount effective to eUcit an immunological response. Subsequently, a second antigen is administered to the animal.
The first antigen and the second antigen in these embodiments can be the same or different, and are preferably the same. Preferred antigens include bacterial and viral antigens, such as HIV antigens (e.g., gpl20, gpl40, gpl60, p24gag and p55gag), hepatitis C virus antigens, influenza A virus antigens, meningitis B bacterial antigens, and streptococcus B bacterial antigens. The second antigen is preferably adsorbed to the microparticles described herein, or is co administered with an adjuvant, such as MF59. The macromolecule can also be coadministered with an adjuvant, if desired. In some preferred embodiments, the macromolecule is aάlministered two or more times before the second antigen, which can also be adrrinistered two or more times.
According to one specific embodiment: (1) the macromolecule is administered (a) at a time of initial adrnuiistration, (b) at a time period ranging 1-8 weeks from the initial administration, and (c) at a time period ranging 4-32 weeks from the initial adrninistration, and (2) the second antigen is administered (a) at a time period ranging from 8-50 weeks from the initial administration and (b) at a time period ranging from 8-100 weeks from the initial administration.
DeUvery of the microparticle compositions of the invention may be performed by any known method, including direct injection (e.g., subcutaneously, intraperitoneally, intravenously or intramuscularly), and such deUvery may also be enhanced by the use of electroporation (see, e.g., U.S. patent appUcation no. 09/499,023, the disclosure of which is hereby incorporated by reference in its entirety). Electroporation is the application of short electrical pulses to cells to increase permeability of the cell membranes, thus faciUtating DNA uptake by ceUs. Recently it has been found that applying an electric field to tissues in vivo significantly increases DNA uptake and gene expression (Mathiesen, I., 1999, Gene Therapy 6:508). Among the tissues targeted for in vivo electroporation have been skin, Uver, tumors, and muscle. For DNA vaccine appUcation, Widera et al. have shown that in vivo electroporation substantiaUy enhances DNA vaccine potency in mice, guinea pigs, and rabbits (Widera, G., et al., 2000, J. Immunol. 164:4635).
The ELVIS vectors of the above-described embodiments are generaUy DNA molecules comprising a promoter that functions in a eukaryotic ceU, a cDNA sequence for which the transcription product is an RNA vector construct (e.g., alphavirus RNA vector repUcon), and a 3' termination region. The RNA vector constructs preferably comprise an RNA genome from a picornavirus, togavirus, flavivirus, coronaviras, paramyxovirus, yeUow fever virus, or alphavirus (e.g., Sindbis virus, Semliki Forest virus, Venezuelan equine encephaUtis virus, or Ross River virus), and more preferably an alphavirus genome, which has been modified by the replacement of one or more structural protein genes with a selected heterologous nucleic acid sequence encoding a gene product of interest. The RNA vector constructs of the present invention generally are obtained by transcription in vitro from a DNA template.
These and other aspects and embodiments of the present invention will readily occur to those of ordinary skill in the art in view of the disclosure herein. Brief Description of the Drawings
Fig. 1 provides a DNA sequence (SEQ ID NO:63) encoding a modified HIV-1 p55gag polypeptide.
Fig. 2 provides a DNA sequence (SEQ ID NO:64) encoding a modified HIV-1 p55gag polypeptide.
Fig. 3 provides a DNA sequence (SEQ ID NO:65) encoding a modified HTV-1 envelope polypeptide.
Fig. 4 provides a DNA sequence (SEQ ID NO:66) encoding a modified HIV-1 envelope polypeptide.
Fig. 5 provides a DNA sequence (SEQ ID NO:67) encoding a modified HIV-1 p55gag polypeptide.
Fig. 6 provides a DNA sequence (SEQ ID NO:68) encoding a modified HIV-1 p55gag polypeptide.
Detailed Description of the Invention
The present invention is based upon the surprising discovery that microparticles with adsorbed nucleic acid molecules, preferably vector constructs capable of expressing a nucleic acid sequence, and more preferably vector constructs comprising a heterologous nucleic acid sequence encoding an antigen, such as pCMV vectors, ELVIS vectors or RNA vector constructs, elicit enhanced immune responses. Additionally, the combination of microparticles with adsorbed nucleic acid molecules (for example, microparticles with adsorbed pCMV vectors, ELVIS vectors or RNA vector constructs) and adjuvants is useful for eUciting enhanced immune responses.
The invention is also based upon the surprising discovery that vector constructs comprising antigen-encoding nucleic acid sequences, such as pCMV vectors, ELVIS vectors or RNA vector constructs, in association with subsequent administration of antigen, eUcit enhanced immune responses.
The practice of the present invention will employ, unless otherwise indicated, conventional methods of chemistry, polymer chemistry, biochemistry, molecular biology, immunology and pharmacology, VΛl±iin the skill of the art. Such techniques are explained fuUy in the Uterature. See, e.g., Remington's Pharmaceutical Sciences, 18th Edition (Easton, Pennsylvania: Mack PubUshing Company, 1990); Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press, Inc.); Handbook of Experimental Immunology, Vols. I-IV (D.M. Weir and C.C. BlackweU, eds., 1986, BlackweU Scientific PubUcations); Sambrook, et al., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Handbook of Surface and Colloidal Chemistry (Birdi, K.S., ed, CRC Press, 1997) and Seymour/Carraher's Polymer Chemistry (4th edition, Marcel Dekker Inc., 1996).
All publications, patents and patent appUcations cited herein, whether supra or infra, are hereby incorporated by reference in their entirety.
As used in this specification and the appended claims, the singular forms "a," "an" and "the" include plural references unless the content clearly dictates otherwise. Thus, for example, the term "microparticle" refers to one or more microparticles, and the Uke.
A. Definitions
In describing the present invention, the foUowing terms will be employed, and are intended to be defined as indicated below.
Unless stated otherwise, all percentages and ratios herein are given on a weight basis.
The term "microparticle" as used herein, refers to a particle of about 10 nmto about 150 μm in diameter, more preferably about 200 nmto about 30 μm in diameter, and most preferably about 500 nm to about 10 μm in diameter. Preferably, the microparticle wiU be of a diameter that permits parenteral or mucosal aclnώiistration without occluding needles and capillaries. Microparticle size is readily determined by techniques weU known in the art, such as photon correlation spectroscopy, laser diffractometry and/or scanning electron microscopy. The term "particle" may also be used to denote a microparticle as defined herein. Microparticle may alternatively refer to a submicron emulsion composition as described herein.
Polymer microparticles for use herein are formed from materials that are sterϋizable, non- toxic and biodegradable. Such materials include, without Umitation, poly(α-hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate. Preferably, microparticles for use with the present invention are polymer microparticles derived from a poly(α-hydroxy acid), in particular, from a poly(lactide) ("PLA") or a copolymer of D,L-lactide and glycoUde or glycoUc acid, such as a poly(D,L-lactide-co-glycoUde) ("PLG" or "PLGA"), or a copolymer of D,L-lactide and caprolactone. The polymer microparticles may be derived from any of various polymeric starting materials which have a variety of molecular weights and, in the case of the copolymers such as PLG, a variety of lactide:glycolide ratios, the selection of which wiU be largely a matter of choice, depending in part on the co administered macromolecule. These parameters are discussed more fully below. Alternatively, microparticles of the invention are comprised in a submicron emulsion.
As used herein, the phrase "oil droplet emulsion" refers to an emulsion comprising a metabolizable oil and an emulsifying agent. The term "submicron emulsion" as used herein refers to an oil droplet emulsion of the invention comprising droplets ranging in size from about 10 nmto about 1000 nm
As used herein, the term "microparticle" may refer to a polymer microparticle as described herein or a submicron emulsion composition as described herein.
The term "detergent" as used herein includes surfactants, dispersing agents, suspending agents, and emulsion stabilizers.. Anionic detergents include, but are not limited to, SDS (sodium dodecyl sulfate), SLS (sodium lauryl sulfate), DSS (disulfosuccinate), sulphated fatty alcohols, and the like. Cationic detergents include, but are not limited to, cetrimide (cetyl trimethyl ammonium bromide, or "CTAB"), benzalkonium chloride, DDA (dimethyl dioctodecyl ammonium bromide), DOTAP(dioleoyl-3-trimethylammonium-propane), and the like. Nonionic detergents include, but are not limited to, PVA, povidone (also known as polyvinylpyrroUdone or PVP), sorbitan esters, polysorbates, polyoxyethylated glycol monoethers, polyoxyethylated alkyl phenols, poloxamers, and the like.
The term "zeta potential" as used herein, refers to the electrical potential that exists across the interface of aU soUds and Uquids, i.e., the potential across the diffuse layer of ions surrounding a charged coUoidal particle. Zeta potential can be calculated from electrophoretic mobilities, i.e., the rates at which colloidal particles travel between charged electrodes placed in contact with the substance to be measured, using techniques weU known in the art.
The term "macromolecule" as used herein refers to, without Umitation, a pharmaceutical, a polynucleotide, a polypeptide, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, an adjuvant, or combinations thereof. Particular macromolecules for use with the present invention are described in more detail below.
The term "pharmaceutical" refers to biologically active compounds such as antibiotics, antiviral agents, growth factors, hormones, and the like, discussed in more detail below.
The term "adjuvant" refers to any substance that assists or modifies the action of a pharmaceutical, including but not limited to immunological adjuvants, which increase or diversify the immune response to an antigen.
A "polynucleotide" is a nucleic acid polymer, which typically encodes abiologicaUy active (e.g., irnmunogenic or therapeutic) protein or polypeptide. Depending on the nature of the polypeptide encoded by the polynucleotide, apolynucleotide can include as Uttle as 10 nucleotides, e.g., where the, polynucleotide encodes an antigen. Furthermore, a "polynucleotide" can include both double- and single-stranded sequences and refers to, but is not limited to, cDNA from viral, procaryotic or eucaryotic mRNA, genomic RNA and DNA sequences from viral (e.g. RNA and DNA viruses and retroviruses) or procaryotic DNA, and especiaUy synthetic DNA sequences. The term also captures sequences that include any of the known base analogs of DNA and RNA. The term further includes modifications, such as deletions, additions and substitutions (generally conservative in nature), to a native sequence, preferably such that the nucleic acid molecule encodes a therapeutic or antigenic protein. These modifications may be deUberate, as through site- directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the antigens.
The terms "polypeptide" and "protein" refer to a polymer of amino acid residues and are not limited to a minimum length of the product. Thus, peptides, oligopeptides, dimers, multimers, and the like, are included within the definition. Both fuU-length proteins and fragments thereof are encompassed by the definition. The terms also include modifications, such as deletions, additions and substitutions (generally conservative in nature), to a native sequence, preferably such that the protein n-aintains the abiUty to eUcit an immunological response or have a therapeutic effect on a subject to which the protein is administered.
By "antigen" is meant a molecule which contains one or more epitopes capable of stimulating a host's immune system to make a cellular antigen-specific immune response when the antigen is presented in accordance with the present invention, or a humoral antibody response. An antigen may be capable of eUciting a cellular or humoral response by itself or when present in combination with another molecule. NormaUy, an epitope wϋl include between about 3-15, preferably about 5-15, and more preferably about 7-15 amino acids. Epitopes of a given protein can be identified using any number of epitope mapping techniques, well known in the art. S ee, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66 (Glenn E. Morris, Ed., 1996) Humana Press, Totowa, New Jersey. For example, linear epitopes may be determined by e.g., concurrently synthesizing large numbers of peptides on soUd supports, the peptides corresponding to portions of the protein molecule, and reacting the peptides with antibodies while the peptides are stiU attached to the supports. Such techniques are known in the art and described in, e.g., U.S. Patent No. 4,708,871; Geysen et al. (1984) Proc. Natl Acad. Sci. USA 81:3998- 4002; Geysen et al. (1986) Molec. Immunol. 23:709-715, all incorporated herein by reference in their entireties. Similarly, conformational epitopes are readily identified by deternώώig spatial conformation of amino acids such as by, e.g., x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols, supra.
The term "antigen" as used herein denotes both subunit antigens, i.e., antigens which are separate and discrete from a whole organism with which the antigen is associated in nature, as weU as killed, attenuated or inactivated bacteria, viruses, parasites or other microbes. Antibodies such as anti-idiotype antibodies, or fragments thereof, and synthetic peptide mimotopes, which can mimic an antigen or antigenic deterrninant, are also captured under the definition of antigen as used herein. Similarly, an oligonucleotide or polynucleotide which expresses a therapeutic or immunogenic protein, or antigenic determinant in vivo, such as in gene therapy and nucleic acid ώimunization appUcations, is also included in the definition of antigen herein.
Further, for purposes of the present invention, antigens can be derived from any of several known viruses, bacteria, parasites and fungi, as weU as any of the various tumor antigens. Furthermore, for purposes of the present invention, an "antigen" refers to a protein which includes modifications, such as deletions, additions and substitutions (generaUy conservative in nature), to the native sequence, so long as the protein maintains the abiUty to eUcit an immunological response. These modifications may be deUberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the antigens. An "immunological response" or "immune response" to an antigen or composition is the development in a subject of a humoral and or a ceUular immune response to molecules present in the composition of interest. For purposes of the present invention, a "humoral immune response" refers to an immune response mediated by antibody molecules, while a "ceUular immune response" is one mediated by T-lymphocytes and/or other white blood cells. One important aspect of ceUular immunity involves an antigen-specific response by cytolytic T-cells ("CTLs"). CTLs have specificity for peptide antigens that are presented in association with proteins encoded by the major histocompatibiUty complex (MHC) and expressed on the surfaces of ceUs. CTLs help induce and promote the intraceUular destruction of intraceUular microbes, or the lysis of ceUs infected with such microbes. Another aspect of ceUular immunity involves an antigen-specific response by helper T-cells. Helper T-cells act to help stimulate the function, and focus the activity of, nonspecific effector ceUs against cells displaying peptide antigens in association with MHC molecules on their surface. A "cellular immune response" also refers to the production of cytokines, chemokines and other such molecules produced by activated T-cells and/or other white blood ceUs, including those derived fromCD4+ and CD8+ T-cells.
A composition, such as an immunogenic composition, or vaccine that elicits a ceUular immune response may serve to sensitize a vertebrate subject by the presentation of antigen in association with MHC molecules at the ceU surface. The ceU-mediated immune response is directed at, or near, ceUs presenting antigen at their surface. In addition, antigen-specific T- lymphocytes can be generated to aEow for the future protection of an immunized host.
The ability of a particular antigen or composition to stimulate a cell-mediated immunological response may be determined by a number of assays, such as by lymphoproliferation (lymphocyte activation) assays, CTL cytotoxic ceU assays, by assaying for T-lymphocytes specific for the antigen in a sensitized subject, or by measurement of cytokine production by T ceUs in response to restimulation with antigen. Such assays are weU known in the art. See, e.g., Erickson et al., J. Immunol. (1993) 151:4189-4199; Doe et al., Bur. J. Immunol. (1994) 24:2369-2376; and the examples below.
Thus, an immunological response as used herein may be one which stimulates the production of CTLs, and/or the production or activation of helper T-cells. The antigen of interest may also elicit an antibody-mediated immune response. Hence, an immunological response may include one or more of the foUowing effects: the production of antibodies by B-cells; and/or the activation of suppressor T-cells and/or γδ T-cells directed specifically to an antigen or antigens present in the composition or vaccine of interest. These responses may serve to neutralize infectivity, and/or mediate antibody-complement, or antibody dependent ceU cytotoxicity (ADCC) to provide protection to an immunized host. Such responses can be determined using standard immunoassays and neutralization assays, weU known in the art.
A composition which contains a selected antigen adsorbed to a microparticle, displays "enhanced immunogenicity" when it possesses a greater capacity to eUcit an immune response than the immune response eUcited by an equivalent amount of the antigen when deUvered without association with the microparticle. Thus, a composition may display "enhanced immunogenicity" because the antigen is more strongly immunogenic by virtue of adsorption to the microparticle, or because a lower dose of antigen is necessary to achieve an immune response in the subject to which it is administered. Such enhanced immunogenicity can be determined by aiiministering the microparticle/antigen composition, and antigen controls to animals and comparing antibody liters against the two using standard assays such as radioimmuno assay and ELISAs, weU known in the art.
The terms "effective amount" or "pharmaceutically effective amount" of a given composition, as provided herein, refer to a nontoxic but sufficient amount of the composition to provide a desired response, such as an immunological response, and corresponding therapeutic effect, or in the case of deUvery of a therapeutic protein, an amount sufficient to effect treatment of the subject, as defined below. As wiU be pointed out below, the exact amount required wiU vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition being treated, and the particular macromolecule of interest, mode of administration, and the like. An appropriate "effective" amount in any individual case may be deteπnined by one of ordinary skill in the art using routine experimentation.
By "vertebrate subject" is meant any member of the subphylum cordata, including, without Umitation, nDimimals such as cattle, sheep, pigs, goats, horses, and humans; domestic animals such as dogs and cats; and birds, including domestic, wild and game birds such as cocks and hens including chickens, turkeys and other gallinaceous birds. The term does not denote a particular age. Thus, both adult and newborn animals are intended to be covered. By "pharmaceutically acceptable" or "pharmacologicaUy acceptable" is meant a material which is not biologicaUy or otherwise undesirable, i.e., the material may be administered to an individual along with the microparticle formulation without causing any undesirable biological effects in the individual or interacting in a deleterious manner with any of the components of the composition in which it is contained.
The term "excipient" refers to substances that are commonly provided within finished dosage forms, and include vehicles, binders, disintegrants, fillers (diluents), lubricants, gUdants (flow enhancers), compression aids, colors, sweeteners, preservatives, suspensing/dispersing agents, filmfonners/coatings, flavors and printing inks.
By "physiological pH" or a "pH in the physiological range" is meant a pH in the range of approximately 7.2 to 8.0 inclusive, more typicaUy in the range of approximately 7.2 to 7.6 inclusive.
As used herein, "treatment" (including variations thereof, for example, "treat" or "treated") refers to any of (i) the prevention of infection or reinfection, as in a traditional vaccine, (n) the reduction or elirnination of symptoms, and (iii) the substantial or complete elimination of the pathogen or disorder in question. Treatment may be effected prophylactically (prior to infection) or therapeutica y (foUowing infection).
As used herein, the phrase "nucleic acid" refers to DNA, RNA, or chimeras formed therefrom
As used herein, the phrase "oUgonucleotide comprising at least one CpG motif' refers to a polynucleotide comprising at least one CpG dinucleotide. OUgonucleotides comprising at least one CpG motif can comprise multiple CpG motifs. These oUgonucleotides are also known as "CpG ohgonucleotides" in the art. As used herein, the phrase "CpG motif refers to a dinucleotide portion of an oUgonucleotide which comprises a cytosine nucleotide foUowed by a guanosine nucleotide. 5-methylcytosine can also be used in place of cytosine.
As used herein, "alphavirus RNA vector repUcon," "RNA vector repUcon," "RNA vector construct," and "repUcon" refer to an RNA molecule which is capable of directing its own ampUfication or self-repUcation in vivo, within a target cell. An alphavirus-derived RNA vector repUcon should contain the foUowing ordered elements: 5' viral sequences required in cis for repUcation (also referred to as 5' CSE), sequences which, when expressed, code for biologicaUy active alphavirus nonstructural proteins (e.g., nsPl, nsP2, nsP3, nsP4), 3' viral sequences required in cis for repUcation (also referred to as 3' CSE), and a polyadenylate tract. An alphavirus-derived RNA vector repUcon also may contain a viral subgenomic "junction region" promoter, sequences from one or more structural protein genes or portions thereof, extraneous nucleic acid molecule(s) which are of a size sufficient to aUow production of viable virus, as weU as heterologous sequence(s) to be expressed.
As used herein, "Eukaryotic Layered Vector Initiation System," "ELVIS, "or "ELVIS vector" refers to an assembly which is capable of directing the expression of a sequence(s) or gene(s) of interest. The eukaryotic layered vector initiation system should contain a 5' promoter which is capable of initiating in vivo (i.e., within a cell) the synthesis of RNA from cDNA, and a viral vector sequence which is capable of directing its own repUcation in a eukaryotic ceU and also expressing a heterologous sequence. In preferred embodiments, the nucleic acid vector sequence is an alphavirus-derived sequence and is comprised of a 5' sequence which is capable of initiating transcription of an alphavirus RNA (also referred to as 5' CSE), as weU as sequences which, when expressed, code for biologicaUy active alphavirus nonstructural proteins (e.g., nsPl, nsP2, nsP3, nsP4), and an alphavirus RNA polymerase recognition sequence (also referred to as 3' CSE). In addition, the vector sequence may include a viral subgenomic "junction region" promoter, sequences from one or more structural protein genes or portions thereof, extraneous nucleic acid molecule(s) which are of a size sufficient to allow optimal amplification, a heterologous sequence to be expressed, one or more restriction sites for insertion of heterologous sequences, as weU as a polyadenylation sequence. The eukaryotic layered vector initiation system may also contain splice recognition sequences, a catalytic ribozyme processing sequence, a nuclear export signal, and a transcription termination sequence.
"Alphavirus vector construct" refers to an assembly which is capable of directing the expression of a sequence or gene of interest. Such vector constructs are generaUy comprised of a 5' sequence which is capable of initiating transcription of an alphavirus RNA (also referred to as 5' CSE), as weU as sequences which, when expressed, code for biologicaUy active alphavirus nonstructural proteins (e.g., nsPl, nsP2, nsP3, nsP4), an alphavirus RNA polymerase recognition sequence (also referred to as 3' CSE), and a polyadenylate tract. In addition, the vector construct may include a viral subgenomic "junction region" promoter, sequences from one or more structural protein genes or portions thereof, extraneous nucleic acid molecule(s) which are of a size sufficient to aUow production of viable virus, a 5' promoter which is capable of initiating the synthesis of viral RNA from cDNA in vitro or in vivo, a heterologous sequence to be expressed, and one or more restriction sites for insertion of heterologous sequences.
As used herein, the phrase "vector construct" generaUy refers to any assembly which is capable of directing the expression of a nucleic acid sequence(s) or gene(s) of interest. The vector construct typicaUy includes transcriptional promoter/enhancer or locus defining element(s), or other elements which control gene expression by other means such as alternate spUcing, nuclear RNA export, post-translational modification of messenger, or post-transcriptional modification of protein. In addition, the vector construct typically includes a sequence which, when transcribed, is operably linked to the sequence(s) or gene(s) of interest and acts as a translation initiation sequence. The vector construct may also optionaUy include a signal which directs pblyadenylation, a selectable marker, as weU as one or more restriction sites and a translation termination sequence. In addition, if the vector construct is placed into a retrovirus, the vector construct may include a packaging signal, long terminal repeats (LTRs), and positive and negative strand primer binding sites appropriate to the retrovirus used (if these are not already present). Examples of vector constructs include ELVIS vectors, which comprise the cDNA complement of RNA vector constructs, RNA vector constructs themselves, alphavirus vector constructs, CMV vector constructs and the like.
One specific type of vector construct is a "plasmid", which refers to a circular double stranded DNA, loop into which additional DNA segments can be Ugated. Specific plasmids described below include pCMV and pSINCP.
According to some embodiments of the present invention, compositions and methods are provided which treat, including prophylacticaUy and/or therapeuticaUy immunize, a host animal against viral, fungal, mycoplasma, bacterial, or protozoan infections, as weU as against tumors. The methods of the present invention are useful for conferring prophylactic and/or therapeutic in_munity to a nτarnmal, preferably a human. The methods of the present invention can also be practiced on mammals, other than humans, including mammals in biomedical research settings. B. General Methods
1. Polymer Microparticles with Adsorbed Macromolecules
Polymer microparticles, including PLA and PLG microparticles, efficiently adsorb biologicaUy active macromolecules. Further, these microparticles adsorb a great variety of molecules, including charged and/or bulky macromolecules. Thus the macromolecule/microparticles used in connection with the present invention can be used as a deUvery system to deUver the biologically active components in order to treat, prevent and/or diagnose a wide variety of diseases.
A wide variety of macromolecules can be delivered in association with the microparticles including, but not limited to, pharmaceuticals such as antibiotics and antiviral agents, nonsteroidal antiinflammatory drugs, analgesics, vasodilators, cardiovascular drugs, psychotiopics, neuroleptics, antidepressants, antiparkinson drugs, beta blockers, calcium channel blockers, bradykinin inhibitors, ACE-inbibitors, vasodilators, prolactin inhibitors, steroids, hormone antagonists, antihistamines, serotonin antagonists, heparin, chemotherapeutic agents, antineoplastics and growth factors, including but not limited to PDGF, EGF, KGF, IGF-1 and IGF-2, FGF, polynucleotides which encode therapeutic or immunogenic proteins, immunogenic proteins and epitopes thereof for use in vaccines, hormones including peptide hormones such as insulin, promsulin, growth hormone, GHRH, LHRH, EGF, somatostatin, SNX-111, BNP, insulinotropin, ANP, FSH, LH, PSH and hCG, gonadal steroid hormones (androgens, estrogens and progesterone), yroid-stimulating hormone, inhibin, cholecystokinin, ACTH, CRF, dynoφhins, endorphins, endothelin, fibronectin fragments, galanin, gastrin, insulinotropin, glucagon, GTP- binding protein fragments, guanylin, the leukokinins, magainin, mastoparans, dermaseptin, systemin, neuromedins, neurotensin, pancreastatin, pancreatic polypeptide, substance P, secretin, thymosin, and the Uke, enzymes, transcription or translation mediators, intermediates in metaboUc pathways, immunomodulators, such as any of the various cytokines including interleukin-1, interleukin-2, interleukin-3, interleukin-4, and gamma-interferon, antigens, and adjuvants.
In some preferred embodiments of the invention, the macromolecule is nucleic acid, more preferably a vector construct such as an ELVIS vector, or RNA vector construct. One particular advantage of the present invention is the abiUty of the microparticles with adsorbed ELVIS vector to generate ceU-mediated immune responses in a vertebrate subject. The ability of the antigen/ microparticles of the present invention to eUcit a ceU-mediated immune response against a selected antigen provides a powerful tool against infection by a wide variety of pathogens. Accordingly, the antigen microparticles of the present invention can be incorporated into vaccine compositions.
Thus, in addition to a conventional antibody response, the systems herein described can provide for, e.g., the association of the expressed antigens with class I MHC molecules such that an in vivo ceUular immune response to the antigen of interest can be mounted which stimulates the production of CTLs to aUow for future recognition of the antigen. Furthermore, the methods may eUcit an antigen-specific response by helper T-cells. Accordingly, the methods of the present invention will find use with any macromolecule for which ceUular and/or humoral immune responses are desired, preferably antigens derived from viral pathogens that may induce antibodies, T-cell helper epitopes and T-cell cytotoxic epitopes. Such antigens include, but are not limited to, those encoded by human and animal viruses and can correspond to either structural or non- structural proteins.
The microparticles of the present invention are particularly useful for irrimunization against intraceUular viruses which normaUy ehcit poor immune responses. For example, the present invention wUl find use for stimulating an immune response against a wide variety of proteins from the herpesvirus farnuy, including proteins derived from herpes simplex virus (HSV) types 1 and 2, such as HSV-1 and HSV-2 glycoproteins gB, gD and gH; antigens derived from varicella zoster virus (VZV), Epstein-Barr virus (EBV) and cytomegalovirus (CMV) including CMV gB and gH; and antigens derived from other human herpesviruses such as HHV6 and HHV7. (See, e.g. Chee et al., Cytomegaloviruses (J.K. McDougaU, ed., Springer-Verlag 1990) pp. 125-169, for a review of the protein coding content of cytomegalovirus; McGeoch et al., J. Gen. Virol (1988) 69:1531- 1574, for a discussion of the various HSV-1 encoded proteins; U.S. Patent No. 5J71,568 for a discussion of HSV-1 and HSV-2 gB and gD proteins and the genes encoding therefor; Baer et al., Nature (1984) 310:207-211, for the identification of protein coding sequences in an EBV genome; and Davison and Scott, J. Gen. Virol. (1986) 67:1759-1816, for areview ofVZV.)
Antigens from the hepatitis family of viruses, including hepatitis A virus (HAV), hepatitis B virus (HBV), hepatitis C virus (HCV), the delta hepatitis virus (HDV), hepatitis E virus (HEV) and hepatitis G virus (HGV), can also be conveniently used in the techniques described herein. By way of example, the viral genomic sequence of HCV is known, as are methods for obtaining the sequence. See, e.g., International Publication Nos. WO 89/04669; WO 90/11089; and WO 90/14436. The HCV genome encodes several viral proteins, including El (also known as E) and E2 (also known as E2/NSI) and an N-terminal nucleocapsid protein (termed "core") (see, Houghton et al., Hepatology (1991) 14:381-388, for a discussion of HCV proteins, including El and E2). Each of these proteins, as weU as antigenic fragments thereof, wiU find use in the present composition and methods.
Similarly, the sequence for the δ-antigen from HDV is known (see, e.g., U.S. Patent No. 5,378,814) and this antigen can also be conveniently used in the present composition and methods. Additionally, antigens derived from HBV, such as the core antigen, the surface antigen, SAg, as weU as the presurface sequences, pre-Sl and pre-S2 (formerly caUed pre-S), as weU as combinations of the above, such as SAg/pre-Sl, SAg/pre-S2, SAg/pre-Sl/pre-S2, and pre-S 1/pre- S2, wUl find use herein. See, e.g., "HBV Vaccines - from the laboratory to Ucense: a case study" in Mackett, M. and WiUiamson, J.D., Human Vaccines and Vaccination, pp. 159-176, for a discussion of HBV structure; and U.S. Patent Nos. 4,722,840, 5,098,704, 5,324,513, incorporated herein by reference in their entireties; Beames et al., J. Virol (1995) 69:6833-6838, Birnbaum et al, ./. Virol (1990) 64:3319-3330; and Zhou et al., J. Virol (1991) 65:5457-5464.
Antigens derived from other viruses wiU also find use in the claimed compositions and methods, such as without Umitation, proteins from members of the families Picornaviridae (e.g., poUoviruses, etc.); CaUciviridae; Togaviridae (e.g., rubeUa virus, dengue virus, etc.); Flaviviridae; Coronaviridae; Reoviridae; Birnaviridae; Rhabodoviridae (e.g., rabies virus, etc.); Filoviridae; Paramyxoviridae (e.g., mumps virus, measles virus, respiratory syncytial virus, etc.); Orthomyxoviridae (e.g., influenza virus types A, B and C, etc.); Bunyaviridae; Arenaviridae; Retroviradae (e.g., HTLV-I; HTLV-II; HIV-1 (also known as HTLV-III, LAV, ARY, hTLR, etc.)), including but not Umited to antigens from the isolates HlVmb, HIVSF., HIVLAV, HIVLAI, HIVMN); HΓV-1C 235, HIV-1US4; HIV-2; simian immunodeficiency virus (SIV) among others. AdditionaUy, antigens may also be derived from human papiUomavirus (HPV) and the tick-bome encephalitis viruses. See, e.g. Virology, 3rd Edition (W.K. Joklik ed. 1988); Fundamental Virology, 2nd Edition (B.N. Fields and D.M. Knipe, eds. 1991), for a description of these and other viruses. More particularly, the gpl20 or gpl40 envelope proteins from any of the above FflV isolates, including members of the various genetic subtypes of HTV, are known and reported (see, e.g., Myers et al., Los Alamos Database, Los Alamos National Laboratory, Los Alamos, New Mexico (1992); Myers et al., Human Retroviruses and Aids, 1990, Los Alamos, New Mexico: Los Alamos National Laboratory; and Modrow et al., J. Virol (1987) 61:570-578, for a comparison of the envelope sequences of a variety of HIV isolates) and antigens derived from any of these isolates wiU find use in the present methods. Furthermore, the invention is equaUy appUcable to other immunogenic proteins derived from any of the various HIV isolates, including any of the various envelope proteins such as gpl60 and gp41, gag antigens such as p24gag and p55gag, as weU as proteins derived from the pol and tat regions. Any of these proteins and antigens may also be modified for use in the present invention. For example, Figures 1, 2, 5, and 6 provide DNA sequences encoding modified gag antigens (SEQ ID NOs: 63, 64, 67, and 68), and Figures 3 and 4 provide DNA sequences encoding modified envelope antigens (SEQ ID NOs: 65 and 66).
Influenza virus is another example of a virus for which the present invention wiU be particularly useful. SpecificaUy, the envelope glycoproteins HA and NA of influenza A are of particular interest for generating an immune response. Numerous HA subtypes of influenza A have been identified (Kawaoka et al., Virology (1990) 179:759-767; Webster et al., "Antigenic variation among type A influenza viruses," p. 127-168. In: P. Palese and D.W. Kingsbury (ed.), Genetics of influenza viruses. Springer-Verlag, New York). Thus, proteins derived from any of these isolates can also be used in the compositions and methods described herein.
The compositions and methods described herein wiU also find use with numerous bacterial antigens, such as those derived from organisms that cause diphtheria, cholera, tuberculosis, tetanus, pertussis, meningitis, and other pathogenic states, including, without limitation, Bordetella pertussis, Neisseria meningitides (A, B, C, Y), Neisseria gonorrhoeae, Helicobacter pylori, and Haemophilus influenza. Hemophilus influenza type B (HIB), Helicobacter pylori, and combinations thereof. Examples of antigens from Neisseria meningitides B are disclosed in the foUowing co-owned patent appUcations: PCT/US99/09346; PCT IB98/01665; and PCT IB99/00103. Examples of parasitic antigens include those derived from organisms causing malaria and Lyme disease.
Additional antigens for use with the invention, some of which are also Usted elsewhere in this appUcation, include the following (references are listed immediately below):
- A protein antigen fromN meningitidis serogroup B, such as those in Refs. 1 to 7 below.
- an outer-membrane vesicle (OMV) preparation fromN. meningitidis serogroup B, such as those disclosed in Refs. 8, 9, 10, 11 etc. below.
- a saccharide antigen fromN meningitidis serogroup A, C, W135 and/or Y, such as the oUgosaccharide disclosed in Ref 12 below from serogroup C (see also Ref. 13).
- a saccharide antigen from Streptococcus pneumoniae [e.g. Refs. 14, 15, 16].
- an antigen from N gonorrhoeae [e.g., Refs. 1, 2, 3].
- an antigen from Chlamydia pneumoniae [e.g., Refs. 17, 18, 19, 20, 21, 22, 23].
- an antigen from Chlamydia trachomatis [e.g. 24].
- an antigen fromhepatitis A virus, such as inactivated virus [e.g., Refs. 25, 26].
- an antigen fromhepatitis B virus, such as the surface and/or core antigens [e.g., Refs. 26, 27].
- an antigen from hepatitis C virus [e.g. Ref. 28].
- an antigen from Bordetella pertussis, such as pertussis holotoxin (PT) and filamentous haeniaglutinin (FHA) from 5. pertussis, optionally also in combination with pertactin and/or agglutinogens 2 and 3 [e.g., Refs. 29 & 30].
- a diphtheria antigen, such as diphtheria toxoid [e.g., chapter 3 of Ref. 31] e.g. the CRMi97 mutant [e.g., Ref. 32].
- a tetanus antigen, such as a tetanus toxoid [e.g., chapter 4 of Ref. 31].
- a protein antigen from Helicobacter pylori such as CagA [e.g. Ref. 33], VacA [e.g. Ref. 33], NAP [e.g. Ref. 34], HopX [e.g. Ref. 35], HopY [e.g. Ref. 35] and/or urease.
- a saccharide antigen from Haemophilus influenzae B [e.g. Ref 13].
- an mngea&om Porphyramonas gingivalis [e.g. Ref 36].
- poUo antigen(s) [e.g. Refs. 37, 38] such as IPV or OPV.
- rabies antigen(s) [e.g. Ref. 39] such as lyophUized inactivated virus [e.g. Ref. 40, Rabavert™).
- measles, mumps and/or rubella antigens [e.g., chapters 9, 10 and 11 of Ref. 31].
- influenza antigen(s) [e.g. chapter 19 of Ref. 31], such as the haenragglutinin and/or neuraminidase surface proteins.
- an antigen from or xe//α catarrhalis [e.g., time 41]. - an antigen from Streptococcus agalactiae (Group B streptococcus) [e.g. Refs. 42, 43]
- an antigen from Streptococcus pyogenes (Group A streptococcus) [e.g. Refs. 43,44, 45].
- an antigen from Staphylococcus aureus [e.g. Ref 46].
- Compositions comprising one or more of these antigens.
Where a saccharide or carbohydrate antigen is used, it is preferably conjugated to a carrier protein in order to enhance immunogenicity [e.g. Refs. 47 to 56]. Preferred carrier proteins are bacterial toxins or toxoids, such as diphtheria or tetanus toxoids. The CRM197 diphtheria toxoid is particularly preferred. Other suitable carrier proteins include N. meningitidis outer membrane protein [e.g. Ref. 57], synthetic peptides [e.g. Refs. 58, 59], heat shock proteins [e.g. Ref. 60], pertussis proteins [e.g. Refs. 61, 62], protein D fromH. Influenzae [e.g. Ref. 63], toxin A or B fromC difficile [e.g. Ref. 64], etc. Where a mixture comprises capsular saccharides from both serogroups A and C, it is preferred that the ratio (w/w) of MenA saccharide:MenC saccharide is greater than 1 (e.g. 2:1, 3:1, 4:1, 5:1, 10:1 or higher). Saccharides from different serogroups ofN. meningitidis may be conjugated to the same or different carrier proteins.
Any suitable conjugation reaction can be used, with any suitable linker where necessary.
Toxic protein antigens may be detoxified where necessary (e.g. detoxification of pertussis toxin by chemical and/or means [Ref. 30].
Where diphtheria antigen is included in the composition it is preferred also to include tetanus antigen and pertussis antigens. Similarly, where a tetanus antigen is included it is preferred also to include diphtheria and pertussis antigens. Similarly, where a pertussis antigen is included it is preferred also to include diphtheria and tetanus antigens.
It is readily apparent that the subject invention can be used to deliver a wide variety of macromolecules and hence to treat, prevent and/or diagnose a large number of diseases. In some embodiments, the macromolecule/microparticle compositions of the present invention can be used for site-specific targeted deUvery. For example, intravenous aαlministration of the macromolecule/microparticle compositions can be used for targeting the lung, Uver, spleen, blood circulation, or bone marrow.
The adsorption of macromolecules to the surface of the adsorbent microparticles (or to submicron emulsions of the present invention) occurs via any bonding-interaction mechanism, including, but not limited to, ionic bonding, hydrogen bonding, covalent bonding, Van der Waals bonding, physical entrapment, and bonding through hydrophiUc/hydrophobic interactions. Those of ordinary skiU in the art may readily select detergents appropriate for the type of macromolecule to be adsorbed.
For example, microparticles manufactured in the presence of charged detergents, such as anionic or cationic detergents, may yield microparticles with a surface having a net negative or a net positive charge, which can adsorb a wide variety of molecules. For example, microparticles manufactured with anionic detergents, such as sodium dodecyl sulfate (SDS), i.e. SDS-PLG microparticles, adsorb positively charged antigens, such as proteins. Sirnuarly, microparticles manufactured with cationic detergents, such as hexadecyltrimethylammonium bromide (CTAB), i.e. CTAB-PLG microparticles, adsorb negatively charged macromolecules, such as DNA. Where the macromolecules to be adsorbed have regions of positive and negative charge, cationic, anionic, nonionic or zwitterioinic detergents may be appropriate.
Biodegradable polymers for manufacturing microparticles for use with the present invention are readily commerciaUy avaUable from, e.g., Boehringer Ingelheim, Germany and B mingham Polymers, Inc., Biriningham, AL. For example, useful polymers for forming the microparticles herein include homopolymers, copolymers and polymer blends derived from the foUowing: polyhydroxybutyric acid (also known as polyhydroxybutyrate); polyhydroxy valeric acid (also known as polyhydroxyvalerate); polyglycolic acid (PGA) (also known as polyglycoUde): polylactic acid (PLA) (also known as polylactide); polydioxanone; polycaprolactone; polyorthoester; and polyanhydride. More preferred are poly( -hydroxy acids), such as poly(L-lactide), poly(D,L- lactide) (both known as "PLA" herein), poly(hydoxybutyrate), copolymers of D,L-lactide and glycoUde, such as poly(D,L-lactide-co-glycoUde) (designated as "PLG" or "PLGA" herein) or a copolymer of D,L-lactide and caprolactone. Particularly preferred polymers for use herein are PLA and PLG polymers. These polymers are available in a variety of molecular weights, and the appropriate molecular weight for a given use is readily determined by one of skiU in the art. Thus, e.g., for PLA, a suitable molecular weight witt be on the order of about 2000 to 5000. For PLG, suitable molecular weights wiU generaUy range from about 10,000 to about 200,000, preferably about 15,000 to about 150,000.
If a copolymer such as PLG is used to form the microparticles, avariety of lactide: glycoUde ratios wiU find use herein and the ratio is largely a matter of choice, depending in part on the coadministered macromolecule and the rate of degradation desired. For example, a 50:50 PLG polymer, containing 50% D,L-lactide and 50% glycoUde, wfll provide a fast resorbing copolymer while 75:25 PLG degrades more slowly, and 85:15 and 90:10, even more slowly, due to the increased lactide component. It is readily apparent that a suitable ratio of lactide:glycoUde is easily determined by one of skiU in the art based, for example, on the nature of the antigen and disorder in question. Moreover, in embodiments of the present invention wherein antigen or adjuvants are entrapped within microparticles, mixtures of microparticles with varying lactide: glycoUde ratios wiU find use herein in order to achieve the desired release kinetics for a given macromolecule and to provide for both a primary and secondary immune response. Degradation rate of the microparticles of the present invention can also be contioUed by such factors as polymer molecular weight and polymer crystaUinity. PLG copolymers with varying lactide:glycoUde ratios and molecular weights are readUy available commerciaUy from a number of sources including from Boehringer Ingelheim, Germany and Birmingham Polymers, Inc., Birmingham, AL. These polymers can also be synthesized by simple polycondensation of the lactic acid component using techniques weU known in the art, such as described in Tabata et al., J. Biomed. Mater. Res. (1988) 22:837-858.
Where used, preferred poly(D,L-lactide-co-glycoUde) polymers are those having a lactide/glycoUde molar ratio ranging from 30:70 to 70:30, more preferably 40:60 to 60:40, and having a molecular weight ranging from 10,000 to 100,000 Daltons, more preferably from 30,000 Daltons to 70,000 Daltons.
The polymer microparticles are prepared using any of several methods weU known in the art. For example, in some embodiments, double emulsion/solvent evaporation techniques, such as those described in U.S. Patent No. 3,523,907 and Ogawa et al., Chem. Pharm. Bull. (1988) 36: 1095-1103, can be used herein to make the microparticles. These techniques involve the formation of a primary emulsion consisting of droplets of polymer solution, which is subsequently mixed with a continuous aqueous phase containing a particle stabilizer/ surfactant.
Alternatively, a water-in-oil-in-water (w/o/w) solvent evaporation system can be used to form the microparticles, as described by OHagan et al., Vaccine (1993) 11:965-969, PCT/US99/17308 (WO 00/06123) to O'Hagan et al. and Jeffery et al., Pharm Res. (1993) 10:362. In this technique, the particular polymer is typically combined with an organic solvent, such as ethyl acetate, dimethylchloride (also caUed methylene chloride and dichloromethane), acetonitrile, acetone, chloroform, and the Uke. The polymer wiU be provided in about a 1-30%, preferably about a 2-15%, more preferably about a 3-10% and most preferably, about a 4-6% solution, in organic solvent. The polymer solution is then combined with an aqueous solution and emulsified to form an o/w emulsion. The aqueous solution can be, for example, deionized water, normal saline, or a buffered solution such as phosphate-buffered saline (PBS) or a sodium citiate/ethylenediaminetetraacetic acid (sodium citrate/ETDA) buffer solution. Preferably, the volume ratio of polymer solution to aqueous liquid ranges from about 5:1 to about 20:1, more preferably about 10:1. Emulsification is conducted using any equipment appropriate for this task, and is typically a high-shear device such as, e.g., an homogenizer.
A volume of the o/w emulsion is then optionaUy preferably combined with a larger volume of an aqueous solution, which preferably contains a cationic, anionic, or nonionic detergent. The volume ratio of aqueous solution to o/w emulsion typically ranges from about 2:1 to 10:1, more typically about 4:1. Examples of anionic, cationic and nonionic detergents appropriate for the . practice of the invention are Usted above and include SDS, CTAB and PVA, respectively. Certain macromolecules may adsorb more readily to microparticles having a combination of stabiUzers and/or detergents, for example, a combination of PVA and DOTAP. Moreover, in some instances, it may be desirable to add detergent to the above organic solution. Where a nonionic detergent such as PVA an emulsion stabilizer is used, it is typically provided in about a 2-15% solution, more typicaUy about a 4-10% solution. Where a cationic or anionic detergent is used, it is typicaUy provided in about a 0.05-5% solution, more typicaUy about a 0.25-1% solution. GeneraUy, a weight to weight detergent to polymer ratio in the range of from about 0.00001:1 to about 0.5:1 wiU be used, more preferably from about 0.0001:1 to about 0.5J, more preferably from about 0.001:1 to about 0.5:1, and even more preferably from about 0.005:1 to about 0.5:1.
The mixture is then homogenized to produce a stable w/o/w double emulsion. Organic solvents are then evaporated. The formulation parameters can be manipulated to allow the preparation of smaU microparticles on the order of 0.05 μm (50 nm) to larger microparticles 50 μm or even larger. See, e.g., jeffery et al., Pharm. Res. (1993) 10:362-368; McGee et al., J. Microencap. (1996). For example, reduced agitation results in larger microparticles, as does an increase in internal phase volume. SmaU particles are produced by low aqueous phase volumes with high concentrations of emulsion stabilizers.
Additional information can be found in U.S. AppUcation Serial No. , Attorney Docket
Nos. PP16502.002, entitled "Microparticles with Adsorbed Macromolecules" filed September 28, 2001.
The formulation parameters can be manipulated to aUow the preparation of smaU microparticles on the order of 0.05 μm (50 nm) to larger microparticles 50 μm or even larger. See, e.g., Jeffery et al., Pharm. Res. (1993) 10:362-368; McGee et al., J. Microencap. (1996). For example, reduced agitation results in larger microparticles, as does an increase in internal phase volume. SmaU particles are produced by low aqueous phase volumes with high concentrations of emulsion stabiUzers.
Microparticles can also be formed using spray-drying and coacervation as described in, e.g., Thomasin et al., J. Controlled Release (1996) 41:131; U.S. Patent No. 2,800,457; Masters, K. (1976) Spray Drying 2nd Ed. Wiley, New York; air-suspension coating techniques, such as pan coating and Wurster coating, as described by Hall et al.5 (1980) The "Wurster Process" in Controlled Release Technologies: Methods, Theory, and Applications (A.F. Kydonieus, ed.), Vol. 2, pp. 133-154 CRC Press, Boca Raton, Florida and Deasy, P.B., Crit. Rev. Ther. Drug Carrier Syst. (1988) S(2):99-139; and ionic gelation as described by, e.g., Lim et al., Science (1980) 210:908-910.
Particle size can be determined by, e.g., laser Ught scattering, using for example, a spectrometer incorporating a heUum-neon laser. GeneraUy, particle size is determined at room temperature and involves multiple analyses of the sample in question (e.g., 5-10 times) to yield an average value for the particle diameter. Particle size is also readily determined using scanning electron microscopy (SEM).
Alternative embodiments of the present invention utilize microparticle preparations comprising a submicron emulsion, which preferably includes an ionic surfactant. For instance, MF59 or others may be used as the base on-containing submicron emulsion, while ionic surfactants may include, but are not limited to, Dioleoyl-3-Trimethylammonium-Propane (DOTAP), Dioleoyl- sn-Glycero-3-Ethylphosphocholine(DEPC) and dioleoyl-phosphatidic acid (DP A), each of which are soluble in squalene. Prototypic ionic emulsions may be formulated by dissolving each of the detergents in squalene/10% Span 85 at concentrations ranging from 4-52 mg/ml squalene. The squalene/surfactant mixtures may be emulsified with 0.5% Tween 80/H2O at 5ml squalene/100 ml H20. A pre-emulsion may be formed by homogenization with a SUverson homogenizer ( 5 minutes, 5000 RPM) and final emulsions may be made by microfluidization (~10,000psi, 5 passes, -vficrofluidizer 110S). Additional discussion concerning submicron emulsions can be found infra.
FoUowing preparation, microparticles can be stored as is or freeze-dried for future use. TypicaUy, in order to adsorb macromolecules to the microparticles, the microparticle preparation is simply mixed with the macromolecule of interest and the resulting formulation can again be lyophiUzed prior to use. GeneraUy, macromolecules are added to the microparticles to yield microparticles with adsorbed macromolecules having a weight to weight ratio of from about 0.0001:1 to 0.25:1 macromolecules to microparticles, preferably, 0.001:1 to 0J, more preferably 0.01 to 0.05. Macromolecule content of the microparticles can be determined using standard techniques.
As noted above, macromolecules for use in connection with the present invention include proteins, preferably antigen molecules, and nucleic acids, preferably vector constructs capable of expressing a nucleic acid sequence, such as CMV-based vectors, ELVIS vectors or RNA vector constructs.
The polymer microparticles of the present invention may have macromolecules entrapped or encapsulated within them, as well as having macromolecules adsorbed thereon. Thus, for example, one of skill in the art may prepare in accordance with the invention microparticles having encapsulated adjuvants with ELVIS vector adsorbed thereon, or microparticles having encapsulated antigen with RNA vector construct adsorbed thereon. The invention contemplates a variety of combinations of nucleic acid macromolecules adsorbed on and entrapped within microparticles, along with other nucleic acids as weU as other antigenic molecules. In some preferred embodiments, the microparticles of the invention have ELVIS vectors or RNA vector constructs adsorbed thereon.
Additionally, any of the embodiments of the microparticles of the invention may be deUvered in conjunction with electroporation.
Once the macromolecule-adsorbed microparticles and/or submicron emulsion microparticles are produced, they are formulated, along with any desired adjuvants, into pharmaceutical compositions including vaccines, to treat, prevent and/or diagnose a wide variety of disorders, as described above. The compositions wiU generaUy include one or more pharmaceuticaUy acceptable excipients. For example, vehicles such as water, saline, glycerol, polyethylene-glycol, hyaluronic acid, ethanol, etc. may be used. Other excipients such as wetting or emulsifying agents, biological buffering substances, and the like, may be present in such vehicles. A biological buffer can be virtuaUy any solution which is pharmacologicaUy acceptable and which provides the formulation with the desired pH, i.e., a pH in the physiological range. Examples of buffer solutions include saline, phosphate buffered saline, Tris buffered saline, Hank's buffered saline, and the like. Other excipients known in the art can also be introduced into the final dosage form, including binders, disintegrants, fiUers (diluents), lubricants, gUdants (flow enhancers), compression aids, colors, sweeteners, preservatives, suspensing/dispersing agents, film formers/coatings, flavors and printing inks.
The compositions of the invention wiU comprise a therapeutically effective amount of one or more macromolecules of interest. That is, an amount of macromolecule/microparticle wiU be included in the compositions, which wiU cause the subject to produce a sufficient response, in order to prevent, reduce, eliminate or diagnose symptoms. The exact amount necessary wiU vary, depending on the subject being treated; the age and general condition of the subject to be treated; the severity of the condition being treated; in the case of an immunological response, the capacity of the subject's immune system to synthesize antibodies; the degree of protection desired and the particular antigen selected and its mode of administration, among other factors. An appropriate effective amount can be readfly deteπnined by one of skill in the art. Thus, a therapeuticaUy effective amount wiU faU in a relatively broad range that can be determined through routine trials. For example, for purposes of the present invention, where the macromolecule is a polynucleotide, an effective dose will typicaUy range from about 1 ng to about 10 mg, more preferably from about 10 ng to about 1 mg, and most preferably about 100 μg to about 1 mg of the macromolecule deUvered per dose; where the macromolecule is an antigen, an effective dose wiU typicaUy range from about 1 μg to about 100 mg, more preferably from about 10 μg to about 1 mg, and most preferably about 50 μg to about 1 mg of the macromolecule deUvered per dose.
Once formulated, the compositions of the invention can be administered parenteraUy, e.g., by injection. The compositions can be injected either subcutaneously, intraperitoneaUy, intravenously or intramuscularly. Other modes of administration include nasal, mucosal, rectal, vaginal, oral and pulmonary adn-imstration, suppositories, and transdermal or transcutaneous appUcations. Dosage treatment may be a single dose schedule or a multiple dose schedule. A multiple dose schedule is one in which a primary course of administration may be with 1-10 separate doses, foUowed by other doses given at subsequent time intervals, chosen to maintain and/or reinforce the therapeutic response, for example at 1-4 months for a second dose, and if needed, a subsequent dose(s) after several months.
In certain embodiments of the invention, a series of one or more injections of a vector construct (which comprises a heterologous nucleic acid sequence encoding an antigen) is foUowed by a series of one or more injections of antigen (also referred to herein as "boosts"). As a specific example, the vector construct may be administered in three injections: (a) at a time of initial administration, (b) at a time period ranging 1-8 weeks from the initial administration, and (c) at a time period ranging 4-32 weeks from the initial administration, while the antigen may be administered in two injections: (a) at a time period ranging from 8-50 weeks from the initial administration and (b) at a time period ranging from 8-100 weeks from the initial administration.
The dosage regimen wUl also, at least in part, be determined by the need of the subject and be dependent on the judgment of the practitioner.
Furthermore, if prevention of disease is desired, the microparticles with adsorbed vector constructs are generaUy administered prior to primary infection with the pathogen of interest. If treatment of disease (other than prevention) is desired, e.g., the reduction of symptoms or recurrences, the microparticles with adsorbed vector constructs are generaUy administered subsequent to primary infection.
2. OU Droplet Emulsions
In other embodiments of the present invention, an oil droplet emulsion (particularly, a submicron emulsion) is prepared comprising a metabolizable oil and an emulsifying agent. Molecules such as an oligonucleotide comprising at least one CpG motif may be combined with the oil droplet emulsion to form an adjuvant.
The oil droplet emulsion preferably comprises a metaboUzable oil and an emulsifying agent, wherein the oil and the emulsifying agent are present in the form of an oil-in-water emulsion having oil droplets substantially aU of which are less than one micron in diameter. Submicron emulsions, with droplets in this preferred size range, show a surprising superiority over other emulsions containing oil and emulsifying agents in which the oil droplets are significantly larger than those provided by the present invention. In preferred embodiments, the emulsion is positively charged as a result of a cationic detergent being used as the emulsifying agent or, alternatively, contains a cationic detergent in addition to the emulsifying agent. This allows for the adsorption of nucleotide antigenic molecules, such as CpG oUgonucleotides or vector constructs.. Alternatively, the use of an anionic detergent allows for the adsorption of molecules such as proteins.
Although individual components of the submicron emulsion compositions of the present invention are generally known, such compositions have not been combined in the same manner. Accordingly, the individual components, although described below both generaUy and in some detail for preferred embodiments, are weU known in the art, and the terms used herein, such as metabolizable oil, emulsifying agent, immunostimulating agent, muramyl peptide, and UpophiUc muramyl peptide, are sufficiently weU known to describe these compounds to one skiUed in the art without further description.
One component of these compositions is a metabolizable, non-toxic oU, preferably one of about 6 to about 30 carbon atoms including, but not Umited to, alkanes, alkenes, alkynes, and their corresponding acids and alcohols, the ethers and esters thereof, and mixtures thereof. The oil can be any vegetable ofl, fish oil, animal oil or syntheticaUy prepared oil which can be metaboUzed by the body of the host animal to which the adjuvant wiU be administered and which is not toxic to the subject. The host animal is typicaUy a mammal, and preferably a human. Mineral oU and similar toxic petroleum distillate oils are expressly excluded from this invention.
The oil component of this invention can also be any long chain alkane, alkene or alkyne, or an acid or alcohol derivative thereof either as the free acid, its salt or an ester such as a mono-, or di- or triester, such as the triglycerides and esters of 1,2-propanediol or similar poly-hydroxy alcohols. Alcohols can be acylated employing amino- or poly-functional acid, for example acetic acid, propanoic acid, citric acid or the like. Ethers derived from long chain alcohols which are oils and meet the other criteria set forth herein can also be used.
The individual alkane, alkene or alkyne moiety and its acid or alcohol derivatives will generaUy have about 6 to about 30 carbon atoms. The moiety can have a straight or branched chain structure. It can be fuUy saturated or have one or more double or triple bonds. Where mono or poly ester- or ether-based oils are employed, the limitation of about 6 to about 30 carbons appUes to the individual fatty acid or fatty alcohol moieties, not the total carbon count.
Any metabolizable oil, particularly from an animal, fish or vegetable source, can be used herein. It is essential that the oU be metaboUzed by the host to which it is adiriinistered, otherwise the oil component can cause abscesses, granulomas or even carcinomas, or (when used in veterinary practice) can make the meat of vaccinated birds and animals unacceptable for human consumption due to the deleterious effect the unmetabolized oU can have on the consumer.
For a detaUed description of such submicron emulsions, see International PubUcation No. WO 90/14837, and commonly owned International Patent AppUcation PCT/USOO/03331.
The oil component of these adjuvants and immunogenic compositions wiU be present in an amount from about 0.5%o to about 20% by volume but preferably no more than about 15%, especially in an amount of about 1% to about 12%. It is most preferred to use from about 1% to about 4% oU.
The aqueous portion of these submicron emulsion compositions is preferably buffered saline or, more preferably, unadulterated water. Because these compositions are intended for parenteral administration, it is preferable to make up final buffered solutions used as immunogenic compositions so that the tonicity, i.e., osmolaUty, is essentiaUy the same as normal physiological fluids in order to prevent post-administration swelling or rapid absorption of the composition because of differential ion concentrations between the composition and physiological fluids. It is also preferable to buffer the saline in order to maintain pH compatible with normal physiological conditions. Also, in certain instances, it can be necessary to maintain the pH at a particular level in order to ensure the stability of certain composition components such as the glycopeptides.
Any physiologicaUy acceptable buffer can be used herein, but phosphate buffers are preferred. Other acceptable buffers such acetate, tris, bicarbonate, carbonate, or the Uke can be used as substitutes for phosphate buffers. The pH of the aqueous component wfll preferably be between about 6.0-8.0.
When the submicron emulsion is initiaUy prepared, however, unadulterated water is preferred as the aqueous component of the emulsion. Increasing the salt concentration makes it more difficult to achieve the desired smaU droplet size. When the final immunogenic compositions is prepared from the adjuvant, the antigenic material can be added in a buffer at an appropriate osmolality to provide the desired immunogenic composition.
The quantity of the aqueous component employed in these compositions wUl be that amount necessary to bring the value of the composition to unity. That is, a quantity of aqueous component sufficient to make 100% will be mixed, with the other components Usted above, in order to bring the compositions to volume.
A substantial number of emulsif ing and suspending agents are generaUy used in the pharmaceutical sciences. These include naturaUy derived materials such as gums from trees, vegetable protein, sugar-based polymers such as alginates and ceUulose, and the like. Certain oxypolymers or polymers having a hydroxide or other hydrophiUc substituent on the carbon backbone have surfactant activity, for example, povidone, polyvinyl alcohol, and glycol ether-based mono- and poly-functional compounds. Long chain fatty-acid-derived compounds form a third substantial group of emulsifying and suspending agents which could be used in this invention. Any of the foregoing surfactants are useful so long as they are non-toxic.
Specific examples of suitable emulsifying agents (also referred to as surfactants or detergents) which can be used in accordance with the present invention are disclosed in commonly owned International patent appUcation PCT/US00/0331. Surfactants are generally divided into four basic types: anionic, cationic, zwitterionic, and nonionic. Examples of anionic detergents include, but are not limited to, alginic acid, capryUc acid, cholic acid, l-decanesulfonic acid, deoxycholic acid, 1-dodecanesulfonic acid, N-lauroylsarcosine, and taurochoUc acid, and the Uke. Cationic detergents include, but are not limited to, cetrimide (hexadecyllximemylammonium bromide, or CTAB), benzalkonium chloride, dimethyl dioctodecyl ammonium (DDA) bromide, DOTAP, dodecyltrimethylammonium bromide, benzyldimethylhexadecyl ammonium chloride, cetylpyridinium chloride, methylbenzethonium chloride, and 4-picoline dodecyl sulfate, and the Uke. Examples of zwitterionic detergents include, but are not United to, 3-[(3-cholamidopropyl)- dimethylammomo]-l-propanesulfonate (commonly abbreviated CHAPS), 3-[(cholamidopropyl)- dimethylammomo]-2-hyα!roxy-l-propanesulfonate (generaUy abbreviated CHAPSO) N-dodecyl- N,N-d methyl-3-arnmomo-l-propanesulfonate, and lyso-α-phosphatidylcholine, and the like. Examples of nonionic detergents include, but are not Umited to, decanoyl-N-methylglucamide, diethylene glycol monopentyl ether, n-dodecyl β-D-glucopyranoside, ethylene oxide condensates of fatty alcohols (e.g., sold under the trade name Lubrol), polyoxyethylene ethers of fatty acids (particularly Cι2-C2o fatty acids), polyoxyethylene sorbitan fatty acid ethers (e.g., sold under the trade name Tween), and sorbitan fatty acid ethers (e.g., sold under the trade name Span), and the Uke.
A particularly useful group of surfactants are the sorbitan-based non-ionic surfactants, such as the commercially available SPAN® or ARLACEL®, usuaUy with a letter or number designation which distinguishes between the various mono-, di- and triester substituted sorbitans. A related group of surfactants comprises polyoxyethylene sorbitan monoesters and polyoxyethylene sorbitan triesters, commerciaUy available under the mark TWEEN®. The TWEEN® surfactants can be combined with a related sorbitan monoester or triester surfactants to promote emulsion stability.
The size of the oU droplets can be varied by changing the ratio of detergent to oil (increasing the ratio decreases droplet size, operating pressure (increasing operating pressure reduces droplet size), temperature (increasing temperature decreases droplet size), and adding an amphipathic immunostimulating agent (adding such agents decreases droplet size). Actual droplet size wiU vary with the particular detergent, oil, and immunostimulating agent (if any) and with the particular operating conditions selected. Droplet size can be verified by use of sizing instruments, such as the commercial Sub-Micron Particle Analyzer (Model N4MD) manufactured by the Coulter Corporation, and the parameters can be varied using the guidelines set forth above until substantiaUy all droplets are less than 1 micron in diameter, preferably less than 0.8 microns in diameter, and most preferably less than 0.5 microns in diameter. By substantiaUy all is meant at least about 80% (by number), preferably at least about 90%, more preferably at least about 95%, and most preferably at least about 98%. The particle size distribution is typically Gaussian, so that the average diameter is smaUer than the stated limits.
A preferred oU droplet emulsion is MF59. MF59 can be made according to the procedures described in, for example, Ott et al, Vaccine Design: The Subunit And Adjuvant Approach, 1995, M.F. PoweU and M.J. Newman, Eds., Plenum Press, New York, p. 277-296; Singh etal, Vaccine, 1998, 16, 1822-1827; Ott et al, Vaccine, 1995, 13, 1557-1562; and Valensi et α/., J. Immunol, 1994, 153, 4029-39, the disclosures of which are incorporated herein by reference in their entirety.
Other oU droplet emulsions include, for example, SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and Ribi® adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial ceU wall components from the group consisting of monophosphoryUpid A (MPL), trehalose dirnycolate (TDM), and ceU waU skeleton (CWS), preferably MPL + CWS (DetoxJ) (for a further discussion of suitable submicron ou-in- water emulsions for use herein, see commonly owned, patent appUcation no. 09/015,736, filed on January 29, 1998).
After preparing the microparticles of the invention, whether of the polymer type or the submicron emulsion type, macromolecules such as polypeptides and vector constructs may be adsorbed thereto as previously discussed. The submicron emulsion microparticles of the present invention may also have macromolecules entrapped or encapsulated within them, as well as having macromolecules adsorbed thereon. Thus, for example, one of skiU in the art may prepare in accordance with the invention microparticles having encapsulated adjuvants with ELVIS vector adsorbed thereon,. or microparticles having encapsulated antigen with RNA vector construct adsorbed thereon. The invention contemplates a variety of combinations of nucleic acid macromolecules adsorbed on and entrapped within microparticles, along with other nucleic acids as weU as other antigenic molecules. Preferably, the microparticles of the invention have ELVIS vectors or RNA vector constructs adsorbed thereon. AdditionaUy, any of the embodiments of the microparticles of the invention may be delivered in conjunction with electroporation.
3. ELVIS vectors
ELVIS vectors are Eukaryotic Layered Vector Initiation Systems, which are generally described in U.S. Patents 5,814,482 and 6,015,686, cited above, as weU as in International Patent AppUcations WO 97/38087 and WO 99/18226. In one embodiment, an ELVIS vector is derived from the genome of an alphavirus, more preferably from Sindbis virus (SIN), Semliki Forest virus (SFV), Venezuelan equine encephahtis virus (VEE), or Ross River virus (RRV). The alphavirus is an RNA virus of approximately 11-12 kb in length, which contains a 5' cap and a 3' polyadenylate tail. The mature infectious virus is composed of the genomic RNA enveloped by the nucleocapsid and envelope proteins. Alphavirus infection of host ceUs occurs by a receptor specific event and culminates in release of genomic RNA into the cytoplasm During viral repUcation, the viral- encoded envelope glycoproteins El and E2 are synthesized and embedded in the host ceU membrane, through which progeny virions bud and release to the outside of the host cell.
RepUcation of the viral genome begins with the genomic RNA strand serving as the template for synthesis of a complementary negative RNA strand. The negative RNA strand then serves as a template for fuU-length genomic RNA, and for an internally initiated positive-strand subgenomic RNA. The nonstructural proteins are translated from the genomic strand, whUe the structural proteins are translated from the subgenomic strand. AU the viral genes are expressed first as polyproteins, then post-translationally processed into individual proteins by proteolytic cleavage.
An alphavirus vector repUcon may be buUt by replacing certain portions of the viral genome (e.g., structural protein genes) with a selected heterologous nucleic acid sequence. Thus, in certain embodiments, an alphavirus replicon vector may comprise a 5' sequence capable of initiating transcription of an alphavirus, a nucleotide sequence encoding the alphavirus nonstructural proteins, an alphaviral junction region promoter, an alphavirus RNA polymerase recognition site, and a 3' polyadenylate tract. AdditionaUy, the alphavirus vector repUcon may be contained as a cDNA copy within an alphavirus vector construct. Such vector constructs typicaUy comprise a 5' promoter capable of initiating synthesis of RNA from cDNA positioned upstream and operably associated with the vector cDNA, such that transcription produces the vector replicon RNA. The vector construct also may contain and a 3' sequence controlling transcription termination. A heterologous nucleic acid sequence may be present upstream or downstream of the viral junction region.
The ELVIS vector capitalizes on the mechanism of RNA virus repUcation to achieve deUvery of a heterologous nucleotide sequence of interest by using a double-layered approach (for example, based on the above-described alphavirus vector construct). In general, an ELVIS vector provides a layered expression system capable of ampUfying the amount of RNA encoding the gene product of interest because the first layer initiates transcription of a second layer. Thus, a typical ELVIS vector comprises a 5' promoter capable of initiating synthesis of RNA from cDNA, a cDNA complement of a construct capable of autonomous repUcation in a ceU, and which construct is also capable of expressing a heterologous nucleic acid sequence, and a 3' sequence controlling transcription termination. The construct capable of autonomous repUcation and expression of the selected nucleic acid sequence may be an alphavirus vector construct. Thus, the first layer of the DNA ELVIS vector transcribes the RNA alphavirus vector construct, from which expression of the selected heterologous nucleic acid sequence is achieved.
An alphavirus-based ELVIS vector may be constructed by first preparing a cDNA complementary to an alphavirus genome. The cDNA corresponding to the genomic RNA is then deleted of sequences encoding one or more viral structural proteins which then may be replaced with heterologous DNA encoding the gene-product of interest, thereby preventing packaging of mature virus and enabling amplification of the heterologous sequence. The modified cDNA containing the heterologous sequence is then inserted within the first layer of the ELVIS vector. Upon entry into the cell and nucleus, the ELVIS vector wiU be transcribed and the resulting mRNA molecules, which are RNA vectors capable of self-repUcation, wiU begin to repUcate and translate polypeptides, including the heterologous gene of interest.
While atypical Sindbis-derived alphavirus vector construct is preferred, other alphavirus species may be readily used according to the teachings provided herein. Alternatively, vectors derived from any RNA virus may be utilized, particularly those from positive-stranded viruses.
The construction of an ELVIS vector, in general, is described in U.S. Patents 5,814,482 and 6,015,686. Briefly, RNA is obtained from an RNA virus, then cDNA is synthesized by PCR ampUfication using appropriate primers for particular genes or portions of the RNA virus, which primers may also contain additional restriction sites as necessary. The cDNA fragments are then cloned into a plasmid and transformed into an appropriate host such as E. coli. Positive colonies are grown for plasmid purification, and then plasmids are assembled into the desired ELVIS vector with a portion having heterologous DNA such as a reporter gene (e.g., GFP) or a desired gene coding for an antigen. Example 3 below describes a particular preferred ELVIS vector (pSINCP) used in accordance with the instant invention.
4. RNA and pCMV vector constructs
In other embodiments of the present invention, an RNA vector construct or RNA repUcon vector is used directly, without the requirement for introduction of DNA into a ceU and transport to the nucleus where transcription would occur. By using the RNA vector for direct deUvery into the cytoplasm of the host ceU, autonomously repUcation and translation of the heterologous nucleic acid sequence occurs efficiently. In this embodiment, the RNA vector construct or RNA repUcon vector is obtained by in vitro transcription from a DNA-based vector construct. Preferably, the RNA vector construct or RNA repUcon vector is derived from the genome of an alphavirus, more preferably from Sindbis virus (SIN), SemUki Forest virus (SFV), Venezuelan equine encephahtis virus (VEE), or Ross River virus (RRV). In other embodiments, the RNA vector construct is derived from a virus other than an alphavirus. Preferably, such other viruses used for the derivation of RNA vector constructs are positive-stranded RNA viruses, and more preferably they are picornaviruses, flaviviruses, rabiviruses, or coronaviruses. Compositions and methods for in vitro transcription of alphavirus-based RNA vectors is provided in detail elsewhere (see U.S. Patent 5,842,723 and Polo et al., 1999, PNAS 96:4598-603). The RNA vector is then adsorbed to a microparticle of the invention for deUvery as detailed herein. While a typical alphavirus RNA vector from SIN, SFV, VEE or RRV is preferred, similar vectors derived from other alphavirus species may be readily substituted.
In other embodiments of the present invention, pCMV vector constructs are used. Such vector constructs are weU known in the art. A particularly preferred pCMV vector contains the immediate-early enhancer/promoter of CMV and a bovine growth hormone terminator. It is described in detaU in Chapman, B. S., et al. 1991. "Effect of intron A fromhuman cytomegalovirus (Towne) immediate-early gene on heterologous expression in mammalian ceUs." Nucleic Acids Res. 19:3979-86.
5. Adjuvants
Adjuvants may optionally be used to enhance the effectiveness of the pharmaceutical compositions, with Thl stimulating adjuvants being particularly preferred. The adjuvants may be administered concurrently with the microparticles of the present invention, e.g., in the same composition or in separate compositions. Alternatively, an adjuvant may be administered prior or subsequent to the microparticle compositions of the present invention. In another embodiment, the adjuvant, such as an immunological adjuvant, may be encapsulated in the microparticle. Adjuvants, just as any macromolecules, may be encapsulated within the microparticles using any of the several methods known in the art. See, e.g., U. S. Patent No. 3,523,907; Ogawa et al., Chem Pharm. Bull. (1988) 36J095-1103; O'Hagan et al., Vaccine (1993) 11:965-969 and Jefferey et al., Pharm. Res. (1993) 10:362. Alternatively, adjuvants may be adsorbed on the microparticle as described above for any macromolecule. Alternatively, adjuvants may comprise the oil droplet emulsions of the present invention.
Immunological adjuvants include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc.; (2) other oil-in water emulsion formulations (with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial ceU waU components), such as for example (a) MF59 (International Publication No. WO90/14837; Chapter 10 in Vaccine design: the subunit an adjuvant approach, eds. PoweU & 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 110Y microfluidizer (Mcrofluidics, 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 ceU waU components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell watt skeleton (CWS), preferably MPL + CWS (Detox™) (for a further discussion of suitable submicron oil-in-water emulsions for use herein, see commonly owned, patent appUcation no. 09/015,736, filed on January 29, 1998); (3) saponin adjuvants, such as Quil A, or QS21 (e.g., Stimulon™ (Cambridge Bioscience, Worcester, MA)) may be used or particle generated therefrom such as ISCOMs (immunostimulating complexes), which ICOMS may be devoid of additional detergent e.g., WO00/07621; (4) Complete Freunds Adjuvant (CFA) and Incomplete Freunds Adjuvant (IFA); (5) cytokines, such as interleukins (e.g. IL-1, IL-2, IL-4, IL- 5, IL-6, IL-7, IL-12 (W099/44636), etc.), interferons (e.g. gamma interferon), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), etc.; (6) monophosphoryl Upid A (MPL) or 3-O-deacylated MPL (3dMPL) e.g. GB-2220221, EP-A-0689454, optionaUy in the substantial absence of alum when used with pneumococcal saccharides e.g. WO00/56358; (7) combinations of 3dMPL with, for example, QS21 and/or on-in-water emulsions, e.g., EP-A-0835318, EP-A- 0735898, EP-A-0761231; (8) oUgonucleotides comprising CpG motifs (Roman et al., Nat. Med, 1997, 3, 849-854; Weiner et al., PNAS USA, 1997, 94, 10833-10837; Davis et al., J. Immunol 1988, 160, 870-876; Chu et al., J. Exp. Med, 1997, 186, 1623-1631; Lipford et al., Eur. J. Immunol. 1997, 27, 2340-2344; Moldoveanu et al., Vaccine, 1988, 16, 1216-1224, Krieg et al., Nature, 1995, 374, 546-549; Klinman et al., PNAS USA, 1996, 93, 2879-2883: BaUas et al., J. Immunol, 1996, 157, 1840-1845; Cowdery et al., J. Immunol, 1996, 156, 4570-4575; Halpern et al., Cell. Immunol, 1996, 167, 72-78; Yamamoto et al., Jpn. J. Cancer Res., 1988, 79, 866-873; Stacey et al., J. Immunol, 1996, 157, 2116-2122; Messina et al., J. Immunol, 1991, 147, 1759- 1764; Yi et al., J. Immunol, 1996, 157, 4918-4925; Yi et al., J. Immunol, 1996, 157, 5394-5402; Yi et ύ., J. Immunol, A998, 160, 4755-4761; and Yi et al., ./ Immunol, 1998, 160, 5898-5906; International patent appUcations WO96/02555, W098/16247, WO98/18810, WO98/40100, W098/55495, W098/37919 and W098/52581) i.e. containing at least one CG dinucleotide, with 5 methylcytosine optionally being used in place of cytosine; (9) a polyoxyethylene ether or a polyoxyethylene ester e.g. W099/52549; (10) a polyoxyethylene sorbitan ester surfactant in combination with an octoxynol (WOO 1/21207) or a polyoxyethylene alkyl ether or ester surfactant in combination with at least one additional non-ionic surfactant such as an octoxynol (WO01/21152); (11) a saponin and an iiimiunostimulatory oUgonucleotide (e.g., a CpG oligonucleotide) (WO00/62800); (12 ) an immunostimulant and a particle of metal salt e.g. WO00/23105; (13) a saponin and an oil-in-water emulsion e.g. W099/11241; (14) a saponin (e.g., QS21) + 3dMPL + IL-12 (optionaUy + a sterol) e.g. W098/57659; (15) detoxified mutants of a bacterial ADP-ribosylating toxin such as a cholera toxin (CT), a pertussis toxin (PT), or an E. coli heat-labtte toxin (LT), particularly LT-K63 (where lysine is substituted for the wild-type amino acid at position 63) LT-R72 (where arginine is substituted for the wUd-type amino acid at position 72), CT-S109 (where serine is substituted for the wUd-type amino acid at position 109), and PT- K9/G129 (where lysine is substituted for the wild-type amino acid at position 9 and glycine substituted at position 129) (see, e.g., International PubUcation Nos. W093/13202 and W092/19265); and (16) other substances that act as immunostimulating agents to enhance the effectiveness of the composition.. Alum (especiaUy aluminum phosphate and/or hydroxide) and MF59 are preferred.
Muramyl peptides include, but are not limited to, N-acetyl-muramyl-L-threonyl-D- isoglutamine (thr-MDP), N-acteyl-normuramyl-L-alanyl-D-isogluatme (nor-MDP), N- acetylmuramyl-L-alanyl-D-isogluatmmy huydroxyphosphoryloxy)-ethylamine (MTP-PE), etc.
For additional examples of adjuvants, see Vaccine Design, The Subunit and the Adjuvant Approach, PoweU, M.F. and Newman, MJ, eds., Plenum Press, 1995)
Thus, an optional additional component of the compositions of the present invention preferably is an adjuvant such as aluminum salts or an oUgonucleotide which comprises at least one CpG motif. As used herein, the phrase "CpG motif' refers to a dinucleotide portion of an oligonucleotide which comprises a cytosine nucleotide followed by a guanosine nucleotide. Such oUgonucleotides can be prepared using conventional oUgonucleotide synthesis weU known to the slatted artisan. Preferably, the oUgonucleotides of the invention comprise a modified backbone, such as a phosphorothioate or peptide nucleic acid, so as to confer nuclease resistance to the oligonucleotide. Modified backbones are well known to those skilled in the art. Preferred peptide nucleic acids are described in detaU in U.S. Patent Numbers 5,821,060, 5,789,573, 5,736,392, and 5,721,102, Japanese Patent No. 10231290, European Patent No. 839,828, and PCT PubUcation Numbers WO 98/42735, WO 98/42876, WO 98/36098, WO 98/27105, WO 98/20162, WO 98/16550, WO 98/15648, WO 98/04571, WO 97/41150, WO 97/39024, and WO 97/38013, the disclosures of which are incorporated herein by reference in their entirety.
The oUgonucleotide preferably comprises between about 6 and about 100 nucleotides, more preferably between about 8 and about 50 nucleotides, most preferably between about 10 and about 40 nucleotides. In addition, the oUgonucleotides of the invention can comprise substitutions of the sugar moieties and nitrogenous base moieties. Preferred oUgonucleotides are disclosed in, for example, Krieg et al, Proc. Natl. Acad. Sci. USA, 1998, 95, 12631-12636, Klinman et al, Proc. Natl. Acad. Sci. USA, 1996, 93, 2879-2883, Weiner et al, Proc. Natl. Acad. Sci. USA, 1991, 94, 10833-10837, Ch et al, J. Exp. Med, 1997, 186, 1623-1631, Brazolot-MiUan et al. , Proc. Natl. Acad. Sci. USA, 1998, 95, 15553-15558, BaUas et al, J. Immunol, 1996, 157, 1840- 1845, Cowdery etal, J. Immunol, 1996, 156, 4570-4575, Halpern tα/., Cell. Immunol, 1996, 167, 72-78, Yamamoto et al., Jpn. J. Cancer Res., 1988, 79, 866-873, Stacey etal, J. Immunol, 1996, 157, 2116-2122, Messina et al., J. Immunol, 1991, 147, 1759-1764, Yi etal, ! Immunol, 1996, 157, 4918-4925, Yi et al, J. Immunol, 1996, 157, 5394-5402, Yi et al, J. Immunol, 1998, 160, 4755-4761, Roman et at, Nat. Med, 1991, 3, 849-854, Davis et al, J. Immunol, 1998, 160, 870-876, Lipford et al, Ear. J. Immunol, 1997, 27, 2340-2344, Moldoveanu et al, Vaccine, 1988, 16, 1216-1224, Yi etal, J. Immunol, 1998, 160, 5898-5906, PCT PubUcation WO 96/02555, PCT PubUcation WO 98/16247, PCT PubUcation WO 98/18810, PCT PubUcation WO 98/40100, PCT PubUcation WO 98/55495, PCT PubUcation WO 98/37919, and PCT PubUcation WO 98/52581, the disclosures of which are incorporated herein by reference in their entirety. It is to be understood that the oUgonucleotides of the invention comprise at least one CpG motif but can contain a pluraUty of CpG motifs.
Preferred oUgonucleotides comprise nucleotide sequences such as, for example, tccatgacgttcctgacgtt (SEQ ID NOJ), ataatcgacgttcaagcaag (SEQ ID NO:2), ggggtcaacgttgagggggg (SEQ ID NO:3), tctcccagcgtgcgccat (SEQ ID NO:4), gagaacgctcgaccttcgat (SEQ ID NO: 5), tccatgtcgttcctgatgct (SEQ ID NO: 6), tccatgacgttcctgatgct (SEQ ID NO:7), gctagacgttagcgt (SEQ ID NO:8), atcgactctcgagcgttctc (SEQ ID NO:9), gaaccttccatgctgttccg (SEQ ID NO: 10), gctagatgttagcgt (SEQ ID NO: 11), tcaacgtt (SEQ ID NO: 12), gcaacgtt (SEQ ID NO: 13), tcgacgtc (SEQ ID NO: 14), tcagcgct (SEQ ID NO: 15), tcaacgct (SEQ ID NOJ6), tcatcgat (SEQ ID NOJ7), tcttcgaa (SEQ ID NOJ 8), tgactgtgaacgttcgagatga (SEQ ID NOJ9), tgactgtgaacgttagcgatga (SEQ ID NO:20), tgactgtgaacgttagagcgga (SEQ ID NO:21), gtttgcgcaacgttgttgccat (SEQ ID NO:22), atggcaacaacgttgcgcaaac (SEQ ID NO:23), cattggaaaacgttcttcgggg (SEQ ID NO:24), ccccgaagaacgttttccaatg (SEQ ID NO:25), attgacgtcaat (SEQ ID NO:26), ctttccattgacgtcaatgggt (SEQ ID NO:27), and tccatacgttcctgacgtt (SEQ ID NO:28). In preferred embodiments of the invention, the oligonucleotide comprises a CpG motif flanked by two purines at the 5' side of the motif and two pyrimidines at the 3' side of the motif. It is to be understood, however, that any oligonucleotide comprising a CpG motif can be used in the present invention as long as the oligonucleotide induces an increase in Thl lymphocyte stimulation when combined with the microparticle compositions described herein.
6. Antigens
The present invention is also directed to immunogenic compositions comprising the microparticles described above with adsorbed macromolecules, preferably vector constructs encoding antigens and/or antigen per se. Generally, an antigen stimulates the proliferation of T- lymphocytes, preferably Thl lymphocytes, with receptors for the antigen and can react with the lymphocytes to initiate the series of responses designated cell-mediated in-munity. An antigen may thus induce a CTL response, and/or a humoral response, and may induce cytokine production.
An epitope is within the scope of this definition of antigen. An epitope is that portion of an antigenic molecule or antigenic complex that determines its immunological specificity. Commonly, an epitope is a peptide or polysaccharide in naturally occurring antigens. In artificial antigens it can be a low molecular weight substance such as an arsaniUc acid derivative. An epitope will react specifically in vivo or in vitro with homologous antibodies or T lymphocytes. Alternative descriptors are antigenic determinant, antigenic structural grouping and haptenic grouping.
In preferred embodiments of the invention, the antigenic substance is derived from a virus such as, for example, human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), herpes simplex virus (HSV), cytomegalovirus (CMV), influenza virus (flu), and rabies virus. Preferably, the antigenic substance is selected from the group consisting of HSV glycoprotein gD , HIV glycoprotein gpl20, HIV p55 gag, and polypeptides from he pol and tat regions. In other preferred embodiments of the invention, the antigenic substance is derived from a bacterium such as, for example, Helicobacter pylori, Haemophilus influenza, cholera, diphtheria, tetanus, Neisseria meningitidis, and pertussis. In other preferred embodiments of the invention, the antigenic substance is from a parasite such as, for example, a malaria parasite. In another preferred embodiment of the present invention, the antigen is adsorbed to the surface of a microparticle of the present invention.
Antigens can be produced by methods known in the art or can be purchased from commercial sources. Antigens within the scope of this invention include whole inactivated virus particles, isolated virus proteins and protein subunits, whole cells and bacteria, cell membrane and cell wall proteins, and the like. Some preferred antigens are described below.
Herpes simplex virus (HSV) rgD2 is a recombinant protein produced in genetically engineered Chinese hamster ovary ceUs. This protein has the normal anchor region truncated, resulting in a glycosylated protein secreted into tissue culture medium The gD2 can be purified in the CHO medium to greater than 90% purity. Human immunodeficiency virus (HIV) env-2-3 is a recombinant form of the HIV enveloped protein produced in genetically engineered Saccharomyces cerevisae. This protein represents the entire protein region of HTV gpl20 but is non-glycosylated and denatured as purified from the yeast. HTV gpl20 is a fully glycosylated, secreted form of gpl20 produced in CHO ceUs in a fashion similar to the gD2 above. Additional HSV antigens suitable for use in immunogenic compositions are described in PCT PubUcations WO 85/04587 and WO 88/02634, the disclosures of which are incorporated herein by reference in their entirety. Mixtures of gB and gD antigens, which are truncated surface antigens lacking the anchor regions, are particularly preferred.
Additional HIV antigens suitable for use in immunogenic compositions are described in U.S. appUcations serial no. 490,858, filed March 9, 1990, and pubUshed European appUcation number 181150 (May 14, 1986), as weU as U.S. appUcations serial nos. 60/168,471; 09/475,515; 09/475,504; and 09/610,313, the disclosures of which are incorporated herein by reference in their entirety.
Cytomegalovirus antigens suitable for use in immunogenic compositions are described in U.S. Patent No. 4,689,225, U.S. appUcation serial number 367,363, filed June 16, 1989 and PCT Publication WO 89/07143, the disclosures of which are incorporated herein by reference in their entirety.
Hepatitis C antigens suitable for use in immunogenic compositions are described in PCT/US 88/04125, published European application number 318216 (May 31, 1989), pubUshed Japanese appUcation number 1-500565 filed November 18, 1988, Canadian appUcation 583,561, and EPO 388,232, disclosures of which are incorporated herein by reference in their entirety. A different set of HCV antigens is described in European patent appUcation 90/302866.0, filed March 16, 1990, and U.S. application serial number 456,637, filed December 21, 1989, and PCT/US90/01348, the disclosures of which are incorporated herein by reference in their entirety.
Immunogenic compositions of the invention can be used to immunize birds and mammals against diseases and infection, including without Umitation cholera, diphtheria, tetanus, pertussis, influenza, measles, meningitis, mumps, plague, poUomyeUtis, rabies, Rocky Mountain spotted fever, rubeUa, smaUpox, typhoid, typhus, feline leukemia virus, and yeUow fever.
Certain immunogenic compositions of the invention wttl employ an effective amount of an antigen. For example, there may be included an amount of antigen which, in combination with an adjuvant, will cause the subject to produce a specific and sufficient immunological response, so as to impart protection to the subject from the subsequent exposure to a virus, bacterium, fungus, mycoplasma, or parasite.
In other embodiments, a composition comprising an antigen wiU be used to boost the unmunological response of a previously acϊministered vector construct, which preferably comprises a heterologous nucleic acid sequence that encodes the antigen. More preferably, the antigen is associated with (e.g., adsorbed to) the microparticles described herein and/or the antigen is coadministered with an adjuvant.
No single dose designation can be assigned which wfll provide specific guidance for each and every antigen which can be employed in this invention. The effective amount of antigen wiU be a function of its inherent activity and purity and is empirically determined by those of ordinary skiU in the art via routine experimentation. It is contemplated that the adjuvant compositions of this invention can be used in conjunction with whole cell or viral immunogenic compositions as weU as with purified antigens or protein subunit or peptide immunogenic compositions prepared by recombinant DNA techniques or synthesis.
Where the antigen is provided in connection with an emulsion, because the adjuvant compositions of the invention are stable, the antigen and emulsion can typicaUy be mixed by simple shaking. Other techniques, such as passing a mixture of the adjuvant and solution or suspension of the antigen rapidly through a small opening (such as a hypodermic needle), readily provide a useful immunogenic composition.
The immunogenic compositions according to the present invention comprise about 1 nanogramto about 1000 microgra s of nucleic acid, preferably DNA such as, for example, CpG ohgonucleotides. In some preferred embodiments, the immunogenic compositions contain about 10 nanograms to about 800 micrograms of nucleic acid. In some preferred embodiments, the immunogenic compositions contain about 0J to about 500 micrograms of nucleic acid. In some preferred embodiments, the immunogenic compositions contain about 1 microgramto about 10 milligrams of nucleic acid. In some preferred embodiments, the immunogenic compositions contain about 250 micrograms to about 1 milligram of nucleic acid. In some preferred embodiments, the immunogenic compositions contain about 500 micrograms to about 1 miUigram of nucleic acid. One skiUed in the art can readily formulate an immunogenic composition comprising any desired amount of nucleic acid. The immunogenic compositions according to the present invention are provided sterile and pyrogen free. The immunogenic compositions can be conveniently administered in unit dosage form and can be prepared by any of the methods weU known in the pharmaceutical art, for example, as described in Remington 's Pharmaceutical Sciences (Mack Pub. Co., Easton, PA, 1980), the disclosure of which is incorporated herein by reference in its entirety.
The present invention is also directed to methods of stimulating an immune response in a host animal comprising administering to the animal one or more immunogenic compositions described above in an amount effective to induce an immune response. The host animal is preferably a mammal, more preferably a human. Preferred routes of administration include, but are not limited to, intramuscular, intraperitoneal, intradermal, subcutaneous, intravenous, intraarterial, intraoccular and oral as weU as transdermal or by inhalation or suppository. Most preferred routes of administration include intramuscular, intraperitoneal, intradermal and subcutaneous injection. According to some embodiments of the present invention, the immunogenic compositions are administered to a host animal using a needleless injection device, which are wett known and widely available. One having ordinary skill in the art can, foUowing the teachings herein, use needleless injection devices to dehver immunogenic compositions to cells of an individual. Additionally, the embodiments of the invention may be deUvered together with electroporation.
The present invention is also directed to methods of immunizing a host animal against a viral, bacterial, or parasitic infection comprising administering to the animal one or more immunogenic compositions described above in an amount effective to induce a protective response. The host animal is preferably a mammal, more preferably a human. Preferred routes of administration are described above. While prophylactic or therapeutic treatment of the host animal can be directed to any pathogen, preferred pathogens, including, but not limited to, the viral, bacterial and parasitic pathogens described above.
The present invention is also directed to methods of inducing an immune response in a host animal comprising administering to the animal one or more immunogenic compositions described above in an amount effective to induce an immune response. The host animal is preferably a mammal, more preferably a human. Preferred routes of administration are described above. One skilled in the art is readily famiUar with immune responses and measurements thereof. The present invention contemplates the use of polymer microparticles or submicron emulsion microparticles with adsorbed macromolecule to eUcit an immune response alone, or in combination with another macromolecule. That is, the invention encompasses microparticles with adsorbed nucleic acid, submicron emulsions with adsorbed nucleic acid or immunostimulating molecule, and the combination of microparticles with adsorbed nucleic acid together with submicron emulsions with adsorbed nucleic acid or irnmunostimulating molecule. Electroporation may also be used to improve delivery of the nucleic acid.
As demonstrated by the foUowing Examples, the present invention's polymer microparticles with adsorbed macromolecules eUcit strong immune responses. Additionally, the present invention's submicron emulsion microparticles also eUcit strong immune responses. The combination of the present invention's microparticles with adsorbed macromolecules is therefore a powerful tool for eliciting immune responses.
AU references cited herein are hereby incorporated by reference in their entirety.
C. Experimental
Below are examples of specific embodiments for carrying out the present invention. The Examples are offered for illustrative purposes only, and are not intended to Umit the scope of the present invention in any way. Those slotted in the art wiU recognize modifications that are within the spirit and scope of the invention.
Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be aUowed for.
Example 1 Preparation of Polymer Microparticles with Adsorbed Nucleic Acid PLG-CTAB microparticles were prepared using a modified solvent evaporation process. Briefly, the microparticles were prepared by emulsifying 10 ml of a 5% w/v polymer solution in methylene chloride with 1 ml of T.E. buffer at high speed using an IKA homogenizer. The primary emulsion was then added to 50ml of distilled water containing cetyl trimethyl ammonium bromide (CTAB) (0.5% w/v). This resulted in the formation of a w/o/w emulsion which was stirred at 6000 rp for 12 hours at room temperature, aUo ing the methylene chloride to evaporate. The resulting microparticles were washed twice in distilled water by centrifugation at 10,000 g and freeze dried.
For a typical batch of 100 mg of DNA adsorbed microparticles, 100 mg of PLG-CTAB cationic microparticles were weighed into a glass vial and resuspended with 5 ml volume of 200μg/ml of DNA solution (i.e., the plasmid pCMV or pSINCP containing gpl40 or p55gag) in T.E. Buffer. The suspension was vortexed for a one minute to uniformly disperse the microparticles in the DNA solution. The vial was set on a shaker (slow speed) at 4 C for overnight adsorption. The next day the microparticles were centrifuged down at 8000 rpm on a Beckman centrifuge for 10 minutes and the supernatant was coUected for DNA quantitation. The peUet was washed once with IX TE buffer by resuspending the pellet in 1 X TE buffer, dispersing with a spatula and centrifuging at 8,000 rpm for 10 minutes. The final peUet was resuspended in a n±iimum amount of de-ionized water (about 2 ml) by dispersing the peUet with a spatula, and freeze dried on a bench top lyophϋizer (Labconco) for 24 hours.
The supernatant was assayed for DNA content by reading the absorbance at 260 nm. Amount of DNA adsorbed on the microparticles was calculated by subtracting the amount in the supernatant from the total DNA input (1 mg per 100 mg of microparticles. The total load was estimated by dissolving 5 mg of final formulation in 0.5 M NaOH/1% SDS solution and reading the clear solution after hydrolysis at 260 nm.
Example 2 Preparation of Submicron emulsion Microparticles with Adsorbed Nucleic Acid A submicron emulsion formed fro MF59 and DOTAP was prepared by providing DOTAP (in chloroform) in a beaker and aUowing it to evaporate down to 200ul. Tween (0.5% w/w), Squalene (5.0% w/w) and Span (0.5% w/w) were added and homogenized for 1 minute using an Omni homogenizer with a 10mm probe at 1 OK revs/min in order to provide a homogeneous feedstock for final emulsification. This was passed 5 times through a Microfluidizer Mil OS homogenizer ( crofluidics Co., Newton, MA) at ~800 psi. The zeta potential of the emulsion, which is a measure of net surface charge, was measured on a DELSA 440 SX Zetasizer from Coulter and found to be approximately + 55 mV. DNA (either 1 mg HTV-1 gpl40 DNA or 0.5 mg of p55 gag DNA, present in pCMV or pSFNCP) was adsorbed by incubation with the submicron emulsion overnight at 4°C.
Example 3 Preparation of ELVIS Vectors and Other Vector Constructs for Adsorption to Microparticles Construction of alphavrrus-based ELVIS and replicon vectors was performed using Sindbis virus as a representative example. As wiU be appreciated, the foUowing may be readily appUed to the derivation of vectors from any alphavirus by one of skiU inthe art. Approximately 107 BHK-21 cells were infected with the SINDCchiron strain of Sindbis virus (ATCC deposit VR-2643, April 13, 1999) at a MOI of 1 PFU/ceU. At 24 hours post-infection, after development of CPE, total RNA was isolated from the ceUs using the TRIzol Reagent (GIBCO/BRL) according to the manufacturer's instructions. After purification, viral RNA was dissolved in nuclease-free water, aUquoted, and stored at -80°C for subsequent use in cDNA cloning.
Synthesis of cDNA was accompUshed by PCR ampUfication, using the primer sets shown below (Sindbis nucleotide numbering indicated for each primer):
1 CCACΛ46C7TGATCT TGTACCAGCCTGATGC 11472- 11450 (SEQ ID NO:29) 1 . 1 CCACSA47TCAGCCAGATGAGTGAGGC 10364-10381 (SEQ ID NO:30)
2 CCACMgC77ϋAAπCGACGTACGCCTCAC 10394- 10375 (SEQ ID NO:31) 2. 1 CCAC6AA 77CATATGGGGAAATCATGAGCC 9614-9634 (SEQ ID NO.J2)
3 CCAC SCTTCATAGACCCTCACTGGCTC 9648-9630 (SEQ ID NO:33) 3.1 CCACeWTTAAGATTAGCACCTCAGGACC 8878-8899 (SEQ JJD NO:34)
4 CCAC 6C7TCTACACGGTCCTGAGGTGC 8908-8887 (SEQ ID NO:35) 4.1 CCACGAA 77CGTCCGATCATGGATAACTCC 8294-8315 (SEQ ID NO:36)
5 CCACA4--ΩTGCGCCACCGAGGAC 8347 -8334 (SEQ ID NO:37) 5. 1 CCACSA477TACTGCCATGTGGAGGCC 7797 -7814 (SEQ ID NO:38)
6 CCACC7~CS46TπACCCAACπAAACAGCC 7368-7348 (SEQ ID NO:39) 6.1 CCACS SCTCGCGACAπCMTGTCGAATGC 6426-6446 (SEQ ID NOJO)
7 CCACC7J3 GGAACTCCTCCCAATACTCGTC 6488-6468 (SEQ ID NO:41) 7 . 1 CCACG4SC7rGACCTTGGAGCGCAATGTCC 5843-5862 (SEQ ID NO:42) 8 CCACC7rS4STTTCGACGTGTCGAGCACC 5900-5882 (SEQ ID NO:43) 8.1 CCACGffiCGACCATGGAAGCAATCCGC 4814-4832 (SEQ ID NO:44)
9 CCACC7Tfi46ACGACGGGπATGGTCGAC 4864-4845 (SEQ ID NO:45) 9.1 CCACS SCfCCACGGAGACAGGCACCGC 4246 -4264 (SEQ ID NO:46)
10 CCACCrC SGATCACTπcmCCTAGGCAC 4299-4277 (SEQ ID NO:47)
10 J CCACβ4GC7rGAACTCTCCCGTAGAπTCC 3407-3427 (SEQ ID NO:48)
11 CCACCTt GATCAAGTTGTGTGCCCTTCC 3464-3445 (SEQ ID NO:49) 11.1 CCAC 4ffC7rcCAGGGGATATCATCCTGAC 2742-2761 (SEQ ID NO:50)
12 CCACC7rgiGCTGTCAπACπCATGTCCG 2825-2804 (SEQ ID NO:51) 12.1 CCACβ4ffC TGAACCGCAAACTATACCACAπGC 1976-1999 (SEQ ID NO:52)
13 CCACC7rfi46CπGTACTGCTCCTCπCTG 2042-2023 (SEQ ID NO:53) 13 J CCACβ GC7TGGAGAACGGGTATCGπCC 1029-1047 (SEQ ID NO:54
14 CCACCfCg igCCGGGATGTACGTGCAC 1069-1052 (SEQ ID NO:55) 14.1 CCACfiffiTAπGACGGCGTAGTACACAC 1-20 (SEQ ID NO:56)
Primer pairs 1-5 were used for cloning of the virus structural protein genes, while pairs 6- 14 were for the virus nonstructural protein genes. OUgonucleotides in pairs 1-5 contained additional sequences representing restriction enzyme sites for EcoRl and Hindlll, which are not present in subgenomic RNA of Sindbis virus. OUgonucleotides 6-14 contained sites for Sαcl and Xhol, which are not present in the whole genome of previously sequenced strains of Sindbis virus (these sites are underlined).
Each reverse transcription (RT) reaction was performed in a 50 μl volume using the Superscriptll enzyme (GIBCO/BRL), according to the manufacturer's instructions. Reaction mixtures contained the amount of RNA equivalent to 10s ceUs and 50 p oles of each primer shown below.
Mixturel: primers 1, 3 and 5
Mixture2: primers 2 and 4
Mixture3: primers 6, 9 and 12
Mixture4: primers 8, 11 and 14 RT reactions were frozen and then used subsequently for PCR amplification. PCR reactions were performed using Vent DNA polymerase (NEB) as recommended by the manufacturer. Each 50 μl PCR reaction contained 3 μl of RT mixtures described above and 50 pmoles of primers. A total of 14 reactions were performed (Table 1).
TABLE 1
Figure imgf000056_0001
PCR reactions for fragments 1-5 were performed using the foUowing conditions: 12 cycles of 95°C for 30 seconds, 56DC for 30 seconds and 74°C for 90 seconds. For fragments 6-14, the number of cycles was changed from 12 to 15. A smaU aUquot of each reaction mixture was analyzed by agarose gel electiophoresis to confirm the presence of the fragments-of the expected size. The remaining reaction mixture was extracted with phenol-chloroform and DNA fragments were precipitated using ethanol. For cloning, fragments 1-5 were digested with HindHI and EcoRI, and then Ugated with plasmid pRS2 (pUC19 with additional restriction sites in polyUnker) treated with the same enzymes. Fragments 6-14 were digested with Sacl and Xhol and Ugated with the same pRS2 plasmid treated with Sαcl and Xhol. All recombinant plasmids were transformed into the E. coli XL-1 Blue strain (Stratagene, La JoUa, CA).
In addition, cDNA clones representing the subgenomic promoter region and 3'-end nontranslated regions also were generated using the foUowing primer pairs:
YSIN1F: 5'-GATTCGGTTACTTCCACAGC (SEQ ID NO:57) YSIN1R: 5'-ACTGACGGCTGTGGTCAGTT (SEQ ID NO:58) YSIN2F: 5'-GATGTACTTCCGAGGAACTG (SEQ ID NO:59) YSIN2R: 5'-CCACAAGCTTGAAATGTTAAAAACAAAATTTTGT (SEQ ID NO: 60)
Positive colonies for each transformation were grown for plasmid purification using a QIAGEN kit according to the manufacturer's instructions. The fragments, designated pi -pi 4 correspondingly, were then assembled into the appropriate vector configurations.
The construction of a Eukaryotic Layered Vector Initiation System (ELVIS) and an alphavirus vector construct for in vitro transcription of repUcon vector RNA was accompUshed using the Sindbis virus cDNA clones pl-pl4, plus the subgenomic and 3'-end region fragments as foUows. AnApαl-Mscl fragment, containing the promoter for SP6 RNA polymerase and start of the Sindbis virus genomic RNA, was Ugated with the Mscl-Xho fragment of cloned fragment 14 in Apαl-Xhόl digested plasmid pRS2. The resulting plasmid was named pl5. Next, the Sαcl-EcόRl fragment of p8, the EcoKl-Nsil fragment of p7 and the Nsil-Xhol fragment of p6 were Ugated into Sαcl-Xhol digested pRS2. The resulting plasmid was named pi 6. Next, the Sαcl-Muήl fragment ofpl2, HαsMunl-Nhel fragment ofpll and the Nhel-Xhol fragment ofplO were Ugated into Sαcl- Xhόl digested pRS2 plasmid. The resulting plasmid was named pl7. The Apαl-ApαlA fragment of pl5 and thsApάLl-Mol fragment of pi 3 then were Ugated into Apαl-Xhόl treated pRS2, resulting in the plasmid named pl8. Next, H eApαl-Nsil fragment of pl8 and theAfø'I--ϊ7-øI fragment of pl7 were Ugated together inApαl-Xliol treated pRS2. The resulting plasmid was named pl9. FinaUy, the Apάl-Avrll fragment of pi 9, theJvrll-Sα/GI fragment of p9 and the SαlGl-Bαm l fragment of pi 6 were Ugated together into a previously constructed Sindbis replicon vector expressing the GFP reporter (see Dubensky et al., J. Virol. 70:508-519, 1996; Polo et al., 1999, ibid; and U.S. Patent 5,843,723), that had been digested wtihApal-Bam l to remove the existing nonstructural protein genes. The resulting Sindbis vector construct, which contains sequences derived from the SINDCchiron virus strain and also encodes a GFP reporter, was designated SINCR-GFP (also known as DCSP6SINgfp). Preparation of replicon RNA from this reporter construct, as weU as Sindbis vector constructs expressing various other heterologous sequences (e.g., antigens, described in the specification and below) was performed by linearization of the DNA using Pmel, foUowed by in vitro transcription using bacteriophage SP6 polymerase as described previously (Polo et al., ibid; Dubensky et al., ibid).
Similarly, the same Sindbis sequences were used for assembly into an alphavirus-based Eukaryotic Layered Vector Initiation System (see U.S. 5,814,482 and 6,015,686), in which the transcription of self-ampUfying vector RNA takes place directly within ELVIS plasmid DNA- transfected eukaryotic ceUs via a eukaryotic promoter (e.g., RNA polymerase II promoter). An ELVIS plasmid DNA, which also expressed GFP reporter, was constructed by replacing Sindbis virus derived sequences in an existing ELVIS vector with the corresponding SINCR-GFP sequences from above. Starting with the previously described ELVIS vector pSINl .5 (Hariharan et al., J. Virol. 72:950-958, 1998), the plasmid backbone first was modified by substituting the plasmid backbone with that frompCMVLink (zur Megede et al., J. Virol. 74:2628-2635, 2000) using two Sαcl sites found in each plasmid, to generate the intermediate construct known as ELVIS1.5CB. Next, one of the two Sαcl sites of ELVIS1.5CB (located adjacent to the SIN 3'- end) was eliminated by partially digesting with Sαcl, blunt-ending using T4 DNA polymerase, and then Ugating into the modified site, aPmel linker 5'-GTTTAAAC-3 '. The correct plasmid without the targeted Sαcl site was designated ELVIS 1.5CBdlSac. This intermediate plasmid then was prepared for insertion of the new SIN nonstructural protein genes by digestion with Sαcl and Xhol. The corresponding nonstructural genes were obtained by PCR amplification from SINCR-GFP using the oUgonucleotide primers
5'CCTATGAGCTCGTTTAGTGAACCGTATTGACGGCGTAGTACACAC (SEQ ID NO:61) and 5'CCTATCTCGAGGGTGGTGTTGTAGTATTAGTC (SEQ ID NO:62), foUowed by digestion with Sαcl and Xhol, and ligation, to produce the intermediate construct SINCP-Not. Finally, one of the two Nσtl sites present in this construct was eliminated by partial digest and Klenow fiU-in, to leave only one Notl site in the polylinker. This newly constructed ELVIS vector was designated SINCP (or pSINCP).
Insertion of heterologous sequences (e.g., antigen-encoding genes) into the SINCR or SINCP alphavirus vectors is performed primarily by digestion with-¥7zoI/iVotI oτ XhollXbal, foUowed by Ugation with a desired DNA fragment that also has XhόllNotl oxXhollXbal teπnini. Alternatively, these sites may be blunt-ended or other polylinker sites may be used (or other heterologous sequences may be replaced) to aUow cloning of a greater number of inserts. For example, the HIV-1 p55gag (SF2 strain) and gpl40 env (SF162 strain) encoding genes were inserted into these vectors. SpecificaUy, the codon-optimized HIV p55gagmod sequence (see commonly owned U.S. Patent AppUcation 09/475,515; zur Megede et al., ibid) was inserted by digesting the vectors with XhόllXbal and Ugating in the p55gagmod fragment obtained by digestion with SaϊllXbal. The resulting vectors were designated SINCR-p55gag and SINCP-p55gag. Similarly, codon-optimized HIV gpl40 sequences (described in Example 10 and Barnett et al., 2001. J. Virol. 75:5526-40), were inserted into both the SINCR and SINCP plasmids to generate the constructs SINCR-gpl40 and SINCP-gpl40. Formulation of ELVIS plasmid DNA (pSINCP) and RNA vector repUcons transcribed in vitro from the SINCR plasmids is performed as described elsewhere in the Examples.
Example 4 Immunization of Rhesus Macaques with Antigen with pCMV or pSINCP Plasmids using Microparticles or Submicron emulsions PLG polymer microparticles and MF59 submicron emulsions were formed as described above in previous Examples 1 and 2. Groups of microparticles and submicron emulsions were made in order to analyze the different effects of immunizing rhesus macaques with a plasmid vector construct, pCMV- gpl40 or pCMV-p55gag (see commonly owned U.S. Patent Application 09/475,515), on microparticles or in a submicron emulsion, as weU as comparing the effect of using an ELVIS plasmid, pSINCP-gpl40 or pSINCP-p55gag, constructed as described above. Six groups of animals were immunized with different formulations as follows:
Group 1 used pCMV- gpl40 and pCMV - p55 gag without microparticles or submicron emulsions.
Group 2 used pCMV- gpl40 and pCMV - p55 gag adsorbed on PLG/CTAB microparticles.
Group 3 used pCMV- gpl40 and pCMV - p55 gag adsorbed to anMF59-DOTAP submicron emulsion.
Group 4 used pSINCP- gpl40 and pSINCP - p55 gag without microparticles or submicron emulsions.
Group 5 used pSINCP- gpl40 and pSINCP - p55 gag adsorbed on PLG/CTAB microparticles.
Group 6, a control, used no antigen, no microparticles, and no submicron emulsions.
For each group of animals, 5 rhesus macaques (only 4 for group 6) were immunized with sufficient quantities of material such that the dosage of vector with gpl40 DNA was 1.0 mg each, and vector containing p55 gag DNA was 0.5 mg each, except for the control which had none. The animals were immunized a second time four weeks after the first immunization, and a third time 14 weeks after the first irnmunization. Serum was analyzed at weeks 2 (2wpl), 6 (2wp2), 11-12 weeks (7wp2), and 16 (2wp3). The route of irnmunization was IM TA. FoUowing immunizations, plasma anti-p55gag and anti-gpl40 IgG titers were measured, the results of which appear below in Tables 2 and 3 as geometric mean titers.
Table 2
Figure imgf000061_0001
Table 3
Figure imgf000061_0002
The same group of animals as was used for the preceding animals were analyzed for induction of a CTL response. The effector to target (E:T) ratios ranged from approximately 4: 1 to 100:1. The results of the CTL assay appear below in Tables 4 and 5. A "responder" is a rhesus macaque which showed 10% or more specific lysis of the target at two or more consecutive E:T ratios. Table 4
Figure imgf000062_0001
Table 5
Figure imgf000062_0002
The same animals were also analyzed for lymphoproliferation. This assay measures specific proliferation of T ceUs in vitro in response to restimulation with antigen. Rhesus macaque peripheral blood mononuclear ceUs (PBMC) were purified fromheparinized whole blood by centrifugation on FicoU-Hypaque gradients. PBMC were cultured at the number of 2 x 105 per weU in flat bottom microtiter plates in the presence or absence of 3 rώcrograms/ l of purified recombinant p55gag protein. Six repUcate cultures per condition were initiated. After 4 days of culture tritiated thymidine ([3H]TdR) was added (1 microcurie per weU). Cultures were continued overnight and harvested the foUowing day. Cells were deposited onto glass microfiber filter sheets. Filter sheets were exposed to scintiUation fluid and counted in Uquid scintillation counter. For each condition [3H]TdR incorporation, measured as the mean counts per min (cpm) for the 6 repUcates was calculated. The results appear below in Table 6. Geometric Mean Stimulation Index (GMSI) is calculated as counts per minute (cpm) of p55gag stimulated ceUs divided by cpm of unstimulated ceUs, thus, the larger the GMSI, the more positive the result.
Table 6
Figure imgf000063_0001
The same animals were also analyzed for induction of intraceUular cytokine production. This assay measures specific production of cytokines by T cells in vitro in response to brief restimulation with antigen. Rhesus macaque peripheral blood mononuclear ceUs (PBMC) were purified fromheparizined whole blood by centrifugation on FicoU-Hypaque gradients. Aliquots of 1 x 106 PBMC were stimulated with a pool of synthetic overlapping peptides that span the gag (or env) protein sequence in the presence of a co-stimulatory anti-CD28 monoclonal antibody. Brefeldin A was added to allow the accumulation of newly synthesized cytokines within cells. After overnight incubation PBMC were stained with commercially available, fluorescently labeled monoclonal antibodies for the presence of intraceUular interferon-γ (IFN- γ) and tumoi necrosis factor-α (TNF-α) and for ceU surface CD4 and CD8 markers. Stained ceU samples were analyzed on a flow cytometer and data were acquired for approximately 50,000 - 100,000 PBMC. The frequency of cytokine-positive ceUs was determined for each sample using commerciaUy available software. The results for gpl40 and p55gag are shown in Tables 7 and 8, respectively. The Tables show the number of responding animals, where a responder is defined as an animal scoring greater than 100 CD4 ceUs per 100,000 expressing TNF-α and IFN- γ as measured by intracellular staining. Table 7
Figure imgf000064_0001
Example 5 RNA Vector Constructs RNA vector constructs (e.g., repUcons) may be adsorbed to microparticles for delivery of heterologous nucleic acid sequences to the ceUs of animals. The RNA vector construct generaUy comprises a viral RNA which has had a region of the genomic RNA (e.g., structural protein gene) replaced with the selected heterologous sequence, derived from the DNA coding sequence for the gene-product of interest. Representative examples of RNA vector constructs include, but are not Umited to, alphavirus RNA vectors (see for example, US Patent 5843723, PCT pubUcation WO 99/18226, and Polo et al., 1999, PNAS 96:4598-4603), picornavirus RNA vectors (see for example, US Patent 6156538, and Vignuzzi et al., 2001, J Gen Virol. 82:1737-47), flavivirus RNA vectors (see for example, Vamavski et al. 1999, Virology 255:366-75), and rabivirus RNA vectors (see for example, Pugachev et al., 2000, J Virol. 74: 10811-5).RNA vector constructs for use in the present invention generaUy may be obtained from plasmid cDNA constructs as a source of starting material, by the standard process of in vitro transcription (see references above). Similarly to plasmid DNA, these RNA vector constructs then may be adsorbed to microparticles of the invention as described elsewhere in the examples . For example, the RNA vector constructs are adsorbed onto the microparticles by incubating 100 mg of cationic microparticles in a 1 mg/ml solution of DNA at 4°C for 6 hr. The microparticles are then separated by centrifugation, the peUet washed with TE buffer, and the microparticles freeze-dried. Reconstitution and deUvery of the PLG-formulated RNA vector constructs is sirnilar to that described for DNA, using for example at least 1 ug, 10 ug, lOOug, or 1000 ug of formulated RNA vector construct for deUvery.
Example 6 Adjuvants in Mice An experiment with mice was performed to analyze the effect of the adjuvant aluminum phosphate (alum) in mice. Polymer microparticles were prepared as above described with or without pCMV-p55gag adsorbed thereon. 10 micrograms of DNA, whether naked or adsorbed to the PLG microparticles was injected in groups of 6 CB6 FI mice on weeks 0 and 6, without or without alum The results, as geometric mean titers of antibody, are shown below in Table 9.
Table 9
Figure imgf000065_0001
Example 7 Electroporation with Microparticles and ELVIS Vectors and RNA Vector Constructs
Electroporation may be used in combination with polymer microp rticles or submicron emulsion microparticles made with any of the nucleic acids described above, such as plasmid DNA^ ELVIS vectors, and RNA vector constructs.
Example 8 Induction of Immune Response in Rhesus with Prime and Boost Immunizations An experiment was performed to determine the effect of priming with DNA and boosting with protein adsorbed to PLG microparticles. Particularly, PLG-SDS microparticles were prepared as 'described above and in commonly owned International patent appUcation PCT/US99/17308, and purified recombinant p55gag protein was adsorbed thereto. A group of animals was immunized with 1 mg pCMV-p55gag. The animals were immunized again at 4 weeks, then again at 8 weeks. The animals were boosted with p55gag protein adsorbed to PLG microparticles at 41 weeks. The results are shown in Table 10 below, which shows antibody titer for responders, induction of helper T ceU lymphoproUferation (mean stimulation index of responders), and induction of CTL (number of responders, based on greater than 10% lysis at two or more consecutive E:T ratios). Results were measured at 14 weeks post 3rd prime for the prime columns, and 2 weeks post boost for the boost columns. Numbers in parentheses indicate the number of responders out of a total of 4 animals.
Table 10
Figure imgf000066_0001
Examnle 9 Induction of Neutralizing Antibodies The sera from the rhesus macaques in Example 5 above were tested for inhibition of two different HIV-1 stiains (SF2 and SF162) using PMBC-grown virus stocks and a CCR5+/CXCR4+/CD4+ T ceU line based assay. Sera were used at a dflution of 1 :20. Inhibitory activity was measured and expressed as a percentage inhibition. The inhibitory activity of the animals' sera prior to immunizations were subtracted from the results for each animal. The results for each of the five animals in each group are shown in Table 11 below.
Table 11
Figure imgf000067_0001
Example 10 Preparation of Plasmids Plasmids encoding HIV-1 p55gag and gpl40env driven by the human cytomegalovirus (CMV) promoter were grown in Escherichia coli strain DH5α, purified using a Qiagen Endofree Plasmid Giga kit (Qiagen, Inc.), and resuspended in 0.9% sodium chloride (Abbott Laboratories, North Chicago, IU.). The pCMV vector used contains the immediate-early enhancer/promoter of CMV and a bovine growth hormone terminator and is described in detail elsewhere (Chapman, B. S., et al. 1991. 'Εffect of intron Afromhuman cytomegalovirus (Towne) immediate-early gene on heterologous expression in mammaUan cells." Nucleic Acids Res. 19:3979-86). The HIV gag plasmid DNA vaccine (pCMVgag) contains a synthetically constructed p55gag gene, with codons reflecting mammaUan usage, derived from the HTV-1 SF2 strain as previously described (zur Megede, J., et al. 2000. "Increased expression and immunogenicity of sequence-modified human immunodeficiency virus type 1 gag gene." J Virol. 74:2628-35). The HIV env plasmid DNA vaccine (pCMVgpl40) consisted of a human tissue plasminogen activator (tPA) signal sequence and the gpl40 from HIV-1 SF162 strain, codon optimized for high level expression in mammaUan cells (Barnett, S. W., et. 2001. "The abiUty of an oUgomeric human immunodeficiency virus type 1 (HIV-1) envelope antigen to elicit neutralizing antibodies against primary HIV-1 isolates is improved foUowing partial deletion of the second hypervariable region." J Virol. 75:5526-40). The SINCP plasmid vector with either HIV-1 p55gag or gpl40env has been described in Example 3 above.
Example 11 Preparation of Proteins The protein and cDNA sequences for the gpl60env.SF162 have been pubUshed in Cheng- Mayer, C, M. Quiroga, J. W. Tung, D. Dina, and J. A. Levy. 1990. 'Niral determinants of human immunodeficiency virus type 1 T-cell or macrophage tropism, cytopathogenicity, and CD4 antigen modulation." J Virol. 64:4390-8. These sequences can be found under Genbank accession number M65024. Recombinant HTV-1 gl40.SF162(dV2) protein was expressed in Chinese hamster ovary cells and purified as previously described (Barnett, S. W., ET. 2001. J Virol. 75:5526-40). Recombinant HTV-1. SF2 p55 gag protein was expressed in yeast and purified by cation exchange chromatography (Chiron Corporation, EmeryviUe, CA). The p55gag cDNA sequence from the SF2 strain of HTV-1 (Genbank accession number K02007) was cloned into aubiquitin expression vector, resulting in the addition of glycine and arginine to the N-terminus of the wild-type sequence. The recombinant p55gag protein was extracted from the yeast ceU peUet using 50 mM phosphate, 6M urea, pH7.9, followed by S-fractogel (cationic) ion exchange chromatography. Elution of the p55gag was obtained with a linear NaCl gradient (peak at 0.4m NaCl). The estimated purity was 90% by SDS-PAGE.
Example 12 DNA-adsorbed polvflactide-co-glvcoUde) PLG microparticles PLG polymer (RG505) was obtained from Boehringer Ingelheim Cationic microparticles were prepared using a modified solvent evaporation process. Briefly, the microparticles were prepared by emulsifying 10 ml of a 5% (wt/vol) polymer solution in methylene chloride withl ml of phosphate-buffered saline (PBS) at high speed using an IKA homogenizer. The primary emulsion was then added to 50 ml of distilled water containing cetyltrimethylammonium bromide (CTAB) (0.5% wt/vol), resulting in the formation of a water-in-oil-in- water emulsion, which was stirred at 6,000 rpmforl2 h at room temperature, aUowing the methylene chloride to evaporate. The resulting microparticles were washed twice in distilled water by centrifugation at 10,000g and freeze-dried. Plasmid DNA from Example 11 was adsorbed onto the microparticles by incubating 100 mg of cationic microparticles 5ml of a 200 microgram/ml solution of DNA at 4°C for 6h The microparticles were then separated by centrifugation, the peUet was washed with TE (Tris-EDTA) buffer, and the microparticles were freeze-dried.
Example 13 Protein-adsorbed PLG microparticles Blank microparticles were prepared by a solvent evaporation technique. Briefly, microparticles were prepared by homogenizing 10ml 6% w/v polymer solution in methylene chloride, with 40 ml of distilled water containing SDS (1 % w/v) at high speed using a 10mm probe. This resulted in an oil in water emulsion, which was stirred at 1000 rpm for 12 hours at room temperature, and the methylene chloride was attowed to evaporate. The resulting microparticles were filtered through a 38ummesh, washed 3 times in distilled water, and freeze- dried. The size distribution of the microparticles was determined using a particles size analyzer (Master sizer, Malvem Instruments, UK).
50mg lyophflized SDS blank particles were incubated with 0.5mg of p55 gag protein from Example 12 in 10ml 25mM Borate buffer pH9 with 6M Urea Particles were left on a lab rocker, (AUquot mixer, Miles labs) at room temperature for 5 hours. The microparticles were separated from the incubation medium by centrifugation, and the SDS peUet was washed once with Borate buffer with 6M Urea then three times with distiUed water, and lyophflized.
The loading level of protein adsorbed to microparticles was deteraiined by dissolving lOmg of the microparticles in 2ml of 5% SDS-0.2M sodium hydroxide solution at room temperature. Protein concentration was measured by BCA protein assay (Pierce, Rockford, Illinois). The Zeta potential for both blank and adsorbed microparticles was measured using a Malvem Zeta analyzer (Malvem Instruments, UK).
Example 14 Preparation of Protein with MF59 Adjuvant Recombinant HIV-1 gl40.SF162(dV2) protein from Example 12 was combined with MF59 adjuvant as previously described (Barnett, S. W., et. 2001. J Virol. 75:5526-40).
Example 15 Immunization Male and female rhesus macaques were housed at Southern Research Institute (Frederick, MD).
Plasmid DNA immunization was performed at weeks 0, 4, and 14. Rhesus were given intramuscular injections of 0.5 mg of pCMVgag from Example 11 (in saline or formulated with PLG/CTAB microparticles as described in Example 13 or formulated with MF59/DOTAP as described in Example 2) and 1.0 mg of pCMVenv from Example 11 (in saline or formulated with PLG/CTAB microparticles as described in Example 13) at 4 separate sites per animal (0.25 mg pCMVgag in upper right arm and upper right leg; 0.5 mg pCMVenv in upper left arm and upper left leg). Alternatively, rhesus were given intramuscular injections of 0.5 mg of pSINCPgag from Example 11 (in saline or formulated with PLG/CTAB microparticles as described in Example 13) and 1.0 mg of pSINCP env from Example 11 (in saline or formulated with PLG/CTAB microparticles as described in Example 13) at 4 separate sites per animal.
Rhesus were boosted by intramuscular injection of 0.2 mg recombinant p55gag protein/PLG microparticles from Example 14 at week 29 and with 0J mg recombinant gpl40env(dV2) protein/MF59 adjuvant from Example 15 at week 38.
Example 16 Antibody responses At various times following immunization, heparinized blood was coUected from anesthetized animals and plasma was recovered by centrifugation. Anti-HIV Gag and Env antibodies were measured by enzyme-linked immunosorbent assay (ELISA) as foUows. WeUs of microtiter plates were coated with recombinant HTV-1. SF2 p55gag protein or recombinant HIV-1. SFl 62 gpl40env protein at 5 rnicrogram/ml in PBS, 50 microUters per weU, and incubated at 4°C overnight. The plates were washed six times with wash buffer (PBS, 0.3% Tween 20) and blocked at 37°C for 1 h with 200 microUters per weU of blocking buffer (PBS, 0.3% Tween 20, 5% goat serum). Test samples were diluted 1 :25 and then seriaUy diluted threefold in blocking buffer. The block solution was aspirated, and then the plates were incubated at room temperature for 1 h with 70 microUters per weU of each plasma dflution. After being washed six times, the plates were incubated for 1 h at 37°C with horseradish peroxidase-conjugated anti-IgG (1:8,000 dilution). FoUowing six washes, the plates were developed with TMB substrate for 15 minutes. The reaction was stopped with 2N HC1 and the optical densities (OD) measured at a wavelength of 450 nm The titer was calculated to be the reciprocal of the dilution at which an OD450 nm of 0.5 was achieved. Table 12. Rhesus Anti-gag plasma antibody titers
Figure imgf000072_0001
) Relative to plasmid immunizations done at weeks 0, 4, and 14 p) Geometric mean for the group
(3) Group arithmetic means and standard errors calculated from log-transformed titers. LL = antilog (arithmetic mean - standard error). UL = antilog (arithmetic mean + standard error)
(4) Recombinant p55gag protein adsorbed to anionic PLG microparticles administered at week 29 Table 13. Rhesus Anti-env plasma antibody titers
Figure imgf000073_0001
(1) Relative to plasmid iinmunizations done at weeks 0, 4, and 14
(2) Geometric mean for the group p) Group arithmetic means and standard errors calculated from log-transformed titers. LL = antilog (arithmetic mean - standard error). UL = antilog (arithmetic mean + standard error)
C4> Recombinant ohgomeric gpl40env(ΔV2) protein MF59 adjuvant administered at week 38 Example 17 Cvtolvtic T Lymphocyte (CTL) responses
A pool of 51 synthetic peptides 20 amino acids (aa) long, overlapping by 10 aa, and spanning p55 gag, and a pool of 66 synthetic peptides 20 aa long, overlapping by 10 aa, and spanning gpl40 were prepared. Rhesus macaque peripheral blood mononuclear ceUs (PBMC) were separated from heparinized blood by centrifugation on FicoU-Paque (Pharmacia Biotech, Piscataway, NJ.) gradients. PBMC were cultured for 8 days in 24-weU plates at 3 x 106 per weU in 1.5 ml of AIM- V/RPMI 1640 (50:50) culture medium (Gibco-BRL,Grand Island, N.Y.) supplemented with 10% fetal bovine serum Gag-specific CTL were stimulated by the addition of the gag peptide pool and env-specific CTL were stimulated by the addition of the env peptide pool. Cultures were supplemented with recombinant human mterleukin-7 (IL-7; 15 ng/ml; R&D Systems, Minneapolis, Minn.). Human recombinant IL-2 (20 IU/ml; Proleul n;Chiron) was added on days 1, 3, and 6. Stable rhesus B-lymphoblastoid ceU lines (B-LCL) were derived by exposing PBMC to herpesvirus papio-containing culture supernatant from the S594 ceU line (Falk, L., et al. 1976. Properties of a baboon lymphotropic herpesvirus related to Epstein-Barr virus. Int J Cancer. 18:798-807. Rabin, H, et al. 1976. Virological studies of baboon (Papio hamadryas) lymphoma: isolation and characterization of foamyviruses. J Med Primatol. 5:13-22.) in the presence of 0.5 microgram/ml cyclosporin A (Sigma, St. Louis, MO). Autologous B-LCL were infected with recombinant vaccinia virus (rVV) encoding HIV-1. SF2 gag-pol (rWgag-pol) or HIV-1. SFl 62 gpl60env (rVVgpl60env) (PFU: cell ratio of 10) and concurrently labeled withNa[51Cr]204 (NEN, Boston, MA) at 25 microcurie per 1 x 10δ B-LCL. After overnight culture at 37°C, rVV-infected, 51Cr- labeled B-LCL were washed and then added (2,500 per round-bottomed well) to duplicate weUs containing threefold serial dilutions of cultured PBMC. Then 105 unlabeled, uninfected B-LCL were added per well to inhibit nonspecific cytolysis. After 4h incubation at 37°C, 50 microUters of culture supernatants were harvested and added to LumaPlates (Packard, Meriden, CT), and radioactivity was counted (counts per minute (cpm)) with a Microbeta 1450 Uquid scintillation counter (WaUac, Gaithersburg, MD). 51Cr released from lysed targets was normalized by using the formula: % Specific 51Cr Release = 100% x (mean experimental cpm - SR)/(MR - SR), where SR = mean cpm from targets alone and MR = mean cpm from targets exposed to Triton X-l 00. An animal was dete iined to have a positive, p55gag-specific response if at two consecutive dilutions of the gag peptide pool-stimulated PBMC the lysis of rWgag-pol-infected B-LCL exceeded lysis of rVVgplβOenv-infected B-LCL by at least 10% and if at two consecutive dilutions of cultured PBMC the lysis of rVVgag-pol-infected B-LCL by the gag peptide pool-stimulated PBMC exceed lysis of rVVgag-pol-infected B-LCL by env peptide pool-stimulated B-LCL by at least 10%. An animal was deterrnined to have a positive, gpl60-specific response if at two consecutive dilutions of the env peptide pool-stimulated PBMC the lysis of rVVgpl60env-infected B-LCL exceeded lysis of rVVgag-pol-infected B-LCL by at least 10% and if at two consecutive dilutions of cultured PBMC the lysis of rVVgplόOenv-infected B-LCL by the env peptide pool-stimulated PBMC exceed lysis of rVVgpl60env-infected B-LCL by gag peptide pool-stimulated B-LCL by at least 10%.
Table 14. Gag-specific CTL
Figure imgf000076_0001
Table 15. Env-specific CTL
Figure imgf000076_0002
Example 18 Lvmphoproliferation assay (LPA) 2 x 105 rhesus PBMC per weU were cultured in the presence or absence of recombinant p55gag protein or the pool of synthetic env peptides. Six repUcate wells were estabUshed for each culture condition. After 4 days of incubation cultures were pulsed overnight with 1 microcurie/weU of [3H]TdR Incorporation of [3H]TdR into ceUs was determined by liquid scintiUation counting (BetaPlate, WaUac, Gaithersburg, MD). Stimulation Index (SI) was calculated as SI = mean cpm (gag or env stimulation)/mean cpm (^stimulated). Table 16. Rhesus Anti-gag lymphoproUferation stimulation indices
Figure imgf000077_0001
(1 Relative to plasmid immunizations done at weeks 0, 4, and 14 Geometric mean for the group
(3) Group arithmetic means and standard errors calculated from log-transformed titers. LL = antilog (arithmetic mean - standard error). UL = antilog (arithmetic mean + standard error)
^ Recombinant p55gag protein adsorbed to anionic PLG microparticles administered at week 29 Table 17. Rhesus anti-env lymphoproUferation stimulation indices
Figure imgf000078_0001
(1) Relative to plasmid immunizations done at weeks 0, 4, and 14
(2) Geometric mean for the group
(3) Group arithmetic means and standard errors calculated from log-transformed titers. LL = antilog (arithmetic mean - standard error). UL = antilog (arithmetic mean + standard error) w Recombinant oUgomeric gpl40env(ΔV2) protein MF59 adjuvant administered at week 38 (5) blank values: assay not performed Example 19 IntraceUular cytokine immunofluorescence and flow cytometry
Rhesus PBMC (1 x 106 per weU) were cultured overnight in the presence of Brefeldin A (Pharmingen, San Diego, CA) and anti-CD28 monoclonal antibody (mAb) (Pharmingen) and in the presence or absence of the gag or env peptide pools. DupUcate weUs were prepared for each condition of stimulation. The next day ceUs were stained with peridinin chlorophyU protein (PerCP)-conjugated anti-CD8 mAb and aUophycocyanin (APC)-conjugated anti-CD4 mAb (Becton Dickinson, San Jose, CA), fixed and permeabflized (Cytofix/Cytoperm, Pharmingen), and stained with fluorescein isothiocyanate (FITC)-conjugated anti-tumor necrosis factor-α (TNF-α) mAb and phycoeiythrin (PE)-conjugated anti-mterferon-γ (IFN-γ) mAb (Pharmingen). Stained ceU samples were analyzed using a FACSCaUbur™ flow cytometer and CeUQuest™ software (Becton Dickinson). The fraction of cells positively stained for IFN-γ and TNF-α was calculated for the CD4+8- and CD8+4- T ceU subsets. The number of gag- or env-specific ceUs was calculated by subtraction of the average IFN-γ/TNF-α fraction found in the unstimulated control wells from the average IFN-γ/TNF-α fraction found in the gag- or env-stimulated weUs.
For a given T cell subset (CD4+8- or CD8+4-) and antigen (gag or env) a response was designated as positive if the fraction of antigen-specific cells was at least 0.1%.
Table 18. Gag-specific IFNγ+/TNFα+ CD4+ T ceUs
Figure imgf000080_0002
^Positive: Frequency > 0.1%
Table 19. Env-specific IFNγ+/TNFα+ CD4+ T ceUs
Figure imgf000080_0003
^Positive: Frequency > OJ %
Table 20. Gag-specific IFNγ+/TNFα÷ CD8+ T ceUs
Induction of p55gag-specific IFNγ+/TNFα+ CD8+ T ceUs by vaccines
Figure imgf000080_0001
Figure imgf000080_0004
*Positive: Frequency > 0.1% Table 21. Env-snecific IFNγ+/TNFα+ CD8+ T ceUs
Figure imgf000081_0001
^Positive: Frequency > 0.1%
Although preferred embodiments of the subject invention have been described in some detail, it is understood that obvious variations can be made without departing from the spirit and the scope of the invention as defined by the appended claims.
REFERENCES:
Ref. 1 - International patent appUcation W099/24578.
Ref. 2 -International patent appUcation W099/36544.
Ref. 3 - International patent appUcation WO99/57280.
Ref. 4 - International patent appUcation WO00/22430.
Ref. 5 - TetteUn et al., (2000) Science 287:1809-1815.
Ref. 6 - International patent appUcation W096/29412.
Ref. 7 - Pizza el al. (2000) Science 287:1816-1820.
Ref. 8 - International patent appUcation PCT/IB01/00166.
Ref. 9 -Bjune et al. (1991) Lancet 338(8775): 1093-1096.
Ref. 10 -Fukasawa et al. (1990) Vaccine 17:2951-2958.
Ref. 11 -Rosenqvist et al. (1998) Dev. Biol. Stand. 92:323-333.
Ref. 12- Costantino et al. (1992) Vaccine 10:691-698.
Ref 13- Costantino et al. (1999) Vaccine 17:1251-1263.
Ref. 14- Watson (2000) Padiatr Infect Dis J 19:331-332.
Ref. 15- Rubin (2000) Pediatr Clin North Am 47:269-285, v.
Ref. 16- Jedrzejas (2001) MicrobiolMol Biol Rev 65:187-207.
Ref. 17 -International patent appUcation filed on 3rd July 2001 claiming priority from
GB-0016363.4].
Ref 18 -Kalman et al. ( 1999) Nature Genetics 21 :385-389.
Ref 19- Read et al. (2000) Nucleic Acids Res 28:1397-406.
Ref. 20- Shirai et al. (2000) J. Infect. Dis. 181(Suppl 3):S524-S527.
Ref. 21 -International patent appUcation W099/27105.
Ref. 22 -International patent appUcation WO00/27994.
Ref. 23 -International patent appUcation WO00/37494.
Ref. 24 -International patent appUcation W099/28475.
Ref. 25 -BeU (2000) Pediatr Infect Dis J 19: 1187-1188.
Ref. 26 -Iwarson (1995) APMIS 103:321-326.
Ref. 27 -GerUch et al. (1990) Vaccine 8 Suppl:S63-68 & 79-80.
Ref. 28 -Hsu et al. (1999) Clin Liver Dis 3:901-915. Re 29 -Gustafsson et al. (1996) N. Engl J. Med. 334:349-355.
Ref. 30 -Rappuoli et al. (1991) TIBTECH 9:232-238.
Ref. 31 -Vaccines (1988) eds. Plotkin & Mortimer. ISBN 0-7216-1946-0.
Ref. 32 -Del Guidice et al. (1998) Molecular Aspects of Medicine 19:1-70.
Ref. 33 -International patent appUcation W093/18150.
Ref. 34 -International patent appUcation W099/53310.
Ref. 35 -International patent appUcation WO98/04702.
Ref. 36 -Ross et al. (2001) Vaccine 19:4135-4142.
Ref. 37 -Sutter et al. (2000) Pediatr Clin North Am 47:287-308.
Ref. 38 -Zimmerman & Spann (1999) Am Fam Physician 59:113-118, 125-126.
Ref. 39 -Dreesen (1997) Vaccine 15 Suppl:S2-6.
Ref. 40 -MMWRMorb Mortal WldyRep 1998 Jan 16;47(1):12, 19.
Ref. 41 -McMichael (2000) Vaccine 19 Suppl S101-107.
Ref. 42 -Schuchat (1999) Lancet 353(9146):51-6.
Ref. 43 -GB patent appUcations 0026333.5, 0028727.6 & 0105640.7.
Ref. 44 -Dale (1999) Infect Dis Clin North Am 13:227-43, viii.
Ref 45 -Ferretti et al. (2001) PNAS USA 98:4658-4663.
Ref. 46 -Kuroda et al. (2001) Lancet 357(9264): 1225-1240; see also pages 1218-1219.
Ref. 47 -Ramsay et al. (2001) Lancet 357(9251): 195-196.
Ref 48- Lindberg (1999) Vaccine 17 Suppl 2:S28-36.
Ref. 49 -Buttery & Moxon (2000) JR Coll Physicians London 34: 163-168.
Ref. 50 -Ahmad & Chapnick (1999) Infect Dis Clin North Am 13:113-133, m
Ref. 51 -Goldblatt (1998) J. Med. Microbiol 47:563-567.
Ref. 52 -European patent 0477 508.
Ref. 53 -US Patent No. 5,306,492.
Ref. 54 -International patent appUcation W098/42721.
Ref. 55 -Conjugate Vaccines (eds. Cruse et al.) ISBN 3805549326, particularly vol. 10:48-114.
Ref. 56- Hermanson (1996) Bioconjugate Techniques ISBN: 0123423368 & 012342335X.
Ref. 57 -European patent application 0372501.
Ref. 58 -European patent application 0378881. Ref. 59 -European patent appUcation 0427347.
Ref. 60 -International patent appUcation W093/17712.
Ref. 61 -International patent appUcation W098/58668.
Ref. 62 -European patent application 0471177.
Ref 63 -International patent appUcation WOOO/56360.
Ref. 64 -international patent appUcation WO00/61761.

Claims

CLAIMS:
1. A method of raising an immune response in a host animal comprising: administering to the animal a vector construct comprising a heterologous nucleic acid sequence encoding a first antigen in an amount effective to elicit an immunological response, wherein the vector construct is adsorbed onto microparticles comprising (i) a polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride and a polycyanoacrylate and (U) a detergent; and subsequently boosting the immunological response by administering a second antigen to the animal, wherein the first antigen and the second antigen can be the same or different.
2. The method of claim 1, wherein the first antigen and the second antigen are the same.
3. The method of claim 1, wherein the vector construct is selected from a plasmid DNA and an RNA vector construct.
4. The method of claim 3, wherein the plasmid DNA is an ELVIS vector.
5. The method of claim 4, wherein the ELVIS vector comprises a cDNA complement of an RNA vector construct derived from a member selected from the group consisting of alphavirus, picomavirus, togavirus, flavivirus, coronavirus, paramyxovirus, and yeUow fever virus.
6. The method of claim 5, wherein the alphavirus is selected from the group consisting of Sindbis virus, SemUki Forest virus, Venezuelan equine encephalitis virus, or Ross River virus.
7. The method of claim 3, wherein the plasmid DNA comprises a CMV promoter/enhancer.
8. The method of claim 1, wherein the first and second antigens are selected from the group consisting of HTV antigens, hepatitis C virus antigens, and influenza A virus antigens.
9. The method of claim 1, wherein the first and second antigens comprise antigens selected from the group consisting of HIV antigens gpl20, gpl40, gpl60, p24gag and p55gag.
10. The method of claim 1, wherein the first and second antigens comprise HTV p55gag.
11. The method of claim 1, wherein the first and second antigens comprise HIV gpl40.
12. The method of claim 1, wherein the second antigen is adsorbed to microparticles comprising (i) a polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactones a polyorthoester, a polyanhydride, and a polycyanoacrylate and (n) a detergent.
13. The method of claim 1, wherein the second antigen is coadministered with an adjuvant.
14. The method of claim 13, wherein the adjuvant is MF59.
15. The method of claim 1, wherein the polymer comprises a poly(α-hydroxy acid) selected from the group consisting of poly(L-lactide), poly(D.L-lactide) and poly(D,L-lactide-co-glycoUde) and wherein the detergent comprises a cationic detergent selected from CTAB, benzalkonium chloride, DDA and DOTAP.
16. The method of claim 1, wherein the vector construct is administered two or more times before the second antigen is administered.
17. The method of claim 16, wherein the second antigen is also adininistered two or more times.
18. The method of claim 17, wherein the vector construct is administered (a) at atime of initial adrjniώstration, (b) at a time period ranging 1 -8 weeks from the initial aάlministration, and (c) at a time period ranging 4-32 weeks from the initial administration, and wherein the second antigen is administered (a) at atime period ranging from 8-50 weeks from the initial administration and (b) at atime period ranging from 8-100 weeks from the initial aάhainistration.
19. The method of claim 1, wherein the animal is a inammal selected from rhesus macaque and a human.
20. The method of claim 1, wherein the vector construct and the second antigen are administered subcutaneously, intiaperitoneaUy, intradermaUy, intravenously or intramuscularly.
21. The method of claim 20, wherein the vector construct and the second antigen are administered intramuscularly.
22. The method of claim 1, wherein the vector construct is coadministered with an adjuvant.
23. The method of claim 1, wherein said immune response comprises a Thl immune response.
24. The method of claim 1, wherein said immune response comprises a CTL immune response.
25. The method of claim 1, wherein said immune response is raised against a viral, bacterial, or parasitic infection.
26. A microparticle with an adsorbent surface to which a first biologicaUy active macromolecule has been adsorbed comprising: a microparticle selected from the group consisting of (a) a polymer microparticle comprising: (i) a polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate; and a detergent; and (b) a submicron emulsion comprising: (i) a metabolizable oil; and (U) one or more emmsifying agents; and the first biologicaUy active macromolecule, wherein the first biologicaUy active macromolecule is a nucleic acid molecule comprising at least one vector construct selected from the group consisting of an ELVIS vector and an RNA vector construct.
27. The microparticle of claim 26, wherein said submicron emulsion is selected as said microparticle.
28. The microparticle of claim 26, wherein said polymer microparticle is selected as said microparticle.
29. The microparticle of claim 28, wherein the polymer microparticle comprises a poly(α- hydroxy acid) selected from the group consisting of poly(L-lactide), poly(D,L-lactide) and poly(D,L-lactide-co-glycoUde).
30. The microparticle of claim 28, wherein the polymer comprises poly(D,L-lactide-co- glycoUde).
31. The microparticle of claim 28, further comprising a second biologicaUy active macromolecule entrapped wilhinthe microparticle, wherein the second biologicaUy active macromolecule is a member selected from the group consisting of apolynucleotide, a polynucleoside, a pharmaceutical, a polypeptide, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, and an adjuvant.
32. The microparticle of claim 28, wherein said vector construct is an ELVIS vector.
33. The microparticle of claim 28, wherein said vector construct is an ELVIS vector comprising a cDNA complement of an RNA vector construct derived from a member selected fromthe group consisting of alphavirus, picomavirus, togavirus, flavivirus, coronavirus, paramyxovirus, and yeUow fever virus, and wherein said RNA vector construct further comprises a selected heterologous nucleotide sequence.
34. The microparticle of claim 33, wherein said ELVIS vector is derived from an alphavirus selected fromthe group consisting of Sindbis virus, SemUki Forest virus, Venezuelan equine encephalitis virus, or Ross River virus.
35. The microparticle of claim 28, wherein said vector construct is an RNA vector construct derived from a member selected fromthe group consisting of alphavirus, picomavirus, togavirus, flavivirus, coronavirus, paramyxovirus, and yeUow fever virus, and wherein said RNA vector construct comprises a selected heterologous nucleotide sequence.
36. The microparticle of claim 35, wherein said RNA vector construct is derived from an alphavirus selected fromthe group consisting of Sindbis virus, SemUki Forest virus, Venezuelan equine encephaUtis virus, or Ross River virus.
37. The microparticle of claim 32, wherein said vector construct comprises a heterologous nucleic acid sequence encoding a member selected from the group consisting of a pharmaceutical, a polypeptide, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, and an adjuvant.
38. The microparticle of claim 37, wherein said heterologous nucleic acid sequence encodes an antigen.
39. The microparticle of claim 38, wherein said antigen is a member selected from the group consisting of HIV gpl20, HTV gpl40, HTV p24gag, HTV p55gag, and Influenza A hemagglutinin antigen.
40. The microparticle of claim 32, wherein said vector construct is a vector selected from the group consisting of the ELVIS vectors pSINCP-gpl40 and pSINCP-p55gag.
41. The microparticle of claim 28, further comprising at least one second biologically active macromolecule adsorbed on the surface thereof, wherein the second biologically active macromolecule is at least one member selected fromthe group consisting of a polypeptide, a polynucleotide, a polynucleoside, an antigen, a pharmaceutical, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, and an adjuvant.
42. The microparticle of claim 41, wherein the second biologicaUy active macromolecule is an antigen.
43. The microparticle of claim 42, wherein the second biologicaUy active macromolecule is an antigen selected fromthe group consisting of HIV gpl20, HIV gpl40, HIV p24gag, HIV p55gag, and Influenza A hemagglutinin antigen.
44. The microparticle of claim 41, wherein the second biologicaUy active macromolecule is a polynucleotide which encodes HIV gpl40.
45. The microparticle of claim 41, wherein the second biologicaUy active macromolecule is an adjuvant.
46. The microparticle of claim 45, wherein the adjuvant is an alun±mm salt.
47. A microparticle composition comprising a microparticle of claim 26 and a pharmaceuticaUy acceptable excipient.
48. The microparticle composition of claim 47, further comprising an adjuvant.
49. The microparticle composition of claim 48, wherein the adjuvant is a member selected fro the group consisting of a CpG oUgonucleotide.
50. The microparticle composition of claim 48, wherein the adjuvant is an du-ninumsalt which is aluminum phosphate.
51. A method of producing a microparticle having an adsorbent surface to which a vector construct capable of expressing a selected nucleic acid sequence is adsorbed, said method comprising the steps of:
(a) emulsifying a mixture of a polymer solution and a detergent to form an emulsion, wherein the polymer solution comprises a polymer selected fromthe group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate, wherein the polymer is present at a concentration of about 1% to about 30% in an organic solvent, and wherein the detergent is present in the mixture at a weight to weight detergent to polymer ratio of from about 0.00001:1 to about 0.5:1;
(b) removing the organic solvent fromthe emulsion, to form said microparticle; and
(c) adsorbing the vector construct to the surface of the microparticle, wherein said vector construct selected fromthe group consisting of an ELVIS vector and an RNA vector construct.
52. The method of claim 51, wherein the vector construct is an ELVIS vector or an RNA vector construct, and comprises a heterologous nucleic acid sequence encoding a member selected from the group consisting of a pharmaceutical, a polypeptide, a hormone, an enzyme, a transcription or translation mediator, an intermediate in a metaboUc pathway, an immunomodulator, an antigen, and an adjuvant.
53. The method of claim 52, wherein the heterologous nucleic acid sequence encodes an antigen selected fromthe group consisting of HTV gpl20, HIV gpl40, HIV p24gag, HIV p55gag, and Influenza A hemagglutinin antigen.
54. The method of claim 53, wherein the antigen is HIV gpl40.
55. A microparticle made according to the method of claim 51.
56. A microparticle composition comprising the microparticle of claim 55 and a pharmaceutically acceptable excipient.
57. A method of inducing an immune response in a host animal comprising administering to said animal the microparticle composition of claim 47.
58. The method of claim 57 wherein said mammal is a human.
59. A method of immunizing a host animal against a viral, bacterial, or parasitic infection comprising administering to said animal the microparticle composition of any of claim 47.
60. The method of claim 59 wherein said mammal is a human.
61. A method of inducing a TM immune response in a host animal comprising administering to said animal the microparticle composition of claim 47.
62. The method of claim 61 wherein said mammal is a human.
63. A method of inducing a CTL immune response in a host animal comprising adrninistering to said animal the microparticle composition of claim 47.
64. The method of claim 63 wherein said mammal is a human.
65. A method of deUvering a therapeuticaUy effective amount of a macromolecule to a host animal comprising the step of aclministering to the vertebrate subject a microparticle composition of claim 47.
66. The method of claim 65 wherein said mammal is a human.
67. A method of treating a host animal having a viraL bacterial, or parasitic infection comprising administering to said animal the microparticle composition of claim 47 in an amount effective to reduce the level of infection thereof.
68. The method of claim 67 wherein said mammal is a human.
69. Use of a microparticle composition of claim 47 for treatment of a disease.
70. Use of a microparticle composition of claim 47 for a vaccine.
71. Use of a microparticle composition of claim 47 for raising an immune response.
72. The microparticle of claim 39, wherein said heterologous nucleic acid sequence encodes an HIV gag polypeptide and comprises a sequence having at least 90% identity to a sequence selected fromthe group consisting of nucleotides 844-903 of SEQ ID NOs:63, nucleotides 841- 900 of SEQ ID NO:64, nucleotides 1513-2547 of SEQ ID NO:65, nucleotides 1210-1353 of SEQ ID NO:66, nucleotides 1213-1353 of SEQ ID NO:67, and nucleotides 82-1512 of SEQ ID NO:68.
73. The microparticle of claim 39, wherein said heterologous nucleic acid sequence encodes an HIV envelope polypeptide and comprises a sequence having at least 90% identity to a sequence selected fromthe group consisting of nucleotides 844-903 of SEQ ID NOs:63, nucleotides 841- 900 of SEQ ID NO:64, nucleotides 1513-2547 of SEQ ID NO:65, nucleotides 1210-1353 of SEQ ID N0.66, nucleotides 1213-1353 of SEQ ID NO:67, and nucleotides 82-1512 of SEQ ID NO:68.
74. The method of claim 53, wherein said heterologous nucleic acid sequence encodes an HIV gag polypeptide and comprises a sequence having at least 90% identity to a sequence selected fromthe group consisting of nucleotides 844-903 of SEQ ID NOs:63, nucleotides 841-900 of SEQ ID NO:64, nucleotides 1513-2547 of SEQ ID NO:65, nucleotides 1210-1353 of SEQ ID NO:66, nucleotides 1213-1353 of SEQ ID NO:67, and nucleotides 82-1512 of SEQ ID NO:68.
75. The method of claim 53, wherein said heterologous nucleic acid sequence encodes an HIV envelope polypeptide and comprises a sequence having at least 90% identity to a sequence selected fromthe group consisting of nucleotides 844-903 of SEQ ID NOs:63, nucleotides 841- 900 of SEQ ID NO:64, nucleotides 1513-2547 of SEQ ID NO:65, nucleotides 1210-1353 of SEQ ID NO:66, nucleotides 1213-1353 of SEQ ID NO:67, and nucleotides 82-1512 of SEQ ID NO:68.
76. The microparticle of claim 27, wherein (a) the oil is a terpenoid and (b) the one or more emulsifying agents comprise one or more non-ionic detergents and one or more cationic detergents.
77. The microparticle of claim 76, wherein the oil is squalene and the one or more emulsifying agents comprise: a polyoxyethylene sorbitan fatty acid ester, a sorbitan fatty acid ester, and DOTAP.
PCT/US2001/030540 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids WO2002026209A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
BR0114305-0A BR0114305A (en) 2000-09-28 2001-09-28 Microparticles for release of heterologous nucleic acids
EP01975584A EP1322287B1 (en) 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids
CN018164595A CN1468089B (en) 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids
AU9489701A AU9489701A (en) 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids
DE60118228T DE60118228T2 (en) 2000-09-28 2001-09-28 MICROPARTICLES FOR THE ADMINISTRATION OF HETEROLOGIC NUCLEIC ACID
CA002421683A CA2421683C (en) 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids
JP2002530039A JP2004518631A (en) 2000-09-28 2001-09-28 Microparticles for delivery of heterologous nucleic acids
MXPA03002640A MXPA03002640A (en) 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids.
ZA2003/01744A ZA200301744B (en) 2000-09-28 2003-03-03 Microparticles for delivery of the heterologous nucleic acids

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US23610500P 2000-09-28 2000-09-28
US60/236,105 2000-09-28
US31590501P 2001-08-30 2001-08-30
US60/315,905 2001-08-30

Publications (2)

Publication Number Publication Date
WO2002026209A2 true WO2002026209A2 (en) 2002-04-04
WO2002026209A3 WO2002026209A3 (en) 2003-01-03

Family

ID=26929462

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/030540 WO2002026209A2 (en) 2000-09-28 2001-09-28 Microparticles for delivery of the heterologous nucleic acids

Country Status (16)

Country Link
US (2) US20030138453A1 (en)
EP (1) EP1322287B1 (en)
JP (1) JP2004518631A (en)
CN (1) CN1468089B (en)
AT (1) ATE320792T1 (en)
AU (1) AU9489701A (en)
BR (1) BR0114305A (en)
CA (1) CA2421683C (en)
DE (1) DE60118228T2 (en)
DK (1) DK1322287T3 (en)
ES (1) ES2260291T3 (en)
MX (1) MXPA03002640A (en)
PT (1) PT1322287E (en)
SG (1) SG137691A1 (en)
WO (1) WO2002026209A2 (en)
ZA (1) ZA200301744B (en)

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004032860A2 (en) 2002-10-07 2004-04-22 Chiron Corporation Hiv vaccine formulations
WO2004060396A2 (en) 2002-12-27 2004-07-22 Chiron Corporation Immunogenic compositions containing phospholpid
WO2005020964A1 (en) * 2003-06-02 2005-03-10 Chiron Corporation Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
WO2005049080A1 (en) * 2003-11-17 2005-06-02 Sanofi Pasteur Vaccine composition admixed with an alkylphosphatidylcholine
WO2004096136A3 (en) * 2003-04-25 2005-06-02 Chiron Corp Compositions comprising cationic microparticles and hcv e1e2 dna and methods of use thereof
JP2006502979A (en) * 2002-06-20 2006-01-26 サイトス バイオテクノロジー アーゲー Packaged virus-like particles for use as adjuvants: methods of preparation and uses
EP1628680A2 (en) * 2003-05-15 2006-03-01 Chiron Corporation Hiv polynucleotides and polypeptides derived from botswana mj4
US7501134B2 (en) 2002-02-20 2009-03-10 Novartis Vaccines And Diagnostics, Inc. Microparticles with adsorbed polypeptide-containing molecules
WO2011149564A1 (en) 2010-05-28 2011-12-01 Tetris Online, Inc. Interactive hybrid asynchronous computer game infrastructure
WO2012006380A2 (en) 2010-07-06 2012-01-12 Novartis Ag Cationic oil-in-water emulsions
WO2012006359A1 (en) 2010-07-06 2012-01-12 Novartis Ag Delivery of self-replicating rna using biodegradable polymer particles
US8158133B2 (en) 2004-12-02 2012-04-17 Vetoquinol Pharmaceutical composition useful for vaccines
WO2013006834A1 (en) 2011-07-06 2013-01-10 Novartis Ag Oil-in-water emulsions that contain nucleic acids
WO2014005958A1 (en) * 2012-07-06 2014-01-09 Novartis Ag Immunogenic compositions and uses thereof
US8691209B2 (en) 2001-09-14 2014-04-08 Cytos Biotechnology Ag Packaging of immunostimulatory substances into virus-like particles: method of preparation and use
KR20150013133A (en) * 2012-03-23 2015-02-04 아미크로베, 인코포레이티드. Compositions and uses of antimicrobial materials with tissue-compatible properties
EP2591114B1 (en) 2010-07-06 2016-06-08 GlaxoSmithKline Biologicals SA Immunisation of large mammals with low doses of rna
US9393215B2 (en) 2005-12-02 2016-07-19 Novartis Ag Nanoparticles for use in immunogenic compositions
US9393295B2 (en) 2008-04-28 2016-07-19 Novartis Ag Nanoparticles for use in pharmaceutical compositions
EP2590676B1 (en) 2010-07-06 2016-08-17 GlaxoSmithKline Biologicals SA Virion-like delivery particles for self-replicating rna molecules
US9433674B2 (en) 2005-11-14 2016-09-06 Centre Nationale De Recherche Scientifique Composition comprising a colloidal synthetic bioresorbable vector and a viral vector
US9636410B2 (en) 2011-07-06 2017-05-02 Glaxosmithkline Biologicals Sa Cationic oil-in-water emulsions
EP3243526A1 (en) * 2010-07-06 2017-11-15 GlaxoSmithKline Biologicals S.A. Delivery of rna to trigger multiple immune pathways
WO2018232257A1 (en) 2017-06-15 2018-12-20 Infectious Disease Research Institute Nanostructured lipid carriers and stable emulsions and uses thereof
EP2591103B1 (en) 2010-07-06 2019-08-28 GlaxoSmithKline Biologicals SA Delivery of rna to different cell types
US10975388B2 (en) 2016-12-14 2021-04-13 Ligandal, Inc. Methods and compositions for nucleic acid and protein payload delivery
WO2022051022A1 (en) 2020-09-04 2022-03-10 Infectious Disease Research Institute Co-lyophilized rna and nanostructured lipid carrier
WO2022051024A1 (en) 2020-09-04 2022-03-10 Infectious Disease Research Institute Genetically-adjuvanted rna vaccines
US11285189B2 (en) 2017-04-06 2022-03-29 Amicrobe, Inc. Compositions and uses of locally-applied antimicrobial synthetic cationic polypeptide(s) with enhanced performance and safety
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
CN114917334A (en) * 2022-07-21 2022-08-19 天津三江永利生物科技有限公司 Water-in-oil-in-water type composite vaccine adjuvant, and preparation method and application thereof
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
WO2024052882A1 (en) 2022-09-09 2024-03-14 Access To Advanced Health Institute Immunogenic vaccine composition incorporating a saponin

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7935805B1 (en) * 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
EP1141314A2 (en) 1998-12-31 2001-10-10 Chiron Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
US8221761B1 (en) * 1999-02-26 2012-07-17 Novartis Ag Enhancement of bactericidal activity of neisseria antigens with oligonucleotides containing CG motifs
US6977245B2 (en) 1999-04-12 2005-12-20 The United States Of America As Represented By The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US7666674B2 (en) 2001-07-27 2010-02-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of sterically stabilized cationic liposomes to efficiently deliver CPG oligonucleotides in vivo
US7354909B2 (en) * 2001-08-14 2008-04-08 The United States Of America As Represented By Secretary Of The Department Of Health And Human Services Method for rapid generation of mature dendritic cells
US7615227B2 (en) * 2001-12-20 2009-11-10 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce angiogenesis
US8466116B2 (en) 2001-12-20 2013-06-18 The Unites States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce epithelial cell growth
AU2003219805B2 (en) * 2002-02-15 2009-06-04 Eisai Inc. Electroporation methods for introducing bioactive agents into cells
US8263091B2 (en) * 2002-09-18 2012-09-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of treating and preventing infections in immunocompromised subjects with immunostimulatory CpG oligonucleotides
ES2615029T3 (en) * 2003-01-14 2017-06-05 Glaxosmithkline Biologicals Sa Microparticles comprising polynucleotides adsorbed on the surface of microparticles or trapped inside them
US20040235770A1 (en) * 2003-04-02 2004-11-25 Coley Pharmaceutical Group, Ltd. Immunostimulatory nucleic acid oil-in-water formulations and related methods of use
GB0416120D0 (en) * 2004-07-19 2004-08-18 Health Prot Agency Stable compositions containing OMV's
US9107813B2 (en) * 2004-11-15 2015-08-18 Derek T. O'Hagan Immunogenic compositions containing anthrax antigen, biodegradable polymer microparticles, and polynucleotide-containing immunological adjuvant
EP2482818A4 (en) * 2009-09-29 2014-04-09 Eyegate Pharmaceuticals Inc Positively-charged poly (d,l-lactide-co-glycolide) nanoparticles and fabrication methods of the same
KR20140022025A (en) * 2011-03-25 2014-02-21 셀렉타 바이오사이언시즈, 인크. Osmotic mediated release synthetic nanocarriers

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998033487A1 (en) * 1997-01-30 1998-08-06 Chiron Corporation Use of microparticles with adsorbed antigen to stimulate immune responses
WO2001081609A2 (en) * 2000-03-22 2001-11-01 Chiron Corporation Compositions and methods for generating an immune response utilizing alphavirus-based vector systems

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL134977C (en) * 1962-07-11
FR2695563B1 (en) * 1992-09-11 1994-12-02 Pasteur Institut Microparticles carrying antigens and their use for the induction of humoral or cellular responses.
US5928647A (en) * 1993-01-11 1999-07-27 Dana-Farber Cancer Institute Inducing cytotoxic T lymphocyte responses
US5603960A (en) * 1993-05-25 1997-02-18 O'hagan; Derek T. Preparation of microparticles and method of immunization
US6015686A (en) * 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
GB9412273D0 (en) * 1994-06-18 1994-08-10 Univ Nottingham Administration means
US5842723A (en) * 1996-11-25 1998-12-01 Burex Automotive America, Inc. Flexible connection arrangement for connecting a pipe of an exhaust system to an engine, especially in a motor vehicle
US5783567A (en) * 1997-01-22 1998-07-21 Pangaea Pharmaceuticals, Inc. Microparticles for delivery of nucleic acid
US6884435B1 (en) * 1997-01-30 2005-04-26 Chiron Corporation Microparticles with adsorbent surfaces, methods of making same, and uses thereof
PT1042001E (en) * 1997-12-16 2002-09-30 Chiron Corp USE OF MICROPARTICLES COMBINED WITH SUBMICronic EMULSES OIL-IN-WATER
CA2360347C (en) * 1998-12-31 2013-05-07 Chiron Corporation Improved expression of hiv polypeptides and production of virus-like particles
US7935805B1 (en) * 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
EP1574210B1 (en) * 1999-02-26 2016-04-06 Novartis Vaccines and Diagnostics, Inc. Microemulsions with adsorbed macromolecules
ES2278786T3 (en) * 2000-09-28 2007-08-16 Novartis Vaccines And Diagnostics, Inc. COMPOSITIONS OF MICROPARTICLES AND MANUFACTURING PROCEDURES OF THE SAME.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998033487A1 (en) * 1997-01-30 1998-08-06 Chiron Corporation Use of microparticles with adsorbed antigen to stimulate immune responses
WO2001081609A2 (en) * 2000-03-22 2001-11-01 Chiron Corporation Compositions and methods for generating an immune response utilizing alphavirus-based vector systems

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J. KAZZAZ, ET AL.: "Induction of cytotoxic T-lymphocite activity in mice with HIV p24 gag protein adsorbed to the surface of poly(lactide-co glycolide) microparticles" PROCEED. INT'L. SYMP. CONTROL. REL. BIOACT. MATER., July 1999 (1999-07), pages 1046-1047, XP002216995 *
K. S. DENIS-MIZE: "Plasmid DNA adsorbed onto cationic microparticles mediates target gene expression and antigen presentation by dendritic cells" GENE THERAPY, vol. 7, 2000, pages 2105-2112, XP002216996 *

Cited By (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8691209B2 (en) 2001-09-14 2014-04-08 Cytos Biotechnology Ag Packaging of immunostimulatory substances into virus-like particles: method of preparation and use
US9950055B2 (en) 2001-09-14 2018-04-24 Kuros Biosciences Ag Packaging of immunostimulatory substances into virus-like particles: method of preparation and use
US7501134B2 (en) 2002-02-20 2009-03-10 Novartis Vaccines And Diagnostics, Inc. Microparticles with adsorbed polypeptide-containing molecules
JP2006502979A (en) * 2002-06-20 2006-01-26 サイトス バイオテクノロジー アーゲー Packaged virus-like particles for use as adjuvants: methods of preparation and uses
US8609109B2 (en) 2002-10-07 2013-12-17 Novartis Vaccines And Diagnostics, Inc. HIV vaccine formulations
EP1565583A2 (en) * 2002-10-07 2005-08-24 Chiron Corporation Hiv vaccine formulations
WO2004032860A2 (en) 2002-10-07 2004-04-22 Chiron Corporation Hiv vaccine formulations
EP1565583A4 (en) * 2002-10-07 2007-11-21 Novartis Vaccines & Diagnostic Hiv vaccine formulations
US8541023B2 (en) 2002-12-27 2013-09-24 Novartis Vaccines And Diagnostics, Inc. Immunogenic compositions containing phospholipid
US8034378B2 (en) 2002-12-27 2011-10-11 Novartis Vaccines And Diagnostics, Inc Immunogenic compositions containing phospholipid
WO2004060396A2 (en) 2002-12-27 2004-07-22 Chiron Corporation Immunogenic compositions containing phospholpid
EP2263687A1 (en) 2002-12-27 2010-12-22 Novartis Vaccines&Diagnostics, Inc. Immunogenic compositions containing phospholipid
WO2004096136A3 (en) * 2003-04-25 2005-06-02 Chiron Corp Compositions comprising cationic microparticles and hcv e1e2 dna and methods of use thereof
EP1628680A2 (en) * 2003-05-15 2006-03-01 Chiron Corporation Hiv polynucleotides and polypeptides derived from botswana mj4
EP1628680A4 (en) * 2003-05-15 2008-08-06 Novartis Vaccines & Diagnostic Hiv polynucleotides and polypeptides derived from botswana mj4
EP2179729A1 (en) 2003-06-02 2010-04-28 Novartis Vaccines and Diagnostics, Inc. Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
WO2005020964A1 (en) * 2003-06-02 2005-03-10 Chiron Corporation Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
US9107831B2 (en) 2003-06-02 2015-08-18 Novartis Vaccines And Diagonstics, Inc. Immunogenic compositions containing microparticles comprising adsorbed toxoid and polysaccharide-containing antigens
US7344720B2 (en) 2003-11-17 2008-03-18 Sanofi Pasteur Sa Vaccine composition
US7767658B2 (en) 2003-11-17 2010-08-03 Aventis Pasteur Sa Vaccine composition
WO2005049080A1 (en) * 2003-11-17 2005-06-02 Sanofi Pasteur Vaccine composition admixed with an alkylphosphatidylcholine
US8158133B2 (en) 2004-12-02 2012-04-17 Vetoquinol Pharmaceutical composition useful for vaccines
US9433674B2 (en) 2005-11-14 2016-09-06 Centre Nationale De Recherche Scientifique Composition comprising a colloidal synthetic bioresorbable vector and a viral vector
US9393215B2 (en) 2005-12-02 2016-07-19 Novartis Ag Nanoparticles for use in immunogenic compositions
US9393295B2 (en) 2008-04-28 2016-07-19 Novartis Ag Nanoparticles for use in pharmaceutical compositions
WO2011149564A1 (en) 2010-05-28 2011-12-01 Tetris Online, Inc. Interactive hybrid asynchronous computer game infrastructure
US10532067B2 (en) 2010-07-06 2020-01-14 Glaxosmithkline Biologicals S.A. Delivery of RNA to trigger multiple immune pathways
US11324770B2 (en) 2010-07-06 2022-05-10 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
EP2591114B1 (en) 2010-07-06 2016-06-08 GlaxoSmithKline Biologicals SA Immunisation of large mammals with low doses of rna
US11913001B2 (en) 2010-07-06 2024-02-27 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11905514B2 (en) 2010-07-06 2024-02-20 Glaxosmithkline Biological Sa Immunisation of large mammals with low doses of RNA
EP2590676B1 (en) 2010-07-06 2016-08-17 GlaxoSmithKline Biologicals SA Virion-like delivery particles for self-replicating rna molecules
US11891608B2 (en) 2010-07-06 2024-02-06 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
EP3115061A1 (en) * 2010-07-06 2017-01-11 GlaxoSmithKline Biologicals SA Virion-like delivery particles for self-replicating rna molecules
US11883534B2 (en) 2010-07-06 2024-01-30 Glaxosmithkline Biologicals Sa Immunisation with lipid formulations with RNA encoding immunogens
US11865080B2 (en) 2010-07-06 2024-01-09 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
EP3243526A1 (en) * 2010-07-06 2017-11-15 GlaxoSmithKline Biologicals S.A. Delivery of rna to trigger multiple immune pathways
WO2012006359A1 (en) 2010-07-06 2012-01-12 Novartis Ag Delivery of self-replicating rna using biodegradable polymer particles
US11857681B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Lipid formulations with RNA encoding immunogens
US11857562B2 (en) 2010-07-06 2024-01-02 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11839686B2 (en) 2010-07-06 2023-12-12 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
EP3449910A1 (en) 2010-07-06 2019-03-06 GlaxoSmithKline Biologicals S.A. Cationic oil-in-water emulsions
EP2591117B1 (en) * 2010-07-06 2019-05-08 GlaxoSmithKline Biologicals SA Delivery of self-replicating rna using biodegradable polymer particles
US11786467B2 (en) 2010-07-06 2023-10-17 Glaxosmithkline Biologicals Sa Lipid formulations with immunogens
EP2591103B1 (en) 2010-07-06 2019-08-28 GlaxoSmithKline Biologicals SA Delivery of rna to different cell types
US10487332B2 (en) 2010-07-06 2019-11-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
WO2012006380A2 (en) 2010-07-06 2012-01-12 Novartis Ag Cationic oil-in-water emulsions
EP3611269A1 (en) 2010-07-06 2020-02-19 GlaxoSmithKline Biologicals SA Delivery of self-replicating rna using biodegradable polymer particles
US11773395B1 (en) 2010-07-06 2023-10-03 Glaxosmithkline Biologicals Sa Immunization of large mammals with low doses of RNA
US11766401B2 (en) 2010-07-06 2023-09-26 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with immunogens
US11759475B2 (en) 2010-07-06 2023-09-19 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11739334B2 (en) 2010-07-06 2023-08-29 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11730754B2 (en) 2010-07-06 2023-08-22 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11717529B2 (en) 2010-07-06 2023-08-08 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11707482B2 (en) 2010-07-06 2023-07-25 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11696923B2 (en) 2010-07-06 2023-07-11 Glaxosmithkline Biologicals, Sa Delivery of RNA to trigger multiple immune pathways
US11291635B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biological Sa Virion-like delivery particles for self-replicating RNA molecules
US11291682B2 (en) 2010-07-06 2022-04-05 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690862B1 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US20220125723A1 (en) 2010-07-06 2022-04-28 Glaxosmithkline Biologicals Sa Lipid formulations with viral immunogens
US11690863B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690865B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11596645B2 (en) 2010-07-06 2023-03-07 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11690864B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11638693B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising RNA encoding an immunogen and lipid formulations comprising mole percentage of lipids
US11638694B2 (en) 2010-07-06 2023-05-02 Glaxosmithkline Biologicals Sa Vaccine for eliciting immune response comprising lipid formulations and RNA encoding multiple immunogens
US11655475B2 (en) 2010-07-06 2023-05-23 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
US11666534B2 (en) 2010-07-06 2023-06-06 Glaxosmithkline Biologicals Sa Methods of administering lipid formulations with viral immunogens
US11690861B2 (en) 2010-07-06 2023-07-04 Glaxosmithkline Biologicals Sa Delivery of RNA to trigger multiple immune pathways
US11759422B2 (en) 2010-08-31 2023-09-19 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of immunogen-encoding RNA
US11639370B2 (en) 2010-10-11 2023-05-02 Glaxosmithkline Biologicals Sa Antigen delivery platforms
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
WO2013006834A1 (en) 2011-07-06 2013-01-10 Novartis Ag Oil-in-water emulsions that contain nucleic acids
US9636410B2 (en) 2011-07-06 2017-05-02 Glaxosmithkline Biologicals Sa Cationic oil-in-water emulsions
US9655845B2 (en) 2011-07-06 2017-05-23 Glaxosmithkline Biologicals, S.A. Oil-in-water emulsions that contain nucleic acids
EP3424495A1 (en) 2011-07-06 2019-01-09 GlaxoSmithKline Biologicals S.A. Oil-in-water emulsions that contain nucleic acids
US10183074B2 (en) 2011-07-06 2019-01-22 Glaxosmithkline Biologicals S.A. Cationic oil-in-water emulsions
US11167028B2 (en) 2011-07-06 2021-11-09 Glaxosmithkline Biologicals Sa Cationic oil-in-water emulsions
US10307374B2 (en) 2011-07-06 2019-06-04 Glaxosmithkline Biologicals S.A. Oil-in-water emulsions that contain nucleic acids
US11026890B2 (en) 2011-07-06 2021-06-08 Glaxosmithkline Biologicals Sa Oil-in-water emulsions that contain nucleic acids
KR102141254B1 (en) 2012-03-23 2020-08-04 아미크로베, 인코포레이티드. Compositions and uses of antimicrobial materials with tissue-compatible properties
US11298401B2 (en) 2012-03-23 2022-04-12 Amicrobe, Inc. Compositions and uses of antimicrobial materials with tissue-compatible properties
US12016898B2 (en) 2012-03-23 2024-06-25 Amicrobe, Inc. Compositions and uses of antimicrobial materials with tissue-compatible properties
KR20150013133A (en) * 2012-03-23 2015-02-04 아미크로베, 인코포레이티드. Compositions and uses of antimicrobial materials with tissue-compatible properties
WO2014005958A1 (en) * 2012-07-06 2014-01-09 Novartis Ag Immunogenic compositions and uses thereof
CN104853770A (en) * 2012-07-06 2015-08-19 诺华股份有限公司 Immunogenic compositions and uses thereof
US10975388B2 (en) 2016-12-14 2021-04-13 Ligandal, Inc. Methods and compositions for nucleic acid and protein payload delivery
US11285189B2 (en) 2017-04-06 2022-03-29 Amicrobe, Inc. Compositions and uses of locally-applied antimicrobial synthetic cationic polypeptide(s) with enhanced performance and safety
WO2018232257A1 (en) 2017-06-15 2018-12-20 Infectious Disease Research Institute Nanostructured lipid carriers and stable emulsions and uses thereof
US11141377B2 (en) 2017-06-15 2021-10-12 Infectious Disease Research Institute Nanostructured lipid carriers and stable emulsions and uses thereof
WO2022051024A1 (en) 2020-09-04 2022-03-10 Infectious Disease Research Institute Genetically-adjuvanted rna vaccines
WO2022051022A1 (en) 2020-09-04 2022-03-10 Infectious Disease Research Institute Co-lyophilized rna and nanostructured lipid carrier
CN114917334A (en) * 2022-07-21 2022-08-19 天津三江永利生物科技有限公司 Water-in-oil-in-water type composite vaccine adjuvant, and preparation method and application thereof
WO2024052882A1 (en) 2022-09-09 2024-03-14 Access To Advanced Health Institute Immunogenic vaccine composition incorporating a saponin

Also Published As

Publication number Publication date
DE60118228D1 (en) 2006-05-11
EP1322287A2 (en) 2003-07-02
JP2004518631A (en) 2004-06-24
DK1322287T3 (en) 2006-07-24
CN1468089A (en) 2004-01-14
MXPA03002640A (en) 2003-06-19
WO2002026209A3 (en) 2003-01-03
CA2421683C (en) 2009-09-15
CN1468089B (en) 2011-09-21
AU9489701A (en) 2002-04-08
EP1322287B1 (en) 2006-03-22
BR0114305A (en) 2003-07-01
US20030138453A1 (en) 2003-07-24
ES2260291T3 (en) 2006-11-01
CA2421683A1 (en) 2002-04-04
SG137691A1 (en) 2007-12-28
PT1322287E (en) 2006-08-31
ATE320792T1 (en) 2006-04-15
DE60118228T2 (en) 2006-12-14
ZA200301744B (en) 2005-09-28
US20080026071A1 (en) 2008-01-31

Similar Documents

Publication Publication Date Title
EP1322287B1 (en) Microparticles for delivery of the heterologous nucleic acids
CA2528007C (en) Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
AU2001294898B2 (en) Microparticle compositions and methods for the manufacture thereof
EP1585542B1 (en) Immunogenic compositions containing phospholipid
AU2001294898A1 (en) Microparticle compositions and methods for the manufacture thereof
US20150072016A1 (en) Microparticles with adsorbed polynucleotide-containing species
AU2001294897B2 (en) Microparticles for delivery of the heterologous nucleic acids
RU2295954C2 (en) Microparticles for delivery of heterologous nucleic acids
AU2001294897A1 (en) Microparticles for delivery of the heterologous nucleic acids
CA2765058C (en) Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
PT1585542E (en) Immunogenic compositions containing phospholipid

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 CO 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 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 GQ GW ML MR NE SN TD TG

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

Ref document number: 2003/01744

Country of ref document: ZA

Ref document number: 200301744

Country of ref document: ZA

Ref document number: 270/KOLNP/2003

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2421683

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2001294897

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2002530039

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2003/002640

Country of ref document: MX

Ref document number: 2001975584

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 018164595

Country of ref document: CN

ENP Entry into the national phase

Ref country code: RU

Ref document number: RU A

Ref document number: 2003112234

Country of ref document: RU

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2001975584

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 2001975584

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2001294897

Country of ref document: AU