WO2020237052A1 - Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment - Google Patents

Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment Download PDF

Info

Publication number
WO2020237052A1
WO2020237052A1 PCT/US2020/034000 US2020034000W WO2020237052A1 WO 2020237052 A1 WO2020237052 A1 WO 2020237052A1 US 2020034000 W US2020034000 W US 2020034000W WO 2020237052 A1 WO2020237052 A1 WO 2020237052A1
Authority
WO
WIPO (PCT)
Prior art keywords
vaccine
hiv
administered
weeks
mva
Prior art date
Application number
PCT/US2020/034000
Other languages
French (fr)
Inventor
Michal SARNECKI
Frank TOMAKA
Maria Grazia Pau
Romas Geleziunas
Devi SENGUPTA
Original Assignee
Janssen Vaccines & Prevention B.V.
Gilead Sciences, 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 Janssen Vaccines & Prevention B.V., Gilead Sciences, Inc. filed Critical Janssen Vaccines & Prevention B.V.
Publication of WO2020237052A1 publication Critical patent/WO2020237052A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • 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
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a 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
    • 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
    • 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

Definitions

  • This application contains a sequence listing, which is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file name“125204- 0103_Sequence_Listing”, creation date of May 20, 2020 and having a size of 64 KB.
  • the sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.
  • HIV human immunodeficiency virus
  • ART does not eliminate the viral reservoir, and treatment is associated with an incomplete restoration of the host immune system.
  • ART facilitates CD4 + T cell reconstruction in the blood, there is only a limited improvement in the function of anti-HIV specific CD8 + T cell responses.
  • ART must be taken life-long with near perfect adherence in order to be effective. This places extreme pressure and costs on international donors and over-taxed health systems in developing countries where HIV prevalence rates are highest.
  • ART has both short-term and long-term side effects for users, and drug resistance rates rise as more people are on treatment for longer periods of time.
  • alternative or complementary treatments including a therapeutic vaccine, which could induce a true or“functional” cure of HIV infection and lessen or eliminate the need for lifelong ART for HIV infected individuals, would therefore be of great benefit.
  • the concept of a“functional cure” includes therapeutic strategies that enable host control of the virus without the need for treatment.
  • the invention relates to methods for inducing an immune response against human immunodeficiency virus (HIV) in HIV-infected subjects undergoing antiretroviral therapy (ART) with a first vaccine comprising adenovirus 26 (Ad26) vectors encoding mosaic HIV antigens, a second vaccine comprising Modified Vaccinia Ankara (MVA) vector(s) encoding the mosaic HIV antigens and optionally a third vaccine comprising isolated adjuvanted Clade C gp140 and Mosaic gp140 proteins, and a Toll-like receptor 7 (TLR-7) agonist, or a pharmaceutically acceptable salt thereof, preferably vesatolimod (VES) or a pharmaceutically acceptable salt thereof.
  • a first vaccine comprising adenovirus 26 (Ad26) vectors encoding mosaic HIV antigens
  • a second vaccine comprising Modified Vaccinia Ankara (MVA) vector(s) encoding the mosaic HIV antigens
  • a third vaccine comprising isolated
  • the invention relates to a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
  • Ad26 adenovirus 26 vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
  • an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens (i.e. the HIV antigens of SEQ ID NOs: 1, 2, 3, and 4), and a pharmaceutically acceptable carrier; and (iii) administering to the human subject an effective amount of vesatolimod (VES) or a pharmaceutically acceptable salt thereof.
  • MVA Modified Vaccinia Ankara
  • the method further comprises: re-administering an immunogenically effective amount of the Ad26 vaccine to the human subject.
  • the method further comprises: re-administering an immunogenically effective amount of the MVA vaccine to the subject.
  • vesatolimod or a pharmaceutically acceptable salt thereof is administered to the subject orally, at a dosage of about 3-15 mg, such as about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of
  • vesatolimod or a pharmaceutically acceptable salt thereof is administered to the subject repeatedly.
  • the Ad26 vaccine consists of a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
  • the MVA vaccine consists of a single MVA vector encoding the four HIV antigens of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3 and SEQ ID NO:4.
  • the MVA vaccine consists of more than one MVA vectors together encoding the four HIV antigens.
  • the first, second, third, and fourth Ad26 vectors together are administered intramuscularly at a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors; and the single MVA vector or the more than one MVA vectors together are administered intramuscularly at a total dose of about 1x10 7 to about 5x10 8 infectious units (IU), preferably about 2x10 8 IU, of the MVA vector or vectors.
  • the MVA vector is an MVA-BN vector.
  • the method further comprises administering to the human subject an immunogenically effective amount of a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides, and the gp140 vaccine is administered in combination with the Ad26 vaccine or the MVA vaccine, preferably the gp140 vaccine is administered in combination with the MVA vaccine.
  • a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides
  • the gp140 vaccine is administered in combination with the Ad26 vaccine or the MVA vaccine, preferably the gp140 vaccine is administered in combination with the MVA vaccine.
  • the one or more isolated HIV gp140 envelope polypeptides are selected from the group consisting of two HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10.
  • the gp140 vaccine comprises both HIV gp140 polypeptides having respectively the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10.
  • the gp140 vaccine further comprises, or is co-administered with, an adjuvant.
  • the gp140 vaccine is administered at a total dose of about 125-350 mg, preferably about 250 mg, of the glycoprotein(s) of the HIV gp140 envelope polypeptide(s), per administration.
  • the Ad26 vaccine is re-administered at about 10-14 weeks, e.g., 12 weeks, after the Ad26 vaccine is initially administered.
  • the MVA vaccine is first administered at about 22-26 weeks, e.g., about 24 weeks, after the Ad26 vaccine is initially administered.
  • the MVA vaccine is re-administered at about 34-38 weeks, e.g., 36 weeks, after the Ad26 vaccine is initially administered.
  • vesatolimod is administered biweekly from 26 to 34 weeks after the Ad26 vaccine is initially administered.
  • vesatolimod is further administered biweekly from 38 to 46 weeks after the Ad26 vaccine is initially administered.
  • the invention relates to a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
  • Ad26 adenovirus 26 vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors per administration;
  • step (ii) intramuscularly re-administering to the human subject the Ad26 vaccine in a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re-administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
  • MVA vectors comprising one or more MVA vectors, preferably one or more MVA-BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x10 7 to about 5x10 8 infectious units (IU), preferably about 2x10 8 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further administering a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i); and
  • vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i).
  • the method further comprises: intramuscularly re- administering to the human subject the MVA vaccine, in a total dose of about 1x10 7 to about 5x10 8 infectious units (IU), preferably about 2x10 8 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further re-administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine, and optionally the gp140 vaccine, is re-administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i).
  • IU infectious units
  • the method further comprises: intramuscularly re- administering to the human subject the MVA vaccine, in a total dose of about 1x10 7 to about 5x10 8 infectious units (IU),
  • the method further comprises: orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi- weekly at 38-46 weeks after the Ad26 vaccine is administered in step (i).
  • vesatolimod or a pharmaceutically acceptable salt thereof is orally administered to the human subject biweekly at a total dose of about 3 mg to about 15 mg, such as at about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration, preferably at 26-34 and 38-46 weeks after the initial administration of the Ad26 vaccine.
  • the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at 26 and 28 weeks after the initial administration of the Ad26 vaccine, and a total dose of 8 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • the subject is a chronically HIV-infected subject.
  • the subject initiated ART outside of the acute phase of HIV infection.
  • the subject initiated ART during the acute phase of HIV infection.
  • the invention also relates to a vaccine combination for use in inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected subject undergoing antiretroviral therapy (ART); and use of a vaccine combination in the
  • HIV immunodeficiency virus
  • HIV-infected subject undergoing antiretroviral therapy
  • the vaccine combination comprises: (i) an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO:1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier; (ii) an MVA vaccine comprising one or more Modified Vaccinia Ankara vectors, preferably Modified Vaccinia Ankara Bavarian Nordic (MVA-BN) vectors, together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and (iii) VES or a pharmaceutically acceptable salt thereof.
  • Ad26 adenovirus 26
  • an MVA vaccine comprising one or more Modified Vaccinia Ankara
  • the vaccine combination further comprises a gp140 vaccine comprising at least one of isolated HIV Clade C and Mosaic gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, respectively, an aluminum adjuvant and a pharmaceutically acceptable carrier.
  • a gp140 vaccine comprises both of the HIV Clade C and Mosaic gp140 envelope polypeptides.
  • the invention relates to methods for inducing an immune response against HIV-1 in an HIV-1 infected human subject being treated with antiretroviral therapy (ART), wherein the ART has successfully suppressed replication of HIV in the blood stream of the subject such that the subject can discontinue the ART and still maintains control of viral replication in the blood stream for at least 24 weeks after discontinuation of the ART.
  • Control of viral replication can be measured using methods known in the art, such as by measuring HIV viral load (e.g., HIV RNA levels), in view of the present disclosure.
  • the invention relates to a method of providing an HIV remission in an HIV-infected human subject undergoing antiretroviral therapy (ART), comprising inducing an immune response against HIV in the subject to thereby control HIV infection after discontinuation of the ART, wherein the immune response against HIV is induced using a method according to an embodiment of the invention.
  • ART- free viremic control e.g., controlling the HIV viral load (and possibly reduce the HIV viral reservoir) in HIV patients, is achieved in at least 20%, preferably at least 30%, 40%, 50% of the human subjects, for at least 24 weeks, preferably longer, after a treatment with a method of the invention and discontinuation of the ART.
  • Another general aspect of the invention relates to a method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
  • the method of treatment further comprises discontinuing the ART treatment of step (i), preferably after an immune response against the HIV is induced in the human subject by a method of the invention.
  • the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART.
  • the conjunctive term“and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by“and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or” as used herein. Concurrent applicability of more than one of the options is also understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or.”
  • “subject” means a human, who will be or has been treated by a method according to an embodiment of the invention.
  • adjuvant and “immune stimulant” are used interchangeably herein, and are defined as one or more substances that cause stimulation of the immune system.
  • an adjuvant is used to enhance an immune response to HIV antigens and antigenic HIV polypeptides of the invention.
  • the terms and phrases“in combination,”“in combination with,” “co-delivery,” and“administered together with” in the context of the administration of two or more therapies or components to a subject refers to simultaneous administration of two or more therapies or components, such as a viral expression vector and an isolated antigenic polypeptide.“Simultaneous administration” can be administration of the two components at least within the same day.
  • two components are“administered together with” or “administered in combination with,” they can be administered in separate compositions sequentially within a short time period, such as 24, 20, 16, 12, 8 or 4 hours, or within 1 hour, or within 30 minutes, or within 10 minutes, or within 5 minutes, or within 2 minutes, or they can be administered in a single composition at the same time.
  • two components or therapies are administered in separate compositions.
  • the use of the term“in combination with” does not restrict the order in which therapies or components are administered to a subject.
  • a first therapy or component e.g.
  • viral expression vector can be administered prior to (e.g., 5 minutes to one hour before), concomitantly with or simultaneously with, or subsequent to (e.g., 5 minutes to one hour after) the administration of a second therapy (e.g., isolated HIV antigenic polypeptide).
  • a second therapy e.g., isolated HIV antigenic polypeptide
  • an Ad26 vaccine comprises one or more adenovirus 26 (Ad26) vectors encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4.
  • Ad26 adenovirus 26 vectors encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4.
  • an immunogenically effective amount of the Ad26 vaccine is administered to the subject more than once.
  • an MVA vaccine comprises one or more Modified Vaccinia Ankara vectors, preferably one or more Modified Vaccinia Ankara Bavarian Nordic (MVA-BN) vectors, encoding the four HIV antigens, i.e. HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4.
  • an immunogenically effective amount of the MVA vaccine is administered to the subject more than once.
  • a further vaccine is administered together with the re- administered Ad26 vaccine, or preferably together with the MVA vaccine.
  • the further vaccine is a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides.
  • the gp140 vaccine comprises one or more of isolated HIV Clade C and Mosaic gp140 proteins having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO:10, respectively. More preferably the gp140 vaccine comprises both HIV Clade C and Mosaic gp140 proteins.
  • the human subjects are further administered vesatolimod or a pharmaceutically acceptable salt thereof.
  • An“HIV vaccine” as used herein comprises the Ad26 vaccine and the MVA vaccine used and administered as described herein, and optionally further comprises the gp140 vaccine used and administered as described herein.
  • HIV Human Immunodeficiency Virus
  • HIV Human immunodeficiency virus
  • HIV-1 is the most common strain of HIV virus and is known to be more pathogenic than HIV-2.
  • the terms“human immunodeficiency virus” and“HIV” refer, but are not limited to, HIV-1 and HIV-2.
  • the envelope proteins described herein refer to those present on HIV-1.
  • HIV is categorized into multiple clades with a high degree of genetic divergence.
  • HIV clade or“HIV subtype” refers to related human
  • Group M major strains
  • Group O outer strains
  • Group N is a new HIV-1 isolate that has not been categorized in either group M or O.
  • the terms“chronic set point”,“set point in chronic HIV infection”,“viral load set point”, and“viral set point in chronic HIV infection” refer to the steady state HIV viral load established in the blood of an HIV infected human.
  • the chronic set point can refer to a value of steady state HIV viral load after infection, following the introduction of antiretroviral therapy or treatment, including administration of ART, a TLR7 agonist, and/or an HIV vaccine described herein, or after cessation of antiretroviral therapy or treatment.
  • a chronic set point can be determined in a single HIV infected human or determined as a median chronic set point in a cohort of HIV infected humans.
  • a first chronic set point can be a percentage of a second chronic set point or the second chronic set point can be a multiple of the first chronic set point.
  • a first chronic set point of 100 copies HIV-1 RNA per mL is 10% of a second chronic set point of 1000 copies HIV-1 RNA per mL, and can alternatively be described as a second chronic set point that is 10-fold higher than a first chronic set point.
  • A“viral rebound” refers to the observation that an undetectable HIV viral load in a virologically suppressed HIV infected human after treatment with ART often reverts to a detectable pre-therapy HIV viral load after cessation of ART.
  • the viral rebound can occur within days or weeks, e.g., 4 weeks, after cessation of ART.
  • A“delay in viral rebound” refers to a time period between the expected observation of viral rebound, e.g., 4 weeks, after cessation of ART as compared to the actual observed viral rebound, e.g., 12 weeks, after cessation of another therapy, e.g., administration of ART, a TLR7 agonist, and HIV vaccine according to the method described herein.
  • the delay in viral rebound is 8 weeks after treatment of the ART, the TLR7 agonist, and the HIV vaccine.
  • a delay in viral rebound can be determined in a single HIV infected human or determined as a median delay in viral rebound in a cohort of HIV infected humans.
  • the methods described herein can be used to induce an immune response against one or more clades of HIV.
  • HIV antigen all refer to a polypeptide capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against HIV in a subject.
  • the HIV antigen can be a protein of HIV, a fragment or epitope thereof, or a combination of multiple HIV proteins or portions thereof, that can induce an immune response against HIV in a subject.
  • An HIV antigen is capable of raising in a host a protective immune response, e.g., inducing an immune response against a viral disease or infection, and/or producing an immunity in (i.e., vaccinates) a subject against a viral disease or infection, that protects the subject against the viral disease or infection.
  • a protective immune response e.g., inducing an immune response against a viral disease or infection
  • an immunity in i.e., vaccinates
  • the HIV antigen can comprise a protein or fragment(s) thereof from HIV, such as the HIV gag, pol and env gene products.
  • the HIV antigen can be an HIV-1 or HIV-2 antigen or fragment(s) thereof.
  • HIV antigens include, but are not limited to gag, pol, and env gene products, which encode structural proteins and essential enzymes. Gag, pol, and env gene products are synthesized as polyproteins, which are further processed into multiple other protein products.
  • the primary protein product of the gag gene is the viral structural protein gag polyprotein, which is further processed into MA, CA, SP1, NC, SP2, and P6 protein products.
  • the pol gene encodes viral enzymes (Pol, polymerase), and the primary protein product is further processed into RT, RNase H, IN, and PR protein products.
  • the env gene encodes structural proteins, specifically glycoproteins of the virion envelope.
  • the primary protein product of the env gene is gp160, which is further processed into gp120 and gp41.
  • a heterologous nucleic acid sequence according to the invention preferably encodes a gag, env, and/or pol gene product, or portion thereof.
  • the HIV antigen comprises an HIV Gag, Env, or Pol antigen, or any portion or combination thereof, more preferably an HIV-1 Gag, Env, or Pol antigen, or any portion or combination thereof.
  • an HIV antigen is a mosaic HIV antigen.
  • “mosaic antigen” refers to a recombinant protein assembled from fragments of natural sequences.
  • The“mosaic antigen” can be computationally generated and optimized using a genetic algorithm.
  • Mosaic antigens resemble natural antigens, but are optimized to maximize the coverage of potential T-cell epitopes found in the natural sequences, which improves the breadth and coverage of the immune response.
  • Examples of mosaic HIV Gag-Pol-Env antigens include those described in, e.g., US20120076812, Barouch et al., Nat Med 2010, 16:319-323; Barouch et al., Cell 155:1-9, 2013; and WO 2017/102929, all of which are incorporated herein by reference in their entirety.
  • the mosaic HIV antigens encoded by the vectors according to the invention comprise one or more of the amino acid sequences selected from the group consisting of SEQ ID NOs: 1-4 and 10.
  • Alternative and/or additional HIV antigens could be encoded by the primer vaccine and/or the booster vaccine of the invention in certain embodiments, e.g. to further broaden the immune response.
  • a mosaic HIV antigen can be produced using methods known in the art. See, e.g., US20120076812, Fischer et al, Nat Med, 2007.13(1): p. 100-6; Barouch et al., Nat Med 2010, 16:319-323, all of which are incorporated herein by reference in their entirety.
  • glycoprotein “env polypeptide,”“env glycoprotein,” and“Env” refers to, but is not limited to, the glycoprotein that is expressed on the surface of the envelope of HIV virions and the surface of the plasma membrane of HIV infected cells, or a fragment thereof that can induce an immune response or produce an immunity against HIV in a subject in need thereof.
  • the env gene encodes gp160, which is proteolytically cleaved into gp120 and gp41. More specifically, gp160 trimerizes to (gp160)3 and then undergoes cleavage into the two noncovalently associated fragments gp120 and gp41. Viral entry is subsequently mediated by a trimer of gp120/gp41 heterodimers.
  • Gp120 is the receptor binding fragment and binds to the CD4 receptor on a target cell that has such a receptor, such as, e.g., a T- helper cell.
  • Gp41 which is non-covalently bound to gp120, is the fusion fragment and provides the second step by which HIV enters the cell.
  • Gp41 is originally buried within the viral envelope, but when gp120 binds to a CD4 receptor, gp120 changes its conformation causing gp41 to become exposed, where it can assist in fusion with the host cell.
  • Gp140 is the uncleaved ectodomain of trimeric gp160, i.e., (gp160)3, that has been used as a surrogate for the native state of the cleaved, viral spike.
  • isolated HIV envelope polypeptides e.g., gp160, gp140, gp120, or gp41
  • gp140 protein e.g., gp140 protein
  • gp41 gp140 protein
  • gp41 gp140 protein
  • gp41 gp140 protein
  • gp41 gp140 protein
  • gp41 gp140 protein
  • gp41 gp140 protein
  • trimeric gp140 protein e.g., gp140 protein
  • each of the terms“stabilized trimeric gp140 protein” and “stabilized trimer of gp140” refers to a trimer of gp140 polypeptides that includes a polypeptide sequence that increases the stability of the trimeric structure.
  • the gp140 polypeptides can have or can be modified to include a trimerization domain that stabilizes trimers of gp140.
  • trimerization domains include, but are not limited to, the T4- fibritin“foldon” trimerization domain; the coiled-coil trimerization domain derived from GCN4; and the catalytic subunit of E. coli aspartate transcarbamoylase as a trimer tag.
  • Examples of isolated antigenic polypeptide are stabilized trimeric gp140 such as those described in Nkolola et al 2010, J. Virology 84(7): 3270-3279; Kovacs et al, PNAS 2012, 109(30):12111-6; WO 2010/042942 and WO 2014/107744, all of which are incorporated by reference in their entirety.
  • the“envelope polypeptide” or“envelope glycoprotein” is a mosaic envelope protein comprising multiple epitopes derived from one or more of Env polyprotein sequences of one or more HIV clades.
  • a“gp140 protein” can be a“mosaic gp140 protein” that contains multiple epitopes derived from one or more gp140 protein sequences of one or more HIV clades.
  • a mosaic gp140 protein is a stabilized trimeric gp140 protein.
  • a mosaic gp140 protein is a stabilized trimer of mosaic gp140 protein comprising the amino acid sequence of SEQ ID NO: 10.
  • the envelope polypeptide” or“envelope glycoprotein” is an envelope protein derived from a particular HIV clade, such as HIV clade A, B, or C.
  • a“gp140 protein” can be a“clade C gp140 protein” that contains envelope protein sequence derived from HIV clade C.
  • a clade C gp140 protein is a stabilized trimeric clade C gp140 protein.
  • a clade C gp140 protein is a stabilized trimer of clade C gp140 protein comprising the amino acid sequence of SEQ ID NO: 9.
  • a gp140 polypeptide such as a stabilized trimeric gp140 protein can be administered together with viral expression vectors, e.g., adenovirus 26 and/or MVA vectors.
  • two gp140 proteins are administered to the same subject, preferably a clade C gp140 having the amino acid sequence of SEQ ID NO: 9 and a mosaic gp140 having the amino acid sequence of SEQ ID NO: 10.
  • the two gp140 proteins can be together in one pharmaceutical composition, preferably administered together with an adjuvant, such as aluminum phosphate adjuvant.
  • a preferred dose for the total amount of gp140 for administration to humans is between about 125 and 350 ⁇ g, such as 125, 150, 175, 200, 225, 250, 275, 300, 325, 350 ⁇ g, or any amount in between, preferably about 250 ⁇ g.
  • a suitable dose would for instance be about 125 ⁇ g of each protein, to provide a total dose of 250 ⁇ g of gp140 protein for an administration to a human subject.
  • the amount of a gp140 polypeptide refers to the amount of the gp140 polypeptide measured as glycoprotein.
  • An isolated gp140 protein can be co-delivered or administered in combination with an adenovirus (e.g., Ad26) expression vector or MVA expression vector.
  • Ad26 adenovirus
  • MVA expression vector adenovirus expression vector
  • a gp140 protein and Ad26 or MVA vector are administered separately, as two distinct formulations.
  • a gp140 protein can be administered with Ad26 or MVA together in a single formulation. Simultaneous administration or co- delivery can take place at the same time, within one hour, or within the same day.
  • a gp140 protein can be administered in an adjuvanted formulation.
  • Suitable adjuvants can be, for example, aluminum phosphate or a saponin-based adjuvant, preferably aluminum phosphate adjuvant.
  • Antigenic polypeptides such as gp140 can be produced and isolated using any method known in the art in view of the present disclosure.
  • an antigenic polypeptide can be expressed from a host cell, preferably a recombinant host cell optimized for production of the antigenic polypeptide.
  • a recombinant gene is used to express a gp140 protein containing mutations to eliminate cleavage and fusion activity, preferably an optimized gp140 protein with increased breadth, intensity, depth, or longevity of the antiviral immune response (e.g., cellular or humoral immune responses) generated upon immunization (e.g. , when incorporated into a
  • composition of the invention e.g., vaccine of the invention
  • a subject e.g., a human
  • the optimized gp140 protein can also include cleavage site mutation(s), a factor Xa site, and/or a foldon trimerization domain.
  • a leader/signal sequence can be operably linked to the N- terminal of an optimized gp140 protein for maximal protein expression.
  • the leader/signal sequence is usually cleaved from the nascent polypeptide during transport into the lumen of the endoplasmic reticulum. Any leader/signal sequence suitable for a host cell of interest can be used.
  • An exemplary leader/signal sequence comprises the amino acid sequence of SEQ ID NO: 11.
  • Primer vaccines, and in certain embodiments booster vaccines, used in the methods of the invention comprise one or more adenovirus vectors, particularly human adenovirus 26 vectors (Ad26) encoding one more mosaic HIV antigens.
  • An adenovirus according to the invention belongs to the family of the Adenoviridae, and preferably is one that belongs to the genus Mastadenovirus.
  • the notation“rAd” means recombinant adenovirus, e.g.,“rAd26” refers to recombinant human adenovirus 26.
  • an adenovirus is a human adenovirus serotype 26 (Ad26).
  • Ad26 human adenovirus serotype 26
  • An advantage of human adenovirus serotype 26 is that, so far, significant experience was obtained with such vectors in clinical trials, and this did not reveal that pre-existing neutralizing antibody responses against such vectors would cause substantial interference with desired vaccine-induced responses, e.g. against the encoded antigens in such vectors.
  • the adenovirus vector is a replication deficient recombinant viral vector, such as a replication deficient recombinant adenovirus 26 vector.
  • the recombinant adenovirus vector useful in the invention is derived mainly or entirely from Ad26 (i.e., the vector is rAd26).
  • the adenovirus is replication deficient, e.g., because it contains a deletion in the E1 region of the genome.
  • Ad26 adenoviruses derived from Ad26 used in the invention, it is typical to exchange the E4-orf6 coding sequence of the adenovirus with the E4-orf6 of an adenovirus of human subgroup C such as Ad5.
  • the adenovirus is a human adenovirus of serotype 26, with a deletion in the E1 region into which the nucleic acid encoding one or more mosaic HIV antigens has been cloned, and with an E4 orf6 region of Ad5.
  • rAd26 vectors Preparation of rAd26 vectors is described, for example, in WO 2007/104792 and in Abbink et al., (2007) Virol 81(9): 4654-63, both of which are incorporated by reference herein in their entirety.
  • Exemplary genome sequences of Ad26 are found in GenBank Accession EF 153474 and in SEQ ID NO: 1 of WO 2007/104792, which is herein incorporated by reference in its entirety.
  • an adenovirus vector useful in the invention is produced using a nucleic acid comprising the entire recombinant adenoviral genome (e.g., a plasmid, cosmid, or baculovirus vector).
  • the adenovirus vectors useful in the invention are typically replication deficient.
  • the virus is rendered replication deficient by deletion or inactivation of regions critical to replication of the virus, such as the E1 region.
  • the regions can be substantially deleted or inactivated by, for example, inserting a gene of interest, such as a gene encoding an HIV antigen (usually linked to a promoter) within the region.
  • the vectors of the invention can contain deletions in other regions, such as the E3 region, or insertions of heterologous genes linked to a promoter within such regions.
  • a packaging cell line is typically used to produce sufficient amounts of adenovirus vectors for use in the invention.
  • a packaging cell is a cell that comprises those genes that have been deleted or inactivated in a replication deficient vector, thus allowing the virus to replicate in the cell.
  • Suitable packaging cell lines include, for example, PER.C6, 911, and HEK293.
  • HIV antigens can be expressed in the adenovirus 26 vectors described herein.
  • the heterologous gene encoding the mosaic HIV antigen can be codon-optimized to ensure proper expression in the treated host (e.g., human). Codon-optimization is a technology widely applied in the art.
  • the heterologous gene encoding the mosaic HIV antigen is cloned into the E1 and/or the E3 region of the adenoviral genome.
  • Non-limiting embodiments of codon optimized nucleotide sequences encoding HIV antigens with SEQ ID NOs: 1-4 are provided herein as SEQ ID NOs: 5-8, respectively.
  • one or more adenovirus 26 (Ad26) vectors comprise nucleic acid that encodes one or more HIV antigens, in particular the one or more Ad26 vectors together encode four mosaic HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4.
  • the Ad26 vaccine used in the invention comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
  • these vectors are present in a single composition in a 1:1:1:1 ratio (based on viral particles).
  • the heterologous gene encoding the mosaic HIV antigen can be under the control of (i.e., operably linked to) an adenovirus-derived promoter (e.g., the Major Late Promoter), or can be under the control of a heterologous promoter.
  • adenovirus-derived promoter e.g., the Major Late Promoter
  • suitable heterologous promoters include the cytomegalovirus immediate early (CMV) promoter and the Rous sarcoma virus (RSV) promoter.
  • CMV cytomegalovirus immediate early
  • RSV Rous sarcoma virus
  • the promoter is located upstream of the heterologous gene encoding the mosaic HIV antigen within an expression cassette.
  • the heterologous promoter is a CMV promoter.
  • an MVA vaccine used in the methods of the invention comprises one or more Modified Vaccinia Ankara (MVA) vectors together encoding four mosaic HIV antigens, in particular the HIV antigens of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4.
  • MVA vectors useful in the invention utilize attenuated virus derived from MVA virus, which is characterized by the loss of their capabilities to reproductively replicate in human cell lines.
  • MVA has been generated by more than 570 serial passages on chicken embryo fibroblasts of the dermal vaccinia strain Ankara (Chorioallantois vaccinia virus Ankara virus, CVA; for review see Mayr et al. (1975) Infection 3, 6-14) that was maintained in the dermal vaccinia strain Ankara (Chorioallantois vaccinia virus Ankara virus, CVA; for review see Mayr et al. (1975) Infection 3, 6-14) that was maintained in the
  • MVA derived from MVA-571 seed stock (corresponding to the 571st passage) was registered in Germany as the primer vaccine in a two-stage parenteral smallpox vaccination program.
  • MVA-572 was used in a small dose as a pre-vaccine in Germany during the smallpox eradication program, and MVA-575 was extensively used as a veterinary vaccine.
  • MVA as well as MVA-BN lacks approximately 15% (31 kb from six regions) of the genome compared with ancestral CVA virus. The deletions affect a number of virulence and host range genes, as well as the gene for Type A inclusion bodies.
  • MVA-575 was deposited on December 7, 2000, at the European Collection of Animal Cell Cultures (ECACC) under Accession No. V00120707.
  • MVA was further passaged by Bavarian Nordic and is designated MVA- BN.
  • “Derivatives” or“variants” of MVA refer to viruses exhibiting essentially the same replication characteristics as MVA as described herein, but exhibiting differences in one or more parts of their genomes.
  • MVA-BN as well as a derivative or variant of MVA-BN fails to reproductively replicate in vivo in humans and mice, even in severely immune suppressed mice.
  • MVA-BN or a derivative or variant of MVA- BN has preferably also the capability of reproductive replication in chicken embryo fibroblasts (CEF), but no capability of reproductive replication in the human keratinocyte cell line HaCat (Boukamp et al (1988), J.
  • a derivative or variant of MVA-BN has a virus amplification ratio at least two fold less, more preferably three-fold less than MVA-575 in Hela cells and HaCaT cell lines. Tests and assays for these properties of MVA variants are described in WO 02/42480 (US 2003/0206926) and WO 03/048184 (US 2006/0159699).
  • the term“not capable of reproductive replication” or“no capability of reproductive replication” is, for example, described in WO 02/42480, which also teaches how to obtain MVA having the desired properties as mentioned above.
  • the term applies to a virus that has a virus amplification ratio at 4 days after infection of less than 1 using the assays described in WO 02/42480 or in U.S. Patent No.6,761,893, both of which are incorporated by reference herein in their entirety.
  • the term“fails to reproductively replicate” refers to a virus that has a virus amplification ratio at 4 days after infection of less than 1.
  • Assays described in WO 02/42480 or in U.S. Patent No.6,761,893 are applicable for the determination of the virus amplification ratio.
  • MVA-based vaccines have safety profile as well as availability for large scale vaccine production. Furthermore, in addition to its efficacy, the feasibility of industrial scale manufacturing can be beneficial. Additionally, MVA-based vaccines can deliver multiple heterologous antigens and allow for simultaneous induction of humoral and cellular immunity.
  • MVA vectors useful for the invention can be prepared using methods known in the art, such as those described in WO/2002/042480, WO/2002/24224, US20110159036, US 8197825, etc., the relevant disclosures of which are incorporated herein by reference.
  • replication deficient MVA viral strains can also be suitable for use in the invention, such as strains MVA-572 and MVA-575, or any other similarly attenuated MVA strain. Also suitable can be a mutant MVA, such as the deleted
  • a dCVA comprises del I, del II, del III, del IV, del V, and del VI deletion sites of the MVA genome. The sites are particularly useful for the insertion of multiple heterologous sequences.
  • the dCVA can reproductively replicate (with an amplification ratio of greater than 10) in a human cell line (such as human 293, 143B, and MRC-5 cell lines), which then enable the optimization by further mutation useful for a virus-based vaccination strategy (see, e.g., WO 2011/092029).
  • the MVA vectors are MVA-BN vectors, such as that described in WO 2018/229711, which is incorporated herein by reference.
  • the MVA vector(s) comprise a nucleic acid that encodes one or more HIV antigens having the amino acid sequences selected from the group consisting of SEQ ID NOs: 1-4.
  • the one or more MVA vectors together encode four mosaic HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4.
  • a particularly useful but non-limiting example of an MVA vaccine that can be used for the present invention is MVA-mBN414, as described in example 7 of WO 2018/229711.
  • Nucleic acid sequences encoding the mosaic HIV antigens can be inserted into one or more intergenic regions (IGR) of the MVA.
  • IGR intergenic regions
  • the IGR is selected from IGR07/08, IGR 44/45, IGR 64/65, IGR 88/89, IGR 136/137, and IGR 148/149.
  • less than 5, 4, 3, or 2 IGRs of the recombinant MVA comprise heterologous nucleotide sequences encoding an HIV antigen, such as a mosaic HIV antigen.
  • the heterologous nucleotide sequences can, additionally or alternatively, be inserted into one or more of the naturally occurring deletion sites, in particular into the main deletion sites I, II, III, IV, V, or VI of the MVA genome.
  • less than 5, 4, 3, or 2 of the naturally occurring deletion sites of the recombinant MVA comprise heterologous nucleotide sequences encoding mosaic HIV antigens.
  • the number of insertion sites of MVA comprising heterologous nucleotide sequences encoding HIV antigens can be 1, 2, 3, 4, 5, or more.
  • the heterologous nucleotide sequences are inserted into 4, 3, 2, or fewer insertion sites.
  • the recombinant MVA comprises at least 2, 3, 4, 5, 6, or 7 genes inserted into 2 or 3 insertion sites.
  • the recombinant MVA viruses provided herein can be generated by routine methods known in the art. Methods to obtain recombinant poxviruses or to insert exogenous coding sequences into a poxviral genome are well known to the person skilled in the art. For example, methods for standard molecular biology techniques such as cloning of DNA, DNA and RNA isolation, Western blot analysis, RT-PCR and PCR amplification techniques are described in Molecular Cloning, A laboratory Manual (2nd Ed.) (J. Sambrook et al., Cold Spring Harbor Laboratory Press (1989)), and techniques for the handling and manipulation of viruses are described in Virology Methods Manual (B.W.J. Mahy et al.
  • the DNA sequence to be inserted into the virus can be placed into an E. coli plasmid construct into which DNA homologous to a section of DNA of the MVA has been inserted.
  • the DNA sequence to be inserted can be ligated to a promoter.
  • the promoter-gene linkage can be positioned in the plasmid construct so that the promoter-gene linkage is flanked on both ends by DNA homologous to a DNA sequence flanking a region of MVA DNA containing a non-essential locus.
  • the resulting plasmid construct can be amplified by propagation within E. coli bacteria and isolated.
  • the isolated plasmid containing the DNA gene sequence to be inserted can be transfected into a cell culture, e.g., of chicken embryo fibroblasts (CEFs), at the same time the culture is infected with MVA. Recombination between homologous MVA DNA in the plasmid and the viral genome, respectively, can generate an MVA modified by the presence of foreign DNA sequences.
  • a cell culture e.g., of chicken embryo fibroblasts (CEFs)
  • CEFs chicken embryo fibroblasts
  • a cell of a suitable cell culture such as, e.g., CEF cells
  • a poxvirus can be infected with a poxvirus.
  • the infected cell can be, subsequently, transfected with a first plasmid vector comprising a foreign or heterologous gene or genes, preferably under the transcriptional control of a poxvirus expression control element.
  • the plasmid vector also comprises sequences capable of directing the insertion of the exogenous sequence into a selected part of the poxviral genome.
  • the plasmid vector also contains a cassette comprising a marker and/or selection gene operably linked to a poxviral promoter.
  • Suitable marker or selection genes are, e.g., the genes encoding the green fluorescent protein, b-galactosidase, neomycin-phosphoribosyltransferase or other markers.
  • the use of selection or marker cassettes simplifies the identification and isolation of the generated recombinant poxvirus.
  • a recombinant poxvirus can also be identified by PCR technology. Subsequently, a further cell can be infected with the recombinant poxvirus obtained as described above and transfected with a second vector comprising a second foreign or heterologous gene or genes.
  • the second vector also differs in the poxvirus- homologous sequences directing the integration of the second foreign gene or genes into the genome of the poxvirus.
  • the recombinant virus comprising two or more foreign or heterologous genes can be isolated.
  • the steps of infection and transfection can be repeated by using the recombinant virus isolated in previous steps for infection and by using a further vector comprising a further foreign gene or genes for transfection.
  • a suitable cell can at first be transfected by the plasmid vector comprising the foreign gene and, then, infected with the poxvirus.
  • a suitable cell can at first be transfected by the plasmid vector comprising the foreign gene and, then, infected with the poxvirus.
  • a third alternative is ligation of DNA genome and foreign sequences in vitro and reconstitution of the recombined vaccinia virus DNA genome using a helper virus.
  • a fourth alternative is homologous recombination in E.coli or another bacterial species between a vaccinia virus genome cloned as a bacterial artificial chromosome (BAC) and a linear foreign sequence flanked with DNA sequences homologous to sequences flanking the desired site of integration in the vaccinia virus genome
  • BAC bacterial artificial chromosome
  • the heterologous nucleic acid encoding one or more mosaic HIV antigens can be under the control of (i.e., operably linked to) one or more poxvirus promoters.
  • the poxvirus promoter is a Pr7.5 promoter, a hybrid early/late promoter, or a PrS promoter, a PrS5E promoter, a synthetic or natural early or late promoter, or a cowpox virus ATI promoter.
  • an MVA vector useful for the invention expresses HIV antigens having the amino acid sequences of SEQ ID NO: 1 to SEQ ID NO: 4.
  • Immunogenic compositions are compositions comprising an immunogenically effective amount of a purified or partially purified adenovirus 26 or MVA vector for use in the invention.
  • the adenovirus 26 and MVA vectors can encode any mosaic HIV antigens in view of the present disclosure, and preferably encode one or more HIV antigens selected from the group consisting of SEQ ID NOs: 1-4.
  • the one or more mosaic HIV antigens encoded by the adenovirus 26 vector can be different from, but preferably are the same as the one or more mosaic HIV antigens encoded by the MVA vector.
  • Immunogenic compositions can be formulated as a vaccine, according to methods well known in the art. Such
  • compositions can include adjuvants to enhance immune responses.
  • the optimal ratios of each component in the formulation can be determined by techniques well known to those skilled in the art in view of the present disclosure.
  • an immunogenically effective amount or“immunologically effective amount” means an amount of a composition or vector sufficient to induce a desired immune effect or immune response in a subject in need thereof.
  • an immunogenically effective amount means an amount sufficient to induce an immune response in a subject in need thereof, preferably a safe and effective immune response in a human subject in need thereof.
  • an immunogenically effective amount means an amount sufficient to produce immunity in a subject in need thereof, e.g., provide a therapeutic effect against a disease such as HIV infection.
  • An immunogenically effective amount can vary depending upon a variety of factors, such as the physical condition of the subject, age, weight, health, etc. An immunogenically effective amount can readily be determined by one of ordinary skill in the art in view of the present disclosure.
  • An immunogenically effective amount can be administered in a single step (such as a single injection), or multiple steps (such as multiple injections), or in a single
  • compositions or multiple compositions It is also possible to administer an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to a subject, and subsequently administer another dose of an immunogenically effective amount to
  • prime-boost regimen immunogenically effective amount to the same subject, in a so-called prime-boost regimen.
  • This general concept of a prime-boost regimen is well known to the skilled person in the vaccine field. Further booster administrations can optionally be added to the regimen, as needed.
  • an immunogenically effective amount when used with reference to a recombinant viral vector can range from about 10 6 viral particles (vps), plaque forming units (pfus) or infectious units (IU) to about 10 12 viral particles or infectious units, for example 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , or 10 12 viral particles or infectious units.
  • an immunogenic composition is an Ad26 vaccine used for initial administration to induce an immune response.
  • an Ad26 vaccine comprises an immunogenically effective amount of one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, and a pharmaceutically acceptable carrier.
  • Ad26 adenovirus 26
  • the HIV antigens can be encoded by the same Ad26 vector or different Ad26 vector, such as one, two, three, four or more Ad26 vectors.
  • the immunogenically effective amount of the one or more Ad26 vectors can be about 10 8 , 10 9 , 10 10 , 10 11 , or 10 12 viral particles (vps), preferably about 10 9 to 10 11 viral particles, and more preferably about 10 10 viral particles, such as for instance about 0.5 x 10 10 , 1 x 10 10 , 2 x 10 10 , 3 x 10 10 , 4 x 10 10 , 5 x 10 10 , 6 x 10 10 , 7 x 10 10 , 8 x 10 10 , 9 x 10 10 , or 10 x 10 10 viral particles.
  • vps viral particles
  • the immunogenically effective amount is about 5 x10 9 to about 1 x 10 11 viral particles, preferably about 5 x 10 10 viral particles, such that the one or more Ad26 vectors are administered at a total dose of about 5 x10 9 to about 1 x 10 11 viral particles per immunization step.
  • the immunogenically effective amount can be from one Ad26 vector or multiple Ad26 vectors.
  • a total administered dose of about 5 x10 9 to about 1 x 10 11 viral particles, such as for instance about 5 x 10 10 viral particles, in the Ad26 vaccine can be from four Ad26 vectors each encoding a different mosaic HIV antigen, such as those shown in SEQ ID NOs: 1, 2, 3, and 4.
  • the immunogenically effective amount of Ad26 vectors together encoding SEQ ID NOs: 1, 2, 3, and 4 consists of four adenovirus vectors, namely a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
  • an Ad26 vaccine comprises more than one Ad26 vector
  • the Ad26 vectors can be included in the composition in any ratio to achieve the desired immunogenically effective amount.
  • the immunogenically effective amount of the Ad26 vectors consists of four Ad26 vectors
  • the first, second, third, and fourth Ad26 vectors are administered at a 1:1:1:1 ratio of viral particles (vps).
  • An MVA vaccine that is useful in the invention comprises an immunogenically effective amount of one or more MVA vectors together encoding the four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, and a pharmaceutically acceptable carrier.
  • the HIV antigens expressed by MVA vectors can be encoded by a single MVA vector, or multiple MVA vectors, such as one, two, or more MVA vectors. In certain advantageous embodiments, the HIV antigens are expressed by a single MVA vector.
  • the immunogenically effective amount of the one or more MVA vectors in the MVA vaccine can be about 10 6 , 10 7 , 10 8 , 10 9 , or 10 10 infectious units (IU), preferably about 10 7 to 10 9 IU, and more preferably about 2x 10 8 IU, such as for instance about 0.5 x 10 8 , 1 x 10 8 , 2 x 10 8 , 3 x 10 8 , 4 x 10 8 , or 5 x 10 8 IU.
  • IU infectious units
  • the immunogenically effective amount is about 1 x10 7 to about 5 x 10 8 IU, preferably about 2 x 10 8 IU, such that the one or more MVA vectors are administered at a total dose of about 1 x10 7 to about 5 x 10 8 IU, preferably about 2 x 10 8 IU per immunization step.
  • the immunogenically effective amount can be from one MVA vector or multiple MVA vectors.
  • a total administered dose of about 1 x10 7 to about 5 x 10 8 IU, such as for instance about 1x10 7 , 5x10 7 , 1x10 8 , 2 x 10 8 , 5x 108 IU, or any dose in between, in the MVA vaccine can be from two MVA vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, for example a first MVA vector encoding the HIV antigens of SEQ ID NOs: 1 and 3, and a second MVA vector encoding the HIV antigens of SEQ ID NO: 2 and SEQ ID NO: 4, wherein preferably the first and second MVA vectors are administered at a 1:1 ratio of IU.
  • a total administered dose of about 1 x10 7 to about 5 x 10 8 IU, such as for instance about 1x10 7 , 5x10 7 , 1x10 8 , 2 x 10 8 , 5x 108 IU, or any dose in between, in the MVA vaccine can be from a single MVA vector encoding four HIV antigens having the amino acid sequences of SEQ ID NOs: 1, 2, 3, and 4.
  • an Ad26 vaccine or an MVA vaccine that is administered after the initial administration of the Ad26 vaccine is administered in combination with one or more isolated HIV gp140 envelope polypeptides.
  • an immunogenically effective amount can range from, e.g. about 125 mg to 350 mg, e.g. about 125, 150, 200, 250, 300, or 350 ⁇ g of the one or more isolated HIV envelope polypeptides.
  • a vaccine composition comprising one or more Ad26 vectors or one or more MVA vectors is administered in combination with a gp140 vaccine composition comprising two isolated HIV envelope gp140 polypeptides, one clade C gp140 polypeptide having the amino acid sequence of SEQ ID NO: 9 and one mosaic gp140 polypeptide having the amino acid sequence of SEQ ID NO: 10, each one for instance present in about 125 ⁇ g per administration to a total of about 250 ⁇ g, all measured as glycoprotein.
  • Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil.
  • immunogenic compositions used in the invention can be formulated for administration according to any method known in the art in view of the present disclosure, and are preferably formulated for intramuscular administration.
  • the vaccine compositions of the invention can comprise other antigens.
  • the other antigens used in combination with the adenovirus 26 and/or MVA vectors and/or gp140 polypeptides can be, for example, other HIV antigens and nucleic acids expressing them.
  • the immunogenic compositions useful in the invention can further optionally comprise adjuvants.
  • Adjuvants suitable for co-administration in accordance with the invention should be ones that are potentially safe, well tolerated and effective in people.
  • Non- limiting examples include QS-21, Detox-PC, MPL-SE, MoGM-CSF, TiterMax-G, CRL- 1005, GERBU, TERamide, PSC97B, Adjumer, PG-026, GSK-I, GcMAF, B-alethine, MPC- 026, Adjuvax, CpG ODN, Betafectin, Aluminium salts such as Aluminium Phosphate (e.g. AdjuPhos) or Aluminium Hydroxide, and MF59.
  • Aluminium Phosphate e.g. AdjuPhos
  • Aluminium Hydroxide MF59.
  • a preferred adjuvant for administration together with isolated HIV envelope polypeptides is aluminum phosphate.
  • the total amount of aluminum phosphate administered can range from, e.g. about 10 ⁇ g to about 1000 ⁇ g, e.g. about 200 ⁇ g to 650 ⁇ g, e.g. about 200, 250, 300, 350, 400, 425, 450, 475, 500, 550, or 600 ⁇ g, preferably about 425 ⁇ g of aluminum.
  • the immunogenic compositions used for generating an immune response comprise a pharmaceutically acceptable carrier, such as a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • a pharmaceutically acceptable carrier such as a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material can depend on the route of administration, e.g., intramuscular, subcutaneous, oral, intradermal, cutaneous, intramucosal (e.g., gut), intranasal or intraperitoneal routes.
  • the pharmaceutically acceptable carrier included in the compositions of the invention is suitable for intramuscular administration.
  • TLR 7 is one of the protein members of the family of receptors (toll like receptors) that recognize pathogen associated molecular patterns (PAMPs) on infectious agents and is expressed by the gene, HGNC:15631, in humans.
  • PAMPs pathogen associated molecular patterns
  • the TLR agonist useful for the invention is vesatolimod (VES), also known as GS-9620, which is a small molecule with the IUPAC name of 4-amino-2-butoxy-8-[[3-(pyrrolidin-1-ylmethyl)phenyl]methyl]-5,7-dihydropteridin- 6-one, or a pharmaceutically acceptable salt thereof.
  • VES vesatolimod
  • Vesatolimod has the structure:
  • Vesatolimod can be formulated with conventional carriers and excipients, which will be selected in accordance with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986), herein incorporated by reference in its entirety. Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10.
  • the formulations include those suitable for the foregoing administration routes.
  • the formulations can conveniently be presented in unit dosage form and can be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.), herein incorporated by reference in its entirety. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations of vesatolimod or a pharmaceutically acceptable salt thereof suitable for oral administration can be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient can also be administered as a bolus, electuary or paste.
  • Suitable routes for the administration of vesatolimod or a pharmaceutically acceptable salt thereof include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route can vary with for example the condition of the recipient.
  • An advantage of vesatolimod compounds used in this invention is that they are orally bioavailable and can be dosed orally.
  • the effective dose of vesatolimod depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active disease or condition, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies.
  • the effective dose can be expected to be from about 0.0001 to about 10 mg/kg body weight per day, typically from about 0.001 to about 1 mg/kg body weight per day, more typically from about 0.01 to about 1 mg/kg body weight per day, even more typically from about 0.05 to about 0.5 mg/kg body weight per day.
  • the daily candidate dose for an adult human of approximately 70 kg body weight will range from about 0.05 mg to about 100 mg, or between about 0.1 mg and about 25 mg, or between about 0.4 mg and about 15 mg, and may take the form of single or multiple doses.
  • vesatolimod or a pharmaceutically acceptable salt thereof is orally administered to the human subject biweekly at a total dose of about 3 mg to about 15 mg, such as at about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration.
  • the effective amount of vesatolimod or a pharmaceutically acceptable salt thereof can be orally administered to the human subject using any suitable dosage form.
  • 4 mg of vesatolimod or an equivalent amount of a pharmaceutically acceptable salt thereof can be orally administered with a single 4 mg tablet or two 2 mg tablets; 6 mg of vesatolimod or an equivalent amount of a pharmaceutically acceptable salt thereof can be orally administered with two 3 mg tablets or three 2 mg tablets; etc.
  • VES or a pharmaceutically acceptable salt thereof can be administered 10 doses, preferably every 14 days, with one of the following dosage form and regimen:
  • Ad26 and MVA vaccine compositions described above can be used in the methods of the invention described herein.
  • the methods of the invention relate to inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected subject undergoing antiretroviral therapy.
  • the methods of administering Ad and MVA vaccines according to embodiments of the invention are effective to induce an immune response against one or multiple clades of HIV.
  • the invention relates to a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
  • Ad26 adenovirus 26 vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
  • an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens (i.e. the HIV antigens of SEQ ID NOs: 1, 2, 3, and 4), and a pharmaceutically acceptable carrier;
  • MVA Modified Vaccinia Ankara
  • VES vesatolimod
  • the method further comprises administering to the human subject a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides in combination with the Ad26 vaccine or MVA vaccine in step (ii) and/or (iii) and/or (v).
  • the gp140 vaccine is preferably administered in steps (iii) and (v).
  • the method preferably further comprises administering to the human subject at least one isolated HIV gp140 envelope polypeptide selected from the group consisting of two trimeric HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO :10, in combination with the Ad26 vaccine or MVA vaccine in step (ii) and/or (iii) and/or (v), preferably in steps (iii) and (v).
  • the gp140 vaccine preferably comprises both HIV gp140 polypeptides, respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10.
  • a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART) comprises:
  • Ad26 adenovirus 26 vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors per administration; (ii) re-administering, preferably intramuscularly, to the human subject the Ad26 vaccine in a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re-administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
  • MVA vectors comprising one or more MVA vectors, preferably one or more MVA- BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x10 7 to about 5x10 8 infectious units (IU), preferably about 2x10 8 IU, of the one or more MVA-BN vectors, per administration; optionally, in combination with the MVA vaccine, further administering a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i);
  • vesatolimod at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i);
  • step (i) administration; optionally, in combination with the MVA vaccine, further re- administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine, and optionally the gp140 vaccine, is administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i); and
  • vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 38-46 weeks after the Ad26 vaccine is administered in step (i).
  • step (iii) comprises administering the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier, in combination with the MVA vaccine.
  • step (v) comprises re-administering the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier, in combination with the MVA vaccine.
  • the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at 26 and 28 weeks after the initial administration of the Ad26 vaccine, and a total dose of 8 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 8 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 10 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • any of the vaccine compositions described herein can be used in a method according to the invention.
  • Embodiments of the Ad26 vaccine; MVA vaccine; Ad26 vectors; MVA vectors; HIV antigens encoded by the Ad26 and MVA vectors; isolated gp140 polypeptides, etc. that can be used in the methods of the invention are discussed in detail above and in the illustrative examples below.
  • “inducing an immune response” when used with reference to the methods described herein encompasses causing a desired immune response or effect in a subject in need thereof against an HIV infection, preferably for therapeutic purposes.“Inducing an immune response” also encompasses providing a therapeutic immunity for treating against a pathogenic agent, i.e., HIV.
  • a pathogenic agent i.e., HIV.
  • the term“therapeutic immunity” or“therapeutic immune response” means that the HIV-infected vaccinated subject is able to control an infection with the pathogenic agent, i.e., HIV, against which the vaccination was done.
  • “inducing an immune response” means producing an immunity in a subject in need thereof, e.g., to provide a therapeutic effect against a disease such as HIV infection.
  • “inducing an immune response” refers to causing or improving cellular immunity, e.g., T cell response, against HIV.
  • “inducing an immune response” refers to causing or improving a humoral immune response against HIV.
  • “inducing an immune response” refers to causing or improving a cellular and a humoral immune response against HIV.
  • the administration of the Ad26 and MVA vaccine compositions according to embodiments of the invention will have a therapeutic aim to generate an immune response against HIV after HIV infection or development of symptoms characteristic of HIV infection.
  • the induced immune response in the subject in which ART has successfully suppressed replication of HIV in the blood stream is such that the subject can discontinue the ART and still maintains control of viral replication in the blood stream for at least 24 weeks after discontinuation of the ART.
  • the patient population for treatment according to the methods of the invention described herein is HIV-infected human subjects, particularly HIV-infected human subjects undergoing antiretroviral therapy (ART).
  • the terms“HIV infection” and“HIV-infected” as used herein refer to invasion of a human host by HIV.
  • “an HIV-infected human subject” refers to a human subject in whom HIV has invaded and subsequently replicated and propagated within the human host, thus causing the human host to be infected with HIV or have an HIV infection or symptoms thereof.
  • An“HIV-infected human subject” has been diagnosed with HIV infection, i.e., tests positive in a screen for HIV infection, e.g. using any assay that is US FDA-approved.
  • undergoing antiretroviral therapy refers to a human subject, particularly an HIV-infected human subject, that is being administered, or who has initiated treatment with antiretroviral drugs.
  • the antiretroviral therapy is started prior to the first administration of the Ad26 vaccine , for instance, about 2 to 6 weeks prior, such as about 2, 3, 4, 5, or 6 weeks prior, or 2-48 months prior, such as about 2, 3, 5, 6, 8, 12, 16, 20, 24, 30, 36, 42, or 48 months prior, or longer.
  • the ART is started earlier than about 44-52 weeks, preferably earlier than about 48 weeks prior to the first administration of the Ad26 vaccine.
  • the antiretroviral therapy is continued during
  • ART is considered“suppressive” as used herein if the subject has plasma HIV RNA levels at less than 50 copies/mL for a certain period of time, including the possibility of blips.
  • the term“stable suppressive” ART as used herein means that the suppressive ART regimen is not modified for a certain period of time.
  • a human subject undergoing antiretroviral therapy is on current stable suppressive ART for at least twenty-four weeks, meaning that while receiving the same ART regimen the subject has plasma HIV ribonucleic acid (RNA) levels at less than 50 copies/mL for at least 24 weeks prior to initiation of a regimen according to the invention.
  • the human subject can have one or more blips (i.e., instances) of plasma HIV RNA greater than 50 copies/ml to less than 200 copies/ml within this period, such as within the 24 week period prior to the initiation of the regimen, provided that screening immediately prior to initiation of the regimen is less than 50 copies/ml.
  • An HIV-infected subject can initiate ART during the acute phase of HIV infection, or outside of the acute phase of HIV infection.
  • the subject initiated ART during the acute phase of HIV infection.
  • the term“acute HIV infection” refers to the initial stage of HIV infection. In general, there are three stages of HIV infection: (1) acute HIV infection, (2) clinical latency, and (3) acquired
  • AIDS immunodeficiency syndrome
  • the host typically develops symptoms such as fever, swollen glands, sore throat, rash, muscle and joint aches and pains, headache, etc., as a result of the body’s natural response to the HIV infection.
  • CD4 levels can decrease rapidly, because the HIV uses CD4 to replicate and then subsequently destroys the CD4.
  • Acute HIV infection is also characterized as Fiebig stages I, II, III, and IV as described in Fiebig et al.,“Dynamics of HIV viremia and antibody seroconversion in plasma donors: implications for diagnosis and staging of primary HIV infection.” AIDS (London, England) (2003) 17(13) 1871-1879, which is herein incorporated by reference in its entirety.
  • an HIV-infected human subject that is subjected to a regimen of the invention is characterized as Fiebig stage I, Fiebig stage II, Fiebig stage III, or Fiebig stage IV.
  • Acute HIV infection is typically within two to four weeks after a host is exposed to and infected with HIV and continues for an additional two to four weeks.
  • the acute HIV infection stage lasts until the host creates its own antibodies against HIV, at which point the clinical latency stage begins.
  • HIV is living or developing in the host without causing any symptoms, or only causing mild symptoms. HIV reproduces at very low levels during the clinical latency stage, although the HIV is still active.
  • the clinical latency stage is sometimes also referred to as“chronic HIV infection” or“asymptomatic HIV infection.”
  • Chronic HIV infection is characterized as Fiebig stage VI.
  • an HIV-infected human subject that is subjected to a regimen of the invention is characterized as Fiebig stage V, or Fiebig stage VI.
  • Fiebig stage VI typically begins about 100 days (i.e., about 14 weeks) after a host is exposed to and infected with HIV.
  • a subject infected with HIV that has progressed to Fiebig stage VI can be referred to or described as a“chronically- infected subject,”“a chronic HIV-infected subject,” or“a subject having chronic HIV infection.”
  • a subject initiating ART outside of the acute or early phase of HIV infection is one who has not begun ART before entering Fiebig VI stage. Whether or not a subject has initiated ART prior to entering Fiebig VI stage of HIV infection can be determined by a clinician based on the subject’s available medical history and laboratory data at the time of HIV diagnosis.
  • a subject who initiates ART outside of the acute phase of HIV infection begins treatment with antiretroviral drugs at the earliest at about 12-16 weeks, after being exposed to and infected with HIV, such as about 12, 13, 14, 15, or 16 weeks or later, after exposure and infection with HIV.
  • a subject who initiates ART during acute HIV infection typically begins treatment with antiretroviral drugs at or prior to about 2 weeks to about 8 weeks after being exposed to and infected with HIV, such as about 1, 2, 3, 4, 5, 6, 7, or 8 weeks after exposure and infection.
  • chronic HIV infection is thought to be more difficult to treat than acute HIV infection, at least because a chronically infected HIV subject typically has larger HIV viral reservoirs than an acutely infected subject due to the longer period of infection prior to initiating any treatment.
  • therapies effective to treat acute HIV infection will likewise be effective to treat chronic HIV infection.
  • the HIV-infected subject is a chronically HIV-infected subject.
  • a chronically HIV-infected subject can initiate ART at any phase of infection, such as during the acute phase of HIV infection or outside the acute phase of HIV infection.
  • a chronically HIV-infected subject initiates ART during the acute phase of HIV infection.
  • the HIV-infected human subject is in the acute phase of HIV infection.
  • a subject undergoing ART can be administered or treated with any antiretroviral drugs known in the art in view of the present disclosure.
  • ART are medications that treat HIV, although the drugs do not kill the virus or remove the virus from the body. However, when taken in combination they can prevent the growth of the virus. When the virus is slowed down, so is HIV disease.
  • Antiretroviral drugs are referred to as ARV.
  • Combination ARV therapy (cART) is referred to as highly active ART (HAART).
  • an ART regimen includes at least three antiviral compounds, e.g., two different reverse transcriptase inhibitors plus either a non-nucleoside reverse transcriptase inhibitor or protease inhibitor or integrase inhibitor.
  • antiretroviral drugs used for ART include, but are not limited to nucleoside reverse transcriptase inhibitors (NRTIs, non-limiting examples of which include zidovudine, didanosine, stavudine, lamivudine, abacavir, tenofovir, combivir [combination of zidovudine and lamivudine], trizivir [combination of zidovudine, lamivudine and abacavir],
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • PIs protease inhibitors
  • INSTIs integrase inhibitors
  • FIs entry inhibitors and/or chemokine receptor antagonists
  • an Ad26 vaccine or an MVA vaccine is administered after the Ad26 vaccine is initially administered.
  • the Ad26 vaccine or MVA vaccine is re-administered or administered at about 12-52 weeks, such as, about 8-15 weeks, about 21-27 weeks, and/or about 33-39 weeks, after the Ad26 vaccine is initially administered.
  • One of ordinary skill in the art will be able to vary the exact timing of the vaccines, frequency of administration thereof, dosage thereof, etc., based upon the teachings herein and clinical experience.
  • Ad26 vaccine is administered at least once and MVA vaccine at least once.
  • Ad26 vaccine is re-administered at about 10-14 weeks, and MVA vaccine is first administered 22- 26 weeks, such as 22, 23, 24, 25, or 26 weeks, after the Ad26 vaccine is initially administered.
  • the Ad26 vaccine is re-administered at about 12 weeks after the Ad26 vaccine is initially administered, the MVA vaccine is first administered at about 24 weeks after the Ad26 vaccine is initially administered.
  • the MVA vaccine is re-administered at about 32-40 weeks, such as at about 36 weeks, after the Ad26 vaccine is initially administered.
  • vesatolimod or a pharmaceutically acceptable salt thereof is administered for the first time after the first administration of the MVA vaccine. In other embodiments, additional vesatolimod or a pharmaceutically acceptable salt thereof is administered after the second administration of the MVA vaccine. In certain embodiments, vesatolimod or a pharmaceutically acceptable salt thereof is administered several times in a series of administrations with about two-week intervals. In certain embodiments, the first administration of such series is about two weeks after the first and/or after the second administration of the MVA vaccine.
  • the Ad26 vaccine is re-administered after the Ad26 vaccine is initially administered, and in such embodiments preferably re-administered before the MVA vaccine is first administered.
  • the Ad26 vaccine can be re- administered at about 10-14 weeks after the Ad26 vaccine is initially administered, such as about 10, 11, 12, 13, or 14 weeks, after the Ad26 vaccine is initially administered, preferably at about 12 weeks after the Ad26 vaccine is initially administered.
  • the MVA vaccine is administered after the Ad26 vaccine is re-administered.
  • the MVA vaccine is first administered at about 22 to 26 weeks, such as 22, 23, 24, 25, or 26 weeks, after the Ad26 vaccine is initially administered, preferably at about 24 weeks after the Ad26 vaccine is initially administered.
  • the MVA vaccine can in certain embodiments be re-administered, e.g. at about 32 to 40 weeks, such as 32, 33, 34, 35, 36, 37, 38, 39, or 40 weeks, after the Ad26 vaccine is initially administered.
  • the MVA vaccine is re-administered at about 36 weeks after the Ad26 vaccine is initially administered.
  • the vaccine compositions can be administered by any method known in the art in view of the present disclosure, and administration is typically via intramuscular, intradermal or subcutaneous administration, preferably intramuscular administration.
  • Intramuscular administration can be achieved by using a needle to inject a suspension or solution of the adenovirus and/or MVA vectors, and/or gp140 polypeptides.
  • An alternative is the use of a needleless injection device to administer the composition (using, e.g., Biojector TM ) or a freeze-dried powder containing the vaccine.
  • the vector will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, and Lactated Ringer's Injection.
  • Preservatives, stabilizers, buffers, antioxidants and/or other additives can be included, as required.
  • a slow- release formulation can also be employed.
  • a method of inducing an immune response according to the invention further comprises administering a latent viral reservoir purging agent.
  • Cells latently infected with HIV carry integrated virus that is transcriptionally silent, making it difficult to effectively eradicate HIV infection in treated subjects.
  • “reservoir purging agent” and“latent viral reservoir purging agent” refer to a substance that reduces the latent pool of HIV by reactivating HIV reservoirs, such as by inducing expression of quiescent HIV.
  • latent viral reservoir purging agents suitable for use with the invention include, but are not limited to, histone deacetylase (HDAC) inhibitors and modulators of toll-like receptors (e.g., TLR7), such as those described in WO2016/007765 and WO2016/177833, which are herein incorporated by reference in their entireties.
  • HDAC histone deacetylase
  • TLR7 modulators of toll-like receptors
  • Another general aspect of the invention relates to a method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
  • the method of treatment further comprises discontinuing the ART treatment of step (i), preferably after an immune response is induced by a method of the invention.
  • the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART.
  • subjects undergo interruption (also referred to as discontinuation, used interchangeably herein) of ART after completion of a regimen according to embodiments of the invention.
  • subjects can undergo antiretroviral analytical treatment interruption (ARV ATI) after completion of a regimen according to embodiments of the invention.
  • ARV ATI antiretroviral analytical treatment interruption
  • “Antiretroviral analytical treatment interruption” and“ARV ATI” as used in the invention refer to discontinuation of treatment with antiretroviral drugs in order to assess viral suppression and viremic control in the absence of continued ART.
  • subjects can undergo ARV ATI, i.e., ART can be discontinued, for example when the subject has plasma HIV RNA levels at less than 50 copies/mL for at least about 52 weeks, but a subject can still undergo ARV ATI even if the subject has one or more blips (i.e., instances) of plasma HIV RNA greater than 50 copies/ml to less than 200 copies/ml within this period, provided that the screening immediately prior to ARV ATI shows less than 50 copies/ml of plasma HIV RNA.
  • HIV viral load e.g., plasma HIV RNA levels
  • the ART can be stopped at about 10- 14 weeks, such as 10, 11, 12, 13, or 14 weeks after the last MVA vaccine is administered.
  • the last MVA vaccine is administered at about 34-38 weeks after the Ad26 vaccine is initially administered.
  • the ART can be stopped at about 46 to 50 weeks, such as 46, 47, 48, 59, or 60 weeks, after the Ad26 vaccine is initially administered, and preferably about 60 weeks after the Ad26 vaccine is initially administered.
  • a boosted protease inhibitor can be administered in place of the NNRTI for about 1-2 weeks prior to stopping ART to reduce the risk of developing NNRTI resistance. It is also possible to administer an activator (e.g. a histone deacetylase inhibitor or TLR7 modulator) during the ATI stage to activate any (e.g. latent) HIV reservoir and thereby improve the immune response.
  • an activator e.g. a histone deacetylase inhibitor or TLR7 modulator
  • Subjects undergoing ARV ATI can be monitored, e.g., by measuring plasma HIV RNA levels.
  • monitoring after the initiation of ARV ATI can occur up to two times per week during the first six weeks when rebound viremia is most likely to occur.
  • “Rebound viremia” is for example defined as plasma HIV RNA levels of greater than 1,000 copies/ml after ARV ATI.
  • ART can be re-initiated in subjects with rebound viremia.
  • a subject treated according to the methods of the invention will maintain viremic control after ART interruption.
  • “maintain viremic control” is in exemplary embodiments defined as at least 24 weeks with plasma HIV RNA of less than 50 copies/mL after ARV ATI.
  • The“maintained viremic control” criterion is in certain exemplary embodiments still deemed to be met if there are one or more instances of plasma HIV RNA greater than 50 copies/ml to less than 1000 copies/ml, as long as the subject does not have plasma HIV RNA levels above 1000 copies/ml on two consecutive determinations at least one week apart.
  • compositions and vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention among individuals with fully suppressed HIV will result in a measurable immune response and maintain viremic control after ARV ATI.
  • subjects can discontinue ART after being treated according to a method of the invention. Discontinuation of ART can be for long periods of time (e.g., at least 24 weeks, preferably longer, e.g. at least about 28, 32, 36, 40, 44, 48, 52 weeks, 16 months, 18, 20, 22, 24 months, or even longer). Such periods of time in which ART is stopped or discontinued are referred to as a“holiday” or“ART holiday” or“treatment holiday”. In other
  • vaccine and TLR7 therapy according to the methods of the invention can provide HIV remission, meaning that viral suppression is maintained in the absence of ART.
  • a human subject that received the vaccines and vesatolimod or a pharmaceutically acceptable salt thereof according to the invention discontinues ART and maintains viral suppression for at least 24 weeks after discontinuing ART.
  • an Ad26 vaccine comprising one or more adenovirus 26 vectors is administered (e.g., intramuscularly) in an amount of about 100 ⁇ l to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 10 8 to 10 12 virus particles/ml.
  • the initial Ad26 vaccination is optionally repeated and is followed by an MVA vaccine comprising one more MVA vectors administered (e.g., intramuscularly) in an amount of about 100 ⁇ l to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 10 6 to 10 9 pfu/ml.
  • vesatolimod or a pharmaceutically acceptable salt thereof is administered, preferably followed by a further administration of the MVA vaccine, and further administrations of vesatolimod or a pharmaceutically acceptable salt thereof.
  • an Ad26 vaccine comprising one or more adenovirus 26 vectors is administered (e.g., intramuscularly) in an amount of about 100 ⁇ l to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 10 8 to 10 12 virus particles/ml.
  • the initial Ad26 vaccination is followed by re- administration of the Ad26 vaccine comprising one more adenovirus 26 vectors administered (e.g., intramuscularly) in an amount of about 100 ⁇ l to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 10 8 to 10 12 virus particles/ml.
  • administration of the MVA vaccine e.g.
  • intramuscularly in an amount of about 100 ⁇ l to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 10 6 to 10 9 pfu/ml, in some embodiments in combination with one or more isolated HIV gp140 polypeptides in an amount of about 100 ⁇ l to about 2 ml, preferably about 0.5 ml, of a solution, to a total dose per administration of about 250 mg polypeptide and aluminum phosphate adjuvant (425 microgram ( ⁇ g) aluminum per dose).
  • the administration of the MVA vaccine is followed by repeated administration of vesatolimod or a pharmaceutically acceptable salt thereof. In preferred embodiments this is followed by another administration of the MVA vaccine, and thereafter further repeated administrations of vesatolimod or a pharmaceutically acceptable salt thereof.
  • the invention also relates to a vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination for use in inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), wherein the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination comprises an Ad26 vaccine and an MVA vaccine and vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention.
  • HIV human immunodeficiency virus
  • ART antiretroviral therapy
  • the invention yet further relates to use of a vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination in the manufacture of a medicament for inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), wherein the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination comprises an Ad26 vaccine and an MVA vaccine and vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention.
  • HAV human immunodeficiency virus
  • ART antiretroviral therapy
  • immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART) can be applied to the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combinations for use and/or uses of the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination in the manufacture of a medicament for inducing an immune response against HIV in an HIV-infected subject undergoing ART.
  • HIV immunodeficiency virus
  • ART antiretroviral therapy
  • a clinical improvement of a treated HIV-infected human above a comparator HIV-infected human treated with a standard of care is expected.
  • the clinical improvement can include one or more of a lower peak viral load, a lower chronic set point, or an increased delay in viral rebound.
  • the method as described herein has an effect on treatment of the HIV infection, for example, as determined by a lower peak viral load as compared to standard therapies, e.g., ART only.
  • a lower peak viral load as compared to standard therapies, e.g., ART only.
  • comparison of a first peak viral load in a first HIV-infected human and a second peak viral load in a second HIV-infected human is measured during the same time period.
  • the measurement is performed after cessation of all antiviral therapies.
  • the viral load is maintained at an undetectable level in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • a first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second peak viral load in a second HIV-infected human after treatment with ART only.
  • the second peak viral load in a second HIV-infected human after treatment with ART only is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART only is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART only is about 1000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • a first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • the second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 20 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • a first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine is about 100 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the method as described herein has an effect on treatment of the HIV infection, for example, as determined by a lower chronic set point as compared to standard therapies, e.g., ART.
  • a lower chronic set point as compared to standard therapies, e.g., ART.
  • comparison of a first chronic set point in a first HIV-infected human and a second chronic set point in a second HIV-infected human is measured at the same time point.
  • the measurement is performed after cessation of all antiviral therapies.
  • a first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second chronic set point in a second HIV-infected human after treatment with ART only.
  • the second chronic set point in a second HIV-infected human after treatment with ART only is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second chronic set point in a second HIV- infected human after treatment with ART is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first chronic set point in a first HIV- infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second chronic set point in a second HIV-infected human after treatment with ART only is about 10 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • a first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • the second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 2 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • a first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • the second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine is about 10 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the instant method can increase the delay in viral rebound as compared to standard therapies after cessation of all antiviral therapies.
  • the viral load does not rebound in an HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
  • the previously HIV-infected human maintains an undetectable viral load after cessation of antiviral therapies for at least 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years, 5 years, or at least 10 years or longer after antiviral therapies have ceased.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is longer than a second delay in viral rebound in a second HIV-infected human after treatment with ART only.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is from about 1 day to about 10 years, e.g., from about 1 week to about 1 year, from about 2 weeks to about 1 year, from about 3 weeks to about 1 year, from about 1 month to about 1 year, from about 2 months to about 1 year, from about 3 months to about 1 year, from about 3 months to about 2 years, etc., longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART only.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is greater than 1 day, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years, 5 years, 10 years or longer compared to a second delay in viral rebound in a second HIV- infected human after treatment with ART only.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 month, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 1.5 years, about 2 years, about 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART only.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 3 months longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART only.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is longer than a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is from about 1 day to about 10 years, e.g., from about 1 week to about 1 year, from about 2 weeks to about 1 year, from about 3 weeks to about 1 year, from about 1 month to about 1 year, from about 2 months to about 1 year, from about 3 months to about 1 year, from about 3 months to about 2 years, etc., longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is greater than 1 day, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 month, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 month, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 1.5 years, about 2 years, about 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 3 months longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is longer than a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is from about 1 day to about 10 years, e.g., from about 1 week to about 1 year, from about 2 weeks to about 1 year, from about 3 weeks to about 1 year, from about 1 month to about 1 year, from about 2 months to about 1 year, from about 3 months to about 1 year, from about 3 months to about 2 years, etc., longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is greater than 1 day, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 month, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 month, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 1.5 years, about 2 years, about 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 month longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine.
  • Embodiment 1 is a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
  • Ad26 adenovirus 26 vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
  • an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and
  • MVA Modified Vaccinia Ankara
  • vesatolimod or a pharmaceutically acceptable salt thereof (VES).
  • Embodiment 1a is the method of Embodiment 1, wherein step (i) is conducted before step (ii).
  • Embodiment 1b is the method of Embodiment 1 or 1a, wherein step (ii) is conducted before step (iii).
  • Embodiment 1c is the method of any one of Embodiments 1 to 1b, wherein the immunogenically effective amount of the MVA vaccine is administered at about 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is initially administered.
  • Embodiment 1d is the method of any one of Embodiments 1 to 1c, wherein the effective amount of vesatolimod or the pharmaceutically acceptable salt thereof is
  • Embodiment 1e is the method of Embodiment 1d, wherein the effective amount of vesatolimod or the pharmaceutically acceptable salt thereof is administered biweekly at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 2 is the method of any one of Embodiments 1-1e, further comprising re-administering to the human subject an immunogenically effective amount of the Ad26 vaccine.
  • Embodiment 2a is the method of Embodiment 2, wherein the immunogenically effective amount of the Ad26 vaccine is re-administered at about 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is initially administered.
  • Embodiment 3 is the method of any one of Embodiments 1 to 2a, further comprising re-administering to the human subject an immunogenically effective amount of the MVA vaccine.
  • Embodiment 3a is the method of Embodiment 3, wherein the MVA vaccine is re- administered at about 34 to 38 weeks, preferably 36 weeks, after the Ad26 vaccine is initially administered.
  • Embodiment 4 is the method of any one of Embodiments 1-3a, wherein the Ad26 vaccine comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
  • Embodiment 5 is the method of any one of Embodiments 1 to 4, wherein the MVA vaccine consists of a single MVA vector encoding the four HIV antigens.
  • Embodiment 5a is the method of Embodiment 5, wherein the single MVA vector is an MVA-BN vector.
  • Embodiment 6 is the method of any one of Embodiments 1 to 5a, wherein the one or more Ad26 vectors together are administered at a total dose of about 5x10 9 to about 1x10 11 viral particles (vp) of the one or more Ad26 vectors, per administration.
  • vp viral particles
  • Embodiment 6a is the method of Embodiment 6, wherein the one or more Ad26 vectors together are administered at a total dose of about 5x10 10 vp of the one or more Ad26 vectors, per administration.
  • Embodiment 7 is the method of any one of Embodiments 1 to 6a, wherein the one or more MVA vectors together are administered at a total dose of about 1x10 7 to about 5x10 8 infectious units (IU) of the one or more MVA vectors, per administration.
  • IU infectious units
  • Embodiment 7a is the method of Embodiment 7, wherein the one or more MVA vectors together are administered at a total dose of about 2 x 10 8 IU of the one or more MVA vectors, per administration.
  • Embodiment 8 is the method of any one of Embodiments 1-7a, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 3-15 mg of vesatolimod, such as about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration.
  • Embodiment 8a is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 4 mg to about 6 mg of vesatolimod per administration.
  • Embodiment 8b is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 6 mg to about 8 mg of vesatolimod per administration.
  • Embodiment 8c is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 8 mg to about 10 mg of vesatolimod per administration.
  • Embodiment 8d is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 10 mg to about 12 mg of vesatolimod per administration.
  • Embodiment 8e is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 12 mg to about 14 mg of vesatolimod per administration.
  • Embodiment 9 is the method of any one of Embodiments 1-8e, wherein vesatolimod or a pharmaceutically acceptable salt thereof is administered multiple times at about 26-46 weeks, preferably at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks, after the Ad26 vaccine is initially administered.
  • Embodiment 9a is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 9b is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 9c is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 9d is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
  • Embodiment 9e is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 9f is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 8 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 10 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 10 is the method of any one of Embodiments 1-9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 11 is the method of any one of Embodiments 1-9, wherein vesatolimod or a pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26 and 28 weeks after the Ad26 vaccine is initially administered and at a total dose of 8 mg per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 12 is the method of any one of Embodiments 1-11, further comprising administering to the human subject an immunogenically effective amount of a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides.
  • Embodiment 12a is the method of Embodiment 12, wherein the gp140 vaccine is administered in combination with the Ad26 vaccine, preferably the re-administering of the Ad26 vaccine.
  • Embodiment 12b is the method of Embodiment 12, wherein the gp140 vaccine is administered in combination with the MVA vaccine.
  • Embodiment 12c is the method of Embodiment 12b, wherein the gp140 vaccine is administered in combination with the initial administration of the MVA vaccine.
  • Embodiment 12d is the method of Embodiment 12b, wherein the gp140 vaccine is administered in combination with the re-administration of the MVA vaccine.
  • Embodiment 12e is the method of Embodiment 12b, wherein the gp140 vaccine is administered in combination with the initial administration of the MVA vaccine and the re- administration of the MVA vaccine.
  • Embodiment 13 is the method of any one of Embodiments 12-12e, wherein the gp140 vaccine comprises two isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, respectively.
  • Embodiment 14 is the method of any one of Embodiments 12 to 13, wherein the one or more isolated HIV gp140 envelope polypeptides are administered at a total dose of about 125-350 mg of the total glycoproteins of the HIV gp140 envelope polypeptide(s), per administration.
  • Embodiment 14a is the method of any one of Embodiments 12 to 13, wherein the one or more isolated HIV gp140 envelope polypeptides are administered at a total dose of about 250 mg of the total glycoproteins of the HIV gp140 envelope polypeptide(s), per administration.
  • Embodiment 15 is a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
  • Ad26 adenovirus 26 vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors per administration;
  • step (ii) re-administering to the human subject the Ad26 vaccine in a total dose of about 5x10 9 to about 1x10 11 viral particles (vp), preferably about 5x10 10 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re- administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
  • an MVA vaccine comprising one or more MVA vectors, preferably one or more MVA-BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x10 7 to about 5x10 8 infectious units (IU), preferably about 2x10 8 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further administering to the human subject a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i); and
  • vesatolimod or the pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg of vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i); preferably at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is administered in step (i).
  • Embodiment 15a is the method of Embodiment 15, wherein the Ad26 vaccine comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
  • Embodiment 15b is the method of Embodiment 15 or 15a, wherein the MVA vaccine consists of a single MVA vector encoding the four HIV antigens.
  • Embodiment 15c is the method of Embodiment 15b, wherein the single MVA vector is an MVA-BN vector.
  • Embodiment 15d is the method of any one of Embodiments 15-15c, wherein:
  • the Ad26 vaccine is initially administered in a total dose of about 5x10 10 vp of the Ad26 vectors per administration;
  • the Ad26 vaccine is re-administered in a total dose of about 5x10 10 vp of the Ad26 vectors per administration at 12 weeks after the Ad26 vaccine is initially administered;
  • the MVA vaccine is initially administered in a total dose of about 2x10 8 IU of the one or more MVA vectors per administration at 24 weeks after the Ad26 vaccine is initially administered;
  • vesatolimod or the pharmaceutically acceptable salt thereof is administered at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is administered in step (i).
  • Embodiment 15e is the method of any one of Embodiment 15 to 15d, wherein each of the Ad26 vaccine and the MVA vaccine is intramuscularly administered, and vesatolimod or the pharmaceutically acceptable salt thereof is orally administered.
  • Embodiment 15f is the method of any one of Embodiments 15 to 15e, wherein the MVA vaccine is administered in combination with the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier.
  • Embodiment 15g is the method of Embodiment 15f, wherein the gp140 vaccine is administered intramuscularly.
  • Embodiment 15h is the method of Embodiment 15f or 15g, wherein the gp140 vaccine is administered at a total dose of about 250 mg of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration.
  • Embodiment 16 is the method of any one of Embodiments 15-15h, further comprising:
  • the MVA vaccine in a total dose of about 1x10 7 to about 5x10 8 infectious units (IU), preferably about 2x10 8 IU, of the one or more MVA vectors, per administration;
  • the gp140 vaccine optionally, in combination with the MVA vaccine, further re-administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration,
  • MVA vaccine and optionally the gp140 vaccine, is re- administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i).
  • Embodiment 16a is the method of Embodiment 16, wherein the MVA vaccine is re-administered in a total dose of about 2x10 8 IU of the one or more MVA vectors per administration at 36 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 16b is the method of Embodiment 16 or 16a, wherein the MVA vaccine is re-administered in combination with the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier.
  • Embodiment 16c is the method of Embodiment 16b, wherein the gp140 vaccine is administered intramuscularly.
  • Embodiment 16d is the method of Embodiment 16b or 16c, wherein the gp140 vaccine is re-administered at a total dose of about 250 mg of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration.
  • Embodiment 17 is the method of any one of Embodiments 15 to 16d, further comprising: re-administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof in a total dose of about 3 mg to about 15 mg per administration at 38- 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 17a is the method of Embodiment 17, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered.
  • Embodiment 17b is the method of Embodiment 17a, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered multiple times at about 26-46 weeks, preferably at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks, after the Ad26 vaccine is initially administered.
  • Embodiment 17c is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per
  • Embodiment 17d is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per
  • Embodiment 17e is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 17f is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
  • Embodiment 17g is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 17h is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 17i is the method of Embodiment 17b, wherein vesatolimod or a pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26 and 28 weeks after the Ad26 vaccine is initially administered and at a total dose of 8 mg per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 17j is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 8 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 10 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
  • Embodiment 18 is the method of any one of Embodiments 1 to 17i, wherein the human subject has initiated the ART during the acute phase of HIV infection.
  • Embodiment 18a is the method of any one of Embodiments 1 to 17i, wherein the human subject has initiated ART outside of the acute phase of HIV infection.
  • Embodiment 18b is the method of any one of Embodiments 1-18a, wherein the human subject has a chronic HIV infection.
  • Embodiment 19 is the method of any one of Embodiments 1 to 18b, wherein the human subject is on a suppressive ART during the treatment.
  • Embodiment 20 is the method of Embodiment 19, wherein the suppressive ART is discontinued after the treatment.
  • Embodiment 21 is a method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
  • Embodiment 22 is the method of Embodiment 21, further comprising
  • step (i) discontinuing the ART treatment of step (i) after step (ii).
  • Embodiment 23 is the method of Embodiment 22, wherein the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART.
  • Embodiment 24 is a combination for use in inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the combination comprising:
  • Ad26 adenovirus 26 vectors
  • an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors, preferably one or more MVA-BN vectors, together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and
  • MVA Modified Vaccinia Ankara
  • a composition comprising vesatolimod or a pharmaceutically acceptable salt thereof (VES).
  • VES pharmaceutically acceptable salt thereof
  • Embodiment 25 is the combination of Embodiment 24, further comprising a gp140 vaccine comprising at least one of two isolated HIV gp140 envelope polypeptides respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, and a pharmaceutically acceptable carrier.
  • Embodiment 25a is the combination of Embodiment 24, wherein the MVA vaccine consists of a single MVA vector, preferably a single MVA-BN vector, encoding the four HIV antigens.
  • Embodiment 25b is the combination of Embodiment 25, wherein the gp140 vaccine comprises the two isolated HIV gp140 envelope polypeptides.
  • Embodiment 26 is the combination of any one of Embodiments 25 to 25b, further comprising an adjuvant, preferably an aluminum adjuvant.
  • Embodiment 27 is the combination of Embodiment 26, wherein the adjuvant is formulated together with the gp140 vaccine or is to be co-administered with the gp140 vaccine.
  • Embodiment 28 is a kit comprising the combination of any one of Embodiments 24-27 and instructions on using the combination to induce an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART).
  • HIV human immunodeficiency virus
  • ART antiretroviral therapy
  • EXAMPLE 1 Study of HIV vaccine regimens in HIV-infected humans undergoing antiretroviral therapy (ART)
  • Screening for eligible participants will be performed within 70 days before administration of the study intervention.
  • the inclusion criteria for enrolling participants in this study include, for example,
  • HIV-1 infection including route and estimated duration of HIV infection, pre-ART viral set point, and ARV history).
  • HIV assay and either a negative/indeterminate HIV conventional Ab test or a negative/indeterminate Western Blot; or
  • ART is defined as a combination therapy regimen including more than 2 compounds, e.g.2x nucleoside reverse transcriptase inhibitors plus either non-nucleoside reverse transcriptase inhibitor or integrase inhibitor. Allowed ART as part of the current ARV regimen include nucleoside reverse transcriptase inhibitors (NRTIs), raltegravir, dolutegravir, rilpivirine, bictegravir, and maraviroc.
  • NRTIs nucleoside reverse transcriptase inhibitors
  • raltegravir dolutegravir
  • rilpivirine bictegravir
  • maraviroc maraviroc.
  • the study will enroll 90 participants randomized in a 1:1:1 ratio to 2 vaccine groups and 1 matching placebo group.
  • the study population will include HIV infected adults who are on suppressive ART for at least 48 weeks prior to screening and who have maintained undetectable viremia (HIV RNA ⁇ 50 cps/mL) for 312 weeks and maintain CD4 counts >450 cells/mm 3 prior to initiation of vaccine/placebo administration.
  • the study comprises of a screening period of 10 weeks, a 48-week treatment period (Stage 1) and a 48 week ATI period (Stage 2 and Stage 3).
  • a long-term extension period (Stage 4, 104 weeks [approximately 2 years] after week 96) might be added for participants in the active treatment groups, based on efficacy outcomes at week 72. Details on the assessments during this optional LTE phase will be defined at that time.
  • Week X refers to X weeks after the initial administration of the Ad26 vaccine at week 0.
  • Subjects receive four doses of study vaccine: adenovirus 26 vectors encoding mosaic HIV antigens (Ad26 . Mos4.HIV) or placebo is administered at weeks 0 and 12; and either (i) MVA vectors encoding mosaic HIV antigens (MVA-BN-HIV) (or placebo), or (ii) MVA-BN-HIV in combination with a mixture of adjuvanted HIV gp140 polypeptides (or placebo) is administered at weeks 24 and 36.
  • adenovirus 26 vectors encoding mosaic HIV antigens (Ad26 . Mos4.HIV) or placebo is administered at weeks 0 and 12
  • MVA vectors encoding mosaic HIV antigens
  • MVA-BN-HIV MVA vectors encoding mosaic HIV antigens
  • placebo placebo
  • Vesatolimod is administered orally at a dose of 6 mg (3 x 2-mg tablets) per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46, or at a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 26 and 28 and a dose of 8 mg (2 x 4 mg tablets) at weeks of 30, 32, 34, 38, 40, 42, 44, and 46.
  • vesatolimod is administered orally at (a) a dose of 4 mg (1 x 4 mg tablets) per administration at weeks 26 and 28, and a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 30, 32, 34, 38, 40, 42, 44, and 46; (b) a dose of 4 mg (1 x 4 mg tablets) per administration at weeks 26, 28 and 30, and a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 32, 34, 38, 40, 42, 44, and 46; (c) a dose of 4 mg (1 x 4 mg tablets) per administration at weeks 26, 28, 30 and 32, and a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 34, 38, 40, 42, 44, and 46; (d) a dose of 10 mg (5 x 2 mg tablets) per administration at weeks 26, 28 and 30, and a dose of 12 mg (6 x 2 mg tablets) per administration at weeks 32, 34, 38, 40, 42, 44, and 46; or (e) a dose of 8 mg (4 x)
  • Ad26.Mos4.HIV is composed of the following four vaccine products supplied pre- mixed in the same vial and administered in a 1:1:1:1 ratio of viral particles (vps): Ad26.Mos1Env, Ad26.Mos2SEnv, Ad26.Mos1Gag-Pol, and Ad26.Mos2Gag-Pol expressing HIV mosaic Env1 (SEQ ID NO: 1), mosaic Env2S (SEQ ID NO: 2), mosaic GagPol1 (SEQ ID: NO 3), and mosaic GagPol2 (SEQ ID NO: 4) genes, respectively; administered at a total dose of about 5 x 10 10 viral particles (vp) in 0.5 mL injection;
  • MVA-BN-HIV is an MVA-BN expressing Mos1.Gag-Pol (SEQ ID NO: 3),
  • gp140 HIV bivalent vaccine adjuvanted, containing 0.16 mg/mL Clade C gp140 (SEQ ID NO: 9), 0.15 mg/mL Mosaic gp140 (SEQ ID NO: 10) and 0.85 mg /mL aluminum (Al); administered at 125 microgram (mcg) of Clade C gp140 glycoprotein, 125 mcg Mosaic gp140 glycoprotein (corresponding with 80 mcg and 75 mcg of protein, respectively), and aluminum adjuvant (425 mcg Al) per 0.5 mL dose;
  • Vaccine placebo is 0.9% sodium chloride (0.5 mL injection);
  • VES placebo is lactose, administered at a total dose corresponding to the same number of tablets used for the VES treatment.
  • Subjects receive the study vaccines administered by intramuscular administration and VES oral dosing or placebo according to the schedule in Table 1 below:
  • Subjects in both the test and control groups continue to receive standard ART (e.g. at least three antiviral compounds, e.g. two nucleoside reverse transcriptase inhibitors plus either non-nucleoside reverse transcriptase inhibitor or protease inhibitor or integrase inhibitor) for HIV treatment during the study.
  • standard ART e.g. at least three antiviral compounds, e.g. two nucleoside reverse transcriptase inhibitors plus either non-nucleoside reverse transcriptase inhibitor or protease inhibitor or integrase inhibitor
  • Blood and optionally genital secretions are taken at specific clinical visits to assess immune responses (cellular and humoral immune responses) and viremic control throughout the study.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • AIDS & HIV (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Methods for inducing an immune response against Human Immunodeficiency Virus (HIV) in HIV-infected subjects undergoing antiretroviral therapy (ART) are described. The methods involve an adenovirus vector primer vaccine, booster vaccines of an adenovirus vector vaccine, a Modified Vaccinia Ankara virus (MV A) vector vaccine, optionally in combination with isolated HIV envelope polypeptides, and a TLR7 agonist such as vesatolimod or a pharmaceutically acceptable salt thereof.

Description

TITLE OF THE INVENTION
Methods for Inducing an Immune Response Against Human Immunodeficiency Virus
Infection in Subjects Undergoing Antiretroviral Treatment CROSS REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
[001] This application claims the benefit of U.S. Provisional Application No.
62/851,258, filed on May 22, 2019, the entire content of which is hereby incorporated by reference in its entirety.
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
[002] This application contains a sequence listing, which is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file name“125204- 0103_Sequence_Listing”, creation date of May 20, 2020 and having a size of 64 KB. The sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.
BACKGROUND OF THE INVENTION
[003] The number of new HIV infections and the number of acquired immunodeficiency syndrome (AIDS) related deaths are declining. Nevertheless, globally, an estimated 36.7 million people were living with human immunodeficiency virus (HIV) in 2016
(http://www.unaids.org/en/resources/fact-sheet), which is an increase from previous years as a result of the wider availability of life-saving antiretroviral therapies (ART).
[004] Despite its proven success at suppressing viral replication saving lives, there are significant challenges to initiating and maintaining ART for all of those HIV-infected patients that need it in the world. For example, ART does not eliminate the viral reservoir, and treatment is associated with an incomplete restoration of the host immune system. In particular, while ART facilitates CD4+ T cell reconstruction in the blood, there is only a limited improvement in the function of anti-HIV specific CD8+ T cell responses. Also, ART must be taken life-long with near perfect adherence in order to be effective. This places extreme pressure and costs on international donors and over-taxed health systems in developing countries where HIV prevalence rates are highest. Moreover, ART has both short-term and long-term side effects for users, and drug resistance rates rise as more people are on treatment for longer periods of time. Thus, alternative or complementary treatments, including a therapeutic vaccine, which could induce a true or“functional” cure of HIV infection and lessen or eliminate the need for lifelong ART for HIV infected individuals, would therefore be of great benefit. The concept of a“functional cure” includes therapeutic strategies that enable host control of the virus without the need for treatment.
[005] Accordingly, there is a need for improved methods of treating HIV-infected subjects, particularly novel therapies that seek to achieve a functional cure of HIV, including a therapeutic vaccine that preferably would improve immune responses to HIV and possibly allow at least some treated subjects to discontinue ART while maintaining viremic control. BRIEF SUMMARY OF THE INVENTION
[006] The invention relates to methods for inducing an immune response against human immunodeficiency virus (HIV) in HIV-infected subjects undergoing antiretroviral therapy (ART) with a first vaccine comprising adenovirus 26 (Ad26) vectors encoding mosaic HIV antigens, a second vaccine comprising Modified Vaccinia Ankara (MVA) vector(s) encoding the mosaic HIV antigens and optionally a third vaccine comprising isolated adjuvanted Clade C gp140 and Mosaic gp140 proteins, and a Toll-like receptor 7 (TLR-7) agonist, or a pharmaceutically acceptable salt thereof, preferably vesatolimod (VES) or a pharmaceutically acceptable salt thereof.
[007] In one general aspect, the invention relates to a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) administering to the human subject an immunogenically effective amount of an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
(ii) administering to the human subject an immunogenically effective amount of an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens (i.e. the HIV antigens of SEQ ID NOs: 1, 2, 3, and 4), and a pharmaceutically acceptable carrier; and (iii) administering to the human subject an effective amount of vesatolimod (VES) or a pharmaceutically acceptable salt thereof.
[008] In one embodiment, the method further comprises: re-administering an immunogenically effective amount of the Ad26 vaccine to the human subject. [009] In certain embodiments, the method further comprises: re-administering an immunogenically effective amount of the MVA vaccine to the subject.
[0010] In certain embodiments, vesatolimod or a pharmaceutically acceptable salt thereof is administered to the subject orally, at a dosage of about 3-15 mg, such as about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of
vesatolimod, per administration.
[0011] In certain embodiments, vesatolimod or a pharmaceutically acceptable salt thereof is administered to the subject repeatedly.
[0012] In certain embodiments, the Ad26 vaccine consists of a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
[0013] In some embodiments, the MVA vaccine consists of a single MVA vector encoding the four HIV antigens of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3 and SEQ ID NO:4.
[0014] In other embodiments, the MVA vaccine consists of more than one MVA vectors together encoding the four HIV antigens.
[0015] In certain embodiments, the first, second, third, and fourth Ad26 vectors together are administered intramuscularly at a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors; and the single MVA vector or the more than one MVA vectors together are administered intramuscularly at a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the MVA vector or vectors. Preferably, the MVA vector is an MVA-BN vector.
[0016] In some embodiments, the method further comprises administering to the human subject an immunogenically effective amount of a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides, and the gp140 vaccine is administered in combination with the Ad26 vaccine or the MVA vaccine, preferably the gp140 vaccine is administered in combination with the MVA vaccine. Preferably the one or more isolated HIV gp140 envelope polypeptides are selected from the group consisting of two HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10. More preferably, the gp140 vaccine comprises both HIV gp140 polypeptides having respectively the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10. Preferably, the gp140 vaccine further comprises, or is co-administered with, an adjuvant. [0017] In certain embodiments, the gp140 vaccine is administered at a total dose of about 125-350 mg, preferably about 250 mg, of the glycoprotein(s) of the HIV gp140 envelope polypeptide(s), per administration.
[0018] In particular embodiments, the Ad26 vaccine is re-administered at about 10-14 weeks, e.g., 12 weeks, after the Ad26 vaccine is initially administered.
[0019] In certain embodiments, the MVA vaccine is first administered at about 22-26 weeks, e.g., about 24 weeks, after the Ad26 vaccine is initially administered.
[0020] In certain embodiments, the MVA vaccine is re-administered at about 34-38 weeks, e.g., 36 weeks, after the Ad26 vaccine is initially administered.
[0021] In certain embodiments, vesatolimod, is administered biweekly from 26 to 34 weeks after the Ad26 vaccine is initially administered.
[0022] In certain embodiments, vesatolimod, is further administered biweekly from 38 to 46 weeks after the Ad26 vaccine is initially administered.
[0023] In another general aspect, the invention relates to a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) intramuscularly administering to the human subject an Ad26 vaccine
comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration;
(ii) intramuscularly re-administering to the human subject the Ad26 vaccine in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re-administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
(iii) intramuscularly administering to the human subject an MVA vaccine
comprising one or more MVA vectors, preferably one or more MVA-BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further administering a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i); and
(iv) orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i).
[0024] In certain embodiments, the method further comprises: intramuscularly re- administering to the human subject the MVA vaccine, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further re-administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine, and optionally the gp140 vaccine, is re-administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i).
[0025] In certain embodiments, the method further comprises: orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi- weekly at 38-46 weeks after the Ad26 vaccine is administered in step (i).
[0026] According to embodiments of the invention, vesatolimod or a pharmaceutically acceptable salt thereof is orally administered to the human subject biweekly at a total dose of about 3 mg to about 15 mg, such as at about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration, preferably at 26-34 and 38-46 weeks after the initial administration of the Ad26 vaccine.
[0027] In certain embodiments, the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[0028] In certain embodiments, the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[0029] In certain embodiments, the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[0030] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
[0031] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at 26 and 28 weeks after the initial administration of the Ad26 vaccine, and a total dose of 8 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[0032] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered. [0033] In some embodiments, the subject is a chronically HIV-infected subject. In some embodiments, the subject initiated ART outside of the acute phase of HIV infection. In preferred embodiments, the subject initiated ART during the acute phase of HIV infection.
[0034] The invention also relates to a vaccine combination for use in inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected subject undergoing antiretroviral therapy (ART); and use of a vaccine combination in the
manufacture of a medicament for inducing an immune response against a human
immunodeficiency virus (HIV) in an HIV-infected subject undergoing antiretroviral therapy (HIV).
[0035] In one embodiment, the vaccine combination comprises: (i) an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO:1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier; (ii) an MVA vaccine comprising one or more Modified Vaccinia Ankara vectors, preferably Modified Vaccinia Ankara Bavarian Nordic (MVA-BN) vectors, together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and (iii) VES or a pharmaceutically acceptable salt thereof.
[0036] In another embodiment, the vaccine combination further comprises a gp140 vaccine comprising at least one of isolated HIV Clade C and Mosaic gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, respectively, an aluminum adjuvant and a pharmaceutically acceptable carrier. Preferably the gp140 vaccine comprises both of the HIV Clade C and Mosaic gp140 envelope polypeptides.
[0037] In certain embodiments, the invention relates to methods for inducing an immune response against HIV-1 in an HIV-1 infected human subject being treated with antiretroviral therapy (ART), wherein the ART has successfully suppressed replication of HIV in the blood stream of the subject such that the subject can discontinue the ART and still maintains control of viral replication in the blood stream for at least 24 weeks after discontinuation of the ART. Control of viral replication can be measured using methods known in the art, such as by measuring HIV viral load (e.g., HIV RNA levels), in view of the present disclosure.
[0038] In certain embodiments, the invention relates to a method of providing an HIV remission in an HIV-infected human subject undergoing antiretroviral therapy (ART), comprising inducing an immune response against HIV in the subject to thereby control HIV infection after discontinuation of the ART, wherein the immune response against HIV is induced using a method according to an embodiment of the invention. Preferably, an ART- free viremic control, e.g., controlling the HIV viral load (and possibly reduce the HIV viral reservoir) in HIV patients, is achieved in at least 20%, preferably at least 30%, 40%, 50% of the human subjects, for at least 24 weeks, preferably longer, after a treatment with a method of the invention and discontinuation of the ART.
[0039] Another general aspect of the invention relates to a method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
(i) treating the human subject with an antiretroviral therapy (ART); and
(ii) inducing an immune response against the HIV in the human subject using a method of the invention. [0040] In certain embodiments, the method of treatment further comprises discontinuing the ART treatment of step (i), preferably after an immune response against the HIV is induced in the human subject by a method of the invention. Preferably, the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART. DETAILED DESCRIPTION OF THE INVENTION
[0041] Various publications, articles and patents are cited or described in the background and throughout the specification; each of these references is herein incorporated by reference in its entirety. Discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is for the purpose of providing context for the invention. Such discussion is not an admission that any or all of these matters form part of the prior art with respect to any inventions disclosed or claimed.
[0042] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention pertains. Otherwise, certain terms used herein have the meanings as set forth in the specification. All patents, published patent applications and publications cited herein are incorporated by reference as if set forth fully herein.
[0043] It must be noted that as used herein and in the appended claims, the singular forms “a,”“an,” and“the” include plural reference unless the context clearly dictates otherwise.
[0044] Unless otherwise indicated, the term“at least” preceding a series of elements is to be understood to refer to every element in the series. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the invention.
[0045] Throughout this specification and the claims which follow, unless the context requires otherwise, the word“comprise”, and variations such as“comprises” and
“comprising”, will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integer or step. When used herein the term“comprising” can be substituted with the term“containing” or “including” or sometimes when used herein with the term“having”.
[0046] When used herein“consisting of” excludes any element, step, or ingredient not specified in the claim element. When used herein,“consisting essentially of” does not exclude materials or steps that do not materially affect the basic and novel characteristics of the claim. Any of the aforementioned terms of“comprising”,“containing”,“including”, and “having”, whenever used herein in the context of an aspect or embodiment of the invention can be replaced with the term“consisting of” or“consisting essentially of” to vary scopes of the disclosure.
[0047] As used herein, the conjunctive term“and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by“and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or” as used herein. Concurrent applicability of more than one of the options is also understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or.”
[0048] As used herein,“subject” means a human, who will be or has been treated by a method according to an embodiment of the invention.
[0049] The terms“adjuvant” and "immune stimulant" are used interchangeably herein, and are defined as one or more substances that cause stimulation of the immune system. In some embodiments, an adjuvant is used to enhance an immune response to HIV antigens and antigenic HIV polypeptides of the invention.
[0050] As used herein, the terms and phrases“in combination,”“in combination with,” “co-delivery,” and“administered together with” in the context of the administration of two or more therapies or components to a subject refers to simultaneous administration of two or more therapies or components, such as a viral expression vector and an isolated antigenic polypeptide.“Simultaneous administration” can be administration of the two components at least within the same day. When two components are“administered together with” or “administered in combination with,” they can be administered in separate compositions sequentially within a short time period, such as 24, 20, 16, 12, 8 or 4 hours, or within 1 hour, or within 30 minutes, or within 10 minutes, or within 5 minutes, or within 2 minutes, or they can be administered in a single composition at the same time. In the typical embodiment, two components or therapies are administered in separate compositions. The use of the term“in combination with” does not restrict the order in which therapies or components are administered to a subject. For example, a first therapy or component (e.g. viral expression vector) can be administered prior to (e.g., 5 minutes to one hour before), concomitantly with or simultaneously with, or subsequent to (e.g., 5 minutes to one hour after) the administration of a second therapy (e.g., isolated HIV antigenic polypeptide).
[0051] The invention relates to methods of inducing an immune response against human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral treatment (ART). According to embodiments of the invention, an Ad26 vaccine comprises one or more adenovirus 26 (Ad26) vectors encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4. In some embodiments of the invention, an immunogenically effective amount of the Ad26 vaccine is administered to the subject more than once. According to the invention, an MVA vaccine comprises one or more Modified Vaccinia Ankara vectors, preferably one or more Modified Vaccinia Ankara Bavarian Nordic (MVA-BN) vectors, encoding the four HIV antigens, i.e. HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4. In some embodiments of the invention, an immunogenically effective amount of the MVA vaccine is administered to the subject more than once. In certain embodiments of the invention, a further vaccine is administered together with the re- administered Ad26 vaccine, or preferably together with the MVA vaccine. Preferably, the further vaccine is a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides. Preferably the gp140 vaccine comprises one or more of isolated HIV Clade C and Mosaic gp140 proteins having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO:10, respectively. More preferably the gp140 vaccine comprises both HIV Clade C and Mosaic gp140 proteins. In all embodiments of the invention, the human subjects are further administered vesatolimod or a pharmaceutically acceptable salt thereof. An“HIV vaccine” as used herein comprises the Ad26 vaccine and the MVA vaccine used and administered as described herein, and optionally further comprises the gp140 vaccine used and administered as described herein.
[0052] Human Immunodeficiency Virus (HIV)
[0053] Human immunodeficiency virus (HIV) is a member of the genus Lentivirinae, which is part of the family of Retroviridae. Two species of HIV infect humans: HIV-1 and HIV-2. HIV-1 is the most common strain of HIV virus and is known to be more pathogenic than HIV-2. As used herein, the terms“human immunodeficiency virus” and“HIV” refer, but are not limited to, HIV-1 and HIV-2. In preferred embodiments, the envelope proteins described herein refer to those present on HIV-1.
[0054] HIV is categorized into multiple clades with a high degree of genetic divergence. As used herein, the term“HIV clade” or“HIV subtype” refers to related human
immunodeficiency viruses classified according to their degree of genetic similarity. There are currently three groups of HIV-1 isolates: M, N and O. Group M (major strains) consists of at least ten clades, A through J. Group O (outer strains) can consist of a similar number of clades. Group N is a new HIV-1 isolate that has not been categorized in either group M or O.
[0055] The terms“chronic set point”,“set point in chronic HIV infection”,“viral load set point”, and“viral set point in chronic HIV infection” refer to the steady state HIV viral load established in the blood of an HIV infected human. The chronic set point can refer to a value of steady state HIV viral load after infection, following the introduction of antiretroviral therapy or treatment, including administration of ART, a TLR7 agonist, and/or an HIV vaccine described herein, or after cessation of antiretroviral therapy or treatment. A chronic set point can be determined in a single HIV infected human or determined as a median chronic set point in a cohort of HIV infected humans. When comparing two chronic set points, a first chronic set point can be a percentage of a second chronic set point or the second chronic set point can be a multiple of the first chronic set point. For example, a first chronic set point of 100 copies HIV-1 RNA per mL is 10% of a second chronic set point of 1000 copies HIV-1 RNA per mL, and can alternatively be described as a second chronic set point that is 10-fold higher than a first chronic set point.
[0056] A“viral rebound” refers to the observation that an undetectable HIV viral load in a virologically suppressed HIV infected human after treatment with ART often reverts to a detectable pre-therapy HIV viral load after cessation of ART. The viral rebound can occur within days or weeks, e.g., 4 weeks, after cessation of ART. A“delay in viral rebound” refers to a time period between the expected observation of viral rebound, e.g., 4 weeks, after cessation of ART as compared to the actual observed viral rebound, e.g., 12 weeks, after cessation of another therapy, e.g., administration of ART, a TLR7 agonist, and HIV vaccine according to the method described herein. In the above hypothetical example, the delay in viral rebound is 8 weeks after treatment of the ART, the TLR7 agonist, and the HIV vaccine. A delay in viral rebound can be determined in a single HIV infected human or determined as a median delay in viral rebound in a cohort of HIV infected humans.
[0057] According to embodiments of the invention, the methods described herein can be used to induce an immune response against one or more clades of HIV.
[0058] HIV antigens
[0059] As used herein, the terms“HIV antigen,”“antigenic polypeptide of an HIV,” “HIV antigenic polypeptide,”“HIV antigenic protein,”“HIV immunogenic polypeptide,” and “HIV immunogen” all refer to a polypeptide capable of inducing an immune response, e.g., a humoral and/or cellular mediated response, against HIV in a subject. The HIV antigen can be a protein of HIV, a fragment or epitope thereof, or a combination of multiple HIV proteins or portions thereof, that can induce an immune response against HIV in a subject. An HIV antigen is capable of raising in a host a protective immune response, e.g., inducing an immune response against a viral disease or infection, and/or producing an immunity in (i.e., vaccinates) a subject against a viral disease or infection, that protects the subject against the viral disease or infection. For example, the HIV antigen can comprise a protein or fragment(s) thereof from HIV, such as the HIV gag, pol and env gene products.
[0060] According to embodiments of the invention, the HIV antigen can be an HIV-1 or HIV-2 antigen or fragment(s) thereof. Examples of HIV antigens include, but are not limited to gag, pol, and env gene products, which encode structural proteins and essential enzymes. Gag, pol, and env gene products are synthesized as polyproteins, which are further processed into multiple other protein products. The primary protein product of the gag gene is the viral structural protein gag polyprotein, which is further processed into MA, CA, SP1, NC, SP2, and P6 protein products. The pol gene encodes viral enzymes (Pol, polymerase), and the primary protein product is further processed into RT, RNase H, IN, and PR protein products. The env gene encodes structural proteins, specifically glycoproteins of the virion envelope. The primary protein product of the env gene is gp160, which is further processed into gp120 and gp41. A heterologous nucleic acid sequence according to the invention preferably encodes a gag, env, and/or pol gene product, or portion thereof. According to a preferred embodiment, the HIV antigen comprises an HIV Gag, Env, or Pol antigen, or any portion or combination thereof, more preferably an HIV-1 Gag, Env, or Pol antigen, or any portion or combination thereof.
[0061] According to preferred embodiments of the invention, an HIV antigen is a mosaic HIV antigen. As used herein,“mosaic antigen” refers to a recombinant protein assembled from fragments of natural sequences. The“mosaic antigen” can be computationally generated and optimized using a genetic algorithm. Mosaic antigens resemble natural antigens, but are optimized to maximize the coverage of potential T-cell epitopes found in the natural sequences, which improves the breadth and coverage of the immune response.
[0062] Examples of mosaic HIV Gag-Pol-Env antigens include those described in, e.g., US20120076812, Barouch et al., Nat Med 2010, 16:319-323; Barouch et al., Cell 155:1-9, 2013; and WO 2017/102929, all of which are incorporated herein by reference in their entirety.
[0063] Preferably, the mosaic HIV antigens encoded by the vectors according to the invention comprise one or more of the amino acid sequences selected from the group consisting of SEQ ID NOs: 1-4 and 10. Alternative and/or additional HIV antigens could be encoded by the primer vaccine and/or the booster vaccine of the invention in certain embodiments, e.g. to further broaden the immune response.
[0064] In view of the present disclosure, a mosaic HIV antigen can be produced using methods known in the art. See, e.g., US20120076812, Fischer et al, Nat Med, 2007.13(1): p. 100-6; Barouch et al., Nat Med 2010, 16:319-323, all of which are incorporated herein by reference in their entirety.
[0065] Envelope Polypeptide
[0066] As used herein, each of the terms“envelope polypeptide,”“envelope
glycoprotein,”“env polypeptide,”“env glycoprotein,” and“Env” refers to, but is not limited to, the glycoprotein that is expressed on the surface of the envelope of HIV virions and the surface of the plasma membrane of HIV infected cells, or a fragment thereof that can induce an immune response or produce an immunity against HIV in a subject in need thereof.
[0067] The env gene encodes gp160, which is proteolytically cleaved into gp120 and gp41. More specifically, gp160 trimerizes to (gp160)3 and then undergoes cleavage into the two noncovalently associated fragments gp120 and gp41. Viral entry is subsequently mediated by a trimer of gp120/gp41 heterodimers. Gp120 is the receptor binding fragment and binds to the CD4 receptor on a target cell that has such a receptor, such as, e.g., a T- helper cell. Gp41, which is non-covalently bound to gp120, is the fusion fragment and provides the second step by which HIV enters the cell. Gp41 is originally buried within the viral envelope, but when gp120 binds to a CD4 receptor, gp120 changes its conformation causing gp41 to become exposed, where it can assist in fusion with the host cell. Gp140 is the uncleaved ectodomain of trimeric gp160, i.e., (gp160)3, that has been used as a surrogate for the native state of the cleaved, viral spike.
[0068] According to certain embodiments of the invention, isolated HIV envelope polypeptides (e.g., gp160, gp140, gp120, or gp41), preferably gp140 protein, and more preferably stabilized trimeric gp140 protein, can be administered, to enhance the immunity induced by expression vectors (e.g., adenovirus 26 and/or MVA vectors) alone.
[0069] As used herein, each of the terms“stabilized trimeric gp140 protein” and “stabilized trimer of gp140” refers to a trimer of gp140 polypeptides that includes a polypeptide sequence that increases the stability of the trimeric structure. The gp140 polypeptides can have or can be modified to include a trimerization domain that stabilizes trimers of gp140. Examples of trimerization domains include, but are not limited to, the T4- fibritin“foldon” trimerization domain; the coiled-coil trimerization domain derived from GCN4; and the catalytic subunit of E. coli aspartate transcarbamoylase as a trimer tag.
[0070] Examples of isolated antigenic polypeptide are stabilized trimeric gp140 such as those described in Nkolola et al 2010, J. Virology 84(7): 3270-3279; Kovacs et al, PNAS 2012, 109(30):12111-6; WO 2010/042942 and WO 2014/107744, all of which are incorporated by reference in their entirety.
[0071] In some embodiments of the invention, the“envelope polypeptide” or“envelope glycoprotein” is a mosaic envelope protein comprising multiple epitopes derived from one or more of Env polyprotein sequences of one or more HIV clades. For example, as used herein a“gp140 protein” can be a“mosaic gp140 protein” that contains multiple epitopes derived from one or more gp140 protein sequences of one or more HIV clades. Preferably, a mosaic gp140 protein is a stabilized trimeric gp140 protein.
[0072] In a preferred embodiment, a mosaic gp140 protein is a stabilized trimer of mosaic gp140 protein comprising the amino acid sequence of SEQ ID NO: 10.
[0073] In some embodiments of the invention, the envelope polypeptide” or“envelope glycoprotein” is an envelope protein derived from a particular HIV clade, such as HIV clade A, B, or C. For example, as used herein a“gp140 protein” can be a“clade C gp140 protein” that contains envelope protein sequence derived from HIV clade C. Preferably, a clade C gp140 protein is a stabilized trimeric clade C gp140 protein.
[0074] In a preferred embodiment, a clade C gp140 protein is a stabilized trimer of clade C gp140 protein comprising the amino acid sequence of SEQ ID NO: 9.
[0075] According to certain embodiments of the invention, a gp140 polypeptide, such as a stabilized trimeric gp140 protein can be administered together with viral expression vectors, e.g., adenovirus 26 and/or MVA vectors.
[0076] In certain embodiments of the invention, two gp140 proteins are administered to the same subject, preferably a clade C gp140 having the amino acid sequence of SEQ ID NO: 9 and a mosaic gp140 having the amino acid sequence of SEQ ID NO: 10. The two gp140 proteins can be together in one pharmaceutical composition, preferably administered together with an adjuvant, such as aluminum phosphate adjuvant. A preferred dose for the total amount of gp140 for administration to humans is between about 125 and 350 µg, such as 125, 150, 175, 200, 225, 250, 275, 300, 325, 350 µg, or any amount in between, preferably about 250 µg. If clade C gp140 and mosaic gp140 are both administered, a suitable dose would for instance be about 125 µg of each protein, to provide a total dose of 250 µg of gp140 protein for an administration to a human subject. As used herein, unless indicated otherwise, the amount of a gp140 polypeptide refers to the amount of the gp140 polypeptide measured as glycoprotein.
[0077] An isolated gp140 protein can be co-delivered or administered in combination with an adenovirus (e.g., Ad26) expression vector or MVA expression vector. According to a preferred embodiment, a gp140 protein and Ad26 or MVA vector are administered separately, as two distinct formulations. Alternatively, a gp140 protein can be administered with Ad26 or MVA together in a single formulation. Simultaneous administration or co- delivery can take place at the same time, within one hour, or within the same day.
Furthermore, a gp140 protein can be administered in an adjuvanted formulation. Suitable adjuvants can be, for example, aluminum phosphate or a saponin-based adjuvant, preferably aluminum phosphate adjuvant.
[0078] Antigenic polypeptides such as gp140 can be produced and isolated using any method known in the art in view of the present disclosure. For example, an antigenic polypeptide can be expressed from a host cell, preferably a recombinant host cell optimized for production of the antigenic polypeptide. According to an embodiment of the invention, a recombinant gene is used to express a gp140 protein containing mutations to eliminate cleavage and fusion activity, preferably an optimized gp140 protein with increased breadth, intensity, depth, or longevity of the antiviral immune response (e.g., cellular or humoral immune responses) generated upon immunization (e.g. , when incorporated into a
composition of the invention, e.g., vaccine of the invention) of a subject (e.g., a human). The optimized gp140 protein can also include cleavage site mutation(s), a factor Xa site, and/or a foldon trimerization domain. A leader/signal sequence can be operably linked to the N- terminal of an optimized gp140 protein for maximal protein expression. The leader/signal sequence is usually cleaved from the nascent polypeptide during transport into the lumen of the endoplasmic reticulum. Any leader/signal sequence suitable for a host cell of interest can be used. An exemplary leader/signal sequence comprises the amino acid sequence of SEQ ID NO: 11.
[0079] Adenovirus Vector
[0080] Primer vaccines, and in certain embodiments booster vaccines, used in the methods of the invention comprise one or more adenovirus vectors, particularly human adenovirus 26 vectors (Ad26) encoding one more mosaic HIV antigens. An adenovirus according to the invention belongs to the family of the Adenoviridae, and preferably is one that belongs to the genus Mastadenovirus. As used herein, the notation“rAd” means recombinant adenovirus, e.g.,“rAd26” refers to recombinant human adenovirus 26.
[0081] According to embodiments of the invention, an adenovirus is a human adenovirus serotype 26 (Ad26). An advantage of human adenovirus serotype 26 is that, so far, significant experience was obtained with such vectors in clinical trials, and this did not reveal that pre-existing neutralizing antibody responses against such vectors would cause substantial interference with desired vaccine-induced responses, e.g. against the encoded antigens in such vectors. Preferably, the adenovirus vector is a replication deficient recombinant viral vector, such as a replication deficient recombinant adenovirus 26 vector.
[0082] In certain embodiments, the recombinant adenovirus vector useful in the invention is derived mainly or entirely from Ad26 (i.e., the vector is rAd26). In some embodiments, the adenovirus is replication deficient, e.g., because it contains a deletion in the E1 region of the genome. For the adenoviruses derived from Ad26 used in the invention, it is typical to exchange the E4-orf6 coding sequence of the adenovirus with the E4-orf6 of an adenovirus of human subgroup C such as Ad5. This allows propagation of such adenoviruses in well- known complementing cell lines that express the E1 genes of Ad5, such as for example 293 cells, PER.C6 cells, and the like (see, e.g. Havenga, et al., 2006, J Gen Virol 87: 2135-43; WO 03/104467). However, such adenoviruses will not be capable of replicating in non- complementing cells that do not express the E1 genes of Ad5. Thus, in certain embodiments, the adenovirus is a human adenovirus of serotype 26, with a deletion in the E1 region into which the nucleic acid encoding one or more mosaic HIV antigens has been cloned, and with an E4 orf6 region of Ad5.
[0083] The preparation of recombinant adenoviral vectors is well known in the art.
Preparation of rAd26 vectors is described, for example, in WO 2007/104792 and in Abbink et al., (2007) Virol 81(9): 4654-63, both of which are incorporated by reference herein in their entirety. Exemplary genome sequences of Ad26 are found in GenBank Accession EF 153474 and in SEQ ID NO: 1 of WO 2007/104792, which is herein incorporated by reference in its entirety. Typically, an adenovirus vector useful in the invention is produced using a nucleic acid comprising the entire recombinant adenoviral genome (e.g., a plasmid, cosmid, or baculovirus vector).
[0084] The adenovirus vectors useful in the invention are typically replication deficient. In these embodiments, the virus is rendered replication deficient by deletion or inactivation of regions critical to replication of the virus, such as the E1 region. The regions can be substantially deleted or inactivated by, for example, inserting a gene of interest, such as a gene encoding an HIV antigen (usually linked to a promoter) within the region. In some embodiments, the vectors of the invention can contain deletions in other regions, such as the E3 region, or insertions of heterologous genes linked to a promoter within such regions.
Mutations in the E3 region of the adenovirus need not be complemented by the cell line, since E3 is not required for replication.
[0085] A packaging cell line is typically used to produce sufficient amounts of adenovirus vectors for use in the invention. A packaging cell is a cell that comprises those genes that have been deleted or inactivated in a replication deficient vector, thus allowing the virus to replicate in the cell. Suitable packaging cell lines include, for example, PER.C6, 911, and HEK293.
[0086] According to embodiments of the invention, HIV antigens can be expressed in the adenovirus 26 vectors described herein. Optionally, the heterologous gene encoding the mosaic HIV antigen can be codon-optimized to ensure proper expression in the treated host (e.g., human). Codon-optimization is a technology widely applied in the art. Typically, the heterologous gene encoding the mosaic HIV antigen is cloned into the E1 and/or the E3 region of the adenoviral genome. Non-limiting embodiments of codon optimized nucleotide sequences encoding HIV antigens with SEQ ID NOs: 1-4 are provided herein as SEQ ID NOs: 5-8, respectively.
[0087] According to embodiments of the invention, one or more adenovirus 26 (Ad26) vectors comprise nucleic acid that encodes one or more HIV antigens, in particular the one or more Ad26 vectors together encode four mosaic HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4. In certain embodiments, the Ad26 vaccine used in the invention comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4. In certain embodiments these vectors are present in a single composition in a 1:1:1:1 ratio (based on viral particles).
[0088] The heterologous gene encoding the mosaic HIV antigen can be under the control of (i.e., operably linked to) an adenovirus-derived promoter (e.g., the Major Late Promoter), or can be under the control of a heterologous promoter. Examples of suitable heterologous promoters include the cytomegalovirus immediate early (CMV) promoter and the Rous sarcoma virus (RSV) promoter. Preferably, the promoter is located upstream of the heterologous gene encoding the mosaic HIV antigen within an expression cassette. In a preferred embodiment, the heterologous promoter is a CMV promoter.
[0089] MVA vectors
[0090] In some embodiments, an MVA vaccine used in the methods of the invention comprises one or more Modified Vaccinia Ankara (MVA) vectors together encoding four mosaic HIV antigens, in particular the HIV antigens of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4. MVA vectors useful in the invention utilize attenuated virus derived from MVA virus, which is characterized by the loss of their capabilities to reproductively replicate in human cell lines.
[0091] MVA has been generated by more than 570 serial passages on chicken embryo fibroblasts of the dermal vaccinia strain Ankara (Chorioallantois vaccinia virus Ankara virus, CVA; for review see Mayr et al. (1975) Infection 3, 6-14) that was maintained in the
Vaccination Institute, Ankara, Turkey for many years and used as the basis for vaccination of humans. In 1976, MVA derived from MVA-571 seed stock (corresponding to the 571st passage) was registered in Germany as the primer vaccine in a two-stage parenteral smallpox vaccination program. As a result of the passaging used to attenuate MVA, there are a number of different strains or isolates, depending on the number of passages conducted in CEF cells. For example, MVA-572 was used in a small dose as a pre-vaccine in Germany during the smallpox eradication program, and MVA-575 was extensively used as a veterinary vaccine. MVA as well as MVA-BN lacks approximately 15% (31 kb from six regions) of the genome compared with ancestral CVA virus. The deletions affect a number of virulence and host range genes, as well as the gene for Type A inclusion bodies. MVA-575 was deposited on December 7, 2000, at the European Collection of Animal Cell Cultures (ECACC) under Accession No. V00120707.
[0092] Strains of MVA having enhanced safety profiles for the development of safer products, such as vaccines or pharmaceuticals, have been developed, for example by
Bavarian Nordic. MVA was further passaged by Bavarian Nordic and is designated MVA- BN. A representative sample of MVA-BN was deposited on August 30, 2000 at the
European Collection of Cell Cultures (ECACC) under Accession No. V00083008. MVA-BN is further described in WO 02/42480 (US 2003/0206926) and WO 03/048184 (US
2006/0159699), both of which are incorporated by reference herein in their entirety.
[0093] “Derivatives” or“variants” of MVA refer to viruses exhibiting essentially the same replication characteristics as MVA as described herein, but exhibiting differences in one or more parts of their genomes. For example, MVA-BN as well as a derivative or variant of MVA-BN fails to reproductively replicate in vivo in humans and mice, even in severely immune suppressed mice. More specifically, MVA-BN or a derivative or variant of MVA- BN has preferably also the capability of reproductive replication in chicken embryo fibroblasts (CEF), but no capability of reproductive replication in the human keratinocyte cell line HaCat (Boukamp et al (1988), J. Cell Biol.106: 761-771), the human bone osteosarcoma cell line 143B (ECACC Deposit No.91112502), the human embryo kidney cell line 293 (ECACC Deposit No.85120602), and the human cervix adenocarcinoma cell line HeLa (ATCC Deposit No. CCL-2). Additionally, a derivative or variant of MVA-BN has a virus amplification ratio at least two fold less, more preferably three-fold less than MVA-575 in Hela cells and HaCaT cell lines. Tests and assays for these properties of MVA variants are described in WO 02/42480 (US 2003/0206926) and WO 03/048184 (US 2006/0159699).
[0094] The term“not capable of reproductive replication” or“no capability of reproductive replication” is, for example, described in WO 02/42480, which also teaches how to obtain MVA having the desired properties as mentioned above. The term applies to a virus that has a virus amplification ratio at 4 days after infection of less than 1 using the assays described in WO 02/42480 or in U.S. Patent No.6,761,893, both of which are incorporated by reference herein in their entirety.
[0095] The term“fails to reproductively replicate” refers to a virus that has a virus amplification ratio at 4 days after infection of less than 1. Assays described in WO 02/42480 or in U.S. Patent No.6,761,893 are applicable for the determination of the virus amplification ratio.
[0096] The advantages of MVA-based vaccine include their safety profile as well as availability for large scale vaccine production. Furthermore, in addition to its efficacy, the feasibility of industrial scale manufacturing can be beneficial. Additionally, MVA-based vaccines can deliver multiple heterologous antigens and allow for simultaneous induction of humoral and cellular immunity.
[0097] MVA vectors useful for the invention can be prepared using methods known in the art, such as those described in WO/2002/042480, WO/2002/24224, US20110159036, US 8197825, etc., the relevant disclosures of which are incorporated herein by reference.
[0098] In another aspect, replication deficient MVA viral strains can also be suitable for use in the invention, such as strains MVA-572 and MVA-575, or any other similarly attenuated MVA strain. Also suitable can be a mutant MVA, such as the deleted
chorioallantois vaccinia virus Ankara (dCVA). A dCVA comprises del I, del II, del III, del IV, del V, and del VI deletion sites of the MVA genome. The sites are particularly useful for the insertion of multiple heterologous sequences. The dCVA can reproductively replicate (with an amplification ratio of greater than 10) in a human cell line (such as human 293, 143B, and MRC-5 cell lines), which then enable the optimization by further mutation useful for a virus-based vaccination strategy (see, e.g., WO 2011/092029).
[0099] In a preferred embodiment of the invention, the MVA vectors are MVA-BN vectors, such as that described in WO 2018/229711, which is incorporated herein by reference.
[00100] According to embodiments of the invention, the MVA vector(s) comprise a nucleic acid that encodes one or more HIV antigens having the amino acid sequences selected from the group consisting of SEQ ID NOs: 1-4. Preferably, the one or more MVA vectors together encode four mosaic HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4. A particularly useful but non-limiting example of an MVA vaccine that can be used for the present invention is MVA-mBN414, as described in example 7 of WO 2018/229711. [00101] Nucleic acid sequences encoding the mosaic HIV antigens can be inserted into one or more intergenic regions (IGR) of the MVA. In certain embodiments, the IGR is selected from IGR07/08, IGR 44/45, IGR 64/65, IGR 88/89, IGR 136/137, and IGR 148/149. In certain embodiments, less than 5, 4, 3, or 2 IGRs of the recombinant MVA comprise heterologous nucleotide sequences encoding an HIV antigen, such as a mosaic HIV antigen. The heterologous nucleotide sequences can, additionally or alternatively, be inserted into one or more of the naturally occurring deletion sites, in particular into the main deletion sites I, II, III, IV, V, or VI of the MVA genome. In certain embodiments, less than 5, 4, 3, or 2 of the naturally occurring deletion sites of the recombinant MVA comprise heterologous nucleotide sequences encoding mosaic HIV antigens.
[00102] The number of insertion sites of MVA comprising heterologous nucleotide sequences encoding HIV antigens can be 1, 2, 3, 4, 5, or more. In certain embodiments, the heterologous nucleotide sequences are inserted into 4, 3, 2, or fewer insertion sites.
Preferably, two insertion sites are used. In certain embodiments, three insertion sites are used. Preferably, the recombinant MVA comprises at least 2, 3, 4, 5, 6, or 7 genes inserted into 2 or 3 insertion sites.
[00103] The recombinant MVA viruses provided herein can be generated by routine methods known in the art. Methods to obtain recombinant poxviruses or to insert exogenous coding sequences into a poxviral genome are well known to the person skilled in the art. For example, methods for standard molecular biology techniques such as cloning of DNA, DNA and RNA isolation, Western blot analysis, RT-PCR and PCR amplification techniques are described in Molecular Cloning, A laboratory Manual (2nd Ed.) (J. Sambrook et al., Cold Spring Harbor Laboratory Press (1989)), and techniques for the handling and manipulation of viruses are described in Virology Methods Manual (B.W.J. Mahy et al. (eds.), Academic Press (1996)). Similarly, techniques and know-how for the handling, manipulation and genetic engineering of MVA are described in Molecular Virology: A Practical Approach (A.J. Davison & R.M. Elliott (Eds.), The Practical Approach Series, IRL Press at Oxford University Press, Oxford, UK (1993) (see, e.g., Chapter 9: Expression of genes by Vaccinia virus vectors)) and Current Protocols in Molecular Biology (John Wiley & Son, Inc. (1998) (see, e.g., Chapter 16, Section IV: Expression of proteins in mammalian cells using vaccinia viral vector)).
[00104] For the generation of the various recombinant MVAs disclosed herein, different methods can be applicable. The DNA sequence to be inserted into the virus can be placed into an E. coli plasmid construct into which DNA homologous to a section of DNA of the MVA has been inserted. Separately, the DNA sequence to be inserted can be ligated to a promoter. The promoter-gene linkage can be positioned in the plasmid construct so that the promoter-gene linkage is flanked on both ends by DNA homologous to a DNA sequence flanking a region of MVA DNA containing a non-essential locus. The resulting plasmid construct can be amplified by propagation within E. coli bacteria and isolated. The isolated plasmid containing the DNA gene sequence to be inserted can be transfected into a cell culture, e.g., of chicken embryo fibroblasts (CEFs), at the same time the culture is infected with MVA. Recombination between homologous MVA DNA in the plasmid and the viral genome, respectively, can generate an MVA modified by the presence of foreign DNA sequences.
[00105] According to a preferred embodiment, a cell of a suitable cell culture such as, e.g., CEF cells, can be infected with a poxvirus. The infected cell can be, subsequently, transfected with a first plasmid vector comprising a foreign or heterologous gene or genes, preferably under the transcriptional control of a poxvirus expression control element. As explained above, the plasmid vector also comprises sequences capable of directing the insertion of the exogenous sequence into a selected part of the poxviral genome. Optionally, the plasmid vector also contains a cassette comprising a marker and/or selection gene operably linked to a poxviral promoter.
[00106] Suitable marker or selection genes are, e.g., the genes encoding the green fluorescent protein, b-galactosidase, neomycin-phosphoribosyltransferase or other markers. The use of selection or marker cassettes simplifies the identification and isolation of the generated recombinant poxvirus. However, a recombinant poxvirus can also be identified by PCR technology. Subsequently, a further cell can be infected with the recombinant poxvirus obtained as described above and transfected with a second vector comprising a second foreign or heterologous gene or genes. In case, this gene shall be introduced into a different insertion site of the poxviral genome, the second vector also differs in the poxvirus- homologous sequences directing the integration of the second foreign gene or genes into the genome of the poxvirus. After homologous recombination has occurred, the recombinant virus comprising two or more foreign or heterologous genes can be isolated. For introducing additional foreign genes into the recombinant virus, the steps of infection and transfection can be repeated by using the recombinant virus isolated in previous steps for infection and by using a further vector comprising a further foreign gene or genes for transfection. [00107] Alternatively, the steps of infection and transfection as described above are interchangeable, i.e., a suitable cell can at first be transfected by the plasmid vector comprising the foreign gene and, then, infected with the poxvirus. As a further alternative, it is also possible to introduce each foreign gene into different viruses, co-infect a cell with all the obtained recombinant viruses and screen for a recombinant including all foreign genes. A third alternative is ligation of DNA genome and foreign sequences in vitro and reconstitution of the recombined vaccinia virus DNA genome using a helper virus. A fourth alternative is homologous recombination in E.coli or another bacterial species between a vaccinia virus genome cloned as a bacterial artificial chromosome (BAC) and a linear foreign sequence flanked with DNA sequences homologous to sequences flanking the desired site of integration in the vaccinia virus genome
[00108] The heterologous nucleic acid encoding one or more mosaic HIV antigens can be under the control of (i.e., operably linked to) one or more poxvirus promoters. In certain embodiments, the poxvirus promoter is a Pr7.5 promoter, a hybrid early/late promoter, or a PrS promoter, a PrS5E promoter, a synthetic or natural early or late promoter, or a cowpox virus ATI promoter.
[00109] In certain embodiments of the invention, an MVA vector useful for the invention expresses HIV antigens having the amino acid sequences of SEQ ID NO: 1 to SEQ ID NO: 4.
[00110] Immunogenic Compositions
[00111] Immunogenic compositions are compositions comprising an immunogenically effective amount of a purified or partially purified adenovirus 26 or MVA vector for use in the invention. The adenovirus 26 and MVA vectors can encode any mosaic HIV antigens in view of the present disclosure, and preferably encode one or more HIV antigens selected from the group consisting of SEQ ID NOs: 1-4. The one or more mosaic HIV antigens encoded by the adenovirus 26 vector can be different from, but preferably are the same as the one or more mosaic HIV antigens encoded by the MVA vector. Immunogenic compositions can be formulated as a vaccine, according to methods well known in the art. Such
compositions can include adjuvants to enhance immune responses. The optimal ratios of each component in the formulation can be determined by techniques well known to those skilled in the art in view of the present disclosure.
[00112] As used herein,“an immunogenically effective amount” or“immunologically effective amount” means an amount of a composition or vector sufficient to induce a desired immune effect or immune response in a subject in need thereof. In one embodiment, an immunogenically effective amount means an amount sufficient to induce an immune response in a subject in need thereof, preferably a safe and effective immune response in a human subject in need thereof. In another embodiment, an immunogenically effective amount means an amount sufficient to produce immunity in a subject in need thereof, e.g., provide a therapeutic effect against a disease such as HIV infection. An immunogenically effective amount can vary depending upon a variety of factors, such as the physical condition of the subject, age, weight, health, etc. An immunogenically effective amount can readily be determined by one of ordinary skill in the art in view of the present disclosure.
[00113] An immunogenically effective amount can be administered in a single step (such as a single injection), or multiple steps (such as multiple injections), or in a single
composition or multiple compositions. It is also possible to administer an immunogenically effective amount to a subject, and subsequently administer another dose of an
immunogenically effective amount to the same subject, in a so-called prime-boost regimen. This general concept of a prime-boost regimen is well known to the skilled person in the vaccine field. Further booster administrations can optionally be added to the regimen, as needed.
[00114] As general guidance, an immunogenically effective amount when used with reference to a recombinant viral vector can range from about 106 viral particles (vps), plaque forming units (pfus) or infectious units (IU) to about 1012 viral particles or infectious units, for example 106, 107, 108, 109, 1010, 1011, or 1012 viral particles or infectious units.
[00115] In one embodiment, an immunogenic composition is an Ad26 vaccine used for initial administration to induce an immune response. According to embodiments of the invention, an Ad26 vaccine comprises an immunogenically effective amount of one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, and a pharmaceutically acceptable carrier. The HIV antigens can be encoded by the same Ad26 vector or different Ad26 vector, such as one, two, three, four or more Ad26 vectors.
[00116] The immunogenically effective amount of the one or more Ad26 vectors can be about 108, 109, 1010, 1011, or 1012 viral particles (vps), preferably about 109 to 1011 viral particles, and more preferably about 1010 viral particles, such as for instance about 0.5 x 1010, 1 x 1010, 2 x 1010, 3 x 1010, 4 x 1010, 5 x 1010, 6 x 1010, 7 x 1010, 8 x 1010, 9 x 1010, or 10 x 1010 viral particles. In certain embodiments, the immunogenically effective amount is about 5 x109 to about 1 x 1011 viral particles, preferably about 5 x 1010 viral particles, such that the one or more Ad26 vectors are administered at a total dose of about 5 x109 to about 1 x 1011 viral particles per immunization step.
[00117] The immunogenically effective amount can be from one Ad26 vector or multiple Ad26 vectors. For example, a total administered dose of about 5 x109 to about 1 x 1011 viral particles, such as for instance about 5 x 1010 viral particles, in the Ad26 vaccine can be from four Ad26 vectors each encoding a different mosaic HIV antigen, such as those shown in SEQ ID NOs: 1, 2, 3, and 4.
[00118] In a particular embodiment, the immunogenically effective amount of Ad26 vectors together encoding SEQ ID NOs: 1, 2, 3, and 4 consists of four adenovirus vectors, namely a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
[00119] In such embodiments where an Ad26 vaccine comprises more than one Ad26 vector, the Ad26 vectors can be included in the composition in any ratio to achieve the desired immunogenically effective amount. Preferably, when the immunogenically effective amount of the Ad26 vectors consists of four Ad26 vectors, the first, second, third, and fourth Ad26 vectors are administered at a 1:1:1:1 ratio of viral particles (vps).
[00120] An MVA vaccine that is useful in the invention comprises an immunogenically effective amount of one or more MVA vectors together encoding the four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, and a pharmaceutically acceptable carrier. The HIV antigens expressed by MVA vectors can be encoded by a single MVA vector, or multiple MVA vectors, such as one, two, or more MVA vectors. In certain advantageous embodiments, the HIV antigens are expressed by a single MVA vector.
[00121] The immunogenically effective amount of the one or more MVA vectors in the MVA vaccine can be about 106, 107, 108, 109, or 1010 infectious units (IU), preferably about 107 to 109 IU, and more preferably about 2x 108 IU, such as for instance about 0.5 x 108, 1 x 108, 2 x 108, 3 x 108, 4 x 108, or 5 x 108 IU. In certain embodiments, the immunogenically effective amount is about 1 x107 to about 5 x 108 IU, preferably about 2 x 108 IU, such that the one or more MVA vectors are administered at a total dose of about 1 x107 to about 5 x 108 IU, preferably about 2 x 108 IU per immunization step. [00122] The immunogenically effective amount can be from one MVA vector or multiple MVA vectors. For example, in some embodiments, a total administered dose of about 1 x107 to about 5 x 108 IU, such as for instance about 1x107, 5x107, 1x108, 2 x 108, 5x 108 IU, or any dose in between, in the MVA vaccine can be from two MVA vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, for example a first MVA vector encoding the HIV antigens of SEQ ID NOs: 1 and 3, and a second MVA vector encoding the HIV antigens of SEQ ID NO: 2 and SEQ ID NO: 4, wherein preferably the first and second MVA vectors are administered at a 1:1 ratio of IU. In more preferred embodiments, a total administered dose of about 1 x107 to about 5 x 108 IU, such as for instance about 1x107, 5x107, 1x108, 2 x 108, 5x 108 IU, or any dose in between, in the MVA vaccine can be from a single MVA vector encoding four HIV antigens having the amino acid sequences of SEQ ID NOs: 1, 2, 3, and 4.
[00123] In some embodiments of the invention, an Ad26 vaccine or an MVA vaccine that is administered after the initial administration of the Ad26 vaccine, is administered in combination with one or more isolated HIV gp140 envelope polypeptides. According to embodiments of the invention, when used with reference to the total amount of the gp140 vaccine administered as part of an immunization, such as at least one of the isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 (clade C gp140 polypeptide) and SEQ ID NO: 10 (mosaic gp140 polypeptide), an immunogenically effective amount can range from, e.g. about 125 mg to 350 mg, e.g. about 125, 150, 200, 250, 300, or 350 µg of the one or more isolated HIV envelope polypeptides. In certain
embodiments, a vaccine composition comprising one or more Ad26 vectors or one or more MVA vectors is administered in combination with a gp140 vaccine composition comprising two isolated HIV envelope gp140 polypeptides, one clade C gp140 polypeptide having the amino acid sequence of SEQ ID NO: 9 and one mosaic gp140 polypeptide having the amino acid sequence of SEQ ID NO: 10, each one for instance present in about 125 µg per administration to a total of about 250 µg, all measured as glycoprotein.
[00124] The preparation and use of immunogenic compositions are well known to those of ordinary skill in the art. Liquid pharmaceutical compositions generally include a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil.
Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol can also be included. The immunogenic compositions used in the invention, can be formulated for administration according to any method known in the art in view of the present disclosure, and are preferably formulated for intramuscular administration.
[00125] The vaccine compositions of the invention can comprise other antigens. The other antigens used in combination with the adenovirus 26 and/or MVA vectors and/or gp140 polypeptides can be, for example, other HIV antigens and nucleic acids expressing them.
[00126] The immunogenic compositions useful in the invention can further optionally comprise adjuvants. Adjuvants suitable for co-administration in accordance with the invention should be ones that are potentially safe, well tolerated and effective in people. Non- limiting examples include QS-21, Detox-PC, MPL-SE, MoGM-CSF, TiterMax-G, CRL- 1005, GERBU, TERamide, PSC97B, Adjumer, PG-026, GSK-I, GcMAF, B-alethine, MPC- 026, Adjuvax, CpG ODN, Betafectin, Aluminium salts such as Aluminium Phosphate (e.g. AdjuPhos) or Aluminium Hydroxide, and MF59.
[00127] For example, a preferred adjuvant for administration together with isolated HIV envelope polypeptides is aluminum phosphate. According to embodiments of the invention, when used with reference to the total amount of aluminum phosphate in a gp140 vaccine composition comprising one or more HIV envelope polypeptides, the total amount of aluminum phosphate administered can range from, e.g. about 10 µg to about 1000 µg, e.g. about 200 µg to 650 µg, e.g. about 200, 250, 300, 350, 400, 425, 450, 475, 500, 550, or 600 µg, preferably about 425 µg of aluminum.
[00128] The immunogenic compositions used for generating an immune response according to embodiments of the invention comprise a pharmaceutically acceptable carrier, such as a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material can depend on the route of administration, e.g., intramuscular, subcutaneous, oral, intradermal, cutaneous, intramucosal (e.g., gut), intranasal or intraperitoneal routes.
Preferably, the pharmaceutically acceptable carrier included in the compositions of the invention is suitable for intramuscular administration.
[00129] TLR 7 Agonist Vesatolimod
[00130] Toll like receptor 7 (TLR 7) is one of the protein members of the family of receptors (toll like receptors) that recognize pathogen associated molecular patterns (PAMPs) on infectious agents and is expressed by the gene, HGNC:15631, in humans. [00131] In a particular embodiment, the TLR agonist useful for the invention is vesatolimod (VES), also known as GS-9620, which is a small molecule with the IUPAC name of 4-amino-2-butoxy-8-[[3-(pyrrolidin-1-ylmethyl)phenyl]methyl]-5,7-dihydropteridin- 6-one, or a pharmaceutically acceptable salt thereof. Vesatolimod is an agonist of the TLR 7 receptor leading to induction of the IFN response, cytokines and chemokines (National Center for Biotechnology Information. PubChem Database. Vesatolimod, CID=46241268, https://pubchem.ncbi.nlm.nih.gov/compound/46241268 (accessed on Apr.23, 2019)).
[00132] Vesatolimod has the structure:
Figure imgf000029_0001
.
[00133] Vesatolimod can be formulated with conventional carriers and excipients, which will be selected in accordance with ordinary practice. Tablets will contain excipients, glidants, fillers, binders and the like. Aqueous formulations are prepared in sterile form, and when intended for delivery by other than oral administration generally will be isotonic. All formulations will optionally contain excipients such as those set forth in the Handbook of Pharmaceutical Excipients (1986), herein incorporated by reference in its entirety. Excipients include ascorbic acid and other antioxidants, chelating agents such as EDTA, carbohydrates such as dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10.
[00134] The formulations include those suitable for the foregoing administration routes. The formulations can conveniently be presented in unit dosage form and can be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.), herein incorporated by reference in its entirety. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. [00135] Formulations of vesatolimod or a pharmaceutically acceptable salt thereof suitable for oral administration can be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient can also be administered as a bolus, electuary or paste.
[00136] Suitable routes for the administration of vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route can vary with for example the condition of the recipient. An advantage of vesatolimod compounds used in this invention is that they are orally bioavailable and can be dosed orally.
[00137] The effective dose of vesatolimod depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses) or against an active disease or condition, the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. The effective dose can be expected to be from about 0.0001 to about 10 mg/kg body weight per day, typically from about 0.001 to about 1 mg/kg body weight per day, more typically from about 0.01 to about 1 mg/kg body weight per day, even more typically from about 0.05 to about 0.5 mg/kg body weight per day. For example, the daily candidate dose for an adult human of approximately 70 kg body weight will range from about 0.05 mg to about 100 mg, or between about 0.1 mg and about 25 mg, or between about 0.4 mg and about 15 mg, and may take the form of single or multiple doses.
[00138] According to embodiments of the invention, vesatolimod or a pharmaceutically acceptable salt thereof is orally administered to the human subject biweekly at a total dose of about 3 mg to about 15 mg, such as at about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration.
[00139] The effective amount of vesatolimod or a pharmaceutically acceptable salt thereof can be orally administered to the human subject using any suitable dosage form. For example, 4 mg of vesatolimod or an equivalent amount of a pharmaceutically acceptable salt thereof can be orally administered with a single 4 mg tablet or two 2 mg tablets; 6 mg of vesatolimod or an equivalent amount of a pharmaceutically acceptable salt thereof can be orally administered with two 3 mg tablets or three 2 mg tablets; etc.
[00140] In certain embodiments, VES or a pharmaceutically acceptable salt thereof, can be administered 10 doses, preferably every 14 days, with one of the following dosage form and regimen:
(a) 1 x 4 mg tablets for doses 1-2 and 3 x 2 mg tablets for doses 3-10;
(b) 1 x 4 mg tablets for doses 1-3 and 3 x 2 mg tablets for doses 4-10;
(c) 1 x 4 mg tablets for doses 1-4 and 3 x 2 mg tablets for doses 5-10;
(d) 3 x 2 mg tablets for doses 1-10;
(e) 3 x 2 mg tablets for doses 1 and 2 and 2 x 4 mg tablets for doses 3-10;
(f) 5 x 2 mg tablets for doses 1-3 and 6 x 2 mg tablets for doses 4-10;
(g) 4 x 2 mg tablets for doses 1-3 and 5 x 2 mg tablets for doses 4-10; or
(h) other suitable dosage form and regimen.
[00141] Method of Inducing an Immune Response Against HIV Infection
[00142] The Ad26 and MVA vaccine compositions described above can be used in the methods of the invention described herein. The methods of the invention relate to inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected subject undergoing antiretroviral therapy. The methods of administering Ad and MVA vaccines according to embodiments of the invention are effective to induce an immune response against one or multiple clades of HIV.
[00143] The invention relates to a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) administering to the human subject an immunogenically effective amount of an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
(ii) optionally re-administering an immunogenically effective amount of the Ad26 vaccine to the human subject;
(iii) administering to the human subject an immunogenically effective amount of an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens (i.e. the HIV antigens of SEQ ID NOs: 1, 2, 3, and 4), and a pharmaceutically acceptable carrier;
(iv) administering to the human subject an effective amount of vesatolimod (VES) or a pharmaceutically acceptable salt thereof; and
(v) optionally, re-administering the MVA vaccine to the human subject.
[00144] In some embodiments of this aspect of the invention, the method further comprises administering to the human subject a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides in combination with the Ad26 vaccine or MVA vaccine in step (ii) and/or (iii) and/or (v). In such embodiments, the gp140 vaccine is preferably administered in steps (iii) and (v). In such embodiments, the method preferably further comprises administering to the human subject at least one isolated HIV gp140 envelope polypeptide selected from the group consisting of two trimeric HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO :10, in combination with the Ad26 vaccine or MVA vaccine in step (ii) and/or (iii) and/or (v), preferably in steps (iii) and (v). In such embodiments, the gp140 vaccine preferably comprises both HIV gp140 polypeptides, respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10.
[00145] In certain embodiments, a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), comprises:
(i) administering, preferably intramuscularly, to the human subject an Ad26
vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration; (ii) re-administering, preferably intramuscularly, to the human subject the Ad26 vaccine in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re-administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
(iii) administering, preferably intramuscularly, to the human subject an MVA
vaccine comprising one or more MVA vectors, preferably one or more MVA- BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA-BN vectors, per administration; optionally, in combination with the MVA vaccine, further administering a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i);
(iv) administering, preferably orally, to the human subject vesatolimod or a
pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i);
(v) re-administering, preferably intramuscularly, to the human subject the MVA vaccine, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per
administration; optionally, in combination with the MVA vaccine, further re- administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine, and optionally the gp140 vaccine, is administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i); and
(vi) re-administering, preferably orally, to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg, such as at a total dose of about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 38-46 weeks after the Ad26 vaccine is administered in step (i).
[00146] In a preferred embodiment, step (iii) comprises administering the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier, in combination with the MVA vaccine.
[00147] In another preferred embodiment, step (v) comprises re-administering the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier, in combination with the MVA vaccine.
[00148] In certain embodiments, the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00149] In certain embodiments, the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00150] In certain embodiments, the method comprises orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00151] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
[00152] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 6 mg of vesatolimod per administration at 26 and 28 weeks after the initial administration of the Ad26 vaccine, and a total dose of 8 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00153] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00154] In certain embodiments, vesatolimod or the pharmaceutically acceptable salt thereof is administered at a total dose of about 8 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 10 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00155] Any of the vaccine compositions described herein can be used in a method according to the invention. Embodiments of the Ad26 vaccine; MVA vaccine; Ad26 vectors; MVA vectors; HIV antigens encoded by the Ad26 and MVA vectors; isolated gp140 polypeptides, etc. that can be used in the methods of the invention are discussed in detail above and in the illustrative examples below.
[00156] According to embodiments of the invention,“inducing an immune response” when used with reference to the methods described herein encompasses causing a desired immune response or effect in a subject in need thereof against an HIV infection, preferably for therapeutic purposes.“Inducing an immune response” also encompasses providing a therapeutic immunity for treating against a pathogenic agent, i.e., HIV. As used herein, the term“therapeutic immunity” or“therapeutic immune response” means that the HIV-infected vaccinated subject is able to control an infection with the pathogenic agent, i.e., HIV, against which the vaccination was done. In one embodiment,“inducing an immune response” means producing an immunity in a subject in need thereof, e.g., to provide a therapeutic effect against a disease such as HIV infection. In certain embodiments,“inducing an immune response” refers to causing or improving cellular immunity, e.g., T cell response, against HIV. In certain embodiments,“inducing an immune response” refers to causing or improving a humoral immune response against HIV. In certain embodiments,“inducing an immune response” refers to causing or improving a cellular and a humoral immune response against HIV. Typically, the administration of the Ad26 and MVA vaccine compositions according to embodiments of the invention will have a therapeutic aim to generate an immune response against HIV after HIV infection or development of symptoms characteristic of HIV infection. In certain embodiments, the induced immune response in the subject in which ART has successfully suppressed replication of HIV in the blood stream is such that the subject can discontinue the ART and still maintains control of viral replication in the blood stream for at least 24 weeks after discontinuation of the ART.
[00157] The patient population for treatment according to the methods of the invention described herein is HIV-infected human subjects, particularly HIV-infected human subjects undergoing antiretroviral therapy (ART). The terms“HIV infection” and“HIV-infected” as used herein refer to invasion of a human host by HIV. As used herein,“an HIV-infected human subject” refers to a human subject in whom HIV has invaded and subsequently replicated and propagated within the human host, thus causing the human host to be infected with HIV or have an HIV infection or symptoms thereof. An“HIV-infected human subject” has been diagnosed with HIV infection, i.e., tests positive in a screen for HIV infection, e.g. using any assay that is US FDA-approved.
[00158] As used herein,“undergoing antiretroviral therapy” refers to a human subject, particularly an HIV-infected human subject, that is being administered, or who has initiated treatment with antiretroviral drugs. According to embodiments of the invention, the antiretroviral therapy (ART) is started prior to the first administration of the Ad26 vaccine , for instance, about 2 to 6 weeks prior, such as about 2, 3, 4, 5, or 6 weeks prior, or 2-48 months prior, such as about 2, 3, 5, 6, 8, 12, 16, 20, 24, 30, 36, 42, or 48 months prior, or longer. In certain embodiments the ART is started earlier than about 44-52 weeks, preferably earlier than about 48 weeks prior to the first administration of the Ad26 vaccine. In a subject undergoing antiretroviral therapy, the antiretroviral therapy is continued during
administration of the regimen of the invention. ART is considered“suppressive” as used herein if the subject has plasma HIV RNA levels at less than 50 copies/mL for a certain period of time, including the possibility of blips. The term“stable suppressive” ART as used herein means that the suppressive ART regimen is not modified for a certain period of time.
[00159] In certain embodiments, a human subject undergoing antiretroviral therapy is on current stable suppressive ART for at least twenty-four weeks, meaning that while receiving the same ART regimen the subject has plasma HIV ribonucleic acid (RNA) levels at less than 50 copies/mL for at least 24 weeks prior to initiation of a regimen according to the invention. However, the human subject can have one or more blips (i.e., instances) of plasma HIV RNA greater than 50 copies/ml to less than 200 copies/ml within this period, such as within the 24 week period prior to the initiation of the regimen, provided that screening immediately prior to initiation of the regimen is less than 50 copies/ml.
[00160] An HIV-infected subject can initiate ART during the acute phase of HIV infection, or outside of the acute phase of HIV infection. In a preferred embodiment, the subject initiated ART during the acute phase of HIV infection. The term“acute HIV infection” refers to the initial stage of HIV infection. In general, there are three stages of HIV infection: (1) acute HIV infection, (2) clinical latency, and (3) acquired
immunodeficiency syndrome (AIDS). During acute HIV infection, the host typically develops symptoms such as fever, swollen glands, sore throat, rash, muscle and joint aches and pains, headache, etc., as a result of the body’s natural response to the HIV infection. During the acute stage of infection, large amounts of the HIV virus are produced in the host, and CD4 levels can decrease rapidly, because the HIV uses CD4 to replicate and then subsequently destroys the CD4. Once the natural immune response of the host brings the level of HIV in the host to a stable level, also known as viral set point, CD4 count begins to increase, but likely not to pre-infection levels. Acute HIV infection is also characterized as Fiebig stages I, II, III, and IV as described in Fiebig et al.,“Dynamics of HIV viremia and antibody seroconversion in plasma donors: implications for diagnosis and staging of primary HIV infection.” AIDS (London, England) (2003) 17(13) 1871-1879, which is herein incorporated by reference in its entirety. In certain embodiments, an HIV-infected human subject that is subjected to a regimen of the invention, is characterized as Fiebig stage I, Fiebig stage II, Fiebig stage III, or Fiebig stage IV.
[00161] Acute HIV infection is typically within two to four weeks after a host is exposed to and infected with HIV and continues for an additional two to four weeks. The acute HIV infection stage lasts until the host creates its own antibodies against HIV, at which point the clinical latency stage begins. During the clinical latency stage, HIV is living or developing in the host without causing any symptoms, or only causing mild symptoms. HIV reproduces at very low levels during the clinical latency stage, although the HIV is still active. The clinical latency stage is sometimes also referred to as“chronic HIV infection” or“asymptomatic HIV infection.” Chronic HIV infection is characterized as Fiebig stage VI. In certain
embodiments, an HIV-infected human subject that is subjected to a regimen of the invention, is characterized as Fiebig stage V, or Fiebig stage VI.
[00162] Chronic HIV infection (i.e., Fiebig stage VI) typically begins about 100 days (i.e., about 14 weeks) after a host is exposed to and infected with HIV. A subject infected with HIV that has progressed to Fiebig stage VI can be referred to or described as a“chronically- infected subject,”“a chronic HIV-infected subject,” or“a subject having chronic HIV infection.” A subject initiating ART outside of the acute or early phase of HIV infection is one who has not begun ART before entering Fiebig VI stage. Whether or not a subject has initiated ART prior to entering Fiebig VI stage of HIV infection can be determined by a clinician based on the subject’s available medical history and laboratory data at the time of HIV diagnosis.
[00163] As used herein, a subject who initiates ART outside of the acute phase of HIV infection (i.e., during chronic HIV infection) begins treatment with antiretroviral drugs at the earliest at about 12-16 weeks, after being exposed to and infected with HIV, such as about 12, 13, 14, 15, or 16 weeks or later, after exposure and infection with HIV. In contrast, a subject who initiates ART during acute HIV infection typically begins treatment with antiretroviral drugs at or prior to about 2 weeks to about 8 weeks after being exposed to and infected with HIV, such as about 1, 2, 3, 4, 5, 6, 7, or 8 weeks after exposure and infection. Thus, chronic HIV infection is thought to be more difficult to treat than acute HIV infection, at least because a chronically infected HIV subject typically has larger HIV viral reservoirs than an acutely infected subject due to the longer period of infection prior to initiating any treatment. Subjects who began ART during acute HIV infection and have plasma HIV RNA levels of less than 50 copies/ml for at least 24 weeks, preferably at least 48 weeks, have low HIV viral reservoirs and/or lower involvement of their viral reservoirs with HIV, and therefore have a higher chance for maintained viral suppression in the absence of ART, i.e., HIV remission. However, due to the differences in progression of acute and chronic HIV infection, it is not certain as to whether therapies effective to treat acute HIV infection will likewise be effective to treat chronic HIV infection.
[00164] In some embodiments of the invention, the HIV-infected subject is a chronically HIV-infected subject. A chronically HIV-infected subject can initiate ART at any phase of infection, such as during the acute phase of HIV infection or outside the acute phase of HIV infection. Preferably, a chronically HIV-infected subject initiates ART during the acute phase of HIV infection. In preferred embodiments of the invention, the HIV-infected human subject is in the acute phase of HIV infection.
[00165] A subject undergoing ART can be administered or treated with any antiretroviral drugs known in the art in view of the present disclosure. ART are medications that treat HIV, although the drugs do not kill the virus or remove the virus from the body. However, when taken in combination they can prevent the growth of the virus. When the virus is slowed down, so is HIV disease. Antiretroviral drugs are referred to as ARV. Combination ARV therapy (cART) is referred to as highly active ART (HAART). Typically, an ART regimen includes at least three antiviral compounds, e.g., two different reverse transcriptase inhibitors plus either a non-nucleoside reverse transcriptase inhibitor or protease inhibitor or integrase inhibitor.
[00166] One of ordinary skill in the art will be able to determine the appropriate antiretroviral treatment, frequency of administration, dosage of the ART, etc. so as to be compatible with simultaneous administration of the regimens of the invention. Examples of antiretroviral drugs used for ART include, but are not limited to nucleoside reverse transcriptase inhibitors (NRTIs, non-limiting examples of which include zidovudine, didanosine, stavudine, lamivudine, abacavir, tenofovir, combivir [combination of zidovudine and lamivudine], trizivir [combination of zidovudine, lamivudine and abacavir],
emtricitabine, truvada [combination of emtricitabine and tenofovir], and epzicom
[combination of abacavir and lamivudine]), non-nucleoside reverse transcriptase inhibitors (NNRTIs, non-limiting examples of which include nevirapine, delavirdine, efavirenz, etravirine, and rilpivirine), protease inhibitors (PIs, non-limiting examples of which include saquinavir, indinavir, ritonavir, nelfinavir, amprenavir, lopinavir/ritonavir, atazanavir, fosamprenavir, tipranavir, darunavir), integrase inhibitors (INSTIs, non-limiting examples including raltegravir, elvitegravir, and dolutegravir), and fusion inhibitors, entry inhibitors and/or chemokine receptor antagonists (FIs, CCR5 antagonists; non-limiting examples including enfuvirtide, aplaviroc, maraviroc, vicriviroc, and cenicriviroc).
[00167] According to embodiments of the invention, an Ad26 vaccine or an MVA vaccine is administered after the Ad26 vaccine is initially administered. In certain embodiments of the invention, the Ad26 vaccine or MVA vaccine is re-administered or administered at about 12-52 weeks, such as, about 8-15 weeks, about 21-27 weeks, and/or about 33-39 weeks, after the Ad26 vaccine is initially administered. One of ordinary skill in the art will be able to vary the exact timing of the vaccines, frequency of administration thereof, dosage thereof, etc., based upon the teachings herein and clinical experience.
[00168] According to embodiments of the invention, Ad26 vaccine is administered at least once and MVA vaccine at least once. According to embodiments of the invention, Ad26 vaccine is re-administered at about 10-14 weeks, and MVA vaccine is first administered 22- 26 weeks, such as 22, 23, 24, 25, or 26 weeks, after the Ad26 vaccine is initially administered. In certain embodiments, the Ad26 vaccine is re-administered at about 12 weeks after the Ad26 vaccine is initially administered, the MVA vaccine is first administered at about 24 weeks after the Ad26 vaccine is initially administered. In certain embodiments, the MVA vaccine is re-administered at about 32-40 weeks, such as at about 36 weeks, after the Ad26 vaccine is initially administered. In certain embodiments, vesatolimod or a pharmaceutically acceptable salt thereof is administered for the first time after the first administration of the MVA vaccine. In other embodiments, additional vesatolimod or a pharmaceutically acceptable salt thereof is administered after the second administration of the MVA vaccine. In certain embodiments, vesatolimod or a pharmaceutically acceptable salt thereof is administered several times in a series of administrations with about two-week intervals. In certain embodiments, the first administration of such series is about two weeks after the first and/or after the second administration of the MVA vaccine.
[00169] In some preferred embodiments, the Ad26 vaccine is re-administered after the Ad26 vaccine is initially administered, and in such embodiments preferably re-administered before the MVA vaccine is first administered. For example, the Ad26 vaccine can be re- administered at about 10-14 weeks after the Ad26 vaccine is initially administered, such as about 10, 11, 12, 13, or 14 weeks, after the Ad26 vaccine is initially administered, preferably at about 12 weeks after the Ad26 vaccine is initially administered.
[00170] In other preferred embodiments, the MVA vaccine is administered after the Ad26 vaccine is re-administered. In certain embodiments, the MVA vaccine is first administered at about 22 to 26 weeks, such as 22, 23, 24, 25, or 26 weeks, after the Ad26 vaccine is initially administered, preferably at about 24 weeks after the Ad26 vaccine is initially administered. The MVA vaccine can in certain embodiments be re-administered, e.g. at about 32 to 40 weeks, such as 32, 33, 34, 35, 36, 37, 38, 39, or 40 weeks, after the Ad26 vaccine is initially administered. In certain preferred embodiments, the MVA vaccine is re-administered at about 36 weeks after the Ad26 vaccine is initially administered.
[00171] Further administrations are possible, and embodiments of the disclosed methods also contemplate administration of such additional immunizations with immunogenic compositions containing Ad26 vectors, MVA vectors, and/or HIV gp140 polypeptides. Any of the Ad26 vectors, MVA vectors, and HIV gp140 polypeptides described herein can be used in additional immunizations.
[00172] The vaccine compositions can be administered by any method known in the art in view of the present disclosure, and administration is typically via intramuscular, intradermal or subcutaneous administration, preferably intramuscular administration. Intramuscular administration can be achieved by using a needle to inject a suspension or solution of the adenovirus and/or MVA vectors, and/or gp140 polypeptides. An alternative is the use of a needleless injection device to administer the composition (using, e.g., BiojectorTM) or a freeze-dried powder containing the vaccine.
[00173] Other modes of administration, such as intravenous, cutaneous, intradermal, oral, intratracheal, or nasal are also envisaged. For intravenous, cutaneous or subcutaneous injection, the vector will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of ordinary skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, and Lactated Ringer's Injection. Preservatives, stabilizers, buffers, antioxidants and/or other additives can be included, as required. A slow- release formulation can also be employed.
[00174] In certain embodiments, a method of inducing an immune response according to the invention further comprises administering a latent viral reservoir purging agent. Cells latently infected with HIV carry integrated virus that is transcriptionally silent, making it difficult to effectively eradicate HIV infection in treated subjects. As used herein,“reservoir purging agent” and“latent viral reservoir purging agent” refer to a substance that reduces the latent pool of HIV by reactivating HIV reservoirs, such as by inducing expression of quiescent HIV. Examples of latent viral reservoir purging agents suitable for use with the invention include, but are not limited to, histone deacetylase (HDAC) inhibitors and modulators of toll-like receptors (e.g., TLR7), such as those described in WO2016/007765 and WO2016/177833, which are herein incorporated by reference in their entireties. The latent viral reservoir purging agent can be administered before, after, or co-administered (i.e., administered in combination) with one or more of the vaccine immunizations described herein. The vaccination of a combination of adenovirus 26 vectors encoding Gag, Pol and Env antigens as a prime, followed by MVA vectors encoding such antigens as a boost, in combination with TLR7 stimulation has shown to result in improved virologic control and delayed viral rebound following discontinuation of antiretroviral therapy in rhesus monkey model studies, demonstrating the potential of therapeutic vaccination combined with innate immune stimulation to aim at functional cure for HIV infection (Borducchi E.N., et al, 2016, Nature 540: 284-287 (doi: 10/1038/nature20583)), the content of which is incorporated herein by reference in its entirety. [00175] Accordingly, another general aspect of the invention relates to a method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
(i) treating the human subject with an antiretroviral therapy (ART); and
(ii) inducing an immune response against the HIV in the human subject using a method according to an embodiment of the invention.
[00176] In certain embodiments, the method of treatment further comprises discontinuing the ART treatment of step (i), preferably after an immune response is induced by a method of the invention. Preferably, the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART.
[00177] In certain embodiments, subjects undergo interruption (also referred to as discontinuation, used interchangeably herein) of ART after completion of a regimen according to embodiments of the invention. In some embodiments, subjects can undergo antiretroviral analytical treatment interruption (ARV ATI) after completion of a regimen according to embodiments of the invention.“Antiretroviral analytical treatment interruption” and“ARV ATI” as used in the invention refer to discontinuation of treatment with antiretroviral drugs in order to assess viral suppression and viremic control in the absence of continued ART. Typically, subjects can undergo ARV ATI, i.e., ART can be discontinued, for example when the subject has plasma HIV RNA levels at less than 50 copies/mL for at least about 52 weeks, but a subject can still undergo ARV ATI even if the subject has one or more blips (i.e., instances) of plasma HIV RNA greater than 50 copies/ml to less than 200 copies/ml within this period, provided that the screening immediately prior to ARV ATI shows less than 50 copies/ml of plasma HIV RNA. HIV viral load, e.g., plasma HIV RNA levels, can be measured using known methods in view of the present disclosure, for example, using the Abbott RealTime HIV-1 viral load assay, or the Roche Cobas Taqman HIV-1 viral load assay.
[00178] According to embodiments of the invention, the ART can be stopped at about 10- 14 weeks, such as 10, 11, 12, 13, or 14 weeks after the last MVA vaccine is administered. In certain embodiments, the last MVA vaccine is administered at about 34-38 weeks after the Ad26 vaccine is initially administered. In these embodiments, the ART can be stopped at about 46 to 50 weeks, such as 46, 47, 48, 59, or 60 weeks, after the Ad26 vaccine is initially administered, and preferably about 60 weeks after the Ad26 vaccine is initially administered. In other embodiments, for subjects who are on non-nucleoside reverse transcriptase inhibitor (NNRTI)-based ART, a boosted protease inhibitor can be administered in place of the NNRTI for about 1-2 weeks prior to stopping ART to reduce the risk of developing NNRTI resistance. It is also possible to administer an activator (e.g. a histone deacetylase inhibitor or TLR7 modulator) during the ATI stage to activate any (e.g. latent) HIV reservoir and thereby improve the immune response.
[00179] Subjects undergoing ARV ATI can be monitored, e.g., by measuring plasma HIV RNA levels. As a non-limiting example, monitoring after the initiation of ARV ATI can occur up to two times per week during the first six weeks when rebound viremia is most likely to occur.“Rebound viremia” is for example defined as plasma HIV RNA levels of greater than 1,000 copies/ml after ARV ATI. ART can be re-initiated in subjects with rebound viremia. Preferably, a subject treated according to the methods of the invention will maintain viremic control after ART interruption. As used herein,“maintain viremic control” is in exemplary embodiments defined as at least 24 weeks with plasma HIV RNA of less than 50 copies/mL after ARV ATI. The“maintained viremic control” criterion is in certain exemplary embodiments still deemed to be met if there are one or more instances of plasma HIV RNA greater than 50 copies/ml to less than 1000 copies/ml, as long as the subject does not have plasma HIV RNA levels above 1000 copies/ml on two consecutive determinations at least one week apart.
[00180] Typically (not using the methods of the instant invention) human HIV-infected subjects have a return of viremia after 2-3 weeks following ART interruption. Without wishing to be bound by any theories, it is believed that therapy using the vaccine
compositions and vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention among individuals with fully suppressed HIV will result in a measurable immune response and maintain viremic control after ARV ATI. In some embodiments, subjects can discontinue ART after being treated according to a method of the invention. Discontinuation of ART can be for long periods of time (e.g., at least 24 weeks, preferably longer, e.g. at least about 28, 32, 36, 40, 44, 48, 52 weeks, 16 months, 18, 20, 22, 24 months, or even longer). Such periods of time in which ART is stopped or discontinued are referred to as a“holiday” or“ART holiday” or“treatment holiday”. In other
embodiments, vaccine and TLR7 therapy according to the methods of the invention can provide HIV remission, meaning that viral suppression is maintained in the absence of ART. In certain embodiments of the invention, a human subject that received the vaccines and vesatolimod or a pharmaceutically acceptable salt thereof according to the invention, discontinues ART and maintains viral suppression for at least 24 weeks after discontinuing ART.
[00181] In one exemplary regimen of the invention, an Ad26 vaccine comprising one or more adenovirus 26 vectors is administered (e.g., intramuscularly) in an amount of about 100 µl to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 108 to 1012 virus particles/ml. The initial Ad26 vaccination is optionally repeated and is followed by an MVA vaccine comprising one more MVA vectors administered (e.g., intramuscularly) in an amount of about 100 µl to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 106 to 109 pfu/ml. After administration of the MVA vaccine, vesatolimod or a pharmaceutically acceptable salt thereof is administered, preferably followed by a further administration of the MVA vaccine, and further administrations of vesatolimod or a pharmaceutically acceptable salt thereof.
[00182] In another exemplary regimen of the invention, an Ad26 vaccine comprising one or more adenovirus 26 vectors is administered (e.g., intramuscularly) in an amount of about 100 µl to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 108 to 1012 virus particles/ml. The initial Ad26 vaccination is followed by re- administration of the Ad26 vaccine comprising one more adenovirus 26 vectors administered (e.g., intramuscularly) in an amount of about 100 µl to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 108 to 1012 virus particles/ml. This is followed by administration of the MVA vaccine (e.g. intramuscularly) in an amount of about 100 µl to about 2 ml, preferably about 0.5 ml, of a solution containing concentrations of about 106 to 109 pfu/ml, in some embodiments in combination with one or more isolated HIV gp140 polypeptides in an amount of about 100 µl to about 2 ml, preferably about 0.5 ml, of a solution, to a total dose per administration of about 250 mg polypeptide and aluminum phosphate adjuvant (425 microgram (µg) aluminum per dose). The administration of the MVA vaccine is followed by repeated administration of vesatolimod or a pharmaceutically acceptable salt thereof. In preferred embodiments this is followed by another administration of the MVA vaccine, and thereafter further repeated administrations of vesatolimod or a pharmaceutically acceptable salt thereof.
[00183] The skilled artisan (e.g., practitioner) will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. [00184] The invention also relates to a vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination for use in inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), wherein the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination comprises an Ad26 vaccine and an MVA vaccine and vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention. The invention yet further relates to use of a vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination in the manufacture of a medicament for inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), wherein the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination comprises an Ad26 vaccine and an MVA vaccine and vesatolimod or a pharmaceutically acceptable salt thereof according to embodiments of the invention. All aspects and embodiments of the invention as described herein with respect to methods of inducing an immune response against a human
immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART) can be applied to the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combinations for use and/or uses of the vaccine and vesatolimod or a pharmaceutically acceptable salt thereof combination in the manufacture of a medicament for inducing an immune response against HIV in an HIV-infected subject undergoing ART.
[00185] A clinical improvement of a treated HIV-infected human above a comparator HIV-infected human treated with a standard of care is expected. The clinical improvement can include one or more of a lower peak viral load, a lower chronic set point, or an increased delay in viral rebound.
[00186] In some embodiments, the method as described herein has an effect on treatment of the HIV infection, for example, as determined by a lower peak viral load as compared to standard therapies, e.g., ART only. As is commonly understood in the art, comparison of a first peak viral load in a first HIV-infected human and a second peak viral load in a second HIV-infected human is measured during the same time period. In some embodiments, the measurement is performed after cessation of all antiviral therapies. In some embodiments, the viral load is maintained at an undetectable level in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
[00187] In some embodiments, a first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second peak viral load in a second HIV-infected human after treatment with ART only. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART only is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART only is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some
embodiments, the second peak viral load in a second HIV-infected human after treatment with ART only is about 1000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
[00188] In some embodiments, a first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 20 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
[00189] In some embodiments, a first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second peak viral load in a second HIV-infected human after treatment with ART and the HIV vaccine is about 100 times higher than the first peak viral load in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
[00190] In some embodiments, the method as described herein has an effect on treatment of the HIV infection, for example, as determined by a lower chronic set point as compared to standard therapies, e.g., ART. As is commonly understood in the art, comparison of a first chronic set point in a first HIV-infected human and a second chronic set point in a second HIV-infected human is measured at the same time point. In some embodiments, the measurement is performed after cessation of all antiviral therapies.
[00191] In some embodiments, a first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second chronic set point in a second HIV-infected human after treatment with ART only. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART only is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second chronic set point in a second HIV- infected human after treatment with ART only is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first chronic set point in a first HIV- infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART only is about 10 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. [00192] In some embodiments, a first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART and the TLR7 agonist is about 2 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine.
[00193] In some embodiments, a first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is lower than a second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine is higher, e.g., from about 1.2 to about 10000 times, from about 2 to about 10000 times, from about 5 to about 10000 times, from about 10 to about 10000 times, higher, than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine is about 1.2, about 1.5, about 2, about 3, about 4, about 5, about 10, about 20, about 50, about 100, about 200, about 500, about 1000, about 2000, about 5000, or about 10000 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In some embodiments, the second chronic set point in a second HIV-infected human after treatment with ART and the HIV vaccine is about 10 times higher than the first chronic set point in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. [00194] The instant method can increase the delay in viral rebound as compared to standard therapies after cessation of all antiviral therapies. In some embodiments, the viral load does not rebound in an HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine. In the case of no rebound, the previously HIV-infected human maintains an undetectable viral load after cessation of antiviral therapies for at least 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years, 5 years, or at least 10 years or longer after antiviral therapies have ceased.
[00195] In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is longer than a second delay in viral rebound in a second HIV-infected human after treatment with ART only. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is from about 1 day to about 10 years, e.g., from about 1 week to about 1 year, from about 2 weeks to about 1 year, from about 3 weeks to about 1 year, from about 1 month to about 1 year, from about 2 months to about 1 year, from about 3 months to about 1 year, from about 3 months to about 2 years, etc., longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART only. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is greater than 1 day, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years, 5 years, 10 years or longer compared to a second delay in viral rebound in a second HIV- infected human after treatment with ART only. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 month, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 1.5 years, about 2 years, about 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART only. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 3 months longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART only. [00196] In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is longer than a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is from about 1 day to about 10 years, e.g., from about 1 week to about 1 year, from about 2 weeks to about 1 year, from about 3 weeks to about 1 year, from about 1 month to about 1 year, from about 2 months to about 1 year, from about 3 months to about 1 year, from about 3 months to about 2 years, etc., longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is greater than 1 day, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 month, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 month, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 1.5 years, about 2 years, about 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 3 months longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the TLR7 agonist.
[00197] In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is longer than a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is from about 1 day to about 10 years, e.g., from about 1 week to about 1 year, from about 2 weeks to about 1 year, from about 3 weeks to about 1 year, from about 1 month to about 1 year, from about 2 months to about 1 year, from about 3 months to about 1 year, from about 3 months to about 2 years, etc., longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is greater than 1 day, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 month, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 week, about 2 weeks, about 3 weeks, about 1 month, about 2 month, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 1.5 years, about 2 years, about 3 years or longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine. In some embodiments, a first delay in viral rebound in a first HIV-infected human after treatment with ART, a TLR7 agonist, and an HIV vaccine is about 1 month longer compared to a second delay in viral rebound in a second HIV-infected human after treatment with ART and the HIV vaccine.
[00198] The following examples of the invention are to further illustrate the nature of the invention. It should be understood that the following examples do not limit the invention and the scope of the invention is to be determined by the appended claims. EMBODIMENTS
[00199] The invention provides also the following non-limiting embodiments.
[00200] Embodiment 1 is a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) administering to the human subject an immunogenically effective amount of an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
(ii) administering to the human subject an immunogenically effective amount of an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and
(iii) administering to the human subject an effective amount of vesatolimod or a pharmaceutically acceptable salt thereof (VES).
[00201] Embodiment 1a is the method of Embodiment 1, wherein step (i) is conducted before step (ii).
[00202] Embodiment 1b is the method of Embodiment 1 or 1a, wherein step (ii) is conducted before step (iii).
[00203] Embodiment 1c is the method of any one of Embodiments 1 to 1b, wherein the immunogenically effective amount of the MVA vaccine is administered at about 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is initially administered.
[00204] Embodiment 1d is the method of any one of Embodiments 1 to 1c, wherein the effective amount of vesatolimod or the pharmaceutically acceptable salt thereof is
administered at about 26-34 weeks after the Ad26 vaccine is initially administered.
[00205] Embodiment 1e is the method of Embodiment 1d, wherein the effective amount of vesatolimod or the pharmaceutically acceptable salt thereof is administered biweekly at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is initially administered.
[00206] Embodiment 2 is the method of any one of Embodiments 1-1e, further comprising re-administering to the human subject an immunogenically effective amount of the Ad26 vaccine.
[00207] Embodiment 2a is the method of Embodiment 2, wherein the immunogenically effective amount of the Ad26 vaccine is re-administered at about 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is initially administered.
[00208] Embodiment 3 is the method of any one of Embodiments 1 to 2a, further comprising re-administering to the human subject an immunogenically effective amount of the MVA vaccine.
[00209] Embodiment 3a is the method of Embodiment 3, wherein the MVA vaccine is re- administered at about 34 to 38 weeks, preferably 36 weeks, after the Ad26 vaccine is initially administered.
[00210] Embodiment 4 is the method of any one of Embodiments 1-3a, wherein the Ad26 vaccine comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
[00211] Embodiment 5 is the method of any one of Embodiments 1 to 4, wherein the MVA vaccine consists of a single MVA vector encoding the four HIV antigens.
[00212] Embodiment 5a is the method of Embodiment 5, wherein the single MVA vector is an MVA-BN vector.
[00213] Embodiment 6 is the method of any one of Embodiments 1 to 5a, wherein the one or more Ad26 vectors together are administered at a total dose of about 5x109 to about 1x1011 viral particles (vp) of the one or more Ad26 vectors, per administration.
[00214] Embodiment 6a is the method of Embodiment 6, wherein the one or more Ad26 vectors together are administered at a total dose of about 5x1010 vp of the one or more Ad26 vectors, per administration.
[00215] Embodiment 7 is the method of any one of Embodiments 1 to 6a, wherein the one or more MVA vectors together are administered at a total dose of about 1x107 to about 5x108 infectious units (IU) of the one or more MVA vectors, per administration.
[00216] Embodiment 7a is the method of Embodiment 7, wherein the one or more MVA vectors together are administered at a total dose of about 2 x 108 IU of the one or more MVA vectors, per administration.
[00217] Embodiment 8 is the method of any one of Embodiments 1-7a, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 3-15 mg of vesatolimod, such as about 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg or 15 mg, of vesatolimod, per administration.
[00218] Embodiment 8a is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 4 mg to about 6 mg of vesatolimod per administration.
[00219] Embodiment 8b is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 6 mg to about 8 mg of vesatolimod per administration.
[00220] Embodiment 8c is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 8 mg to about 10 mg of vesatolimod per administration. [00221] Embodiment 8d is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 10 mg to about 12 mg of vesatolimod per administration.
[00222] Embodiment 8e is the method of Embodiment 8, wherein vesatolimod, or the pharmaceutically acceptable salt thereof, is administered at a total dose of about 12 mg to about 14 mg of vesatolimod per administration.
[00223] Embodiment 9 is the method of any one of Embodiments 1-8e, wherein vesatolimod or a pharmaceutically acceptable salt thereof is administered multiple times at about 26-46 weeks, preferably at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks, after the Ad26 vaccine is initially administered.
[00224] Embodiment 9a is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00225] Embodiment 9b is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00226] Embodiment 9c is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00227] Embodiment 9d is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine. [00228] Embodiment 9e is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00229] Embodiment 9f is the method of Embodiment 9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 8 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 10 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00230] Embodiment 10 is the method of any one of Embodiments 1-9, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00231] Embodiment 11 is the method of any one of Embodiments 1-9, wherein vesatolimod or a pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26 and 28 weeks after the Ad26 vaccine is initially administered and at a total dose of 8 mg per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00232] Embodiment 12 is the method of any one of Embodiments 1-11, further comprising administering to the human subject an immunogenically effective amount of a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides.
[00233] Embodiment 12a is the method of Embodiment 12, wherein the gp140 vaccine is administered in combination with the Ad26 vaccine, preferably the re-administering of the Ad26 vaccine.
[00234] Embodiment 12b is the method of Embodiment 12, wherein the gp140 vaccine is administered in combination with the MVA vaccine.
[00235] Embodiment 12c is the method of Embodiment 12b, wherein the gp140 vaccine is administered in combination with the initial administration of the MVA vaccine.
[00236] Embodiment 12d is the method of Embodiment 12b, wherein the gp140 vaccine is administered in combination with the re-administration of the MVA vaccine. [00237] Embodiment 12e is the method of Embodiment 12b, wherein the gp140 vaccine is administered in combination with the initial administration of the MVA vaccine and the re- administration of the MVA vaccine.
[00238] Embodiment 13 is the method of any one of Embodiments 12-12e, wherein the gp140 vaccine comprises two isolated HIV gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, respectively.
[00239] Embodiment 14 is the method of any one of Embodiments 12 to 13, wherein the one or more isolated HIV gp140 envelope polypeptides are administered at a total dose of about 125-350 mg of the total glycoproteins of the HIV gp140 envelope polypeptide(s), per administration.
[00240] Embodiment 14a is the method of any one of Embodiments 12 to 13, wherein the one or more isolated HIV gp140 envelope polypeptides are administered at a total dose of about 250 mg of the total glycoproteins of the HIV gp140 envelope polypeptide(s), per administration.
[00241] Embodiment 15 is a method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) administering to the human subject an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration;
(ii) re-administering to the human subject the Ad26 vaccine in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re- administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
(iii) administering to the human subject an MVA vaccine comprising one or more MVA vectors, preferably one or more MVA-BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further administering to the human subject a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i); and
(iv) administering to the human subject vesatolimod or a pharmaceutically
acceptable salt thereof at a total dose of about 3 mg to about 15 mg of vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i); preferably at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is administered in step (i).
[00242] Embodiment 15a is the method of Embodiment 15, wherein the Ad26 vaccine comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
[00243] Embodiment 15b is the method of Embodiment 15 or 15a, wherein the MVA vaccine consists of a single MVA vector encoding the four HIV antigens.
[00244] Embodiment 15c is the method of Embodiment 15b, wherein the single MVA vector is an MVA-BN vector.
[00245] Embodiment 15d is the method of any one of Embodiments 15-15c, wherein:
(i) the Ad26 vaccine is initially administered in a total dose of about 5x1010 vp of the Ad26 vectors per administration;
(ii) the Ad26 vaccine is re-administered in a total dose of about 5x1010 vp of the Ad26 vectors per administration at 12 weeks after the Ad26 vaccine is initially administered; (iii) the MVA vaccine is initially administered in a total dose of about 2x108 IU of the one or more MVA vectors per administration at 24 weeks after the Ad26 vaccine is initially administered; and
(iv) vesatolimod or the pharmaceutically acceptable salt thereof is administered at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is administered in step (i).
[00246] Embodiment 15e is the method of any one of Embodiment 15 to 15d, wherein each of the Ad26 vaccine and the MVA vaccine is intramuscularly administered, and vesatolimod or the pharmaceutically acceptable salt thereof is orally administered.
[00247] Embodiment 15f is the method of any one of Embodiments 15 to 15e, wherein the MVA vaccine is administered in combination with the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier.
[00248] Embodiment 15g is the method of Embodiment 15f, wherein the gp140 vaccine is administered intramuscularly.
[00249] Embodiment 15h is the method of Embodiment 15f or 15g, wherein the gp140 vaccine is administered at a total dose of about 250 mg of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration.
[00250] Embodiment 16 is the method of any one of Embodiments 15-15h, further comprising:
re-administering to the human subject the MVA vaccine, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per administration;
optionally, in combination with the MVA vaccine, further re-administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration,
wherein the MVA vaccine, and optionally the gp140 vaccine, is re- administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i).
[00251] Embodiment 16a is the method of Embodiment 16, wherein the MVA vaccine is re-administered in a total dose of about 2x108 IU of the one or more MVA vectors per administration at 36 weeks after the Ad26 vaccine is initially administered. [00252] Embodiment 16b is the method of Embodiment 16 or 16a, wherein the MVA vaccine is re-administered in combination with the gp140 vaccine, the aluminum adjuvant and the pharmaceutically acceptable carrier.
[00253] Embodiment 16c is the method of Embodiment 16b, wherein the gp140 vaccine is administered intramuscularly.
[00254] Embodiment 16d is the method of Embodiment 16b or 16c, wherein the gp140 vaccine is re-administered at a total dose of about 250 mg of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration.
[00255] Embodiment 17 is the method of any one of Embodiments 15 to 16d, further comprising: re-administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof in a total dose of about 3 mg to about 15 mg per administration at 38- 46 weeks after the Ad26 vaccine is initially administered.
[00256] Embodiment 17a is the method of Embodiment 17, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered.
[00257] Embodiment 17b is the method of Embodiment 17a, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered multiple times at about 26-46 weeks, preferably at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks, after the Ad26 vaccine is initially administered.
[00258] Embodiment 17c is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per
administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00259] Embodiment 17d is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, and 30 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per
administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00260] Embodiment 17e is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 4 mg of vesatolimod per administration at 26, 28, 30 and 32 weeks after the Ad26 vaccine is initially administered to the human subject, and at a total dose of 6 mg of vesatolimod per administration at 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00261] Embodiment 17f is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 6 mg of vesatolimod per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 after the initial administration of the Ad26 vaccine.
[00262] Embodiment 17g is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 10 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 12 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00263] Embodiment 17h is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00264] Embodiment 17i is the method of Embodiment 17b, wherein vesatolimod or a pharmaceutically acceptable salt thereof is orally administered at a total dose of 6 mg per administration at about 26 and 28 weeks after the Ad26 vaccine is initially administered and at a total dose of 8 mg per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00265] Embodiment 17j is the method of Embodiment 17b, wherein vesatolimod or the pharmaceutically acceptable salt thereof is orally administered at a total dose of about 8 mg of vesatolimod per administration at 26, 28 and 30 weeks after the initial administration of the Ad26 vaccine, and a total dose of 10 mg of vesatolimod per administration at 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
[00266] Embodiment 18 is the method of any one of Embodiments 1 to 17i, wherein the human subject has initiated the ART during the acute phase of HIV infection.
[00267] Embodiment 18a is the method of any one of Embodiments 1 to 17i, wherein the human subject has initiated ART outside of the acute phase of HIV infection.
[00268] Embodiment 18b is the method of any one of Embodiments 1-18a, wherein the human subject has a chronic HIV infection. [00269] Embodiment 19 is the method of any one of Embodiments 1 to 18b, wherein the human subject is on a suppressive ART during the treatment.
[00270] Embodiment 20 is the method of Embodiment 19, wherein the suppressive ART is discontinued after the treatment.
[00271] Embodiment 21 is a method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
(i) treating the human subject with an antiretroviral therapy (ART); and (ii) inducing an immune response against the HIV in the human subject using a method of any one of Embodiments 1-20.
[00272] Embodiment 22 is the method of Embodiment 21, further comprising
discontinuing the ART treatment of step (i) after step (ii).
[00273] Embodiment 23 is the method of Embodiment 22, wherein the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART.
[00274] Embodiment 24 is a combination for use in inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the combination comprising:
(i) an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors
together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
(ii) an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors, preferably one or more MVA-BN vectors, together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and
(iii) a composition comprising vesatolimod or a pharmaceutically acceptable salt thereof (VES).
[00275] Embodiment 25 is the combination of Embodiment 24, further comprising a gp140 vaccine comprising at least one of two isolated HIV gp140 envelope polypeptides respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, and a pharmaceutically acceptable carrier.
[00276] Embodiment 25a is the combination of Embodiment 24, wherein the MVA vaccine consists of a single MVA vector, preferably a single MVA-BN vector, encoding the four HIV antigens. [00277] Embodiment 25b is the combination of Embodiment 25, wherein the gp140 vaccine comprises the two isolated HIV gp140 envelope polypeptides.
[00278] Embodiment 26 is the combination of any one of Embodiments 25 to 25b, further comprising an adjuvant, preferably an aluminum adjuvant.
[00279] Embodiment 27 is the combination of Embodiment 26, wherein the adjuvant is formulated together with the gp140 vaccine or is to be co-administered with the gp140 vaccine.
[00280] Embodiment 28 is a kit comprising the combination of any one of Embodiments 24-27 and instructions on using the combination to induce an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART). EXAMPLES
[00281] EXAMPLE 1: Study of HIV vaccine regimens in HIV-infected humans undergoing antiretroviral therapy (ART)
[00282] Clinical studies in humans are conducted to investigate the effect of 2 different vaccine regimens and a TLR7 agonist in HIV 1 infected adults who started ART during acute HIV infection. Study vaccinations (e.g., with Ad26 vectors, MVA vectors and adjuvanted isolated HIV gp140 polypeptides) and TLR7 agonist (e.g., Vesatolimod) administration will occur in addition to ART.
[00283] Objectives
[00284] This study will evaluate whether the combination of 2 different vaccine regimens and a TLR7 agonist in HIV-1 infected adults who started ART during acute HIV infection will be safe and immunogenic and will lead to sustained virologic control after initial viral rebound following ART interruption.
[00285] Study Population
[00286] Screening for eligible participants will be performed within 70 days before administration of the study intervention. The inclusion criteria for enrolling participants in this study include, for example,
1. must have confirmed HIV-1 infection (including route and estimated duration of HIV infection, pre-ART viral set point, and ARV history).
2. must have initiated ART within 14 days of documented acute HIV-1 infection defined by: - confirmed HIV detectable by PCR and a negative or indeterminate EIA; or
- a positive 4th gen. HIV assay and either a negative/indeterminate HIV conventional Ab test or a negative/indeterminate Western Blot; or
- confirmed HIV detectable by PCR within 21 days of a negative HIV NAAT; or - documented criteria for Fiebig I-IV stages.
3. must be on suppressive ART for at least 48 weeks prior to screening. ART is defined as a combination therapy regimen including more than 2 compounds, e.g.2x nucleoside reverse transcriptase inhibitors plus either non-nucleoside reverse transcriptase inhibitor or integrase inhibitor. Allowed ART as part of the current ARV regimen include nucleoside reverse transcriptase inhibitors (NRTIs), raltegravir, dolutegravir, rilpivirine, bictegravir, and maraviroc.
4. must have a plasma HIV RNA <50 cps/mL at screening and at least one documented evidence of plasma HIV RNA <50 cps/mL after the last ART change.
° One blip of HIV RNA ³50 and <200 cps/mL within 12 weeks before screening is acceptable, provided that the most recent HIV RNA is <50 cps/mL. In case of ART change at screening, 1 blip of HIV RNA ³50 and <200 cps/mL is allowed after 6 weeks on new ART regimen, provided that HIV RNA is <50 cps/mL for the repeat test.
[00287] Vaccination and Experimental Design
[00288] This is a multi-center, randomized, parallel-group, placebo-controlled, double- blind, Phase 1/2a clinical study to investigate the safety, tolerability, efficacy,
immunogenicity, and effect on HIV viremic control after ARV ATI of 2 different vaccine regimens and a TLR7 agonist in HIV 1 infected adults who started ART during acute HIV infection. Study vaccinations and TLR7 administration will occur in addition to ART.
[00289] The study will enroll 90 participants randomized in a 1:1:1 ratio to 2 vaccine groups and 1 matching placebo group. The study population will include HIV infected adults who are on suppressive ART for at least 48 weeks prior to screening and who have maintained undetectable viremia (HIV RNA <50 cps/mL) for ³12 weeks and maintain CD4 counts >450 cells/mm3 prior to initiation of vaccine/placebo administration.
[00290] The study comprises of a screening period of 10 weeks, a 48-week treatment period (Stage 1) and a 48 week ATI period (Stage 2 and Stage 3). A long-term extension period (Stage 4, 104 weeks [approximately 2 years] after week 96) might be added for participants in the active treatment groups, based on efficacy outcomes at week 72. Details on the assessments during this optional LTE phase will be defined at that time. As used herein, “week X” refers to X weeks after the initial administration of the Ad26 vaccine at week 0.
[00291] Dosage and Administration
[00292] Subjects receive four doses of study vaccine: adenovirus 26 vectors encoding mosaic HIV antigens (Ad26.Mos4.HIV) or placebo is administered at weeks 0 and 12; and either (i) MVA vectors encoding mosaic HIV antigens (MVA-BN-HIV) (or placebo), or (ii) MVA-BN-HIV in combination with a mixture of adjuvanted HIV gp140 polypeptides (or placebo) is administered at weeks 24 and 36. Vesatolimod is administered orally at a dose of 6 mg (3 x 2-mg tablets) per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46, or at a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 26 and 28 and a dose of 8 mg (2 x 4 mg tablets) at weeks of 30, 32, 34, 38, 40, 42, 44, and 46. Alternatively, vesatolimod is administered orally at (a) a dose of 4 mg (1 x 4 mg tablets) per administration at weeks 26 and 28, and a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 30, 32, 34, 38, 40, 42, 44, and 46; (b) a dose of 4 mg (1 x 4 mg tablets) per administration at weeks 26, 28 and 30, and a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 32, 34, 38, 40, 42, 44, and 46; (c) a dose of 4 mg (1 x 4 mg tablets) per administration at weeks 26, 28, 30 and 32, and a dose of 6 mg (3 x 2 mg tablets) per administration at weeks 34, 38, 40, 42, 44, and 46; (d) a dose of 10 mg (5 x 2 mg tablets) per administration at weeks 26, 28 and 30, and a dose of 12 mg (6 x 2 mg tablets) per administration at weeks 32, 34, 38, 40, 42, 44, and 46; or (e) a dose of 8 mg (4 x 2 mg tablets) per administration at weeks 26, 28 and 30, and a dose of 10 mg (5 x 2 mg tablets) per administration at weeks 32, 34, 38, 40, 42, 44, and 46.
[00293] Study vaccines (Ad26mos and MVA-BN-HIV, and adjuvanted HIV gp140 polypeptides), VES, and placebo with the administered doses are as follows:
(i) Ad26.Mos4.HIV is composed of the following four vaccine products supplied pre- mixed in the same vial and administered in a 1:1:1:1 ratio of viral particles (vps): Ad26.Mos1Env, Ad26.Mos2SEnv, Ad26.Mos1Gag-Pol, and Ad26.Mos2Gag-Pol expressing HIV mosaic Env1 (SEQ ID NO: 1), mosaic Env2S (SEQ ID NO: 2), mosaic GagPol1 (SEQ ID: NO 3), and mosaic GagPol2 (SEQ ID NO: 4) genes, respectively; administered at a total dose of about 5 x 1010 viral particles (vp) in 0.5 mL injection;
(ii) MVA-BN-HIV is an MVA-BN expressing Mos1.Gag-Pol (SEQ ID NO: 3),
Mos2.Gag-Pol (SEQ ID NO: 4), Mos1.Env (SEQ ID NO: 1) and Mos2S.Env (SEQ ID NO: 2); administered at a total dose about 2 x 108 infectious units (IU) in 0.5 mL injection; MVA-BN-HIV has been described in more detail as MVA- mBN414 in example 7 of WO 2018/229711;
(iii) gp140 HIV bivalent vaccine, adjuvanted, containing 0.16 mg/mL Clade C gp140 (SEQ ID NO: 9), 0.15 mg/mL Mosaic gp140 (SEQ ID NO: 10) and 0.85 mg /mL aluminum (Al); administered at 125 microgram (mcg) of Clade C gp140 glycoprotein, 125 mcg Mosaic gp140 glycoprotein (corresponding with 80 mcg and 75 mcg of protein, respectively), and aluminum adjuvant (425 mcg Al) per 0.5 mL dose;
(iv) Vaccine placebo is 0.9% sodium chloride (0.5 mL injection);
(v) Participants will receive 10 doses of VES orally once every 14 days as the
following:
a. total dose of 6 mg (3 x 2 mg tablets) of VES per administration at weeks 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46;
b. total dose of 6 mg (3 x 2 mg tablets) of VES per administration at weeks 26 and 28, and total dose of 8 mg (2 x 4 mg tablets) of VES per administration at weeks 30, 32, 34, 38, 40, 42, 44, and 46;
c. total dose of 4 mg (1 x 4 mg tablets) of VES per administration at weeks 26 and 28, and total dose of 6 mg (3 x 2 mg tablets) of VES per administration at weeks 30, 32, 34, 38, 40, 42, 44, and 46;
d. total dose of 4 mg (1 x 4 mg tablets) of VES per administration at weeks 26, 28 and 30, and total dose of 6 mg (3 x 2 mg tablets) of VES per administration at weeks 32, 34, 38, 40, 42, 44, and 46;
e. total dose of 4 mg (1 x 4 mg tablets) of VES per administration at weeks 26, 28, 30 and 32, and total dose of 6 mg (3 x 2 mg tablets) of VES per administration at weeks 34, 38, 40, 42, 44, and 46;
f. total dose of 10 mg (5 x 2 mg tablets) of VES per administration at weeks 26, 28 and 30, and total dose of 12 mg (6 x 2 mg tablets) of VES per
administration at weeks 32, 34, 38, 40, 42, 44, and 46; or
g. total dose of 8 mg (4 x 2 mg tablets) of VES per administration at weeks 26, 28 and 30, and total dose of 10 mg (5 x 2 mg tablets) of VES per
administration at weeks 32, 34, 38, 40, 42, 44, and 46 (ii) VES placebo is lactose, administered at a total dose corresponding to the same number of tablets used for the VES treatment.
[00294] Subjects receive the study vaccines administered by intramuscular administration and VES oral dosing or placebo according to the schedule in Table 1 below:
Table 1: Schedule for administration of study vaccines
Figure imgf000066_0001
[00295] Subjects in both the test and control groups continue to receive standard ART (e.g. at least three antiviral compounds, e.g. two nucleoside reverse transcriptase inhibitors plus either non-nucleoside reverse transcriptase inhibitor or protease inhibitor or integrase inhibitor) for HIV treatment during the study. Blood and optionally genital secretions are taken at specific clinical visits to assess immune responses (cellular and humoral immune responses) and viremic control throughout the study.
[00296] It is understood that the examples and embodiments described herein are for illustrative purposes only, and that changes could be made to the embodiments described above without departing from the broad inventive concept thereof. It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications within the spirit and scope of the invention as defined by the appended claims. SEQUENCE LISTING
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
REFERENCES
1. US20120076812
2. Barouch et al., Nat Med 2010, 16:319-323
3. Barouch et al., Cell 155:1-9, 2013
4. Fischer et al, Nat Med, 2007.13(1): p.100-6
5. Havenga, et al., 2006, J Gen Virol 87: 2135-43
6. WO 03/104467
7. WO 2007/104792
8. Abbink et al., (2007) Virol 81(9): 4654-63
9. Mayr et al. (1975) Infection 3, 6-14
10. Mayr, A. & Danner, K. (1978), Dev. Biol. Stand.41: 225-234
11. Stickl (1974), Prev. Med.3: 97-101
12. Stickl and Hochstein-Mintzel (1971), Munch. Med. Wochenschr.113: 1149-1153 13. Mayr et al. (1978), Zentralbl. Bacteriol. (B) 167:375-390
14. WO 02/42480 (US 2003/0206926)
15. WO 03/048184 (US 2006/0159699)
16. US20110159036
17. US 8197825
18. WO 2011/092029
19. Nkolola et al 2010, J. Virology 84(7): 3270-3279
20. Kovacs et al, PNAS 2012, 109(30):12111-6
21. WO 2010/042942
22. WO 2014/107744
23. National Center for Biotechnology Information. PubChem Database. Vesatolimod, CID=46241268, https://pubchem.ncbi.nlm.nih.gov/compound/46241268 (accessed on Apr.23, 2019)

Claims

CLAIMS What is claimed is: 1. A method of inducing an immune response against a human
immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) administering to the human subject an immunogenically effective amount of an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier;
(ii) administering to the human subject an immunogenically effective amount of an MVA vaccine comprising one or more Modified Vaccinia Ankara (MVA) vectors together encoding the four HIV antigens and a pharmaceutically acceptable carrier; and
(iii) administering to the human subject an effective amount of vesatolimod or a pharmaceutically acceptable salt thereof (VES).
2. The method of claim 1, further comprising re-administering to the human subject an immunogenically effective amount of the Ad26 vaccine, preferably at about 10-14 weeks, more preferably 12 weeks, after the Ad26 vaccine is initially administered.
3. The method of claim 1 or 2, further comprising re-administering to the human subject an immunogenically effective amount of the MVA vaccine, preferably wherein the MVA vaccine is initially administered at about 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is initially administered, and preferably the MVA vaccine is re-administered at about 34 to 38 weeks, preferably 36 weeks, after the Ad26 vaccine is initially administered.
4. The method of any one of claims 1-3, wherein the Ad26 vaccine comprises a first Ad26 vector encoding the HIV antigen of SEQ ID NO: 1, a second Ad26 vector encoding the HIV antigen of SEQ ID NO: 2, a third Ad26 vector encoding the HIV antigen of SEQ ID NO: 3, and a fourth Ad26 vector encoding the HIV antigen of SEQ ID NO: 4.
5. The method of any one of claims 1 to 4, wherein the MVA vaccine consists of a single MVA vector encoding the four HIV antigens.
6. The method of any one of claims 1 to 5, wherein the one or more Ad26 vectors
together are administered at a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the one or more Ad26 vectors, per administration.
7. The method of any one of claims 1 to 6, wherein the one or more MVA vectors
together are administered at a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2 x 108 IU, of the one or more MVA vectors, per
administration.
8. The method of any one of claims 1-7, wherein vesatolimod, or a pharmaceutically acceptable salt thereof, is administered at a total dose of about 3-15 mg of
vesatolimod, preferably about 4-12 mg of vesatolimod, for example about 6-8 mg of vesatolimod, per administration.
9. The method of any one of claims 1-8, wherein vesatolimod or a pharmaceutically acceptable salt thereof is administered multiple times at about 26-46 weeks, preferably at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks, after the Ad26 vaccine is initially administered.
10. The method of any one of claims 1-9, wherein vesatolimod or a pharmaceutically acceptable salt thereof is administered at a total dose of 6 mg per administration at about 26, 28, 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered.
11. The method of any one of claims 1-9, wherein vesatolimod or a pharmaceutically acceptable salt thereof is administered at a total dose of 6 mg per administration at about 26 and 28 weeks after the Ad26 vaccine is initially administered and at a total dose of 8 mg per administration at 30, 32, 34, 38, 40, 42, 44, and 46 weeks after the Ad26 vaccine is initially administered
12. The method of any one of claims 1-11, further comprising administering to the human subject an immunogenically effective amount of a gp140 vaccine comprising one or more isolated HIV gp140 envelope polypeptides, preferably in combination with the Ad26 vaccine or the MVA vaccine, more preferably in combination with the MVA vaccine.
13. The method of claim 12, wherein the gp140 vaccine comprises two isolated HIV
gp140 envelope polypeptides having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, respectively, and the gp140 vaccine is administered in combination with the MVA vaccine.
14. The method of claim 12 or 13, wherein the one or more isolated HIV gp140 envelope polypeptides are administered at a total dose of about 125-350 mg, preferably about 250 mg, of the total glycoproteins of the HIV gp140 envelope polypeptides, per administration.
15. A method of inducing an immune response against a human immunodeficiency virus (HIV) in an HIV-infected human subject undergoing antiretroviral therapy (ART), the method comprising:
(i) intramuscularly administering to the human subject an Ad26 vaccine comprising one or more adenovirus 26 (Ad26) vectors together encoding four HIV antigens having the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, and a pharmaceutically acceptable carrier, in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration;
(ii) intramuscularly re-administering to the human subject the Ad26 vaccine in a total dose of about 5x109 to about 1x1011 viral particles (vp), preferably about 5x1010 vp, of the Ad26 vectors per administration, wherein the Ad26 vaccine is re- administered 10-14 weeks, preferably 12 weeks, after the Ad26 vaccine is administered in step (i);
(iii) intramuscularly administering to the human subject an MVA vaccine comprising one or more MVA vectors, preferably one or more MVA-BN vectors, encoding the four HIV antigens and a pharmaceutically acceptable carrier, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per administration; optionally, in combination with the MVA vaccine, further administering to the human subject a gp140 vaccine comprising two isolated HIV gp140 envelope polypeptides respectively having the amino acid sequences of SEQ ID NO: 9 and SEQ ID NO: 10, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration, wherein the MVA vaccine and optionally the gp140 vaccine, is administered 22-26 weeks, preferably 24 weeks, after the Ad26 vaccine is administered in step (i); and
(iv) orally administering to the human subject vesatolimod or a pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg of
vesatolimod, per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is administered bi-weekly at 26-34 weeks after the Ad26 vaccine is administered in step (i); preferably, at a total dose of 6 mg of vesatolimod per administration at 26, 28, 30, 32 and 34 weeks after the Ad26 vaccine is administered in step (i), or at a total dose of 6 mg of vesatolimod per administration at 26 and 28 weeks after the Ad26 vaccine is administered in step (i), and at a total dose of 8 mg of vesatolimod per administration at 30, 32 and 34 weeks after the Ad26 vaccine is administered in step (i).
16. The method of claim 15, further comprising:
intramuscularly re-administering to the human subject the MVA vaccine, in a total dose of about 1x107 to about 5x108 infectious units (IU), preferably about 2x108 IU, of the one or more MVA vectors, per administration;
optionally, in combination with the MVA vaccine, further re-administering to the human subject the gp140 vaccine, an aluminum adjuvant and a pharmaceutically acceptable carrier, at a total dose of about 125 mg to 350 mg, preferably about 250 mg, of the glycoproteins of the two isolated HIV gp140 envelope polypeptides per administration,
wherein the MVA vaccine, and optionally the gp140 vaccine, is re- administered 34-38 weeks, preferably 36 weeks, after the Ad26 vaccine is administered in step (i).
17. The method of claim 15 or 16, further comprising:
orally re-administering to the human subject vesatolimod or a
pharmaceutically acceptable salt thereof at a total dose of about 3 mg to about 15 mg per administration, wherein vesatolimod or the pharmaceutically acceptable salt thereof is re-administered bi-weekly at 38-46 weeks after the Ad26 vaccine is administered in step (i); preferably, at a total dose of 6 or 8 mg of vesatolimod per administration at 38, 40, 42, 44 and 46 weeks after the Ad26 vaccine is administered in step (i).
18. The method of any one of claims 1 to 17, wherein the human subject has initiated the ART during the acute phase of HIV infection.
19. The method of any one of claims 1 to 18, wherein the human subject is on a
suppressive ART during the treatment.
20. The method of claim 19, wherein the suppressive ART is stopped after the treatment.
21. A method of treating a human immunodeficiency virus (HIV) infection in a human subject in need thereof, comprising:
(i) treating the human subject with an antiretroviral therapy (ART); and (ii) inducing an immune response against the HIV in the human subject using a method of any one of claims 1-20.
22. The method of claim 21, further comprising: discontinuing the ART treatment of step (i) after step (ii).
23. The method of claim 22, wherein the human subject maintains viral suppression for at least 24 weeks after discontinuing the ART.
PCT/US2020/034000 2019-05-22 2020-05-21 Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment WO2020237052A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962851258P 2019-05-22 2019-05-22
US62/851,258 2019-05-22

Publications (1)

Publication Number Publication Date
WO2020237052A1 true WO2020237052A1 (en) 2020-11-26

Family

ID=71070052

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/034000 WO2020237052A1 (en) 2019-05-22 2020-05-21 Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment

Country Status (2)

Country Link
TW (1) TW202110476A (en)
WO (1) WO2020237052A1 (en)

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002024224A2 (en) 2000-09-21 2002-03-28 Oxxon Pharmaccines Limited Use of replication-deficient poxvirus vector to boost cd4+ t cell immune response to antigen
WO2002042480A2 (en) 2000-11-23 2002-05-30 Bavarian Nordic A/S Modified vaccinia ankara virus variant
WO2003048184A2 (en) 2001-12-04 2003-06-12 Bavarian Nordic A/S Flavivirus ns1 subunit vaccine
WO2003104467A1 (en) 2002-04-25 2003-12-18 Crucell Holland B.V. Means and methods for the production of adenovirus vectors
WO2007104792A2 (en) 2006-03-16 2007-09-20 Crucell Holland B.V. Recombinant adenoviruses based on serotype 26 and 48, and use thereof
WO2010042942A2 (en) 2008-10-10 2010-04-15 Children's Medical Center Corporation Biochemically stabilized hiv-1 env trimer vaccine
US20110159036A1 (en) 2003-03-28 2011-06-30 Bernard Moss MVA expressing modified HIV envelope, GAG, and POL genes
WO2011092029A1 (en) 2010-01-28 2011-08-04 Bavarian Nordic A/S Vaccinia virus mutants containing the major genomic deletions of mva
US20120076812A1 (en) 2008-11-18 2012-03-29 Barouch Dan H Antiviral vaccines with improved cellular immunogenicity
US8197825B2 (en) 1995-07-04 2012-06-12 Gsf-Forschungszentrum Fur Umwelt Und Gesundheit Gmbh Recombinant MVA virus and the use thereof
WO2014107744A1 (en) 2013-01-07 2014-07-10 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (hiv) envelope (env) trimer vaccines and methods of using the same
WO2016007765A1 (en) 2014-07-11 2016-01-14 Gilead Sciences, Inc. Modulators of toll-like receptors for the treatment of hiv
WO2016049287A1 (en) * 2014-09-26 2016-03-31 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
WO2016177833A1 (en) 2015-05-04 2016-11-10 Bionor Immuno As Dosage regimen for hiv vaccine
WO2017102929A1 (en) 2015-12-15 2017-06-22 Janssen Vaccines & Prevention B.V. Human immunodeficiency virus antigens, vectors, compositions, and methods of use thereof
US20180064803A1 (en) * 2016-09-02 2018-03-08 Janssen Vaccines & Prevention B.V. Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
WO2018229711A1 (en) 2017-06-15 2018-12-20 Janssen Vaccines & Prevention B.V. Poxvirus vectors encoding hiv antigens, and methods of use thereof

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8197825B2 (en) 1995-07-04 2012-06-12 Gsf-Forschungszentrum Fur Umwelt Und Gesundheit Gmbh Recombinant MVA virus and the use thereof
WO2002024224A2 (en) 2000-09-21 2002-03-28 Oxxon Pharmaccines Limited Use of replication-deficient poxvirus vector to boost cd4+ t cell immune response to antigen
WO2002042480A2 (en) 2000-11-23 2002-05-30 Bavarian Nordic A/S Modified vaccinia ankara virus variant
US20030206926A1 (en) 2000-11-23 2003-11-06 Paul Chaplin Modified vaccinia ankara virus variant
US6761893B2 (en) 2000-11-23 2004-07-13 Bavarian Nordic A/S Modified vaccinia ankara virus variant
WO2003048184A2 (en) 2001-12-04 2003-06-12 Bavarian Nordic A/S Flavivirus ns1 subunit vaccine
US20060159699A1 (en) 2001-12-04 2006-07-20 Paul Howley Flavivirus ns1 subunit vaccine
WO2003104467A1 (en) 2002-04-25 2003-12-18 Crucell Holland B.V. Means and methods for the production of adenovirus vectors
US20110159036A1 (en) 2003-03-28 2011-06-30 Bernard Moss MVA expressing modified HIV envelope, GAG, and POL genes
WO2007104792A2 (en) 2006-03-16 2007-09-20 Crucell Holland B.V. Recombinant adenoviruses based on serotype 26 and 48, and use thereof
WO2010042942A2 (en) 2008-10-10 2010-04-15 Children's Medical Center Corporation Biochemically stabilized hiv-1 env trimer vaccine
US20120076812A1 (en) 2008-11-18 2012-03-29 Barouch Dan H Antiviral vaccines with improved cellular immunogenicity
WO2011092029A1 (en) 2010-01-28 2011-08-04 Bavarian Nordic A/S Vaccinia virus mutants containing the major genomic deletions of mva
WO2014107744A1 (en) 2013-01-07 2014-07-10 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (hiv) envelope (env) trimer vaccines and methods of using the same
WO2016007765A1 (en) 2014-07-11 2016-01-14 Gilead Sciences, Inc. Modulators of toll-like receptors for the treatment of hiv
WO2016049287A1 (en) * 2014-09-26 2016-03-31 Beth Israel Deaconess Medical Center, Inc. Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
WO2016177833A1 (en) 2015-05-04 2016-11-10 Bionor Immuno As Dosage regimen for hiv vaccine
WO2017102929A1 (en) 2015-12-15 2017-06-22 Janssen Vaccines & Prevention B.V. Human immunodeficiency virus antigens, vectors, compositions, and methods of use thereof
US20180064803A1 (en) * 2016-09-02 2018-03-08 Janssen Vaccines & Prevention B.V. Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
WO2018229711A1 (en) 2017-06-15 2018-12-20 Janssen Vaccines & Prevention B.V. Poxvirus vectors encoding hiv antigens, and methods of use thereof

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
- CITY GILEAD: "Gilead Announces Latest Data in Ongoing HIV Cure Research Program - New Studies Evaluate Agents with Potential Role in Eliminating HIV Viral Reservoir", 23 July 2019 (2019-07-23), pages 2019, XP055711484, Retrieved from the Internet <URL:https://gilead2019tf.q4web.com/files/doc_news/archive/eb4cdb0a-1ac7-4622-981b-7fd62c5aeef6.pdf> [retrieved on 20200703] *
"Expression of genes by Vaccinia virus vectors", 1998, JOHN WILEY & SON, INC.
"Expression of proteins in mammalian cells using vaccinia viral vector", 1986
"The Practical Approach Series", 1993, OXFORD UNIVERSITY PRESS, article "Molecular Virology: A Practical Approach"
"Virology Methods Manual", 1996, ACADEMIC PRESS
ABBINK ET AL., VIROL, vol. 81, no. 9, 2007, pages 4654 - 63
ANGELA TSAI ET AL: "ABSTRACT", JOURNAL OF VIROLOGY, vol. 91, no. 8, 29 March 2017 (2017-03-29), US, XP055711585, ISSN: 0022-538X, DOI: 10.1128/JVI.02166-16 *
BAROUCH ET AL., CELL, vol. 155, 2013, pages 1 - 9
BAROUCH ET AL., NAT MED, vol. 16, 2010, pages 319 - 323
BORDUCCHI E.N. ET AL., NATURE, vol. 540, 2016, pages 284 - 287
BORDUCCHI ERICA N ET AL: "Antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys", NATURE,, vol. 563, no. 7731, 3 October 2018 (2018-10-03), pages 360 - 364, XP036632361, ISSN: 0028-0836, [retrieved on 20181003], DOI: 10.1038/S41586-018-0600-6 *
BOUKAMP ET AL., J. CELL BIOL., vol. 106, 1988, pages 761 - 771
DEVI SENGUPTA ET AL: "SAFETY AND ANALYTIC TREATMENT INTERRUPTION OUTCOMES OF VESATOLIMOD IN HIV CONTROLLERS - CROI Conference", 11 March 2020 (2020-03-11) - 11 March 2020 (2020-03-11), Boston, Massachusetts, XP055711468, Retrieved from the Internet <URL:https://www.croiconference.org/abstract/safety-and-analytic-treatment-interruption-outcomes-of-vesatolimod-in-hiv-controllers/> [retrieved on 20200703] *
FIEBIG ET AL.: "Dynamics of HIV viremia and antibody seroconversion in plasma donors: implications for diagnosis and staging of primary HIV infection", AIDS, vol. 17, no. 13, 2003, pages 1871 - 1879, XP002371394, DOI: 10.1097/00002030-200309050-00005
FISCHER ET AL., NAT MED, vol. 13, no. 1, 2007, pages 100 - 6
FISCHER ET AL., NATMED, vol. 13, no. 1, 2007, pages 100 - 6
HAVENGA ET AL., J GEN VIROL, vol. 87, 2006, pages 2135 - 43
J. SAMBROOK ET AL.: "Molecular Cloning, A laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
KOVACS ET AL., PNAS, vol. 109, no. 30, 2012, pages 12111 - 6
MAYR ET AL., INFECTION, vol. 3, 1975, pages 6 - 14
MAYR ET AL., ZENTRALBL. BACTERIOL. (B, vol. 167, 1978, pages 375 - 390
MAYR, A.DANNER, K., DEV. BIOL. STAND., vol. 41, 1978, pages 225 - 234
NKOLOLA ET AL., J. VIROLOGY, vol. 84, no. 7, 2010, pages 3270 - 3279
RENEE R RAM ET AL: "Activation of HIV-specific CD8+ T-cells from HIV+ donors by vesatolimod", ANTIVIRAL THERAPY- AN OFFICIAL PUBLICATION OF THE INTERNATIONAL SOCIETY FOR ANTIVIRAL RESEARCH, 1 January 2020 (2020-01-01), GB, XP055711498, ISSN: 1359-6535, DOI: 10.3851/IMP3359 *
STICKL, PREV. MED., vol. 3, 1974, pages 97 - 101
STICKLHOCHSTEIN-MINTZEL, MUNCH. MED. WOCHENSCHR., vol. 113, 1971, pages 1149 - 1153

Also Published As

Publication number Publication date
TW202110476A (en) 2021-03-16

Similar Documents

Publication Publication Date Title
US10525123B2 (en) Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
US11207400B2 (en) Compositions and methods for inducing protective immunity against human immunodeficiency virus infection
US20190083620A1 (en) Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
JP7272965B2 (en) Poxvirus vectors encoding HIV antigens and methods of use thereof
US20190022212A1 (en) Methods for safe induction of cross-clade immunity against human immunodeficiency virus infection in human
WO2020237052A1 (en) Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
WO2022272275A1 (en) Combinations of vaccines and neutralizing antibodies for treating human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
NZ767061A (en) Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
NZ767061B2 (en) Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
NZ730841B2 (en) Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
OA18541A (en) Methods and compositions for inducing protective immunity against human immunodeficiency virus infection

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: 20731748

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20731748

Country of ref document: EP

Kind code of ref document: A1