WO2016068919A1 - Combinaison thérapeutique pour le traitement de réservoirs viraux - Google Patents

Combinaison thérapeutique pour le traitement de réservoirs viraux Download PDF

Info

Publication number
WO2016068919A1
WO2016068919A1 PCT/US2014/062965 US2014062965W WO2016068919A1 WO 2016068919 A1 WO2016068919 A1 WO 2016068919A1 US 2014062965 W US2014062965 W US 2014062965W WO 2016068919 A1 WO2016068919 A1 WO 2016068919A1
Authority
WO
WIPO (PCT)
Prior art keywords
hiv
viral
vaccine
dna
reservoirs
Prior art date
Application number
PCT/US2014/062965
Other languages
English (en)
Inventor
Harriet L. Robinson
Suefen KWA
Original Assignee
Geovax, Inc.
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 Geovax, Inc. filed Critical Geovax, Inc.
Publication of WO2016068919A1 publication Critical patent/WO2016068919A1/fr

Links

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2740/16034Use 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

Definitions

  • the present invention relates to compositions and methods useful for reducing viral reservoirs in a subject in need thereof.
  • the present invention relates to compositions and methods useful for reducing reservoirs of HIV in a subject in need thereof.
  • a viral reservoir is a cell or anatomical site in which a replication-competent form of a virus accumulates and persists with more stable kinetic properties than the main pool of actively replicating virus.
  • Viral reservoirs represent a challenge to eradication of viral infection, even when viremia can be suppressed and maintained below detectable levels by means of anti-viral therapy.
  • HIV reservoirs of HIV established early in infection represent a significant challenge to eradication of HIV infection.
  • Potential reservoirs for HIV include resting memory CD4+ T cells, in which latent HIV shows minimal decay even in patients on highly active antiretroviral therapy (HAART).
  • HAART highly active antiretroviral therapy
  • the persistence of virus in this reservoir is consistent with the biology of these cells and the long-term persistence of immunologic memory. While other HIV reservoirs have been reported, the reservoir in resting CD4+ T cells appears to be sufficient for a persistence of HIV infection in the majority of patients on HAART regimens.
  • compositions and methods are provided for reducing viral reservoirs. Methods are provided for reducing viral reservoirs in a person on continuous antiretroviral drugs involving administering a stimulating ( "shock") agent to a subject in need thereof, followed by
  • viral reservoirs are eradicated.
  • the viral reservoir is an HIV reservoir.
  • the present invention is a composition comprising one or more shock agents selected from a DNA plasmid, an HIV DNA vaccine, a CpG oligodeoxynucleotide, a TLR9 receptor agonist, an HDAC inhibitor, an NFkappaB activator, a cytosine methylation inhibitor, or an acetaldehyde dehydrogenase inhibitor.
  • shock agents selected from a DNA plasmid, an HIV DNA vaccine, a CpG oligodeoxynucleotide, a TLR9 receptor agonist, an HDAC inhibitor, an NFkappaB activator, a cytosine methylation inhibitor, or an acetaldehyde dehydrogenase inhibitor.
  • the shock agent is an HIV DNA vaccine.
  • the shock agent is an HIV DNA vaccine comprising one or more safety mutations.
  • the HIV DNA vaccine comprises (i) a prokaryotic origin of replication and (ii) a eukaryotic transcription cassette comprising a vaccine insert encoding
  • Gag protein comprising one or more safety mutations to inactivate zinc fingers
  • a Pol protein comprising one or more safety mutations to (i) inhibit integrase activity and (ii) inhibit polymerase, strand transfer, and/or RNase H activity of reverse transcriptase;
  • the Gag protein has safety mutations that inactivate one or more of the zinc fingers corresponding to clade B C392S, C395S, C413S, and C416S.
  • the Pol protein has safety mutations that delete the integrase sequence.
  • the Pol protein has safety mutations that inactivate reverse transcriptase corresponding to clade B D185N, W266T, and E478Q. In one embodiment, the Pol protein has safety mutations to inhibit protease activity that corresponds to clade B D25A.
  • the clade B HIV is strain HXB2.
  • the HIV DNA vaccine has (i) a prokaryotic origin of replication and (ii) a eukaryotic transcription cassette comprising a vaccine insert encoding
  • HIV-1 clade B Pol protein comprising:
  • the HIV DNA vaccine comprises a sequence encoding human GM-
  • the HIV DNA vaccine co-expresses GM-CSF.
  • the shock agent comprises a CpG oligodeoxynucleotide (CpG)
  • the shock agent comprises CpG ODN, wherein the CpG ODN is CpG-7909.
  • the composition comprises two shock agents, wherein the shock agents are co-administered HIV DNA vaccine and CpG ODN.
  • the shock agent is an HDAC inhibitor and more particularly, the shock agent is romidepsin.
  • the shock agent is an acetaldehyde dehydrogenase inhibitor, and more particularly, the shock agent is disulfiram.
  • the shock agent is a cytosine methylation inhibitor, and more particularly, 5-azacytidine.
  • the present invention comprises one or more kill agents selected from an HIV vaccine, a monoclonal antibody or combinations thereof.
  • the HIV vaccine is selected from an MVA vaccine, an adenoviral vaccine, or a vesicular stomatitis virus (VSV) vaccine.
  • the adenoviral vaccine is a human chimpanzee adenoviral vector vaccine.
  • the HIV vaccine is an MVA HIV vaccine.
  • the kill agent is an MVA HIV vaccine including (i) a sequence encoding an HIV Env antigen inserted into deletion site II of an MVA, wherein the HIV Env antigen comprises gpl20 and the membrane spanning and ectodomain of gp41, but lacks part of all of the cytoplasmic domain, and (ii) a sequence encoding HIV Gag and Pol antigens inserted into deletion site III of an MVA, wherein the sequence encoding the HIV Gag and Pol antigens comprises safety mutations to limit packaging of viral R A and inactivate reverse transcriptase and delete the integrase coding sequence; and wherein the sequences of (i) and (ii) are under the control of promoters compatible with poxvirus expression systems.
  • the safety mutations that inactivate reverse transcriptase and delete the integrase coding sequence correspond to clade B HIV D185N, W266T, and E478Q.
  • the clade B HIV is strain HXB2.
  • the kill agent is an antibody.
  • the kill agent is a monoclonal antibody or a recombinant Ab.
  • the kill agent is a neutralizing antibody or a non-neutralizing antibody.
  • the kill agent is a Ab that also initiates innate immune responses.
  • the kill agent is a neutralizing Ab selected from antibodies such as 10-1074, 10E8, 17B, 2F5, 2G12, 447-52D, 448-D, 4E10, 670D, bl2, BB34, CAP206-CH12, CHOI, CH02, CH03, CH04, CH08, CH30, CH31, CH32, CH33, CH34, HJ16, M66.6, NIH45, NIH46, PG16, PG9, PGT121, PGT128, VRCOl, VRC02, VRC03, or VRC07.
  • antibodies such as 10-1074, 10E8, 17B, 2F5, 2G12, 447-52D, 448-D, 4E10, 670D, bl2, BB34, CAP206-CH12, CHOI, CH02, CH03, CH04, CH08, CH30, CH31, CH32, CH33, CH34, HJ16, M66.6, NIH45, NIH46, PG16
  • the kill agent is a non-neutralizing antibody selected from antibodies such as 120-16, 126-50, 246-D, 4B3, 50-69, 98-43, 98-6, b6, 3D6, A32, or F240.
  • the present invention is a method of reducing viral reservoirs in a subject on antiretroviral treatment in need thereof by: (i) administering one or more shock agents; and (ii) administering one or more kill agents, thereby reducing viral reservoirs.
  • the kill agent is administered between about 1 and about 4 weeks, about 1 and about 2 weeks, or about 1 week after administration of the shock agent.
  • the kill agent is administered between about 1 and about 10 days, about 1 and about 7 days, about 1 and about 5 days, about 1 and about 3 days, about 3 and about 7 days, about 5 and about 6 days, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days after the shock agent.
  • the kill agent is administered within 24 hours, i.e., within one day of the shock agent.
  • the method may comprise one or more additional steps.
  • the method further comprises the step of: (iii) measuring the presence of viral reservoirs for reduction.
  • viral reservoirs are measured at about 1, about 2, about 3, about 4, about 5, about 6, about 7, or about 8 weeks after step (ii).
  • the viral reservoirs are measured about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, or 12 months after step (ii).
  • the viral reservoirs are measured about 1, about 2, about 3, about 4, or about 5 years after step (ii).
  • the method further comprises determining the measuring the temporal levels of reactivated viral R A between step (i) and (ii) in order to determine peak levels of reactivated virus.
  • the kill agent is administered at or about the time of peak virus reactivation.
  • the subject is undergoing antiretroviral therapy prior to, during, and/or after administration of the shock and/or kill agents.
  • the method is repeated two or more times. In a particular embodiment, the method is repeated two or more times at regular intervals. Where the method is repeated, the term "cycle" is used to refer to steps (i) and (ii), collectively. In one embodiment, the method is repeated at least 2 times, at least 3 times, at least 4 times, at least 5 times, or more than 5 times.
  • the method is repeated and about the interval between cycles is about 1 to about 4 weeks, about 2 to about 4 week, about 3 about 4 weeks, about 1 week, about 2 weeks, about 3 weeks, or about 4 weeks.
  • the method is repeated between about 2 and about 8 times, about 4 and about 8 times, about 6 and about 8 times, about 2, about 4, about 6 or about 8 times.
  • the method is repeated more than 8 times.
  • step (i) and (ii) is substantially the same for all shock/kill cycles.
  • the method is repeated until reservoirs are undetectable by reservoir assay.
  • the method is repeated until viral reservoirs are reduced.
  • FIG. 1 provides graphs showing that CpG ODN, but not GpC ODN or GEO-D03 DNA, induce HIV-1 activation in ACH-2 cells.
  • FIG. 1A cells were seeded at lxlO 6 cells/well.
  • FIG. IB cells were seeded at 0.3xl0 6 cells/well.
  • Expression of HIV-Gag was determined by intracellular staining using mouse mAb KC57 (Beckman Coulter) and flow cytometry.
  • FIG. 2 provides graphs showing viral loads (in copies/mL plasma) in patients
  • FIG. 3 provides graphs showing reservoir measurement for all participants.
  • Pre-Vaccine indicates the time point prior to the first DNA vaccination for all participants other than 01-7. Because this sample was not available for participant 01-7, the 01-7 Pre-Vaccine time point is from Week 10, between the second DNA vaccination and the first MVA vaccination.
  • FIG. 4 provides a schematic of an exemplary shock/kill administration regimen for the flushing and killing of cells expressing reactivated HIV to reduce viral reservoirs .
  • compositions and methods are provided for reducing viral reservoirs in a subject in need thereof.
  • reducing viral reservoirs refers to reduction of viral reservoirs compared to the levels of viral reservoirs present before treatment, including total elimination or eradication of viral reservoirs, i.e., reduction to zero or undetectable levels. Other similar terms include “flushing", “clearing” or “eradicating” viral reservoirs.
  • Methods are provided for reducing viral reservoirs in a subject in need thereof, comprising (i) administering one or more stimulating or "shock” agents to stimulate the viral reservoir; and (ii) administering one or more eradication or “kill” agents, thereby reducing viral reservoirs.
  • the viral reservoir may be, for example, an HIV viral reservoir.
  • ART anti-retroviral therapy
  • the HIV DNA vaccine acts as a shock agent and stimulates the reservoirs to express virus, making the infected cells susceptible to killing by the CD8+ T cells elicited by the MVA vaccine, which acts as an eradication or kill agent.
  • the ART drugs reduce or prevent virus spread to uninfected cells
  • shock and kill agents are useful in this two-step approach to reducing viral reservoirs in a subject in need thereof.
  • the virus may be HIV or another viral agent.
  • shock agent refers to a compound capable of stimulating viral reservoirs so as to activate or increase expression of latent virus, resulting in transiently increased viral load.
  • the shock agent may be, without limitation, a small molecule shock agent or a nucleic acid shock agent such as an oligonucleotide, a nucleic acid vaccine or the like.
  • the present methods employ shock agents comprising nucleic acid sequences that induce an immune response to a virus (e.g., HIV-1), including nucleic acid analogs of those sequences and compositions containing those nucleic acids (whether vector plus insert or insert only or vector only; e.g., physiologically acceptable solutions, which may include carriers such as liposomes, calcium, particles (e.g., gold beads) or other reagents used to deliver DNA to cells).
  • a virus e.g., HIV-1
  • nucleic acid analogs of those sequences and compositions containing those nucleic acids whether vector plus insert or insert only or vector only; e.g., physiologically acceptable solutions, which may include carriers such as liposomes, calcium, particles (e.g., gold beads) or other reagents used to deliver DNA to cells).
  • nucleic acid vaccines have improved an side effect profile. Certain shock agents described herein cause substantial systemic and local adverse events. However, the nucleic acid vaccines described herein activate viral reservoirs in the presence of very limited local or systemic reactogenicity.
  • the analogs can be sequences that are not identical to those disclosed herein, but that include the same or similar mutations (e.g., the same point mutation or a similar point mutation) at positions analogous to those included in the present sequences. For example, a given residue or domain can be identified in various HIV clades even though it does not appear at precisely the same numerical position.
  • the analogs can also be sequences that include mutations that, while distinct from those described herein, similarly inactivate a viral gene product (e.g., an HIV gene product).
  • a gene that is truncated to a greater or lesser extent than one of the genes described here, but that is similarly inactivated (e.g., that loses a particular enzymatic or functional activity) is within the scope of the present disclosure.
  • the pathogens and antigens include human immunodeficiency viruses of any clade (e.g. from any known clade or from any isolate (e.g., clade A, AG, B, C, D, E, F, G, H, I, J, K, or L). Additional HIV sequences and mutant sequences are known in the art (e.g., the HIV Sequence Database in Los Alamos and the HIV RT/Protease Sequence Database in Stanford).
  • the vectors include sequences from a pathogen, they can be administered to a patient to elicit an immune response.
  • methods of administering antigen-encoding vectors alone or in combination with one another, are also described herein.
  • These methods can be carried out to either immunize patients (thereby reducing the patient's risk of becoming infected) or to treat patients who have already become infected; when expressed, the antigens may elicit both cell-mediated and humoral immune responses that may substantially prevent the infection (e.g., immunization can protect against subsequent challenge by the pathogen) or limit the extent of the impact of an infection on the patient's health. While in many instances the patient will be a human patient, the disclosure is not so limited. Other animals, including non-human primates, domesticated animals, and livestock can also be treated.
  • compositions described herein regardless of the pathogen or pathogenic subtype (e.g., the HIV clade(s)) they are directed against, can include a nucleic acid vector (e.g., a plasmid).
  • a nucleic acid vector e.g., a plasmid
  • plasmids designated pGAl, pGA2 can be used as the basis for a vaccine or therapy.
  • Such vectors can be engineered using standard recombinant techniques (several of which are illustrated in the examples, below) to include insert sequences that encode viral antigens that, when administered to, and subsequently expressed in, a patient will elicit (e.g., induce or enhance) an immune response that provides the patient with some form of protection against the virus from which the antigens were obtained or derived (e.g., protection against infection, protection against disease, or amelioration of one or more of the signs or symptoms of a disease).
  • the vector administered is a pGA vector
  • it can comprise the sequence of, for example, pGAl (SEQ ID NO: 1 of PCT/US2011/025422) or derivatives thereof (e.g., SEQ ID NOs:2 and 3 of PCT/US2011/025422), or pGA2 (SEQ ID NO:4 of PCT/US2011/025422) or derivatives thereof (e.g., SEQ ID NOs:5 and 6 of PCT/US2011/025422).
  • pGAl SEQ ID NO: 1 of PCT/US2011/025422
  • pGA2 SEQ ID NO:4 of PCT/US2011/025422
  • derivatives thereof e.g., SEQ ID NOs:5 and 6 of PCT/US2011/025422.
  • pGAl is a 3897 bp plasmid that includes a promoter (bp 1-690), the CMV-intron A (bp 691-1638), a synthetic mimic of the tPA leader sequence (bp 1659 - 1721), the bovine growth hormone polyadenylation sequence (bpl761-1983), the lambda TO terminator (bp 1984-2018), the kanamycin resistance gene (bp 2037-2830) and the ColEI origin of replication (bp 2831-3890).
  • the DNA sequence of the pGAl construct is provided in SEQ ID NO:l of PCT/US2011/025422. The indicated restriction sites are useful for cloning antigen- encoding sequences.
  • the Cla I or BspD I sites are used when the 5 ' end of a vaccine insert is cloned upstream of the tPA leader.
  • the Nhe I site is used for cloning a sequence in frame with the tPA leader sequence.
  • the sites listed between Sma I and Bin I are used for cloning the 3' terminus of an antigen-encoding sequence.
  • pGA2 is a 2947 bp plasmid lacking the 947 bp of intron A sequences found in pGAl .
  • pGA2 is the same as pGAl , except for the deletion of intron A sequences.
  • pGA2 is valuable for cloning sequences which do not require an upstream intron for efficient expression, or for cloning sequences in which an upstream intron might interfere with the pattern of splicing needed for good expression.
  • a schematic map of pGA2 with useful restriction sites for cloning vaccine inserts and the DNA sequence of pGA2 is provided in SEQ ID NO:2 of
  • pGA2.1 and pGA2.2 are multiple cloning site derivatives of pGA2.
  • the DNA sequence of pGA2.1 is provided in SEQ ID NO:5 of PCT/US2011/025422 and the sequence of pGA2.2 is provided in SEQ ID NO:6 of
  • Plasmids having "backbone" sequences that differ from those disclosed herein are also within the scope of the disclosure so long as the plasmids retain substantially all of the characteristics necessary to be therapeutically effective (e.g., one can substitute nucleotides, add nucleotides, or delete nucleotides so long as the plasmid, when administered to a patient reactivates the expression of latent proviral DNA.
  • the plasmids retain substantially all of the characteristics necessary to be therapeutically effective (e.g., one can substitute nucleotides, add nucleotides, or delete nucleotides so long as the plasmid, when administered to a patient reactivates the expression of latent proviral DNA.
  • 1-10, 11-20, 21-30, 31-40, 41- 50, 51-60, 61-70, 71-80, 81-90, 91-100, or more than 100 nucleotides can be deleted or replaced.
  • the encoded antigens can be of any HIV clade or subtype or any recombinant form thereof.
  • different clades exhibit isolate diversity, with each isolate within a clade having overall similar diversity from the consensus sequence for the clade (see, e.g., Subbarao et al, AIDS 10(Suppl A) :S 13-23, 1996).
  • most isolates can be used as a reasonable representative of sequences for other isolates of the same clade.
  • nucleic acid shock agents described herein can be practiced with natural variants of genes or nucleic acid molecules that result from recombination events, alternative splicing, or mutations (these variants may be referred to herein simply as "recombinant forms" of HIV).
  • the shock agent is an HIV DNA vaccine including:
  • a eukaryotic transcription cassette comprising a vaccine insert encoding one or more immunogens selected from the group consisting of HIV Gag, Pol, Tat, Rev, Vpu and Env, wherein the insert sequence encoding HIV Gag comprises one or more mutations that disable the zinc fingers and wherein the reverse transcriptase activity of HIV pol is inactivated and the protease activity is inactivated.
  • vectors of the disclosure include plasmids encoding a Gag protein (e.g., a Gag protein in which one or both of the zinc fingers have been inactivated); a Pol protein (e.g., a Pol protein in which integrase, RT, and/or protease activities have been inhibited); a Vpu protein (which may be encoded by a sequence having a mutant start codon); and Env, Tat, and/or Rev proteins (in a wild type or mutant form).
  • a Gag protein e.g., a Gag protein in which one or both of the zinc fingers have been inactivated
  • a Pol protein e.g., a Pol protein in which integrase, RT, and/or protease activities have been inhibited
  • Vpu protein which may be encoded by a sequence having a mutant start codon
  • Env, Tat, and/or Rev proteins in a wild type or mutant form
  • plasmids encoding the antigens just described can be combined with (e.g., mixed with) other plasmids that encode antigens obtained from, or derived from, a different HIV clade (or subtype or recombinant form thereof).
  • the inserts per se are also within the scope of the disclosure.
  • compositions include the following.
  • the antigen can be a wild type or mutant gag sequence (e.g., a gag sequence having a mutation in one or more of the sequences encoding a zinc finger at one or more of the cysteine residues corresponding to clade B HIV positions 392, 395, 413, or 416 to another residue (e.g., serine) or the mutation can change one or more of the cysteine residues
  • the one or more immunogens includes HIV Gag and wherein the
  • HIV gag mutations consist of amino acid mutations corresponding to C392S, C395S, C413S, and C416S of Clade B HIV strain HXB2 that disable the zinc fingers.
  • compositions used in the methods described can also include a vector (e.g., a plasmid vector) encoding: (a) a Gag protein in which one or both zinc fingers have been inactivated; (b) a Pol protein in which (i) the integrase activity has been inhibited by deletion of some or all of the pol sequence, (ii) the polymerase, strand transfer, and/or R ase H activity of reverse
  • a vector e.g., a plasmid vector
  • a vector e.g., a plasmid vector
  • proteolytic activity can be inhibited by introducing a mutation at positions 1641 - 1643 of SEQ ID NO : 8 of WO2011103417 or at an analogous position in the sequence of another HIV clade.
  • the reverse transcriptase of Pol comprises amino acid mutations corresponding to D185N, W266T and E478Q of HIV strain HXB2 that inactivate reverse transcriptase activity.
  • the one or more immunogens includes HIV Gag and wherein the active site of the protease was mutated to limit proteolytic cleavage of viral Gag proteins and the maturation of viral particles.
  • the protease mutation corresponds to D25A of Clade B HIV strain HXB2.
  • the HIV DNA vaccine includes ADA Env, Tat, Rev and Vpu, wherein the Bglll fragment in the ADA Env is deleted leaving tat, rev and vpu coding regions intact.
  • the HIV DNA vaccine includes one or more immunogens that are clade B HIV proteins.
  • the HIV DNA vaccine includes clade B HIV proteins, HIV
  • the vector lacks LTRs, integrase, vif, and vpr sequences of HIV.
  • the HIV DNA vaccine includes cytomegalovirus immediate early promoter (CMVIE) and a polyadenylation sequence.
  • CMVIE cytomegalovirus immediate early promoter
  • the HIV DNA vaccine includes intron A of the CMV promoter. In another embodiment, the HIV DNA vaccine includes a leader sequence.
  • the leader sequence is a polyadenylation sequence which is a bovine growth hormone polyadenylation sequence or a rabbit beta globin polyadenylation sequence.
  • expression of the HIV DNA vaccine forms immature virus- like particles (VLPs) that bud from the plasma membrane of DNA-expressing cells.
  • VLPs virus-like particles
  • HIV DNA vaccine insert encoding these above mentioned clade B zinc finger, reverse transcriptase and protease mutations and is designated herein as a "JS7 insert".
  • An HIV DNA vaccine containing this JS7 insert in plasmid pGA2 is referred to as "GEO-D02".
  • At least one of the two or more sequences can be mutant or mutated so as to limit the encapsidation of viral RNA (preferably, the mutation(s) limit encapsidation appreciably).
  • antigens that remain well expressed e.g., antigens that are expressed about as well as or better than their wild type counterparts, but are less biologically active than their wild type counterparts, are within the scope of the disclosure.
  • Techniques are also available for assessing the immune response. One can, for example, detect anti-viral antibodies or virus-specific T cells.
  • the mutant vectors or vaccine inserts provided result in an increase (e.g., at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 30%, 40%, or 50%) in the avidity of immunogen-specific antibodies, an increase (e.g., by at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 100-fold, 200-fold, 300- fold, 400-fold, or 500-fold) in immunogen-specific antibody titers, an increase (e.g., at least 1- fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11-fold, 12-fold, 13- fold, 14-fold, or 15-fold) in an increase (
  • the mutant constructs can include sequences encoding one or more of the substitution mutants described herein (see, e.g. the Examples) or an analogous mutation in another HIV clade.
  • HIV antigens can be rendered less active by deleting part of the gene sequences that encode them.
  • the compositions of the disclosure can include constructs that encode antigens that, while capable of eliciting an immune response, are mutant (whether encoding a protein of a different length or content than a corresponding wild type sequence) and thereby less able to carry out their normal biological function when expressed in a patient.
  • expression, immunogenicity, and activity can be assessed using standard techniques in molecular biology and immunology.
  • the vectors of the present disclosure can be administered with an adjuvant, including a genetic adjuvant.
  • the nucleic acid vectors regardless of the antigen they express, can optionally include such genetic adjuvants as GM-CSF, IL-15, IL-2, interferon response factors, secreted forms of flt-3, CD40 ligand and mutated caspase genes.
  • Genetic adjuvants can also be supplied in the form of fusion proteins, for example by fusing one or more C3d gene sequences (e.g., 1-3 (or more) C3d gene sequences) to an expressed antigen.
  • the DNA vectors express HIV-1 antigens and GM-CSF, and those constructs can be administered to subjects as described herein.
  • the GM-CSF sequence can be introduced into a variety of different DNA vectors expressing HIV-1 antigens. HIV inserts such as those described herein, and in US-2003-0175292-A1 are particularly useful. Any plasmid within the scope of the disclosure can be tested for expression by transfecting cells, such as 293T cells (a human embryonic kidney cell line) and assessing the level of antigen expression (by, for example, an antigen-capture ELISA or a Western blot).
  • the GM-CSF sequence included in the vectors and the vaccine inserts may be a full- length human GM-CSF or may be a polypeptide that includes a sequence that is at least 95% identical to GM-CSF and has one or more (e.g., two or three) biological activities of GM-CSF (e.g., capable of stimulating macrophage differentiation and proliferation, or activating antigen presenting dendritic cells).
  • the GM-CSF may include one or more mutations (e.g., one or more (e.g., at least two, three, four, five, or six) amino acid substitutions, deletions, or additions)).
  • any mutant GM-CSF proteins also have one or more (e.g., two or three) biological activities of GM-CSF (as described above).
  • Assays for the measurement of the biological activity of GM-CSF proteins are known in the art (see, e.g., U.S. Patent No. 7,371,370; incorporated herein by reference in its entirety).
  • the nucleic acid vectors of the disclosure encode GM-CSF and at least one antigen (which may also be referred to as an immunogen) obtained from, or derived from, any HIV clade or isolate (i.e., any subtype or recombinant form of HIV).
  • the antigen (or immunogen) may be: a structural component of an HIV virus; glycosylated, myristoylated, phosphorylated, or otherwise post-translationally modified; one that is expressed intracellularly, on the cell surface, or secreted (antigens that are not normally secreted may be linked to a signal sequence that directs secretion).
  • the antigen can be all, or an antigenic portion of, Gag, , Pol, Env (e.g., gpl60 or gpl20, or a CCR5-using Env), Tat, Rev, Vpu, Nef, Vif, Vpr, or a VLP (e.g., a polypeptide derived from a VLP that is capable of forming a VLP, including an Env-defective HIV VLP).
  • Env e.g., gpl60 or gpl20, or a CCR5-using Env
  • Tat Rev
  • Vpu Nef
  • Vif Vif
  • Vpr or a VLP (e.g., a polypeptide derived from a VLP that is capable of forming a VLP, including an Env-defective HIV VLP).
  • a VLP e.g., a polypeptide derived from a VLP that is capable of forming a VLP, including an Env
  • the HIV DNA vaccine contains the above-described JS7 insert and also a sequence encoding GM-CSF as described in WO2011103417.
  • This HIV DNA vaccine is also designated "GEO-D03". Multiclade Nucleic Acid Vaccines
  • composition includes either a vector with an insert or an insert alone, and that insert encodes multiple protein antigens
  • one of the antigens can be a wild type or mutant gag sequence, including those described above.
  • a composition includes more than one type of vector or more than one type of insert, at least one of the vectors or inserts (whether encoding a single antigen or multiple antigens) can include a wild type or mutant gag sequence, including those described above or analogous sequences from other HIV clades.
  • the vaccine insert in either or both vectors can encode gag; where both vectors encode gag, the gag sequence in the first vector can be from one HIV clade (e.g., clade B) and that in the second vector can be from another HIV clade (e.g., clade C).
  • the antigen can be wild type or mutant Pol.
  • the sequence can be mutated by deleting or replacing one or more nucleic acids, and those deletions or substitutions can result in a Pol gene product that has less enzymatic activity than its wild type counterpart
  • composition includes either a vector with an insert or an insert alone, and that insert encodes multiple protein antigens
  • one of the antigens can be a wild type or mutant pol sequence, including those described above (these multi-protein-encoding inserts can also encode the wild type or mutant gag sequences described above).
  • compositions include more than one type of vector or more than one type of insert
  • at least one of the vectors or inserts can include a wild type or mutant pol sequence, including those described above (and, optionally, a wild type or mutant gag sequence, including those described above (i.e., the inserts can encode Gag-Pol).
  • the vaccine insert in either or both vectors can encode Pol; where both vectors encode Pol, the Pol sequence in the first vector can be from one HIV clade (e.g., clade B) and that in the second vector can be from another HIV clade (e.g., clade A or G).
  • the Pol sequence in the first vector can be from one HIV clade (e.g., clade B) and that in the second vector can be from another HIV clade (e.g., clade A or G).
  • another portion of the pol sequence that can optionally be altered is the sequence encoding the protease activity (regardless of whether or not sequences affecting other enzymatic activities of Pol have been altered).
  • the antigen can be a wild type or mutant Env, Tat, Rev, Nef, Vif, Vpr, or Vpu.
  • the composition includes either a vector with an insert or an insert alone, and that insert encodes multiple protein antigens
  • one of the antigens can be a wild type or mutant Env.
  • multi-protein expressing inserts can encode wild type or mutant Gag-Pol and Env; they can also encode wild type or mutant Gag-Pol and Env and one or more of Tat, Rev, Nef, Vif, Vpr, or Vpu (each of which can be wild type or mutant).
  • Env, Tat, Rev, Nef, Vif, Vpr, or Vpu can be mutant by virtue of a deletion, addition, or substitution of one or more amino acid residues (e.g., any of these antigens can include a point mutation).
  • one or more amino acids can be deleted from the gpl20 surface and/or gp41 transmembrane cleavage products of Env.
  • one or more amino acids can be deleted from one or more of: the matrix protein (p7), the capsid protein (p24), the nucleocapsid protein (p7) and the C-terminal peptide (p6).
  • amino acids in one or more of these regions can be deleted (this may be especially desired where the vector is a viral vector, such as MVA).
  • one or more amino acids can be deleted from the protease protein (plO), the reverse transcriptase protein (p66/p51), or the integrase protein (p32).
  • compositions of the disclosure can include a vector (e.g., a plasmid or viral vector) that encodes: (a) a Gag protein in which one or more of the zinc fingers has been inactivated to limit the packaging of viral R A; (b) a Pol protein in which (i) the integrase activity has been inhibited by deletion of some or all of the pol sequence and (ii) the polymerase, strand transfer, and/or RNase H activity of reverse transcriptase has been inhibited by one or more point mutations within the pol sequence; and (c) Env, Tat, Rev, and Vpu, with or without mutations.
  • a vector e.g., a plasmid or viral vector
  • the encoded proteins can be obtained or derived from a subtype A, B or C HIV (e.g., HIV-1) or recombinant forms thereof.
  • the compositions include non- identical vectors
  • the sequence in each type of vector can be from a different HIV clade (or subtype or recombinant form thereof).
  • the disclosure features compositions that include plasmid vectors encoding the antigens just described (Gag-Pol, Env etc.), where some of the plasmids include antigens that are obtained from, or derived from, one clade and other plasmids include antigens that are obtained (or derived) from another clade. Mixtures representing two, three, four, five, six, or more clades (including all clades) are within the scope of the disclosure.
  • first and second vectors are included in a composition
  • either vector can be pGAl/JS2, pGAl/JS7, pGAl/JS7.1 , pGA2/JS2, pGA2/JS7, pGA2/JS7.1 (pGAl .1, pGAl .2 or the pGA vectors with other permutations in restrictions sites used for addition of vaccine inserts can be used in place of pGAl, and pGA2.1 or pGA2.2 can be used in place of pGA2).
  • either vector can be pGAl/IC25, pGAl/IC2, pGAl/IC48, pGAl/IC90, pGA2/IC25, pGA2/IC2, pGA2/IC48, or pGA2/IC90 (here again, pGAl . 1 or pGA1.2 can be used in place of pGAl, and pGA2.1 or pGA2.2 can be used in place of pGA2).
  • the encoded proteins can be those of, or those derived from, a subtype C HIV (e.g.,, HIV1) or a recombinant form thereof.
  • the vector can be pGAl/IN2, pGAl .
  • the encoded proteins can also be those of, or those derived from, any of HIV clades (or subtypes) E, F, G, H, I, J, K or L or recombinant forms thereof.
  • An HIV-1 classification system has been published by Los Alamos National Laboratory (HIV Sequence Compendium-2001 , Kuiken ct al, published by Theoretical Biology and Biophysics Group T-10, Los Alamos, NM, (2001 )), more recent HIV sequences are available on the Los Alamos HIV sequence database website.
  • the plasmids can contain the inserts such as JS7, IC25, and IN3 referenced in WO2003076591 and shown in Table 1.
  • plasmids encoding the antigens just described can be combined with (e.g., mixed with) other plasmids that encode antigens obtained from, or derived from, a different HIV clade (or subtype or recombinant form thereof).
  • the inserts per se are also within the scope of the disclosure.
  • the inserts may contain sequences that encode one or more conserved protein sequences and/or may contain one or more designer sequences (e.g., mosaic sequences that contain a sequence from one or more HIV clades).
  • plasmids described above can be administered to any subject as shock agents.
  • plasmids or other vectors that express an IN series of clade C HIV-1 sequences can be administered as a shock agent to a subject who has been infected with an HIV of clade C or another clade.
  • vectors expressing antigens of various clades can be combined to elicit an immune response against more than one clade (this can be achieved whether one vector expresses multiple antigens, or mosaic or conserved element antigens from different clades or multiple vectors express single antigens from different clades), one can tailor the vaccine formulation to best protect a given subject.
  • Useful plasmids may or may not contain a terminator sequence that substantially inhibits transcription (the process by which R A molecules are formed upon DNA templates by complementary base pairing).
  • Useful terminator sequences include the lambda TO terminator and functional fragments or variants thereof. The terminator sequence is positioned within the vector in the same orientation and at the C terminus of any open reading frame that is expressed in prokaryotes (i.e., the terminator sequence and the open reading frame are operably linked).
  • Selectable marker genes are known in the art and include, for example, genes encoding proteins that confer antibiotic resistance on a cell in which the marker is expressed (e.g., resistance to kanamycin, ampicillin, or penicillin).
  • the selectable marker is so-named because it allows one to select cells by virtue of their survival under conditions that, absent the marker, would destroy them.
  • the selectable marker, the terminator sequence, or both (or parts of each or both) can be, but need not be, excised from the plasmid before it is administered to a patient.
  • plasmid vectors can be administered in a circular form, after being linearized by digestion with a restriction endonuclease, or after some of the vector "backbone" has been altered or deleted.
  • the nucleic acid vectors can also include an origin of replication (e.g., a prokaryotic origin of replication) and a transcription cassette that, in addition to containing one or more restriction endonuclease sites, into which an antigen-encoding insert can be cloned, optionally includes a promoter sequence and a polyadenylation signal.
  • an origin of replication e.g., a prokaryotic origin of replication
  • a transcription cassette that, in addition to containing one or more restriction endonuclease sites, into which an antigen-encoding insert can be cloned, optionally includes a promoter sequence and a polyadenylation signal.
  • Promoters known as strong promoters can be used and may be preferred.
  • One such promoter is the cytomegalovirus (CMV) intermediate early promoter, although other (including weaker) promoters may be used without departing from the scope of the present disclosure.
  • CMV cytomegalovirus
  • strong polyadenylation signals may be selected (e.g., the signal derived from a bovine growth hormone (BGH) encoding gene, or a rabbit ⁇ globin polyadenylation signal (Bohm et al, J. Immunol. Methods 193:29-40, 1996; Chapman et al, Nucl. Acids Res. 19:3979- 3986, 1991; Hartikka et al, Hum. Gene Therapy 7: 1205-1217, 1996; Manthorpe et al, Hum. Gene Therapy 4:419-431, 1993; Montgomery et al, DNA Cell Biol. 12:777-783, 1993)).
  • BGH bovine growth hormone
  • the vectors can further include a leader sequence (a leader sequence that is a synthetic homolog of the tissue plasminogen activator gene leader sequence (tPA) is optional in the transcription cassette) and/or an intron sequence, such as a cytomegalovirus (CMV) intron A or an SV40 intron.
  • a leader sequence a leader sequence that is a synthetic homolog of the tissue plasminogen activator gene leader sequence (tPA) is optional in the transcription cassette
  • an intron sequence such as a cytomegalovirus (CMV) intron A or an SV40 intron.
  • CMV cytomegalovirus
  • intron A increases the expression of many antigens from RNA viruses, bacteria, and parasites, presumably by providing the expressed RNA with sequences that support processing and function as a eukaryotic mRNA.
  • Multi- cistronic vectors may be used to express more than one immunogen or an immunogen and an immunostimulatory protein (Iwasaki et al., J. Immunol. 158:4591-4601, 1997a; Wild et al, Vaccine 16:353-360, 1998).
  • vectors encoding one or more antigens from one or more HIV clades or isolates may, but do not necessarily, include a leader sequence and an intron (e.g., the CMV intron A).
  • the vectors of the present disclosure differ in the sites that can be used for accepting antigen-encoding sequences and in whether the transcription cassette includes intron A sequences in the CMVIE promoter. Accordingly, one of ordinary skill in the art may modify the insertion site(s) or cloning site(s) within the plasmid without departing from the scope of the disclosure. Both intron A and the tPA leader sequence have been shown in certain instances to enhance antigen expression (Chapman et al, Nucleic Acids Research 19:3979-3986, 1991).
  • the shock agent can include any viral or bacterial vector that includes an insert described herein.
  • the disclosure also encompasses administration of at least two (e.g., three, four, five, or six) vectors (e.g., plasmid or viral vectors that contain the same vaccine insert (i.e., an insert encoding the same antigens).
  • the patient may receive two types of vectors, and each of those vectors can elicit an immune response against an HIV of a different clade.
  • the disclosure features methods in which a patient receives a composition that includes (a) a first vector comprising a vaccine insert encoding one or more antigens that elicit an immune response against a human
  • immunodeficiency virus of a first subtype or recombinant form and (b) a second vector comprising a vaccine insert encoding one or more antigens that elicit an immune response against an HIV of a second subtype or recombinant form.
  • the first and second vectors can be any of those described herein.
  • the inserts in the first and second vectors can be any of those described herein.
  • the immunodeficiency virus vaccine inserts of the present disclosure were designed to generate non-infectious VLPs (a term that can encompass true VLPs as well as aggregates of viral proteins) from a single DNA. This was achieved using the subgenomic splicing elements normally used by immunodeficiency viruses to express multiple gene products from a single viral R A.
  • the subgenomic splicing patterns are influenced by (i) splice sites and acceptors present in full length viral RNA, (ii) the Rev responsive element (RRE) and (iii) the Rev protein.
  • the splice sites in retroviral RNAs use the canonical sequences for splice sites in eukaryotic mRNAs.
  • the RRE is an approximately 200 bp RNA structure that interacts with the Rev protein to allow transport of viral RNAs from the nucleus to the cytoplasm.
  • the approximately 10 kb RNA of immunodeficiency virus mostly undergoes splicing to the mRNAs for the regulatory genes Tat, Rev, and Nef. These genes are encoded by exons present between RT and Env and at the 3' end of the genome.
  • the singly spliced mRNA for Env and the unspliced mRNA for Gag and Pol are expressed in addition to the multiply spliced mRNAs for Tat, Rev, and Nef.
  • non-infectious VLPs from a single DNA affords a number of advantages to an immunodeficiency virus vaccine.
  • the expression of a number of proteins from a single DNA affords the vaccinated host the opportunity to respond to the breadth of T- and B- cell epitopes encompassed in these proteins.
  • the expression of proteins containing multiple epitopes allows epitope presentation by diverse histocompatibility types. By using whole proteins, one offers hosts of different histocompatibility types the opportunity to raise broad- based T cell responses. This may be essential for the effective containment of immunodeficiency virus infections, whose high mutation rate supports ready escape from immune responses (Evans et al, Nat. Med.
  • Immunogens can also be engineered to be more or less effective for raising antibody or T cells by targeting the expressed antigen to specific cellular compartments. For example, antibody responses are raised more effectively by antigens that are displayed on the plasma membrane of cells, or secreted therefrom, than by antigens that are localized to the interior of cells (Boyle et al, Int. Immunol. 9: 1897-1906, 1997; Inchauspe et al, DNA Cell. Biol. 16: 185-195, 1997). Tc responses may be enhanced by using N-terminal ubiquitination signals which target the DNA encoded protein to the proteosome causing rapid cytoplasmic degradation and more efficient peptide loading into the MHC I pathway (Rodriguez et al, J.
  • the shock agent is a CpG oligodeoxynucleotide (CpG ODN), also referred to as a CpG oligonucleotide.
  • CpG ODN CpG oligodeoxynucleotide
  • TLR9 Toll-like receptor 9 senses unmethylated CpG dinucleotides, a hallmark of microbial DNA, that can be mimicked by synthetic oligonucleotides containing CpG motifs (CpG ODNs).
  • the CpG oligonucleotide is co- administered with an HIV DNA vaccine as the shock agent.
  • TLR9 stimulation by CpG DNA or CpG ODNs triggers intracellular signaling leading to the activation of macrophages, dendritic cells (DC) and B cells, and the production of cytokines, chemokines, and immunoglobulins. Subsequently, cytokines produced by DC, such as IL-12, induce the differentiation of naive T cells into T helper 1 (Thl) and cytotoxic T-cells (CTL).
  • CpG ODNs are short synthetic single-stranded DNA molecules containing unmethylated
  • CpG dinucleotides in particular sequence contexts (CpG motifs).
  • CpG ODNs may possess a partially or completely phosphorothioated (PS) backbone, as opposed to the natural phosphodiester (PO) backbone found in genomic bacterial DNA.
  • PS phosphorothioated
  • PO phosphodiester
  • Three major classes of stimulatory CpG ODNs have been identified based on structural characteristics and activity on human peripheral blood mononuclear cells (PBMCs), in particular B cells and plasmacytoid dendritic cells (pDCs). These three classes are Class A (Type D), Class B (Type K) and Class C.
  • PBMCs peripheral blood mononuclear cells
  • pDCs plasmacytoid dendritic cells
  • CpG-A ODNs are characterized by a PO central CpG-containing palindromic motif and a
  • PS-modified 3' poly-G string induce high IFN-a production from pDCs but are weak stimulators of TLR9-dependent NF- ⁇ signaling and pro-inflammatory cytokine (e.g. IL-6) production.
  • CpG-B ODNs contain a full PS backbone with one or more CpG dinucleotides. They strongly activate B cells and TLR9-dependent NF- ⁇ signaling but weakly stimulate IFN-a secretion.
  • CpG-C ODNs combine features of both classes A and B. They contain a complete PS backbone and a CpG-containing palindromic motif. C-Class CpG ODNs induce strong IFN-a production from pDC as well as B cell stimulation.
  • the CpG oligonucleotide is CpG 7909.
  • CpG 7909 is an immunomodulating synthetic oligonucleotide designed to specifically activate the Toll-like receptor 9 (TLR9).
  • CpG 7909 acting through the TLR9 receptor present in B cells and plasmacytoid dendritic cells, stimulates human B-cell proliferation, enhances antigen-specific antibody production and induces interferon-alpha production, interleukin-10 secretion and natural killer cell activity.
  • the CpG ODN is CpG 2006.
  • CpG 2006 is a Class B synthetic oligonucleotide, also known to specifically activate the Toll-like receptor 9 (TLR9).
  • the CpG ODN is another synthetic oligonucleotide, for example CpG ODN 1585, CpG ODN 2118, CpG ODN 1826, or CpG 1668.
  • the shock agent is an HDAC inhibitor.
  • the HDAC inhibitor is a Class I HDAC inhibitor. In another embodiment, the Class I HDAC inhibitor is selected from HDAC1, HDAC 2, HDAC 3 and HDAC 8. In one embodiment the HDAC inhibitor is a Class II HDAC inhibitor. In another embodiment, the Class II HDAC inhibitor is selected from HDAC4, HDAC 5, HDAC 6, HDAC 7, HDAC9 and HDAC 10.
  • the HDAC inhibitor is a Class III HDAC inhibitor, also known as the sirtuins.
  • the Class III HDAC inhibitor is selected from SIRTl, SIRT 2, SIRT 3, SIRT 4, SIRT 5, SIRT 6, and SIRT 7.
  • the HDAC inhibitor is a Class IV HDAC inhibitor.
  • the Class IV HDAC inhibitor is HDAC11.
  • the HDAC inhibitor is selected from Romidepsin (Istodax, depsipeptide, NSC 630176, FK228 or FR901228), Panobinostat (LBH589), Valproic acid,
  • the shock agent is an NFkappaB activator such as, but not limited to, phorbol esters and TNF-a.
  • the shock agent is a cytosine methylation inhibitor such as, but not limited to 5-azacytidine.
  • the shock agent is an acetaldehyde dehydrogenase inhibitor.
  • the acetaldehyde dehydrogenase inhibitor is selected from 4-amino-
  • Cyanamide Daidzin (antioxidant isoflavone), 4-(Diethylamino)benzaldehyde, Disulfiram, Gossypol, Kynurenine metabolites, Molinate, Nitroglycerin, or Pargyline.
  • the acetaldehyde dehydrogenase inhibitor is disulfiram.
  • combinations of the above described shock agents may be used to optimize the reliability of reservoir activation.
  • the HIV DNA vaccine may be combined with one or more of a CpG oligonucleotide, a TLR9 receptor agonist, an HDAC inhibitor, an NFkappaB activator, a cytosine methylation inhibitor, or an acetaldehyde dehydrogenase inhibitor.
  • a CpG oligodeoxynucleotide can be used in combination with the HIV DNA vaccine to shock viral reservoirs.
  • kill agent refers to a composition useful in killing, destroying, eradicating, or lysing virus-expressing cells.
  • kill agents target and destroy reactivated CD4+ HIV reservoirs.
  • the kill agent may be any suitable agent, such as a viral vector, viral vaccine or antibody, e.g., monoclonal antibody or recombinant antibody.
  • the HIV vaccine is a HIV vaccine that elicits a T cell immune response.
  • Methods of measuring T cell immune response are known in the art.
  • the HIV vaccine is selected from an MVA vaccine, an adenoviral vaccine, or a vesicular stomatitis virus (VSV) vaccine.
  • VSV vesicular stomatitis virus
  • the HIV vaccine that may be used as the kill agent of the present methods is a modified vaccinia Ankara (MVA) vaccine.
  • MVA HIV vaccines are known in the art and have been described in international patent applications WO2002/072754 and WO/2006/026667.
  • MVA has been particularly effective as a vaccine in mouse models (Schneider et al., Nat. Med. 4:397-402, 1998). MVA is a highly attenuated strain of vaccinia virus that was developed toward the end of the campaign for the eradication of smallpox, and it has been safety tested in more than 100,000 people (Mahnel et al, Berl. Munch Tier GmbHl Klischr 107:253-256, 1994; Mayr et al, Monbl. Bakteriol. 167:375-390, 1978). During over 500 passages in chicken cells, MVA lost about 10% of its genome and the ability to replicate efficiently in primate cells.
  • MVA has proved to be a highly effective expression vector (Sutter et al, Proc. Natl. Acad. Sci. U.S.A. 89: 10847-10851,1992), raising protective immune responses in primates for parainfluenza virus (Durbin et al. J. Infect. Dis. 179: 1345-1351, 1999), measles (Stittelaar et al. J. Virol. 74:4236-4243, 2000), and immunodeficiency viruses (Barouch et al, J. Virol. 75:5151-5158, 2001; Ourmanov et al, J. Virol. 74:2740-2751, 2000; Amara et al, J.
  • Vaccinia viruses have been used to engineer viral vectors for recombinant gene expression and as recombinant live vaccines (Mackett et al, Proc. Natl. Acad. Sci. U.S.A.
  • DNA sequences which may encode any of the HIV antigens described herein, can be introduced into the genomes of vaccinia viruses. If the gene is integrated at a site in the vector DNA that is nonessential for the life cycle of the virus, it is possible for the newly produced recombinant vaccinia virus to be infectious (i.e., able to infect cells) and to express the integrated DNA sequences.
  • the viral vectors featured in the compositions and methods of the present disclosure are highly attenuated. Several attenuated strains of vaccinia virus were developed to avoid undesired side effects of smallpox vaccination.
  • the modified vaccinia Ankara (MVA) virus was generated by long-term serial passages of the Ankara strain of vaccinia virus on chicken embryo fibroblasts (CVA; see Mayr et al, Infection 3:6-14, 1975).
  • the MVA virus is publicly available from the American Type Culture Collection (ATCC; No. VR-1508; Manassas, VA).
  • ATCC American Type Culture Collection
  • VA Manassas
  • the desirable properties of the MVA strain have been demonstrated in clinical trials (Mayr et al, Monbl. Bakteriol. 167:375-390, 1978; Stickl et al, Dtsch. Med. Wschr. 99:2386-2392, 1974; see also, Sutter and Moss, Proc. Natl. Acad. Sci. U.S.A.
  • the MVA vectors can be prepared as follows.
  • Insertions may also be introduced into naturally-occurred deletions with modified deletion sites (e.g., restructured deletion III, but not only) to enhance stability of the insertion or introduced between essential genes using sequences flanking the insertion site.
  • modified deletion sites e.g., restructured deletion III, but not only
  • I18R/G1LI18G1 see, for e.g., Wyatt et al,
  • the recombinant vaccinia virus can be isolated by methods known in the art (isolation can be facilitated by use of a detection marker).
  • the DNA to be inserted can be linear or circular (e.g., a plasmid, linearized plasmid, gene, gene fragment, or modified HIV genome).
  • the foreign DNA sequence is inserted between the sequences flanking the naturally-occurring deletion, between the sequences of a modified naturally occurring deletion, or between the sequences marking the boundaries of two essential genes.
  • the sequence can include regulatory sequences (e.g., a promoter, such as the promoter of the vaccinia 11 kDa gene or the 7.5 kDa gene or modified H5).
  • regulatory sequences e.g., a promoter, such as the promoter of the vaccinia 11 kDa gene or the 7.5 kDa gene or modified H5.
  • the DNA construct can be introduced into MVA-infected cells by a variety of methods, including calcium phosphate-assisted transfection (Graham et al, Virol. 52:456-467,
  • Suitable inserts include Gag-Pol and Env sequences from the JS7, IC25 and IN3 inserts described above in Table 1.
  • potential kill agents are antibodies, including for example, neutralizing or non- neutralizing antibodies.
  • the kill agent is a monoclonal antibody or recombinant antibody.
  • the kill agent is a human monoclonal or recombinant antibody.
  • Non-neutralizing antibodies kill by mobilizing innate immune responses such as antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and Complement-mediated (C '-mediated) lysis. While not to be bound by any specific mechanism, it is believed that ADCC-mediating antibodies recognize HIV-1 antigens expressed on the membrane of infected cells and bind the Fey receptor (FcR) of the effector cell population to induce ADCC.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • C '-mediated Complement-mediated
  • Neutralizing Ab can elicit innate immune responses to kill viral reservoirs.
  • the kill agent is a neutralizing Ab such as 10-1074, 10E8, 17B, 2F5,
  • the kill agent is a non-neutralizing antibody such as 120-16, 126-50, 246-D, 4B3, 50-69, 98-43, 98-6, b6, 3D6, A32, or F240
  • the killing agent is an antibody to a gpl20 carbohydrate epitope of HIV.
  • this gpl20 carbohydrate epitope-binding antibody killing agent is any one of a number of broadly neutralizing MAb.
  • the Ab is A32 which is believed to bind to the CI region of gpl20.
  • the methods provided herein employ combination therapy with at least one antiretroviral agent, each of which is administered to the subject in a therapeutically effective amount.
  • ART is employed to prevent, or reduce spread of any reactivated HIV infection that could result from stimulation of viral reservoirs with shock/kill agents.
  • the antiretroviral agent is preferably administered prior to, or during the administration of shock kill agents.
  • the subject is undergoing a regular regimen of antiretroviral therapy prior to and during the administration of the shock and kill agents.
  • antiretroviral agents include any substance that can inhibit, reduce, or eliminate retroviral infection of a cell.
  • ART refers to any antiretroviral therapy including highly active antiretroviral therapy (HAART).
  • HAART is generally considered ART therapy that comprises at least 3 antiretro viral agents.
  • antiretro viral agents include, but are not limited to, the two classes known as reverse transcriptase inhibitors and protease inhibitors, as well as agents that are inhibitors of viral entry.
  • ART comprises the administration of therapeutically effective amounts of at least one reverse transcriptase inhibitor and at least one protease mhibitor in combination with at least one additional antiretroviral agent.
  • at least two reverse transcriptase inhibitors are administered in combination with at least one protease mhibitor.
  • at least two protease inhibitors are administered in combination with at least one reverse transcriptase mhibitor.
  • nucleoside analogs which are a class of compounds that are known to inhibit HIV, and non-nucleoside drugs.
  • Nucleoside analogs are exemplified by didanosine (2',3'-dideoxyinosine or [ddl], available as Videx® from Bristol Myers-Squibb, Waliiiigford, Conn,); zidovudine (3 " '-azido-2',3'-dideoxythymidine or azidothymidine [AZT], available from Glaxo- Wellcome Co,, Research Triangle Park, N,C); zaicitabine (2',3 '-dideoxycytidine [ddC], available as Hivid® from Hoffman-La Roche, Basel, Switzerland); lamivudine 2'-deoxy-3'-thiacytidine [3TC] (Epivir®, available from
  • nucleoside reverse transcriptase inhibitors include abacavir (1592U89, ZiagenTM, available from Glaxo- Wellcome Co.).
  • Non-nucleoside reverse transcriptase inhibitors include abacavir (1592U89, ZiagenTM, available from Glaxo- Wellcome Co.).
  • transcriptase inhibitors include nevirapine (ViramuneTM, available from Boehringer Ingeiheim Pharmaceuticals, Inc.); delaviridine (Rescriptor®, available from Pharmacia & Upjohn,
  • protease inhibitors useful in the present invention include, but are not limited to, Indinavir sulfate (available as CrixivanTM capsules from Merck & Co., Inc., West Point, Pa.), saquinavir (Invirase® and Fortovase®, available from Hoffmnan-La Roche), ritonavir (Norvir®, available from Abbott Laboratories, Abbott Park, 111.); ABT-378 (new name: lopinavir, available from Abbott Laboratories); Amprenavir (AgeneraseTM, available from Glaxo Wellcome, Inc.); and Nelfinavir (Viraeept®), and GWI 1 (available from Glaxo
  • reverse transcriptase and protease inhibitors are not intended to be limiting. It is recognized that any known inhibitor, such as novel integrase inhibitors, as well as those under development, may be used in the methods of the invention. See, for example, the drugs for HIV infection disclosed in Medical Letter 42(Jan, 10, 2000): 1-6, herein incorporated by reference.
  • Suitable human dosages for these compounds can var widely. However, such dosages can readily be determined by those of skill in the art.
  • Therapeutically effective amounts of these drugs are administered during ART.
  • therapeutically effective amount is intended an amount of the antiretroviral agent that is sufficient to decrease the effects of HIV infection, or an amount that is sufficient to favorably influence the pharmacokinetic profi le of one or more of the other antiretroviral agents used in the ART protocol.
  • vorably influence is intended that the antiretroviral agent, when administered in a therapeutically effective amount, affects the metabolism of one or more of the other antiretroviral agents used in ART, such that the bioavailability of the other agent or other agents is increased.
  • the therapeutically effective dose of an antiretroviral agent for purposes of having a favorable influence on the pharmacokinetics of another antiretroviral agent used in the ART protocol is typically lower than the amount to be administered to have a direct therapeutic effect on HIV, such as inhibition of HIV replication.
  • an antiretroviral agent that has undesirable adverse effects at the full dosage required for therapeutic effectiveness against HIV replication can provide a therapeutic benefit a lower doses with fewer adverse side effects.
  • an antiretroviral agent when administered in a therapeutically effective amount to an HIV-infected subject, decreases the effects of HIV infection by, for example, inhibiting replication of HIV, thereby decreasing viral load in the subject undergoing antiretroviral therapy.
  • an antiretroviral agent when administered in a therapeutically effective amount to an HIV-infected subject, favorably influences the
  • the protease inhibitor ritonavir when administered at full doses is a potent inhibitor of HIV in serum-and lymph nodes.
  • adverse reactions are common, such as gastrointestinal intolerance, hyperglycemia, insulin resistance, new onset or worsening diabetes, increased bleeding in hemophiliacs, circumoral and peripheral paresthesias, altered taste, and nausea and vomitmg.
  • Ritonavir can be administered at low doses (for example, 100 to 400 mg bid) with minimal intrinsic antiviral activity to increase the serum concentrations and decrease the dosage frequency of other protease inhibitors (see, Hsu et al. (1998) Clin. Pharmacokinet, 35:275). See, for example, the favorable influence of ritonavir on the protease inhibitor lopinavir (ABT-378) (Eron et al. (1999) ICAAC 39 addendum: 18, Abstract LB-20).
  • IDV can be administered at a dosage of about 800 mg, three times a day
  • D4T can be administered at a dosage of about 30-40 mg, twice a day
  • Nelfinavir can be administered at a dosage of about 1250 mg, twice a day, or 750 mg three times a day.
  • These agents are generally administered in oral formulations, though any suitable means of administration known in the art may be utilized for their delivery.
  • ART ' is administered to an HIV-infected subject to effectively reduce the pool of actively replicating virus to an undetectable amount in plasma samples collected from the subject.
  • undetectable amount in the plasma is intended the amount of actively replicating HIV is less than about 500 RNA molecules/mi, preferably less than about 400 RNA molecules/ml, more preferably less than about 300 RN A molecules/ml, still more preferably less than about 200 RNA molecules/ml, even more preferably less than about 100 RNA molecules/ml, most preferably less than about 50 RNA molecules/ml. Any method known to those skil led in the art may be utilized to measure viral load in the plasma, including the methods described elsewhere herein.
  • plasma viral load can be determined using a branched chain DNA assay (bDNA), which has a lower limit of detection (LLD) of 50 HIV RNA molecules/ml (see Jacobson et al. (1996) Proc. Natl. Acad. Sci. USA 93: 10405-10410; herein incorporated by reference).
  • bDNA branched chain DNA assay
  • LLD lower limit of detection
  • plasma viral RN A is said to be
  • compositions described herein are administered in a multi-step method to reduce or purge viral reservoirs for eradication by patient immune response. Stimulating and killing viral reservoirs under conditions that reduce or clear active virus out of the body provides a two-step approach for flushing or clearing viral reservoirs and preventing viral re-emergence. Slight increases in viral levels have been observed typically 1-2 weeks post DNA vaccinations in a clinical trial where combination DNA/MVA vaccines were administered in conjunction with anti-retro viral therapy (ART). Timing the DNA vaccine's effect of flushing HIV from the reservoirs as a "shock agent” and administration of a "kill agent” such as an antibody or viral vaccine in the presence of ART provides a method to reduce or eliminate viral reservoirs.
  • ART anti-retro viral therapy
  • a DNA vaccine acts as a "shock agent" and stimulates the reservoirs to express virus thereby making the infected cells susceptible to killing by the CD8+ T cells elicited by the killing agent.
  • the ART drugs reduce or prevent virus spread to uninfected cells.
  • use of other shocking and killing agents described herein alone or in combination provide alternate embodiments of this method.
  • the present invention is a method of reducing or eliminating viral reservoirs in an individual in need thereof by:
  • step (i) occurs before step (ii), i.e., the shock agent is administered before the kill agents.
  • the shock and kill agents are administered simultaneously or on the same day, i.e., within the same 24 hour period.
  • the kill agent in step (ii) is administered between about 1 and about 4 weeks, between about 1 and about 2 weeks, or about one week after the shock agent is administered in step (i).
  • the kill agent in step (ii) is administered between about 1 to about 10 days, about 1 to about 7 days, about 1 to about 5 days, about 1 to about 3 days, about 3 to about 7 days, about 5 to about 6 days, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days after
  • the shock agent is administered about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 14 hours, about 16 hours, about 18 hours, about 20 hours, or about 24 hours before the kill agent.
  • the reduction of the viral reservoirs is complete, i.e., the virus is eradicated or eliminated.
  • the method further comprises the step of: (iii) measuring the presence of viral reservoirs for reduction.
  • viral reservoirs are measured at about 1, about 2, about 3, about 4, about 5, about 6, about 7, or about 8 weeks after step (ii). In another embodiment, viral reservoirs are measured at about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, or about 12 months. In a further embodiment, viral reservoirs are measured about 1, about 2, about 3, about 4, or about 5 years after step (ii).
  • viral load is determined after administration of one or more shock agents as an indicator of reservoir activation.
  • the present invention is a method of reducing or eliminating viral reservoirs in a subject in need thereof, comprising:
  • Viral load may be determined, for example, about 2 hours, about 4 hours, about 6 hours, about 8 hours, about 10 hours, about 12 hours, about 14 hours, about 16 hours, about 18 hours, about 20 hours, about 22 hours or about 24 hours after administration of the shock agent.
  • viral load may be determined about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, or about 6 days after administration of the shock agent.
  • viral load may be determined about 1, about 2, about 3, about 4, about 5, about 6 or about 7 weeks or more after administration of the shock agent.
  • viral load is determined in a clinical setting, i.e., by a medical professional or support staff.
  • viral load is determined by the individual, and when peak viral load is detected the individual notifies the healthcare provider to obtain administration of the killing agent.
  • Methods and kits for determining viral load by an individual are known in the art.
  • the peak viral load occurs between about 2 and about 4 hours, about 4 and about 6 hours, about 6 and about 8 hours, about 8 and about 10 hours, about 10 and about 12 hours, about 12 and about 14 hour, about 14 and about 16 hours, about 16 and about 18 hours, about 18 and about 20 hours, about 20 and about 22 hours, or about 24 hours.
  • the peak viral load occurs between about 1 and about 4 weeks, about 1 and about 2 weeks, or about 1 week after administration of the shock agent.
  • the peak viral load occurs between about 1 and about 7 days, about 1 and about 5 days, or about 1 and about 3 days.
  • the peak viral load occurs between about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days.
  • the kill agent is administered at about the time of peak reservoir activation as measured by viral load.
  • the kill agents is administered within about 1, about 2, about 4, about 6, about 8, about 12, about 18, about 20 or about 24 hours of peak viral load.
  • the kill agent in step (iii) is administered about 1-10 day, 1-7 day, 1-
  • the kill agent in step (iii) is administered to the subject about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 10 weeks, about 12 weeks, about 16 weeks, about 20 weeks, or about 24 weeks after administration of shock agent in step (i) or determination of peak viral load in (ii).
  • the method further comprises the step of:
  • reservoirs are measured in (iv) at about 1, about 2, about 3, about 4, about 5, about 6, about 7, or about 8 weeks after step (iii). In another embodiment, reservoirs are measured in step (v) about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, or about 12 months after step (iii). In further embodiments, reservoirs in (iv) are measured at about 1, about 2, about 3, about 4, or about 5 years after step (iii). In one embodiment, the individual is undergoing antiretroviral therapy prior to, during, and after administration of shock and kill agents.
  • the antiretroviral agent is administered prior to and during the administration of shock and kill agents.
  • the shock agent in addition to flushing viral reservoirs, is also capable of priming an immune response.
  • the immune response is biased towards CD4+ T cells, and contributes to control by providing CD4+ T cell help for the CD8+ T cell response.
  • immunostimulatory effects of a vaccine shock agent work in conjunction with, and may increase the potency of, kill agent-stimulated immune responses and ART to reduce or eradicate viral reservoirs.
  • the present methods also provide multiple cycles of shock/kill agent administration to be repeated at regular intervals for continued reduction of the viral reservoir or until such time that viral reservoirs are eliminated.
  • Additional doses of one or more of the shock agents described herein and/or the kill agents described herein may be administered to a subject. Repeating a shock/kill administration cycle provides a protocol that is repeated at regular intervals for continued flushing/eradication of the reservoir.
  • the present invention is a method of reducing or eliminating viral reservoirs in an individual in need by:
  • the method further comprises the step of:
  • the shock/kill cycle is repeated two or more times. In a particular embodiment, the shock/kill cycle is repeated two or more times at regular intervals.
  • reservoirs are measured at about 1, 2, 3, 4, 5, 6, 7, or 8 weeks, about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months, or 1, 2, 3, 4, or 5 years after step (ii).
  • the method is repeated at least twice, at least 3 times, at least 4 times, at least 5 times, at least 6 times, at least 7 times, at least 8 times, or more than 8 times.
  • treatment involves at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8 or more than 8 cycles.
  • about 2-8, about 4-8, or about 6-8 shock/kill cycles are provided.
  • about 1-4-week, 2-4 week, 3-4 week, 1 week, 2 week, 3 week, 4 week or more than 4 week intervals are provided between cycles.
  • 4-week intervals are used between cycles.
  • treatment is continued until such time that reservoirs are reduced or eliminated as measured by methods known in the art to assess viral reservoir levels.
  • the reduction or elimination of reservoirs is measured by TILDA as compared to reservoir levels prior to treatment.
  • the reduction or elimination of reservoirs is measured by integrated DNA test as compared to reservoir levels prior to treatment.
  • reduction of viral reservoir levels is about 2 fold as compared to levels prior to treatment.
  • reduction of viral reservoir levels is about 3 fold, about 4 fold, about 5 fold, about 6 fold, about 7 fold, or more than 7 fold as compared to levels prior to treatment.
  • viral reservoir levels are undetectable after shock/kill treatment.
  • other infectious diseases that need to flush latent reservoirs might be managed or controlled through this method of simultaneous administration of vaccine shock/kill agents containing the corresponding genetic material for infectious diseases other than HIV with or without appropriate concomitant drug therapies.
  • compositions described herein can be administered in a variety of ways including through any parenteral or topical route.
  • shock and kill agents may be administered by intravenous, intraperitoneal, intradermal, subcutaneous or intramuscular methods.
  • Inoculation can be, for example, with a hypodermic needle, needleless delivery devices such as those that propel a stream of liquid into the target site, or with the use of a gene gun that bombards DNA on gold beads into the target site.
  • the shock and kill agents can be administered to a mucosal surface by a variety of methods including, but not limited to, electroporation, intranasal administration (e.g., nose drops or inhalants), or intrarectal or intravaginal administration by solutions, gels, foams, or suppositories.
  • shock and kill agents can be orally administered in the form of a tablet, capsule, chewable tablet, syrup, emulsion, or the like.
  • agents can be administered transdermally, by passive skin patches, iontophoretic means, and the like.
  • any physiologically acceptable medium can be used to administer a shock or kill agent to a subject.
  • suitable pharmaceutically acceptable carriers known in the art include, but are not limited to, sterile water, saline, glucose, dextrose, or buffered solutions.
  • the media may include auxiliary agents such as diluents, stabilizers (i.e., sugars (glucose and dextrose were noted previously) and amino acids), preservatives, wetting agents, emulsifying agents, pH buffering agents, additives that enhance viscosity or syringability, colors, and the like.
  • the medium or carrier will not produce adverse effects, or will only produce adverse effects that are far outweighed by the benefit conveyed.
  • a therapeutically effective amount of a vector (whether considered the first, second, third, etc. vector) can be administered by an intramuscular, a mucosal, or an intradermal route, together with a physiologically acceptable carrier, diluent, or excipient, and, optionally, an adjuvant.
  • a therapeutically effective amount of the same or a different vector can subsequently be administered by an intramuscular or an intradermal route, together with a physiologically acceptable carrier, diluent, or excipient, and, optionally, an adjuvant to boost an immune response.
  • Such components can be readily selected by one of ordinary skill in the art, regardless of the precise nature of the antigens incorporated in the vaccine or the vector by which they are delivered.
  • DNA can be delivered in a variety of ways, any of which can be used to deliver the plasmids of the present disclosure to a subject.
  • DNA can be injected in, for example, saline (e.g., using a hypodermic needle) delivered as a ballistic (by, for example, a gene gun that accelerates DNA-coated beads) or delivered by electroporation.
  • Saline injections deliver DNA into extracellular spaces, whereas gene gun deliveries bombard DNA directly into cells. Electroporations transiently disrupt the integrity of cellular membranes, thereby allowing entry of the DNA.
  • the saline injections require much larger amounts of DNA (typically 100-1000 times more) than the gene gun (Fynan et al, Proc. Natl. Acad.
  • extracellular plasmid DNA has a short half-life of about 10 minutes (Kawabata et al., Pharm. Res. 12:825-830, 1995; Lew et al., Hum. Gene Ther. 6:553, 1995).
  • Vaccination by saline injections can be intramuscular (i.m.), intradermal (i.d.), or mucosal (as described below in more detail); gene gun deliveries can be administered to the skin or to surgically exposed tissue such as muscle.
  • the DNA can be applied to the mucosa or by a parenteral route of inoculation.
  • Intranasal administration of DNA in saline has met with both good (Asakura et al, Scand. J. Immunol. 46:326-330, 1997; Sasaki et al, Infect. Immun. 66:823-826, 1998b) and limited (Fynan et al, Proc. Natl. Acad. Sci. U.S.A. 90: 11478- 82, 1993) success.
  • the gene gun has successfully raised IgG following the delivery of DNA to the vaginal mucosa (Livingston et al, Ann. New York Acad. Sci. 772:265-267 ', 1995). Some success at delivering DNA to mucosal surfaces has also been achieved using liposomes
  • the dose of DNA needed to raise a response depends upon the method of delivery, the host, the vector, and the encoded antigen.
  • the method of delivery may be the most influential parameter. From 10 ⁇ g to 5 mg of DNA is generally used for saline injections of DNA, whereas from 0.2 ⁇ g to 20 ⁇ g of DNA is used more typically for gene gun deliveries of DNA. In general, lower doses of DNA are used in mice (10-100 ⁇ g for saline injections and 0.2 ⁇ g to 2 ⁇ g for gene gun deliveries), and higher doses in primates (100 ⁇ g to 1 mg for saline injections and 2 ⁇ g to 20 ⁇ g for gene gun deliveries). The much lower amount of DNA required for gene gun deliveries reflect the gold beads directly delivering DNA into cells.
  • the method includes a step of measuring the presence of reservoirs after shock/kill treatment.
  • Metrics of viral infection are assayed to determine baseline infection and/or reservoir levels as well as effectiveness of treatment.
  • metrics of viral infection include, but are not limited to viral reservoirs, viral RNA and T Cell levels.
  • Suitable assays to measure viral reservoirs include, but are not limited to viral outgrowth assay, limiting dilution co-culture, measurement of total cell-associated HIV DNA by PCR, quantification of test for integrated DNA by Alu-gag PCR, quantification of 2-LTR circles by PCR, and TILDA assay although other assays for measuring viral reservoirs are known to those in the art.
  • PBMC stimulated cells
  • a PCR test for integrated DNA is used to measure the number of HIV DNA copies per million CD4+ T cells.
  • CD4+ T cells are isolated from patients. The samples are subjected to quantitative PCR analysis, in which a specific sequence indicative of the presence of HIV genome was amplified. The number of HIV genomes per sample is derived from the quantitative PCR results using statistical methods known to those in the art.
  • TILDA Tu/rev Induced Limiting Dilution Assay
  • integrated DNA tests quantify the number of HIV genomes (including both replication-competent genomes and defective genomes)
  • TILDA measures HIV RNA that has been multiply spliced, meaning that it has gone through the first few steps in HIV replication. Detection of multiply-spliced RNA indicates the presence of HIV genome that, upon activation, is capable of producing RNA with correct splicing sites.
  • TILDA is a more accurate indicator of the infectious potential of an HIV-infected individual's reservoir.
  • CD4+ T cells are isolated from the body and stimulated for 12 hours with PMA and ionomycin, a combination of drugs known to be extremely potent in activating latent HIV.
  • the samples of stimulated cells, now containing activated virus, are serially diluted. Multiple dilutions of each sample are then tested for multiply-spliced HIV-1 tatrev RNA
  • msHIV-tatrev RNA by quantitative RT-PCR.
  • TILDA is highly precise, sensitive, and reproducible. It is also much faster than many other techniques and does not require specialized equipment beyond common laboratory instruments.
  • Assays are performed before administration of vaccine and at a reasonable time after administration of vaccine. In one embodiment, these criteria are assayed at about 5 days, about 7 days, about 9 days, about 10 days, about 12 days, about 14 days, about 18 days, about 21 days, or about 24 days after administration of DNA or MVA.
  • the viral RNA is measured 7 days after vaccine administration. In another embodiment, viral RNA and T cells are measured 14 days after vaccine administration.
  • measurements of viral infection are assayed before vaccine administration to determine baseline values. Measurements are repeated after treatment to assess effectiveness of the treating.
  • EXAMPLE 1 CpG ODN, but not GpC ODN or GEO-D03, activated pro viral expression in ACH-2 cells.
  • Unmethylated CpG motifs can activate proviral DNA in the ACH-2 cell line, which harbors a latent provirus.
  • Unmethylated CpG motifs are recognized as activators of a number of cellular signaling pathways, which include pathways such as NFKB and NFAT-1 which affect provirus expression.
  • Certain types of molecules including cytokines and histone deacetylase (HDAC) inhibitors, are capable of inducing activation of latent proviral HIV- 1.
  • CpG ODN, GpC ODN (similar to CpG ODN but without immunostimulatory CpG motifs) and GEO-D03 DNA were tested on ACH-2 cells.
  • the ACH-2 cells were cultured under standard conditions and then exposed to CpG ODN, GpC ODN and GEO-D03.
  • ACH-2 cells were cultured at 37°C in 5% C02 atmosphere in RPMI-1640 (Corning), containing 10% fetal bovine serum, penicillin, and streptomycin. Cells were incubated in a 24 well plate with stimulants diluted in DNA resuspension buffer (DNA RB; 25mM sodium phosphate, 0.2mM EDTA, 120mM sodium chloride, and l%v/v ethanol). Stimulants tested were CpG ODN 2006 (Invivogen), negative control ODN 2137 (Invivogen), DNA vaccine GEO-D03 (GeoVax), DNA RB only or medium only for 20 hours. Cells were harvested and then stained with an antibody specific for HIV-1 Gag protein. The level of Gag expression, which indicates the degree of HIV-1 activation, was quantified by flow cytometry of the stained cells.
  • EXAMPLE 2 Transient increases in viral RNA were associated with the administration of GEO- D02 DNA (pGA2/JS7) to infected and drug-treated humans.
  • ART Anti-Retroviral Therapy
  • FIG. 2 illustrates transient increases in viral load observed during the immunization phase of the trial. Those that appeared to be stimulated by DNA inoculations are indicated with green arrows, and the corresponding participant IDs are circled in green.
  • EXAMPLE 3 Administration Regime to Reduce or Clear HIV from Viral Reservoirs in Patients Infected with HIV
  • a regimen using DNA as a shock and an MVA HIV vaccine as a kill in participants on continuous antiretro viral therapy is shown in Figure 4.
  • the administrations are timed to have the shock effect and reactivation of latent virus coincide with the kill effect of elicited CD8+ responses.
  • Administration of vector in the presence of systemic anti-retro viral therapy prevents or reduces spread of infection to uninfected cells.
  • HIV DNA and MVA62B vectors are administered at 0, 4, 8, and 12 weeks in the presence of antiretro viral therapy.
  • Viral RNA and T cell response are assayed 7 or 14 days after administration of vectors. Reservoirs are assayed at 0 weeks, and 20, 28 and 36 weeks to determine effectiveness of treatment.
  • EXAMPLE 4 An example of the reduction of a viral reservoir of an individual who underwent spontaneous expression of virus at the time of a MVA vaccine response.
  • a spontaneous transient enhancement of viral RNA occurred coincident with the 2 nd MVA administration in in GV-TH-01 in participant 01-7 (see FIG. 2).
  • the transient reactivation of virus occurred when a substantial CD8+ T cell response had been raised.
  • This patient underwent significant decreases in both proviral DNA in CD4+ T cells and the expression of spliced Tat and Rev mRNA from latent proviral DNA (TILDA assay- Tat-Rev induced Limiting Dilution Assay) (Figure 4).
  • TILDA Tat-Rev Induced Limiting Dilution Assay

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des compositions et des procédés permettant de réduire des réservoirs de virus. La stimulation de réservoirs viraux dans des conditions qui réduisent ou éliminent le virus actif du corps fournit une approche à deux étapes de chasse et d'éradication de réservoirs viraux. L'administration d'un agent de choc tel qu'un vaccin à ADN anti-viral réactive les réservoirs viraux et un agent neutralisant, tel qu'un vaccin anti-viral ou un anti-anticorps viral, reconnaît et tue les cellules réactivées en présence de médicaments antirétroviraux systémiques pour réduire ou prévenir l'infection de cellules non infectées. Un schéma d'administrations multiples fournit une stimulation répétée et la chasse de réservoirs viraux pour un traitement efficace.
PCT/US2014/062965 2014-10-29 2014-10-29 Combinaison thérapeutique pour le traitement de réservoirs viraux WO2016068919A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461896939P 2014-10-29 2014-10-29
US61/896,939 2014-10-29

Publications (1)

Publication Number Publication Date
WO2016068919A1 true WO2016068919A1 (fr) 2016-05-06

Family

ID=51946011

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/062965 WO2016068919A1 (fr) 2014-10-29 2014-10-29 Combinaison thérapeutique pour le traitement de réservoirs viraux

Country Status (1)

Country Link
WO (1) WO2016068919A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11278607B2 (en) 2016-01-08 2022-03-22 Geovax, Inc. Compositions and methods for generating an immune response to a tumor associated antigen
US11311612B2 (en) 2017-09-19 2022-04-26 Geovax, Inc. Compositions and methods for generating an immune response to treat or prevent malaria

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072754A2 (fr) 2001-03-08 2002-09-19 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Mva exprimant des genes enveloppe, gag et pol du vih
US20030175292A1 (en) 2000-03-02 2003-09-18 Robinson Harriet L. Compositions and methods for generating an immune response
WO2006026667A2 (fr) 2004-08-27 2006-03-09 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Virus mva de recombinaison exprimant les genes modifies env, gag et pol du vih variantes a/g, b, et c
WO2007035957A2 (fr) * 2005-09-23 2007-03-29 Pathologica, Llc. Methodes de traitement d'infections virales au moyen d'analogues de polyamine
US7371370B2 (en) 2000-09-08 2008-05-13 Massachusetts Institute Of Technology Methods of using G-CSF analog compositions
US20110104789A1 (en) * 2009-10-30 2011-05-05 Yuntao Wu Non-integrating rev-dependent lentiviral vector and methods of using the same
WO2011103417A2 (fr) 2010-02-18 2011-08-25 Emory University Of Technology Transfer Vecteurs exprimant des antigènes du vih et le gm-csf et procédés associés destinés à générer une réponse immunitaire
WO2012018856A2 (fr) * 2010-08-02 2012-02-09 Virxsys Corporation Vaccinothérapie contre le vih associée à une monothérapie antivirale concomitante

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030175292A1 (en) 2000-03-02 2003-09-18 Robinson Harriet L. Compositions and methods for generating an immune response
US7371370B2 (en) 2000-09-08 2008-05-13 Massachusetts Institute Of Technology Methods of using G-CSF analog compositions
WO2002072754A2 (fr) 2001-03-08 2002-09-19 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Mva exprimant des genes enveloppe, gag et pol du vih
WO2003076591A2 (fr) 2002-03-08 2003-09-18 Emory University Compositions et procedes destines a produire une reponse immunitaire
WO2006026667A2 (fr) 2004-08-27 2006-03-09 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Virus mva de recombinaison exprimant les genes modifies env, gag et pol du vih variantes a/g, b, et c
WO2007035957A2 (fr) * 2005-09-23 2007-03-29 Pathologica, Llc. Methodes de traitement d'infections virales au moyen d'analogues de polyamine
US20110104789A1 (en) * 2009-10-30 2011-05-05 Yuntao Wu Non-integrating rev-dependent lentiviral vector and methods of using the same
WO2011103417A2 (fr) 2010-02-18 2011-08-25 Emory University Of Technology Transfer Vecteurs exprimant des antigènes du vih et le gm-csf et procédés associés destinés à générer une réponse immunitaire
WO2012018856A2 (fr) * 2010-08-02 2012-02-09 Virxsys Corporation Vaccinothérapie contre le vih associée à une monothérapie antivirale concomitante

Non-Patent Citations (69)

* Cited by examiner, † Cited by third party
Title
AMARA ET AL., J. VIROL., vol. 76, 2002, pages 7625 - 7631
ANDRE ET AL., J. VIROL., vol. 72, 1998, pages 1497 - 1503
ASAKURA ET AL., SCAND. J. IMMUNOL., vol. 46, 1997, pages 326 - 330
BAROUCH ET AL., J. VIROL., vol. 75, 2001, pages 5151 - 5158
BLANCHARD ET AL., J. GEN. VIROL., vol. 79, 1998, pages 1159 - 1167
BOHM ET AL., J. IMMUNOL. METHODS, vol. 193, 1996, pages 29 - 40
BOYLE ET AL., INT. IMMUNOL., vol. 9, 1997, pages 1897 - 1906
BULLEN ET AL., NAT MED., vol. 20, no. 4, April 2014 (2014-04-01), pages 425 - 429
CHAPMAN ET AL., NUCL. ACIDS RES., vol. 19, 1991, pages 3979 - 3986
CHAPMAN ET AL., NUCLEIC ACIDS RESEARCH, vol. 19, 1991, pages 3979 - 3986
CHEN ET AL., J. VIROL., vol. 72, 1998
CHUN TW ET AL., J EXP MED, vol. 188, 1998, pages 83 - 91
DURBIN ET AL., J. INFECT. DIS., vol. 179, 1999, pages 1345 - 1351
ERON ET AL., ICAAC, vol. 39, 1999
EVANS ET AL., NAT. MED., vol. 5, 1999, pages 1270 - 1276
FELTQUATE ET AL., J. IMMUNOL., vol. 158, 1997, pages 2278 - 2284
FYNAN ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, 1993, pages 11478 - 11482
FYNAN ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, 1993, pages 11478 - 82
GRAESSMANN ET AL., METH. ENZYMOL., vol. 101, 1983, pages 482 - 492
GRAHAM ET AL., VIROL., vol. 52, 1973, pages 456 - 467
HARTIKKA ET AL., HUM. GENE THERAPY, vol. 7, 1996, pages 1205 - 1217
HSU ET AL., CLIN. PHARMACOKINET., vol. 35, 1998, pages 275
INCHAUSPE ET AL., DNA CELL. BIOL., vol. 16, 1997, pages 185 - 195
IWASAKI ET AL., J. IMMUNOL., vol. 158, 1997, pages 4591 - 4601
J. D. SILICIANO: "A long-term latent reservoir for HIV-1: discovery and clinical implications", JOURNAL OF ANTIMICROBIAL CHEMOTHERAPY, vol. 54, no. 1, 16 June 2004 (2004-06-16), pages 6 - 9, XP055178750, ISSN: 0305-7453, DOI: 10.1093/jac/dkh292 *
JACOBSON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 93, 1996, pages 10405 - 10410
JONES ET AL., VACCINE, vol. 15, 1997, pages 814 - 817
KAWABATA ET AL., PHARM. RES., vol. 12, 1995, pages 825 - 830
KUIKEN ET AL.: "HIV Sequence Compendium-2001", 2001, THEORETICAL BIOLOGY AND BIOPHYSICS GROUP T-10
LEW ET AL., HUM. GENE THER., vol. 6, 1995, pages 553
LIVINGSTON ET AL., ANN. NEW YORK ACAD. SCI., vol. 772, 1995, pages 265 - 267
MACKETT ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 79, pages 7415 - 7419
MAHNEL ET AL., BERL. MUNCH TIERARZTL WOCHENSCHR, vol. 107, 1994, pages 253 - 256
MANTHORPE ET AL., HUM. GENE THERAPY, vol. 4, 1993, pages 419 - 431
MASAHIKO ITO ET AL: "Peripheral B Cells as Reservoirs for Persistent HCV Infection", FRONTIERS IN MICROBIOLOGY, vol. 2, 1 January 2011 (2011-01-01), XP055179368, DOI: 10.3389/fmicb.2011.00177 *
MAYR ET AL., INFECTION, vol. 3, 1975, pages 6 - 14
MAYR ET AL., ZENTRALBL. BAKTERIOL., vol. 167, 1978, pages 375 - 390
MCCLUSKIE ET AL., ANTISENSE NUCLEIC ACID DRUG DEV., vol. 8, 1998, pages 401 - 414
MEDICAL LETTER, vol. 42, 10 January 2000 (2000-01-10), pages 1 - 6
MEYER ET AL., J. GEN. VIROL., vol. 72, 1991, pages 1031 - 1038
MONTGOMERY ET AL., DNA CELL BIOL., vol. 12, 1993, pages 777 - 783
NEUMANN ET AL., EMBO J., vol. 1, 1982, pages 841 - 845
OURMANOV ET AL., J. VIROL., vol. 74, 2000, pages 2740 - 2751
PERTMER ET AL., J. VIROL., vol. 70, 1996, pages 6119 - 6125
POIGNARD ET AL., IMMUNITY, vol. 10, 1999, pages 431 - 438
RASMUSSEN TA ET AL., HUM VACC IMMUNOTHER, vol. 9, 2013, pages 993 - 1001
ROBINSON; PERTMER: "Advances in Virus Research", vol. 53, 2000, ACADEMIC PRESS
RODRIGUEZ ET AL., J. VIROL., vol. 71, 1997, pages 8497 - 8503
SASAKI ET AL., INFECT. IMMUN., vol. 66, 1998, pages 823 - 826
SCHAFFNER, PROC. NATL. ACAD. SCI. U.S.A., vol. 77, 1980, pages 2163 - 2167
SCHELLER C ET AL., J BIOL CHEM, vol. 279, 2004, pages 21897 - 21902
SCHIFFNER ET AL., RETROVIROLOGY, vol. 10, 2013, pages 72
SCHNEIDER ET AL., NAT. MED., vol. 4, 1998, pages 397 - 402
SIZEMORE ET AL., SCIENCE, vol. 270, 1995, pages 299 - 302
SIZEMORE ET AL., VACCINE, vol. 15, 1997, pages 804 - 807
SMITH ET AL., BIOTECH. GENET. ENGIN. REV., vol. 2, 1984, pages 383 - 407
STEBBING ET AL., CLIN EXP IMMUNOL., vol. 145, no. 2, August 2006 (2006-08-01), pages 271 - 276
STICKL ET AL., DTSCH. MED. WSCHR., vol. 99, 1974, pages 2386 - 2392
STITTELAAR ET AL., J. VIROL., vol. 74, 2000, pages 4236 - 4243
STRAUBINGER ET AL., METH. ENZYMOL., vol. 101, 1983, pages 512 - 527
SUBBARAO ET AL., AIDS, vol. 10, no. A, 1996, pages 13 - 23
SUTTER ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 89, 1992, pages 10847 - 10851
SUTTER; MOSS, PROC. NATL. ACAD. SCI. U.S.A., vol. 89, 1992, pages 10847 - 10851
TOBERY ET AL., J. EXP. MED., vol. 185, 1997, pages 909 - 920
UCHIJIMA ET AL., J. IMMUNOL., vol. 161, 1998, pages 5594 - 5599
WIGLER ET AL., CELL, vol. 16, 1979, pages 777 - 785
WILD ET AL., VACCINE, vol. 16, 1998, pages 353 - 360
WU ET AL., J. IMMUNOL., vol. 159, 1997, pages 6037 - 6043
WYATT ET AL., RETROVIROLOGY, vol. 6, 2009, pages 416

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11278607B2 (en) 2016-01-08 2022-03-22 Geovax, Inc. Compositions and methods for generating an immune response to a tumor associated antigen
US11413341B2 (en) 2016-01-08 2022-08-16 Geovax, Inc. Vaccinia viral vectors encoding chimeric virus like particles
US11311612B2 (en) 2017-09-19 2022-04-26 Geovax, Inc. Compositions and methods for generating an immune response to treat or prevent malaria
US11857611B2 (en) 2017-09-19 2024-01-02 Geovax, Inc. Compositions and methods for generating an immune response to treat or prevent malaria

Similar Documents

Publication Publication Date Title
US11090379B2 (en) HIV pre-immunization and immunotherapy
US20130078276A1 (en) Vectors expressing hiv antigens and gm-csf and related methods of generating an immune response
US9254319B2 (en) Compositions and methods for generating an immune response
US20210121561A1 (en) Methods of producing cells resistant to hiv infection
US20110171258A1 (en) Polyvalent, primary hiv-1 glycoprotein dna vaccines and vaccination methods
US20160243214A1 (en) Subgenomic replicons of the flavivirus dengue
US20090169503A1 (en) Dna-based vaccination of retroviral-infected individuals undergoing treatment
Lena et al. Co-immunization of rhesus macaques with plasmid vectors expressing IFN-γ, GM-CSF, and SIV antigens enhances anti-viral humoral immunity but does not affect viremia after challenge with highly pathogenic virus
WO2016068919A1 (fr) Combinaison thérapeutique pour le traitement de réservoirs viraux
WO2018195447A1 (fr) Immunogènes du vih à base de mva recombinée et leurs utilisations
US7196186B2 (en) DNA composition and uses thereof
AU2003220111B2 (en) Compositions and methods for generating an immune response
JP2007515386A (ja) Hiv感染個体の治療用免疫化
US20050220816A1 (en) Mutant viral nucleic acids and vaccine containing same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14802250

Country of ref document: EP

Kind code of ref document: A1

WPC Withdrawal of priority claims after completion of the technical preparations for international publication

Ref document number: 61/896,939

Country of ref document: US

Date of ref document: 20160708

Free format text: WITHDRAWN AFTER TECHNICAL PREPARATION FINISHED

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14802250

Country of ref document: EP

Kind code of ref document: A1