WO2020069375A1 - TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE - Google Patents

TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE Download PDF

Info

Publication number
WO2020069375A1
WO2020069375A1 PCT/US2019/053548 US2019053548W WO2020069375A1 WO 2020069375 A1 WO2020069375 A1 WO 2020069375A1 US 2019053548 W US2019053548 W US 2019053548W WO 2020069375 A1 WO2020069375 A1 WO 2020069375A1
Authority
WO
WIPO (PCT)
Prior art keywords
tnf
antibody
cdg
tnfr2
mice
Prior art date
Application number
PCT/US2019/053548
Other languages
French (fr)
Inventor
Lei Jin
Original Assignee
University Of Florida Research Foundation, 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 University Of Florida Research Foundation, Inc. filed Critical University Of Florida Research Foundation, Inc.
Priority to US17/280,986 priority Critical patent/US20210353748A1/en
Priority to EP19865436.0A priority patent/EP3856199A4/en
Publication of WO2020069375A1 publication Critical patent/WO2020069375A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • A61K31/708Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid having oxo groups directly attached to the purine ring system, e.g. guanosine, guanylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7084Compounds having two nucleosides or nucleotides, e.g. nicotinamide-adenine dinucleotide, flavine-adenine dinucleotide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/191Tumor necrosis factors [TNF], e.g. lymphotoxin [LT], i.e. TNF-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/07Bacillus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0043Nose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • mucosal vaccines typically have the benefits of low cost and ease of administration.
  • mucosal vaccines there are very few mucosal vaccines approved for use, which is largely due to problems with developing safe and effective mucosal adjuvants.
  • B. Absolute number of lung DC subsets in C57BL/6 mice administered (i.n.) with saline or 5pg FITC-CDG for 5 hours. n 3.
  • FIG. cDC2 expression of TNFR2 is required for CDG-induced lung cDC2 maturation in vivo by activating RelB.
  • A. C57BL/6 (WT), TNFRL 7 and TNFR2 7 mice were immunized (i.n.) with two doses of PspA or PspA plus CDG (5ug). Serum anti-PspA IgG and BALF IgA were determined by ELISA. n 3.
  • B. WT and TNFR2 7 mice were treated (i.n.) with saline or CDG (5pg) for 16 hours.
  • IRF4 n/ll CDl l c trc mice were adoptively transferred (i.n.) with lung cDC2 sorted from WT or TNFR2 7 mice lung.
  • the recipient mice were administered (i.n.) with saline or CDG (5pg) for l6hrs.
  • D Flow cytometry analysis of TNFR2 expression on lung cDC2 in WT mice administered (i.n) with saline or CDG for l6hrs. n>3.
  • Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test). [0004] FIG 3.
  • cDC2 expression of TNFR2 and RelB is required for Thl and Thl7 responses, but dispensable for CDG-induced antibody response.
  • FIG. 4 Lung TNFR2 cDC2 mediate CDG-induced humoral response.
  • C Sorted TNFR2 + and TNFR2 lung cDC2 from WT mice were labelled with CFSE and adoptively transferred into the MPYS 7 mice. Recipient mice were treated with CDG (i.n.) for l6hrs.
  • G A cartoon illustrating following CDG administration, TNFR2 + cDC2 activate RelB to induce Thl, Th2 and Thl7 responses while TNFR2 cDC2 mediate the antibody response. Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test).
  • FIG. 5 Lung cDC2 consist of two distinct subsets.
  • E. Phenotypic analysis of CD45.1 TNFR2 + and TNFR2 cDC2 transferred into IRF4 fl/fl CDl lc cre mice. n 3.
  • F. Flow cytometry analysis of TNFR2 expression on cDC2 subsets transferred into MPYS 7 mice treated with CDG (i.n.) for l6hrs. n 3.
  • FIG. CDG activates TNFR2 7 cDC2 in vivo to produce TNF.
  • A-B Indicated mice were administered (i.n.) with saline or CDG for 5hrs. TNF production was measured in lung homogenates by ELISA. n>3.
  • C. WT and TNFR2 7 mice were treated with saline or CDG for 16 hours. TNF in lung cDC2 was determined by an intracellular cytokine stain. n 3.
  • WT, IRF4 fl/fl CDl lc cre (A) and Batf3 _/ (B) mice were treated (i.n.) with saline or CDG (5m g for 16 hours.
  • WT, TNFR2 / mice or TNFR2 7 mice receiving (i.n.) WT monocytes or TNFR2 7 monoctyes were immunized with CDG/PspA.
  • H. WT cDC2 adoptively transferred into TNFR2 _/ mice were immunized with CDG/PspA.
  • Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test).
  • FIG 8. TNFR2 cDC2 express mTNF but have few processed antigen.
  • A. WT mice were administered with saline or CDG (5pg) for l6hrs. mTNF expression was determined by Flow cytometry using mouse TNFR2-Fc recombinant protein. n 3.
  • B-C. Flow cytometry analysis of mTNF expression in lung DCs (B) and cDC2 (C) n 3.
  • Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test).
  • FIG. 9 moDCs promote CDG-induced Tfh and GC B cell generation in the lung.
  • WT mice were administered with Ea-OVA (lOpg) or Ea-OVA/CDG (5pg) for l6hrs.
  • C-D. WT, TNFR2 A micc or TNFR2 7 mice receiving (i.n.) WT monocytes were immunized with
  • E. TNFR2 + and TNFR2 cDC2 were adoptively transferred into IRF4 n/ll CDl lc cre mice and immunized with CDG/PspA.
  • FIG. 10 A-B Gating strategy for lung DCs.
  • cDCl are CDl lc+MHCIIhiCD103+CDl lb- CD64-IRF8+
  • cDC2 are CD1 lc+MHCIIhiCDl lb+ CD64-IRF4+
  • moDCs are
  • C. Flow cytometry analysis of lung DCs subsets in IRF4fi/fi and IRF4fi/flCDl lccre mice including the total number of DCs. n 3.
  • D. Flow cytometry analysis of lung DCs subsets in C57BF/6 (WT) and Batf3-/- mice including the total number of DCs. n 3.
  • Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test).
  • FIG. 11 A WT mice were intranasally administered with saline or 5pg FITC-CDG for 5 hours. Fung FITC positive cells were analyzed by Flow cytometry. n>3. B. CD86 expression on
  • CDl lc+MHCIIint Alveolar macrophages (AMs) and lung DCs (CDl lc+MHCIIhi) in WT mice treated (i.n.) with saline or 5mg CDG for 16 hours. n 3.
  • C. Flow cytometry analysis of MPYS expression in AMs (SiglecF+ CDl lc+) from MPYSfl/flLysMcre mice. n 3.
  • C Indicated mice were treated (i.n.) with saline or CDG for 5 hrs. Lung MCP-l, TNF, IL- 1 b and ILl2p70 production were measured by ELISA in lung homogenates. n>3.
  • D.WT, Batf3-/-, and IRF4fl/flCDl lccre mice were immunized (i.n.) with two doses (14 days apart) of PspA or PspA plus CDG (5ug).
  • Anti-PspA IgGl and IgG2c in serum and BALF were determined by ELISA. n>3.
  • EF. Splenocytes (E) or lung cells (F) from immunized WT, Batf3-/-, and IRF4fl/flCDl lccre mice were stimulated with 5pg/ml PspA for 4 days in culture. Cytokines were measured in the supernatant by ELISA. n>3. Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test).
  • Splenocytes (E) or lung cells (F) from immunized WT, TNFR1-/-, and TNFR2-/- mice were stimulated with 5mg/ml PspA for 4 days in culture. Cytokines were measured in the supernatant by ELISA. n>3. Graphs represent means ⁇ standard error from three independent experiments. The significance is represented by and asterisk (*) where p ⁇ 0.05 (unpaired Student’s t test).
  • Isotype control Ab (50pg) or anti-TNFR2 Ab (50pg) was administered (i.n.) 30min later. The lungs were harvested 16 hours later.
  • E. Flow cytometry analysis of lung DCs populations in RelBfl/fl and RelBfl/flCDl lCCre mice. n 3.
  • FIG. 16 A. Flow cytometry analysis of CD26 expression on lung TNFR2+ and TNFR2- cDC2.
  • FIG. 17. A. pre-cDC2 gating strategy in the bone marrow. Naive B6.CD45.1 mice were used.
  • B Experimental scheme for the transfer of pre-cDC2 into IRF4fl/flCDl lccre mice (CD45.2).
  • C Phenotypic analysis of CD45.1 DCs transferred into IRF4fl/flCDl lccre mice.
  • D Experimental scheme for the adoptive transfer of TNFR2+ and TNFR2- lung cDC2 into the MPYS7- mice.
  • FIG. 18C Adult and aged mice were immunized or not as (FIG. 18 A). One month later, mice were infected (i.n) with S.
  • FIG. 19D The
  • FIG. 20 3-month old C57BL/6 (Adult), 18-month old C57BL/6 (Aged) mice were immunized (i.n.) with CDG (5mg)/PspA(2mg) or PBS for l6hrs. Lung moDCs
  • FIG. 21 A cartoon of TNF-Fc fusion protein and its proposed mode of action on moDCs.
  • FIG. 21B A cartoon of the CDG/PspA/TNF-Fc vaccine strategy.
  • FIG. 21C Lung Tfh cells( PD1+CXCR5+) cells were examined in adult and aged mice 14 days after
  • Graphs represent means ⁇ standard error from three
  • FIG. 23 shows sequence information on Mouse TNF.
  • D221N, A223R amino acid modification
  • CXCL13 is a chemokine essential for the formation of germinal center.
  • CCL20 is a T cells recruiting chemokine (FIG. 25A) and Bcl6 (FIG. 25B) is a transcriptional factor important for memory cells development.
  • the present disclosure is based on the discovery of a new lung dendritic cell subset and its novel in vivo model of action that generates efficacious mucosal vaccine responses.
  • the characterization of this new subset of dendritic cells along with its mode of action have allowed the design of vaccine compositions that are tailored to generate efficacious vaccine responses in lung mucosa.
  • the new lung dendritic cells possess a unique marker profile, TNFR2 pRelB PD- LL CX3CR1 + cDC2 (C3D2).
  • a vaccine composition that includes a therapeutically effective amount of an antigen; a CDN; soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment.
  • a TNF conjugated with an moDC targeting moiety is implemented in addition to or in place of TNF and/or the CD64 antibody or antibody fragment.
  • the TNF conjugated with an moDC targeting moiety may include a TNF-Fc (IgG2A) or an anti-CD64 antibody conjugate (antibody-cytokine fusion protein such as anti-CD64-TNF).
  • the CDN may include, but is not limited to, cyclic-di-GMP (CDG), cyclic-di-AMP (CD A), cyclic-di-IMP (CDI), cyclic-AMP-GMP (CDA/G), cyclic-AMP- IMP (CDA/I, and cyclic-GMP-IMP (CDG/I).
  • CDN is CDG.
  • CD64 antibody is monoclonal antibody.
  • a method of eliciting an immune response in a subject that involves administering to the subject a vaccine composition as described herein.
  • the vaccine composition is one that activates monocyte derived dendritic cells (moDCs), to induce lung mucosal antibody responses.
  • MoDCs monocyte derived dendritic cells
  • One unique advantage of the compositions described herein is that allow for the generation of effective vaccine responses on mucosal surface. They also allow for the generation of long-termed memory responses that is critical for vaccine-induced protection.
  • a method of eliciting an immune response that involves administering an antigen; and co-administering a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment.
  • an moDC targeting moiety e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF
  • the compositions are not necessarily administered in the same composition but may be administered separately, either by the same or different mode of administration.
  • kits that includes an inhalative administration device and a therapeutically effective amount of a composition described herein.
  • the kit may include a vaccine composition or a separate antigen composition and adjuvant composition.
  • the one or more compositions may be loaded into the device or provided in a separate container for use in the device.
  • Another embodiment pertains to a method for treating, or preventing, a disease or condition in a subject.
  • the method involves administering an antigen associated with the disease or condition; and co-administering a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment.
  • TNF cyclic dinucleotide
  • a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64
  • a further embodiment pertains to an adjuvant composition that includes a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti- CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment.
  • TNF cyclic dinucleotide
  • CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti- CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment.
  • TNF cyclic dinucleotide
  • CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an
  • the term“about” as used herein means approximately, roughly, around, or in the region of. When the term“about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20% up or down (higher or lower).
  • TNF tumor necrosis factor
  • the TNF sequence used is typically that of the species being treated, that is, human TNF when used in inducing an immune response in humans.
  • TNF unless otherwise indicated, includes both soluble TNF and transmembrane TNF.
  • soluble TNF or“sTNF” are used interchangeably herein to refer to the l7KDa trimeric soluble cytokine form of tumor necrosis factor that is found in blood plasma, i.e., the form that circulates throughout the body and confers TNF with its potent endocrine function.
  • transmembrane TNF or“tmTNF” or“mTNF” are used interchangeably herein to refer to transmembrane TNF.
  • tmTNF binds to only TNFR2.
  • antibody(ies) refers to an immunoglobulin molecule which specifically binds with an antigen.
  • Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins.
  • the term“antibody(ies)” includes whole antibodies of any isotype (e.g., IgG, IgA, IgM, IgE, etc.) or antibody fragments.
  • Antibodies or antibody fragments include Fab fragments, a Fab' fragments, a heavy chain antibodies, single-domain antibodies (sdAb), variable domain of a heavy chain antibodies, VHH, Nanobodies, single-chain variable fragments (scFv), a tandem scFvs, a bispecific T-cell engagers (BITEs), a diabodies, single-chain diabodies, DARTs, triple bodies, or a nanoantibodies.
  • the term“moDC” targeting moiety refers to a component associated with TNF that targets the TNF to moDC cells.
  • the moDC targeting moiety is an anti- CD64 antibody or antibody fragment.
  • the moDC targeting moiety is a fragment crystallizable region (Fc region) of an antibody.
  • the Fc is an Fc of a mouse IgG2a isotype or a human IgGl, IgG3 isotype.
  • A“disease” is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate.
  • a“disorder” in an animal is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.
  • patient “subject,”“individual,” and the like are used interchangeably herein, and refer to any animal, or cells thereof whether in vitro or in situ, amenable to the methods described herein.
  • patient, subject or individual is a human.
  • the term“cancer” or“tumor” as used herein means is intended to include any neoplastic growth in a patient, including an initial tumor and any metastases.
  • the cancer can be of the liquid or solid tumor type.
  • Liquid tumors include tumors of hematological origin (hematological cancer), including, e.g., myelomas (e.g., multiple myeloma), leukemias (e.g., Waldenstrom's syndrome, chronic lymphocytic leukemia, other leukemias), and lymphomas (e g, B-cell lymphomas, non-Hodgkins lymphoma).
  • Solid tumors can originate in organs, and include cancers such as lung, breast, prostate, ovary, colon, kidney, and liver.
  • cyclic dinucleotide or“CDN” refer to secondary signaling molecules including cyclic-di-GMP (CDG), cyclic-di-AMP (CDA), cyclic-di-IMP (CDI), cyclic-AMP- GMP (CDA/G), cyclic-AMP-IMP (CDA/I, and cyclic-GMP-IMP (CDG/I).
  • CDNs trigger interferon in a STING-dependent manner.
  • the term“specifically binds,” is used herein in reference to the interaction of an antibody, a polypeptide, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant, binding domain, or epitope) on the chemical species.
  • a ligand recognizes and binds to a specific receptor structure rather than to proteins generally.
  • an antibody recognizes and binds to a specific protein structure rather than to proteins generally.
  • therapeutic means a treatment and/or prophylaxis.
  • a therapeutic effect is obtained by a diminution, suppression, remission, or eradication of a disease state.
  • therapeutically effective amount refers to the amount of the subject compound that will elicit the biological or clinical response of a tissue, system, or subject that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • therapeutically effective amount includes that amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the signs or symptoms of the disorder or disease being treated.
  • the therapeutically effective amount will vary depending on the compound, the disease and its severity and the age, weight, etc., of the subject to be treated.
  • To“treat” a disease as the term is used herein, means to reduce the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject.
  • the term“vaccine” refers to a substance that induces anti-cancer or anti-pathogen immunity or suppresses the cancer or the pathogen upon later introduction of the cancer or pathogen into the subject.
  • the term“antigen associated with a disease or condition” refers to an antigen that induces an immune response against abnormal cells (e.g. cancer cells or infected cells) or pathogen cells (e.g. microbes) in a subject.
  • the term“vaccine composition” refers to a composition that includes an antigen and one or more adjuvant component(s).
  • immune response includes T cell mediated and/or B-cell mediated immune responses.
  • exemplary immune responses include T cell responses, e.g., cytokine production and cellular cytotoxicity, and B cell responses, e.g., antibody production.
  • immune response includes immune responses that are indirectly affected by T cell activation, e.g., antibody production (humoral responses) and activation of cytokine responsive cells, e.g., macrophages
  • Immune cells involved in the immune response include lymphocytes, such as B cells and T cells (CD4+, CD8+, Thl and Th2 cells); antigen presenting cells (e.g., professional antigen presenting cells such as dendritic cells, macrophages, B lymphocytes, Langerhans cells, and non-professional antigen presenting cells such as
  • an immune response involves activation of moDCs by a newly identified ( TNFR2 pRelB PD-L 1 CX3CRl + cDC2) described herein.
  • immunogen is defined as a molecule that induces an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both.
  • immunogens can be derived from recombinant or genomic DNA. A skilled artisan will understand that any DNA, which comprises a nucleotide sequences or a partial nucleotide sequence encoding a protein that elicits an immune response therefore encodes an“immunogen” as that term is used herein.
  • an immunogen need not be encoded solely by a full length nucleotide sequence of a gene. It is readily apparent that the present invention includes, but is not limited to, the use of partial nucleotide sequences of more than one gene and that these nucleotide sequences are arranged in various combinations to elicit the desired immune response. Moreover, a skilled artisan will understand that an immunogen need not be encoded by a“gene” at all. It is readily apparent that an immunogen can be generated, synthesized or can be derived from a biological sample.
  • composition includes, e.g.,
  • subcutaneous s.c.
  • intravenous i.v.
  • intramuscular i.m.
  • intradermal i.d.
  • Topical administering or administration of a composition includes contacting a body surface of the subject, including the skin, the eye, or the mucosa, with the composition.
  • Tnhalative administering or administration refers to delivery to the upper respiratory tract or lungs.
  • inhalative administration includes delivery via inhaling of a composition through the mouth and/or nose.
  • Intranasal administration includes inhalative administration through the nose.
  • co-administration or“co-administering” as used herein refers to the administration of an active agent before, concurrently, or after the administration of another active agent such that the biological effects of either agents overlap.
  • the combination of agents as taught herein can act synergistically to treat or prevent the various diseases, disorders or conditions described herein. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • CDNs include the bacterial second messengers cyclic di-GMP (CDG), cyclic di-AMP, 3’3’-cyclic GMP-AMP and the mammalian second messenger 2’3’-cyclic GMP-AMP 1 .
  • CDG is the founding member and the most studied CDNs 2 .
  • CDG does not cause acute toxicity in mice l .
  • CDG is a more potent activator of Thl and Th2 immune response than LPS, CpG oligonucleotides (ODN) or aluminum salt based adjuvant in mice 3 .
  • ODN CpG oligonucleotides
  • CDG adjuvanted vaccines protect mice from H5N 1 influenza 4 , Acinetobacter baumannii 5 , Staphylococcus aureus 6 , Klebsiella pneumoniae 7 and Streptococcus pneumoniae 8 ’ 9 .
  • CDG also showed cancer vaccine adjuvant activity in animal models 10, u .
  • MPYS also known as STING and MITA
  • STING and MITA is a receptor for CDNs 12 and a critical player in sensing cytosolic DNA 13, 14 .
  • MPYS 7 mice lose CDNs adjuvant activity 15 .
  • TNF signaling not type I IFN signaling, is essential for the adjuvant activity of CDG in vivo 15, 16 .
  • CDG when administered intranasally, CDG not only induced lung production of TNF, IL- 1 b but also the anti-inflammatory cytokine IL-10 17 . Consequently, CDG adjuvant does not induce over- inflammatory responses in the lung 17 .
  • the precise in vivo mechanism by which TNF mediates CDG adjuvant activity in vivo is unknown.
  • DCs orchestrate vaccine adjuvant responses 18 . They consist of developmentally and functionally distinct subsets that promote either immunogenic or tolerogenic immune responses 19 22 . Lungs have three DC subsets: the CDl03 + conventional DC (cCDl), the
  • cDC2 cDC2 + monocyte-derived CDl lb + CD24 CD64 + DC
  • Activated TNFR2 cDC2 expresses mTNF, which interacts with TNFR2 on moDC to drive moDC maturation promoting CDG adjuvant responses.
  • the methods of the present invention comprise administering an antigen, or a polynucleotide encoding an immunogen, and co-administering one or more adjuvant components, directly to a subject.
  • the one or more adjuvant components include a CDN, sTNF and/or CD64 antibody or antibody fragment.
  • adjuvant components include a CDN and a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF).
  • Administration of the antigen and one or more adjuvant components can comprise, for example, inhalative, intramuscular, intravenous, peritoneal, subcutaneous, and intradermal administration.
  • the antigen and adjuvant component(s) are administered concurrently. In an even more specific example, the antigen and adjuvant component(s) are administered in the same composition.
  • the actual dose and schedule can vary depending on whether the antigen and adjuvant component(s) are administered in combination with other pharmaceutical compositions, or depending on inter-individual differences in pharmacokinetics, drug disposition, and metabolism.
  • One skilled in the art can easily make any necessary adjustments in accordance with the exigencies of the particular situation.
  • Administration of the antigen and adjuvant component(s) may be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of the antigen and adjuvant component(s) may be essentially continuous over a preselected period of time or may be in a series of spaced doses. Both local and systemic administration is contemplated.
  • the amount administered will vary depending on various factors including, but not limited to, the composition chosen, the particular disease, the weight, the physical condition, and the age of the subject, and whether prevention or treatment is to be achieved. Such factors can be readily determined by the clinician employing animal models or other test systems which are well known to the art.
  • the antigen and adjuvant component(s) When the antigen and adjuvant component(s) are prepared for administration, they may be combined with a pharmaceutically acceptable carrier, diluent or excipient to form a pharmaceutical formulation, or unit dosage form.
  • a pharmaceutically acceptable carrier, diluent or excipient to form a pharmaceutical formulation, or unit dosage form.
  • the total active ingredients in such formulations include from 0.1 to 99.9% by weight of the formulation.
  • A“pharmaceutically acceptable” carrier, diluent, excipient, and/or salt is one that is compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • the active ingredient for administration may be present as a powder, as granules, as a solution, as a suspension or as an emulsion.
  • compositions containing the antigen and/or adjuvant component(s) can be prepared by procedures known in the art using well known and readily available ingredients.
  • the antigen and adjuvant component(s) can also be formulated as solutions appropriate for inhalative administration or parenteral administration, for instance by intramuscular, subcutaneous, intradermal or intravenous routes.
  • the pharmaceutical formulations of antigen and adjuvant component(s) can also take the form of an aqueous or anhydrous solution or dispersion, or alternatively the form of an emulsion or suspension.
  • parenteral administration e.g., by injection, for example, bolus injection or continuous infusion
  • the formulations may be presented in unit dose form in ampules, pre-filled syringes, small volume infusion containers or in multi-dose containers with an added
  • the active ingredients may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water
  • Devices for intranasal administration include spray devices. Suitable nasal spray devices are commercially available from Becton Dickinson, Pfeiffer GMBH and Valois. Spray devices for intranasal use do not depend for their performance on the pressure applied by the user.
  • Pressure threshold devices are particularly useful since liquid is released from the nozzle only when a threshold pressure is attained. These devices make it easier to achieve a spray with a regular droplet size. Pressure threshold devices suitable for use with the present invention are known in the art and are described for example in WO 91/13281 and EP 311 863 B. Such devices are currently available from Pfeiffer GmbH and are also described in Bommer, R.
  • intranasal devices produce droplets (measured using water as the liquid) in the range 1 to 500 pm. Below 10 pm there is a risk of inhalation, therefore it is desirable to have no more than about 5% of droplets below 10 pm.
  • Bi-dose delivery may also be implemented for intranasal delivery.
  • Bi-dose devices contain two subdoses of a single vaccine dose, one sub-dose for administration to each nostril.
  • the invention provides in a further aspect a pharmaceutical kit comprising
  • an intranasal administration device containing a vaccine formulation or comprising
  • an intranasal administration device and a separate vaccine formulation(s) for use with that device.
  • the unit content of active ingredient or ingredients contained in an individual dose of each dosage form need not in itself constitute an effective amount for treating the particular indication or disease since the necessary effective amount can be reached by administration of a plurality of dosage units. Moreover, the effective amount may be achieved using less than the dose in the dosage form, either individually, or in a series of administrations.
  • the pharmaceutical formulations may include, as optional ingredients, pharmaceutically acceptable carriers, diluents, solubilizing or emulsifying agents, and salts of the type that are well-known in the art.
  • pharmaceutically acceptable carriers include water and
  • physiologically acceptable buffered saline solutions such as phosphate buffered saline solutions pH 7.0-8.0.
  • the vaccination methods of the invention induce protective immunity against cancer or a pathogen, by generating an immune response directed against the cancer or the pathogen.
  • the methods of the invention induce production of pathogen-specific antibodies.
  • the methods of the invention induce a pathogen-specific cell-mediated immune response.
  • the methods of the invention induce production of pathogen- specific antibodies and a pathogen-specific cell- mediated immune response.
  • the methods of the invention induce production of tumor- specific antibodies.
  • the methods of the invention induce a tumor- specific cell-mediated immune response.
  • the methods of the invention induce production of tumor- specific antibodies and a tumor- specific cell- mediated immune response.
  • Anti-cancer and anti-pathogen immunity can be induced by administering an antigen and adjuvant component(s) described herein, and the induction of anti-cancer or anti-pathogen immunity enables treatment and prevention of a disease associated with cancer or the presence of the pathogen.
  • a decrease in mortality of individuals having a disease, a decrease of the disease markers in the blood, alleviation of detectable symptoms accompanying the disease and such are also included in the therapy or prevention of the disease associated with cancer or infection by the pathogen.
  • Such therapeutic and preventive effects are preferably statistically significant, for example, observed at a significance level of 5% or less, wherein the therapeutic or preventive effect of a vaccine against a disease, is compared to a control without vaccine administration.
  • Student's t-test, the Mann-Whitney U-test or ANOVA may be used for determining statistical significance.
  • Certain embodiments provide a method for treating, or preventing, a disease or condition associated cancer or infection by a pathogen.
  • the antigen and adjuvant(s) and methods of administering same may be administered prophylactic ally or therapeutically to subjects suffering from, or at risk of, or susceptible to, developing the disease or condition, including cancer or infection by a pathogen. Such subjects may be identified using standard clinical methods.
  • prophylactic administration occurs prior to the manifestation of overt clinical symptoms of disease, such that a disease or disorder is prevented or alternatively delayed in its progression.
  • the term“prevent” In the context of the field of medicine, the term“prevent”
  • composition embodiments described herein encompasses any activity which reduces the burden of mortality or morbidity from disease.
  • Prevention can occur at primary, secondary and tertiary prevention levels. While primary prevention avoids the development of a disease, secondary and tertiary levels of prevention encompass activities aimed at preventing the progression of a disease and the emergence of symptoms as well as reducing the negative impact of an already established disease by restoring function and reducing disease-related complications.
  • the induction of anti-cancer or anti-pathogen immunity by composition embodiments described herein can be further confirmed by observing the induction of antibody production against specific immunogens.
  • the one or more antigens are determined to induce anti-cancer or anti-pathogen immunity.
  • antigens include but are not limited to the following:
  • the vaccine formulation can include multiple antigens.
  • one formulation can include CDG+PspA+H7/HA to protect infection from both Influenza and pneumococcal infection in one immunization.
  • CDG directly targets lung cDCl and cDC2, not moDCs.
  • DCs mediate CDG adjuvant activity in vivo 17.
  • All DC subsets express the CDG receptor MPYS, which is an ER-resident protein (Fig 1A).
  • CDG has two phosphate groups preventing it from directly passing through the cell membrane.
  • mice were intranasally administered with FITC-conjugated CDG and FITC+ lung cells were examined after 5hrs.
  • Alveolar macrophages take up CDG but are dispensable for CDG adjuvant activity.
  • AM (CD1 lc + MHC IT nt ) took up most of the fluorescent CDG in vivo (Fig 11A). Unlike DCs, AM did not increase expression of CD86 following CDG treatment (Fig 11B). To determine whether AM are required for CDG responses in vivo, we used the MPYS fl/fl LysM cre mice 17 , which deleted MPYS expression in AM (Fig 11C). The activation of lung DCs by CDG was unaltered in the MPYS ll/ri LysM trc mice (Fig 11D).
  • MPYS ll/ri LysM trc mice produced similar anti-pneumococcal surface protein A (PspA) antibody as the WT upon CDG/PspA immunization (Fig 11E).
  • PspA anti-pneumococcal surface protein A
  • CDG differentially activates lung DC subsets in vivo.
  • DCs subsets are functionally distinct.
  • CDG increased CD86 and CCR7 expression in lung cDCl and cDC2 (Fig 1C & 1D) 17 .
  • both cDCs migrate, bearing processed antigen, to the lung draining lymph nodes (dLNs) (Fig 12A-B).
  • dLNs lung draining lymph nodes
  • cDCl activated RelA
  • Fig 1F cDC2 activated RelB
  • some cDC2 have constitutively activated RelB (Fig 1F).
  • IRF4 fl/fl CDl lc cre and Batf3 _/ mice we used the IRF4 fl/fl CDl lc cre and Batf3 _/ mice.
  • cDCl and cDC2 are controlled by transcriptional factors Batf3 and IRF4 respectively.
  • IRF4 fl/fl CDl lc cre mice lack cDC2 in the lung (Fig 10C) 28-30 while Batf3 -/- mice lack cDCl (Fig 10D) 31, 32 .
  • CDG-induced lung production of TNF, IL- 1 b and IL-l2p70 were dramatically reduced in IRF4 n/ll CDl lc cre mice but not in the Batf3 -/- mice (Fig 12C).
  • CDG-induced MCP-l production was largely absent in the Batf3 -/- mice but not in the IRFd ⁇ CDl lc cre mice (Fig 12C).
  • intranasally administered CDG differentially activates lung cDCl and cDC2 in vivo.
  • CDG indirectly activates moDCs
  • moDCs did not take up CDG, they increased expression of CD86 in response to intranasal administration of CDG (Fig 1C). This was independent of MPYS expression in moDCs as moDCs in MPYS 11/11 LysM trc mice had normal levels of CD86 expression (Fig 11D). Notably, activated moDCs did not increase CCR7 (Fig 1D) and did not migrate to dLNs (Fig 12B). Last, moDCs activated both RelA and RelB in response to CDG (Fig 1G). We concluded that CDG indirectly activate moDCs and activated lung moDCs were not migratory 25, 33 .
  • IRF4 fl/fl CDl lc cre mice (Fig 10C).
  • CDG adjuvant activity was examined in the IRF4 fl/fl CDl lc cre mice. Mice were intranasally administered with PspA alone or with CDG. PspA-specific Ab responses were examined in the blood and bronchoalveolar lavage fluid (BALF). Unlike the WT mice, CDG did not induce anti-PspA Abs in BALF and serum (Fig 1H, 12D-E) from immunized IRF4 fl/fl CDl lc cre mice.
  • IRF4 n/ll CDl lc cre mice were then immunized with CDG/PspA.
  • WT cDC2 generated PspA-specific serum IgG and IgA in the IRF4 fl/fl CDl lc cre mice similar to the WT mice (Fig II).
  • lung cDC2 are critical for CDG adjuvant activity.
  • TNFR2 is essential for CDG adjuvant activity
  • TNF signaling is critical for CDG adjuvant activity in vivo 15, 16 .
  • TNF signals through two TNFR1 and TNFR2.
  • TNFR1 binds transmembrane TNF (tmTNF) and soluble TNF (sTNF) while TNFR2 only binds to tmTNF 34-37 .
  • the lung DC compartment is not altered by the lack of either TNFR1 or TNFR2 (Fig. 13A-B).
  • CDG induced reduced humoral and cellular immune responses in TNFR1 7 mice (Fig 2A, 13C-F).
  • CDG completely lost its adjuvant activity in TNFR2 7 mice (Fig 2A, 13C-F).
  • TNFR2 expression is essential for CDG adjuvant activity.
  • TNFR2 expression on lung cDC2 is required for its maturation
  • cDC2 are critical for CDG adjuvant activity (Fig 1).
  • FIG. 1 We next examined lung cDC2 maturation in the TNFR2 7 mice.
  • CDG did not enhance CD86 or CCR7 expression in lung cDC2 of TNFR2 7 mice in vivo (Fig. 2B & 14A).
  • CDG induced CD86 and CCR7 expression on cDCl of TNFR2 7 mice in vivo (Fig 14A-B).
  • TNFR2 expression on lung cDC2 are required for their maturation in response to CDG in vivo.
  • TNFR2 + lung cDC2 have constitutively activated RelB
  • TNFR2 on lung cDC2 is required CDG-induced cDC2 maturation in vivo (Fig 2C).
  • Fig 2C CDG-induced cDC2 maturation in vivo
  • TNFR2 + cDC2 is required for CDG-induced Thl and TH17 responses but dispensable for the humoral responses
  • cDC2 is a heterogeneous population 21 ⁇ 32 ⁇ 38 ⁇ 39 .
  • lung TNFR2 + pRelB + cDC2 were mature and required for the Thl/Thl7 responses but not humoral responses while the TNFR2 cDC2 was not mature but mediates CDG-induced antibody response.
  • TNFR2 expression could define functionally distinct lung cDC2 subsets.
  • TNFR2 + cDC2 were able to rescue Thl/Thl7 responses in the IRF4 fl/fl CDl lc cre mice (Fig 4F).
  • TNFR2 + cDC2 also rescued Th2 response, which is different from the RclB n/ll CDl lc cre mice (Fig 4F & 3C).
  • Th2 response which is different from the RclB n/ll CDl lc cre mice (Fig 4F & 3C).
  • TNFR2 cDC2 In contrast, adoptive transfer of TNFR2 cDC2 completely restored antibody, but not Th, responses in MPYS 7 (Fig 4D, 15A-B) and IRF4 fl/fl CDl lc cre mice (Fig 4E-F). In fact, levels of anti-PspA IgG and IgA in IRF4 n/ll CDl lc cre mice receiving TNFR2 cDC2 were similar to the WT (Fig 4E).
  • lung cDC2 can be divided into two functionally distinct subsets: TNFR2 + cDC2 and TNFR2 cDC2.
  • the TNFR2 + cDC2 are important for CDG-induced cellular immunity, while TNFR2 cDC2 are responsible for CDG-induced humoral responses
  • TNFR2 + and TNFR2 lung cDC2 are derived from pre-cDC2 but not from each other
  • TNFR2 + cDC2 are positive for BTLA, PDL-l, arginase 1 (Argl).
  • the TNFR2 + cDC2 also have mixed expression of PD-L2 and CD30lb (Fig 5A).
  • the lung TNFR2 cDC2 expressed CX3CR1 (Fig 5A).
  • Both populations express common cDC2 markers as SIRPa, CD26, IRF4 and Zbtb46 (Fig 5 A, 5B & 16A) 40 ’ 41 .
  • both subsets of cDC2 were absent in Flt3 7 mice (Fig 16B-D).
  • TNFR2 + and TNFR2 cDC2 are negative for cDCl markers IRF8, XCR1 and not affected in Batf3 _/ mice confirming their identity as cDC2 (Fig 5B, 16C-D).
  • the CD45.l + TNFR2 + cDC2 expressed BTLA, PD-L1 and had mixed expression of PD-L2 and CD30lb (Fig 5E), similar to the resident TNFR2 + cDC2.
  • the CD45.l + TNFR2 cDC2 only expressed CX3CR1 (Fig 5E).
  • TNFR2 + and TNFR2 cDC2 populations represent different developmental/active stage of cDC2.
  • CDG CD45.1 lung TNFR2 + and TNFR2 cDC2 into MPYS 7 mice
  • the recipient mice were then activated by CDG (i.n.).
  • TNFR2 cDC2 did not upregulate TNFR2 while TNFR2 + cDC2 maintained their expression of TNFR2 (Fig 5F).
  • CDG treatment did not affect PD-L1 expression either (Fig 5F).
  • lung cDC2 consist of two functionally and developmentally distinct subsets, TNFR2 + and TNFR2 cDC2.
  • CDG activates TNFR2-deficient cDC2 in vivo
  • cDC2 produced TNF in TNFR2 7 mice (Fig 6C). Deleting TBK1 in hematopoietic and endothelial lineages (TBK 1 11/11 Vav Crc ) dramatically reduced CDG-induced lung TNF production (Fig 6D) suggesting TBK1 is needed for TNF production by CDG. Indeed, TBK1 was activated in the TNFR2 7 cDC2 (Fig 6E). Last, adoptive transferred TNFR2 cDC2 produced lung TNF in MPYS 7 lung (Fig 4B). We concluded that although TNFR2 cDC2 fail to mature, they were activated by CDG in vivo.
  • TNFR2 7 cDC2 restored CDG responses in IRF4 n/ll CD l l c trc mice (Fig 3A). Yet, TNFR2 7 mice had no CDG responses (Fig 2A). We reasoned that TNFR2 expression on moDCs may be important for CDG responses in vivo. Indeed, we found that CDG induced TNFR2 on moDCs in WT mice (Fig 7E) and moDCs from TNFR2 7 mice did not upregulate CD86 in response to CDG in vivo (Fig 7F). Last, adoptive transfer of WT monocytes into TNFR2 7 mice restored CDG-induced IgG and IgA responses (Fig 7G). Notably, adoptive transfer WT cDC2 into TNFR2 7 mice did not restore CDG-induced antibody responses (Fig 7H). We concluded that moDCs expression of TNFR2 is critical for its maturation and subsequent induction of CDG adjuvant response.
  • CDG induces mTNF expression on TNFR2 cDC2 in vivo
  • TNFR2 + and TNFR2 cDC2 produced TNF upon intranasal CDG treatment (Fig 4B). We asked if the lung TNFR2 cDC2 specifically expressed mTNF.
  • DQ + cDC2 were DQ-Green (Fig 8D). Strikingly, comparing to the TNFR2 + cDC2 subset, very few TNFR2 cDC2 subset were DQ + (Fig 8D) suggesting that the TNFR2 cDC2 either did not take up antigen or were not efficient at antigen processing. In comparison, all DQ + moDCs were TNFR2 + cells (Fig 8D) indicating TNFR2 + moDCs were indeed mature DCs. moDCs promote CDG-induced Tfti and GC B cells generation in the lung
  • Tfh cells and GC B cells play central roles in promoting humoral responses.
  • CDG/PspA immunization i.n.
  • moDCs are activated by TNFR2 cDC2.
  • TNFR2 cDC2 restoring TNFR2 cDC2 in the
  • IRF4 n/ll CDl lc cre mice should restore Tfh cells. Indeed, we found that adoptive transfer of TNFR2 cDC2, but not TNFR2 + cDC2, into IRF4 n/ll CDl lc cre mice generated Tfh cells (Fig 9E). Together, we propose that CDG activates TNFR2 cDC2 that matures moDCs to generate Tfh and GC B cells promoting CDG-induced antibody responses in vivo (Fig 9F).
  • mice Eight to sixteen-week old mice, both males, and females, were used for all experiments.
  • MPYS ' mice (Trnernl73 ⁇ tmlCamb> ) have been described previously 38 ’ 39 .
  • the following strains were obtained from The Jackson Laboratory: Irf fl (#009380) 40 , Batf3 (#Ol3755) 30 , TnfrP (#002818) 41 , Tnfr2- (#002620) 42 , Re IB 1 ' (#0287l9) 43 , CDllC cre (#008068) 44 .
  • Vav trt' -TBK 1 11/11 mice were from Dr. Fitzgerald’s lab. All mice are on a C57BL/6 background. Mice were housed and bred in the Animal Research Facility at Albany Medical College and the University of Florida. All experiments with mice were performed by the regulations and approval of the Institutional Animal Care and Use Committee from Albany Medical College or the University of Florida.
  • mice [0115] Groups of mice (4 per group) were intranasally vaccinated with CDG (5pg, Invivogen, cat# vac-cdg) adjuvanted PspA (2pg, BEI Resources) or PspA alone 17 . Mice were immunized twice at 14 days interval. For intranasal vaccination, animals were anesthetized using isoflurane in an E-Z Anesthesia system (Euthanex Corp, Palmer, PA). PspA, with or without CDG was administered in 20m1 saline. Sera were collected 14 days after the last immunization. The PspA- specific Abs were determined by ELISA.
  • mice were intranasally administered 5pg CDG, then sacrificed after 5hrs by C02 asphyxiation 17 .
  • Lungs were perfused with cold PBS. The harvested lungs were washed with PBS once, then stored in 0.7ml Tissue protein extraction reagent (T-PER) (Thermo Scientific, cat#785l0) containing protease inhibitors (Roche, cat#l 1836153001) at -80°C. Later, the lung was thawed on ice and homogenized with Minilys® (Precellys, 5,000 RPM for 30sec) using Precellys lysing kit (Precellys, cat# KT03961). Lung homogenates were transferred to a l.5ml tube and spun at l4,000g for 30min at 4°C. The supernatant was collected and analyzed for TNL production by ELISA (eBioscience, cat#88-7324).
  • mice were first given CDG (i.n.). Half an hour later, treated mice were administered (i.n.) with 50pg/50pl anti-TNLR2 Ab (BioLegend, TR75-32.4), or isotype control (BioLegend, HTK888). Lung was harvested l6hr later and analyzed by Llow cytometer.
  • mice were intranasally administered with or without CDG (5pg, vaccine-grade). After 20hrs, the lungs were lavaged and perfused with ice-cold PBS. Excised lungs were digested in DMEM containing 200pg/ml DNase I (Roche, 10104159001), 25mg/ml Liberase TM (Roche, 05401119001) at 37°C for 3hrs. Red blood cells were then lysed and a single cell suspension was prepared and analyzed by BDTM LSR II and FACScan flow cytometry.
  • mice were intranasally administered 20pg DQTM-Ovalbumin (DQ-OVA) (Life technologies, D12053) with, or without CDG (5pg, vaccine-grade). After 20hrs, the lungs were lavaged, perfused and harvested. Lung cells were analyzed by BDTM LSR II and FACScan flow cytometry.
  • DQ-OVA DQTM-Ovalbumin
  • CD80 (16-10A1), CD86 (GL1), CD11B (Ml/70), CD11C (N418), FceRIa (MAR-1), MHC II (M5/114.15.2), CDl03(2E7), CD24 (ml/69), CD64 (x54-5/7.l), CCR7 (4B 12), TNF (MP6-XT22), TNFR2 (TR75-89), PD-L1 (10F.9G2), BTLA (8F4), PD-L2 (TY25), CD30lb (URA-l), CX3CR1 (SA011F11), CDl72a (P84), CD44 (IM7).
  • TNFR2 Ab cat# 50128-R112-A
  • TNFR2-Fc mouse TNFR2 extracellular domain, Met l-Gly 258, was fused with the Fc region of human IgGl at the C-terminus, cat# 50128-M02H), human IgGl Fc (cat# 10702-HNAH).
  • Alexa Fluor®488 mouse anti-human IgGl Fc was from Invitrogen (cat# A- 10631).
  • FITC-CDG (2’-Fluo-AHC-CDG ) was from Biolog (cat# F009).
  • Cell sorting was performed on the BD FACSArialll Flow Cytometer and Cell Sorter. After sorting, dendritic cells were CFSE labeled, according to the protocol from the manufacturer (Invitrogen). Intracellular staining
  • mice were intranasally administered saline or cyclic di- GMP (5pg, vaccine-grade). The lungs were lavaged, perfused, and harvested at 5hr post treatment. Excised lungs were washed in PBS and digested in DMEM containing 200pg/ml DNase I (Roche, 10104159001), 25pg/ml Liberase TM (Roche, 05401119001), and Golgi-plug at 37°C for 6hrs. The single lung cell suspension was fixed in Cytofix/perm buffer (BD
  • Pulmonary TNFR2 + and TNFR2 cDC2 were sorted from the lungs of naive donor mice with a FACSArialll flow cytometer. After sorting, dendritic cells were CFSE labeled, according to the protocol from the manufacturer (Invitrogen). Between 500,000 - 1,000,000 cells were administered intranasally into recipient mice. 24 hours later of transfer, recipient mice were intranasally vaccinated with CDG (5pg, Invivogen, cat# vac-cdg) adjuvanted PspA (2pg, BEI Resources) or PspA alone 17 . Recipient mice received two doses of transferred cells and were immunized at 14 days interval. Statistical Analysis
  • C57BF/6J mice were immunized with Influenza nucleoprotein (NP)/TNF-Fc(IgG2A) or NP/TNF D22iN/A223R -Fc(IgG2A) as shown in FIG. 25.
  • CCF20 is a T cells recruiting chemokine and Bcl6 is a transcriptional factor important for memory cells development.
  • the data in FIG. 25A shows that soluble TNF- Fc induced CCF20 expression in moDCs, and that this effect was stronger than tmTNF-Fc treatment.
  • FIG. 25B shows that sTNF-Fc induced Bcl6 expression in moDCs and that this effect was stronger than tmTNF-Fc treatment.
  • IgG2a examples include, but are not limited to, accession nos AAN76044, AAN76043.1, AAN76042.1, AXN93670.2, and AXN93670.2 and
  • IgG3 examples include CAA67886.1, CAC10266.1, CAC10265.1, and CAC10264.1 Also see Human IgGl, IgG3, and IgG3 Hinge-Truncated Mutants Show Different Protection Capabilities against Meningococci Depending on the Target Antigen and Epitope Specificity S. Giuntini, D. M. Granoff, P. T. Beemink, O. Ihle, D. Bratlie, T. E. Michaelsen
  • c-di-GMP is an effective immunomodulator and vaccine adjuvant against pneumococcal infection. Vaccine 2008; 26(36): 4676-4685.
  • Blaauboer SM Gabrielle VD, Jin L. MPYS/STING-mediated TNF-alpha, not type I IFN, is essential for the mucosal adjuvant activity of (3'-5')-cyclic-di-guanosine-monophosphate in vivo. J Immunol 2014; 192(1): 492-502.
  • Nanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvants. J Clin Invest 2015; 125(6): 2532-2546. 17. Blaauboer SM, Mansouri S, Tucker HR, Wang HL, Gabrielle VD, Jin L. The mucosal adjuvant cyclic di-GMP enhances antigen uptake and selectively activates pinocytosis-efficient cells in vivo. Elife 2015; 4.
  • transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 1995; 83(5): 793-802.
  • the type 1 receptor (CDl20a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci U SA 1998; 95(2): 570-575.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Endocrinology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Otolaryngology (AREA)
  • Virology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Described herein are novel vaccine compositions and methods for use thereof in inducing an immune response in a subject especially aged subjects. Specifically exemplified are vaccine compositions that include an antigen; a cyclic dinucleotide; soluble tumor necrosis factor (TNF); or a CD64 antibody or antibody fragment. Optionally, the vaccine composition comprises a TNF conjugated with a moDC targeting moiety in addition to or in place of TNF or CD64 antibody or antibody fragment, or both TNF and CD64 antibody or antibody fragment.

Description

TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE
GOVERNMENT SUPPORT
This invention was made with government support under Grant Nos. All 10606, AI125999, AI132865 awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND
[0001] Typically, pathogens enter the body via mucosal surfaces. Vaccination by mucosal routes is viewed to be more effective at inducing protective immunity against mucosal pathogens, and mucosal vaccines have the benefits of low cost and ease of administration. However, there are very few mucosal vaccines approved for use, which is largely due to problems with developing safe and effective mucosal adjuvants.
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention is illustrated by way of example, and not by way of limitation, in the figures of the accompanying drawings.
[0002] FIG 1. cDC2 play a central role in mediating the adjuvant activity of CDG. A. Flow cytometry analysis of MPYS expression in lung DC subsets. n>3. B. Absolute number of lung DC subsets in C57BL/6 mice administered (i.n.) with saline or 5pg FITC-CDG for 5 hours. n=3. C. Absolute number of CD86+ lung DC subsets in mice administered (i.n.) with saline or 5pg CDG for 16 hours. n>3. D. Absolute number of CCR7+ lung DC subsets in mice administered (i.n.) with saline or 5pg CDG for 16 hours. n>3. E-G. Flow cytometry analysis of pRelA and pRelB in cDCl (E), cDC2 (F), and moDCs (G) from mice treated with saline or CDG for l6hrs. n>3. H. C57BL/6, Batf3_/ , and IRF4n/llCDl l ctrc mice were immunized (i.n.) with two doses (14 days apart) of PspA or PspA plus CDG (5ug). Anti-PspA IgG in serum and IgA in BALF were determined by ELISA. n>3. 1. IRF4n/llCDl l ctrc mice were adoptively transferred (i.n.) with lung cDC2 sorted from WT mice lung and immunized (i.n.) with PspA or CDG/PspA. Serum anti- PspA IgG and BALF IgA were determined by ELISA. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0003] FIG 2. cDC2 expression of TNFR2 is required for CDG-induced lung cDC2 maturation in vivo by activating RelB. A. C57BL/6 (WT), TNFRL7 and TNFR2 7 mice were immunized (i.n.) with two doses of PspA or PspA plus CDG (5ug). Serum anti-PspA IgG and BALF IgA were determined by ELISA. n=3. B. WT and TNFR27 mice were treated (i.n.) with saline or CDG (5pg) for 16 hours. CD86 expression in lung cDC2 were determined by Flow cytometry. n=3. C. IRF4n/llCDl l ctrc mice were adoptively transferred (i.n.) with lung cDC2 sorted from WT or TNFR27 mice lung. The recipient mice were administered (i.n.) with saline or CDG (5pg) for l6hrs. CD86 expression in lung cDC2 were determined by Flow cytometry. n=3. D. Flow cytometry analysis of TNFR2 expression on lung cDC2 in WT mice administered (i.n) with saline or CDG for l6hrs. n>3. E. Flow cytometry analysis of TNFR2 expression on pRelB+ cDC2 from mice treated with saline or CDG. n=3. F. Flow cytometry analysis of pRelB expression on TNFR2+ cDC2 from mice treated with CDG. n=3. G. Flow cytometry analysis of pRelB expression on cDC2 from WT and TNFR2 7 mice. n=3. H. Flow cytometry analysis of CD86 expression on lung cDC2 in RelB 11/11 and RclBn/llCD l 1 CC C mice treated with CDG. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test). [0004] FIG 3. cDC2 expression of TNFR2 and RelB is required for Thl and Thl7 responses, but dispensable for CDG-induced antibody response. A. IRF4fl/flCDl lccre mice were adoptively transferred (i.n.) with lung cDC2 sorted from WT or TNFR27 mice lung and immunized (i.n.) with PspA or CDG/PspA. Serum anti-PspA IgG and BALF anti-PspA IgA were determined by ELISA. n=3. B. RelB 11/11 and RclBl1/nCDl 1CC C mice were immunized (i.n) with CDG/PspA or PspA alone as before. Serum anti-PspA IgG and BALF anti-PspA IgA were determined by ELISA. n=3. C. Lung cells from immunized RelB 11/11 and RclBn/llCDl 1CC C mice were stimulated with 5pg/ml PspA for 4 days in culture. Cytokines were measured in the supernatant by ELISA. n>3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired
Student’s t test).
[0005] FIG. 4: Lung TNFR2 cDC2 mediate CDG-induced humoral response. A. Flow cytometry analysis of TNFR2 expression on lung CDG-FITC+ cDC2 from WT mice. n=3. B. Lung TNF production in CDG treated (i.n.) MPYS 7 mice adoptively transferred with WT TNFR2+ or TNFR2 lung cDC2. n=3 C. Sorted TNFR2+ and TNFR2 lung cDC2 from WT mice were labelled with CFSE and adoptively transferred into the MPYS 7 mice. Recipient mice were treated with CDG (i.n.) for l6hrs. CD86 and CCR7 expression on CFSE positive lung cells were examined by Flow cytometry. n=3. D. Sorted TNFR2+ and TNFR2 lung cDC2 from WT mice were adoptively transferred into the MPYS 7 mice. Recipient mice were immunized (i.n.) with PspA or CDG/PspA twice. Serum anti-PspA IgG were determined by ELISA. n=3. E. Sorted TNFR2+ and TNFR2 lung cDC2 from WT mice were adoptively transferred into
IRF4n/llCDl lccre mice. Recipient mice were immunized (i.n.) with PspA or CDG/PspA. Serum anti-PspA IgG were determined by ELISA. n=3. F. Lung cells from recipient IRF4fl/flCDl lccre mice were stimulated with 5mg/ml PspA for 4 days in culture. Cytokines were measured in the supernatant by ELISA. G. A cartoon illustrating following CDG administration, TNFR2+ cDC2 activate RelB to induce Thl, Th2 and Thl7 responses while TNFR2 cDC2 mediate the antibody response. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0006] FIG. 5: Lung cDC2 consist of two distinct subsets. A. Flow cytometry analysis of lung TNFR2+ vs TNFR2 cDC2 at steady-state. n=3. B-C. Flow cytometry analysis of IRF4 and IRF8 (B) and Zbtb46 (C) in TNFR2+ and TNFR2 cDC2. n=3. D. Flow cytometry analysis in lungs of IRF4n/llCDl lctrc mice reconstituted with CD45.1+ pre-cDC2. n=3. pre-cDC2 were sorted from the bone marrow of B6.CD45.1 mice and transferred (i.n.) into IRF4fl/flCDl lccre mice. n=3. E. Phenotypic analysis of CD45.1 TNFR2+ and TNFR2 cDC2 transferred into IRF4fl/flCDl lccre mice. n=3. F. Flow cytometry analysis of TNFR2 expression on cDC2 subsets transferred into MPYS 7 mice treated with CDG (i.n.) for l6hrs. n=3.
[0007] FIG 6. CDG activates TNFR27 cDC2 in vivo to produce TNF. A-B. Indicated mice were administered (i.n.) with saline or CDG for 5hrs. TNF production was measured in lung homogenates by ELISA. n>3. C. WT and TNFR27 mice were treated with saline or CDG for 16 hours. TNF in lung cDC2 was determined by an intracellular cytokine stain. n=3. D. TBKlfl/fl and TBKlfl/flVavcre mice were administered (i.n.) with saline or CDG for 5hrs. TNF production was measured in lung homogenates by ELISA. n=3. E. Flow cytometry analysis of p-TBKl expression in lung cDC2 from WT and TNFR27 mice treated with saline or CDG for 16 hours. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test). [0008] FIG 7. TNFR2 expression on moDCs is required for CDG adjuvant activity. A-B.
WT, IRF4fl/flCDl lccre (A) and Batf3_/ (B) mice were treated (i.n.) with saline or CDG (5m g for 16 hours. CD86 expression in lung moDC were determined by Flow cytometry. n=3. C. WT and TNFR2 7 cDC2 were adoptively transferred into the IRF4n/llCDl lccre mice. The mice were treated with CDG (i.n.) for l6hrs. CD86 expression on endogenous moDC were examined by Flow cytometry. n=3. D. Flow cytometry analysis of CD86 expression on lung moDCs in RclB 11/11 and RelBfl/flCDl lcCre mice treated with CDG for l6hrs. n=3. E. Flow cytometry analysis of TNFR2 expression on lung moDC of WT mice treated (i.n.) with saline or CDG for l6hrs. n=3. F. Flow cytometry analysis of CD86 expression on lung moDCs from WT or TNFR27 mice treated (i.n.) with saline or CDG for l6hrs. n=3. G. WT, TNFR2 / mice or TNFR27 mice receiving (i.n.) WT monocytes or TNFR2 7 monoctyes were immunized with CDG/PspA. Serum anti-PspA IgG and BALF anti-PspA IgA were determined by ELISA. n=3. H. WT cDC2 adoptively transferred into TNFR2_/ mice were immunized with CDG/PspA. Serum anti-PspA IgG were determined by ELISA. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0009] FIG 8. TNFR2 cDC2 express mTNF but have few processed antigen. A. WT mice were administered with saline or CDG (5pg) for l6hrs. mTNF expression was determined by Flow cytometry using mouse TNFR2-Fc recombinant protein. n=3. B-C. Flow cytometry analysis of mTNF expression in lung DCs (B) and cDC2 (C) n=3. D. Flow cytometry analysis of antigen uptake and processing in lung CD1 lb+ DC from WT mice treated (i.n.) with DQ-OVA (20ug) and CDG (5ug) for 16 hours. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0010] FIG. 9: moDCs promote CDG-induced Tfh and GC B cell generation in the lung. A-
B. WT mice were administered with Ea-OVA (lOpg) or Ea-OVA/CDG (5pg) for l6hrs. YAE+ moDCs (A) and CD86+YAE+ moDCs were determined by Flow cytometry. n=3. C-D. WT, TNFR2 Amicc or TNFR2 7 mice receiving (i.n.) WT monocytes were immunized with
CDG/PspA. At Day 14, CD4+PDl+CXCR5+ Tfh (C) and CDl9+ Bcl6+ B cells (D) were determine in lung by flow cytometry. n=3. E. TNFR2+ and TNFR2 cDC2 were adoptively transferred into IRF4n/llCDl lccre mice and immunized with CDG/PspA. At Day 14, CD4+ Bcl6+ Tfh were determine by flow cytometry. n=3. F. Model: following CDG administration, TNFR2 cDC2 produce mTNF to activate moDCs, which will generate Tfh to mediate the antibody response. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0011] FIG. 10 A-B. Gating strategy for lung DCs. cDCl are CDl lc+MHCIIhiCD103+CDl lb- CD64-IRF8+, cDC2 are CD1 lc+MHCIIhiCDl lb+ CD64-IRF4+, moDCs are
CDl lc+MHCIIhiCDl lb+CD64+. n>3. C. Flow cytometry analysis of lung DCs subsets in IRF4fi/fi and IRF4fi/flCDl lccre mice including the total number of DCs. n=3. D. Flow cytometry analysis of lung DCs subsets in C57BF/6 (WT) and Batf3-/- mice including the total number of DCs. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0012] FIG. 11 A WT mice were intranasally administered with saline or 5pg FITC-CDG for 5 hours. Fung FITC positive cells were analyzed by Flow cytometry. n>3. B. CD86 expression on
CDl lc+MHCIIint Alveolar macrophages (AMs) and lung DCs (CDl lc+MHCIIhi) in WT mice treated (i.n.) with saline or 5mg CDG for 16 hours. n=3. C. Flow cytometry analysis of MPYS expression in AMs (SiglecF+ CDl lc+) from MPYSfl/flLysMcre mice. n=3. D. Absolute number of CD86+ lung DC subsets in WT and MPYSfl/flLysMcre mice administered (i.n.) with saline or 5pg CDG for 16 hours. n=3. E. MPYS-/-, MPYSfl/fl, MPYSfl/flLysMcre, and
MPYSfl/flCDl lccre mice were immunized (i.n.) with two doses (14 days apart) of PspA or PspA plus CDG (5ug). Anti- PspA Abs in serum were determined by ELISA. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0013] FIG 12. A-B. Flow cytometry analysis of antigen uptake and processing in DCs from the lungs (A) and dLNs (B) of WT mice treated (i.n.) with DQ-OVA (20ug) and CDG (5ug) for 16 hours. n=3. C. Indicated mice were treated (i.n.) with saline or CDG for 5 hrs. Lung MCP-l, TNF, IL- 1 b and ILl2p70 production were measured by ELISA in lung homogenates. n>3.
D.WT, Batf3-/-, and IRF4fl/flCDl lccre mice were immunized (i.n.) with two doses (14 days apart) of PspA or PspA plus CDG (5ug). Anti-PspA IgGl and IgG2c in serum and BALF were determined by ELISA. n>3. EF. Splenocytes (E) or lung cells (F) from immunized WT, Batf3-/-, and IRF4fl/flCDl lccre mice were stimulated with 5pg/ml PspA for 4 days in culture. Cytokines were measured in the supernatant by ELISA. n>3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0014] FIG. 13. A-B. Flow cytometry analysis of lung DCs subsets from WT, TNFR1-/-, and TNFR2-/- mice. n=3 C-D. WT, TNFR1-/- and TNFR2-/- mice were immunized (i.n.) with two doses (14 days apart) of PspA or PspA plus CDG (5ug). Anti-PspA IgGl (C) and IgG2c (D) in serum and BALF were determined by ELISA. n>3. E-F. Splenocytes (E) or lung cells (F) from immunized WT, TNFR1-/-, and TNFR2-/- mice were stimulated with 5mg/ml PspA for 4 days in culture. Cytokines were measured in the supernatant by ELISA. n>3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0015] FIG. 14. A. WT, TNFR2-/- mice were treated (i.n.) with saline or CDG (5pg) for 16 hours. CCR7 expression in lung cDCl and cDC2 were determined by Flow cytometry. n=3. B. WT, TNFR2-/- mice were treated (i.n.) with saline or CDG (5pg) for 16 hours. CD86 expression in lung cDCl were determined by Flow cytometry. n=3. C. C57B6/J mice were administered (i.n.) with saline or CDG (5pg). Isotype control Ab (50pg) or anti-TNFR2 Ab (50pg) was administered (i.n.) 30min later. The lungs were harvested 16 hours later. CD86 expression in lung cDC2 was determined by Flow cytometry. n=3. D. Flow cytometry analysis of TNFR2 expression on lung cDCl in WT mice administered (i.n) with saline or CDG for l6hrs. n>3. E. Flow cytometry analysis of lung DCs populations in RelBfl/fl and RelBfl/flCDl lCCre mice. n=3. F. Flow cytometry analysis of CD86 expression on lung cDCl in RelBfl/fl and
RelBfl/flCDl lCCre mice treated with CDG. n=3.
[0016] FIG. 15. A-B. Sorted TNFR2+ and TNFR2- lung cDC2 from WT mice were adoptively transferred into the MPYS-/- mice. Recipient mice were immunized (i.n.) with CDG/PspA. Serum anti-PspA IgGl (A) and IgG2c (B) were determined by ELISA. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0017] FIG. 16. A. Flow cytometry analysis of CD26 expression on lung TNFR2+ and TNFR2- cDC2. B-D. Flow cytometry analysis of lung DCs subsets in WT, Flt3 -/- and Batf3-/- mice. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by and asterisk (*) where p<0.05 (unpaired Student’s t test).
[0018] FIG. 17. A. pre-cDC2 gating strategy in the bone marrow. Naive B6.CD45.1 mice were used. B. Experimental scheme for the transfer of pre-cDC2 into IRF4fl/flCDl lccre mice (CD45.2). C. Phenotypic analysis of CD45.1 DCs transferred into IRF4fl/flCDl lccre mice. D. Experimental scheme for the adoptive transfer of TNFR2+ and TNFR2- lung cDC2 into the MPYS7- mice.
[0019] FIG. 18.A-B. 3-month old C57BF/6( Adult), l8month old C57BF/6 (Aged) mice were immunized (i.n.) with CDG (5pg) and PspA (2pg) twice at 2-weeks intervals. Serum anti-PspA total IgG (FIG. 18A) and BAFF anti-PspA IgA (FIG. 18B) were determined by EFISA at indicated time points after the last immunization. n=3. FIG. 18C. Adult and aged mice were immunized or not as (FIG. 18 A). One month later, mice were infected (i.n) with S. pneumoniae strain D39 (50xFD50,~2.5xl08c.f.u). Animal health was monitored for 2-weeks. n=3. Graphs represent means +/- standard error from three independent experiments. The significance is calculated (unpaired Student’s t test).
[0020] FIG. 19. FIG. 19A. Flow analysis of lung DCs subsets from adult and aged mice.n=3. FIG. 19B-C. Total numbers of lung DC subsets from adult and aged mice. n=3. FIG. 19D.
Adult and aged mice were treated (i.n.) with CDG (5pg) for 20hrs. TNF was determined in lung homogenates by EFISA. n=3. Graphs represent means ± standard error from three independent experiments. The significance is represented by an asterisk (*) where p<0.05 (unpaired Student’s t test). [0021] FIG. 20. 3-month old C57BL/6 (Adult), 18-month old C57BL/6 (Aged) mice were immunized (i.n.) with CDG (5mg)/PspA(2mg) or PBS for l6hrs. Lung moDCs
(CD64+CDl lB+MHCIIhiCDl lC+CD24-) activation were examined by Flow cytometry. n=3.
[0022] FIG. 21. FIG. 21A. A cartoon of TNF-Fc fusion protein and its proposed mode of action on moDCs. FIG. 21B. A cartoon of the CDG/PspA/TNF-Fc vaccine strategy. FIG. 21C. Lung Tfh cells( PD1+CXCR5+) cells were examined in adult and aged mice 14 days after
immunization by Flow cytometry. n=3. FIG. 21D. Mucosal anti-PspAIgA was examined in mice 14 days post immunization. n=3. Graphs represent means ± standard error from three
independent experiments.
[0023] FIG. 22. Aged mice (~l8months old) were immunized (i.n.) with CDG/NP6CGG. Serum anti-NP2 (high-affinity) or anti-NP23 (low-affinity) IgG was examined in mice 1 month post immunization. n=3. Graphs represent means ± standard error from three independent
experiments.
[0024] FIG. 23 shows sequence information on Mouse TNF. There is a two amino acid modification (D221N, A223R) that causes TNF sequence to bind only to TNFR2. One skilled in the art would appreciate that similar strategies could be applied to human TNF sequence.
[0025] FIG. 24. C57BL/6J mice were immunized with Influenza nucleoprotein (NP)TNF- Fc(IgG2A) [sTNF-Fc] or NP/TNFD22iN/A223R-Fc(IgG2A) [tmTNF-Fc] Lung moDCs (FIG. 24A) and TFH cells (FIG. 24B) were analyzed on day 14 by Flow cytometry. n=3. CXCL13 is a chemokine essential for the formation of germinal center.
[0026] FIG 25. A-B. C57BL/6J mice were immunized with Influenza nucleoprotein (NP)/TNF- Fc(IgG2A) [sTNF-Fc] or NP/TNFD22iN/A223R-Fc(IgG2A) [tmTNF-Fc]. Lung were analyzed on day 14 by Flow cytometry. n=3. CCL20 is a T cells recruiting chemokine (FIG. 25A) and Bcl6 (FIG. 25B) is a transcriptional factor important for memory cells development.
DETAILED DESCRIPTION
[0027] The present disclosure is based on the discovery of a new lung dendritic cell subset and its novel in vivo model of action that generates efficacious mucosal vaccine responses. The characterization of this new subset of dendritic cells along with its mode of action have allowed the design of vaccine compositions that are tailored to generate efficacious vaccine responses in lung mucosa. The new lung dendritic cells possess a unique marker profile, TNFR2 pRelB PD- LL CX3CR1+ cDC2 (C3D2).
[0028] According to one embodiment, provided is a vaccine composition that includes a therapeutically effective amount of an antigen; a CDN; soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment. In another embodiment, a TNF conjugated with an moDC targeting moiety is implemented in addition to or in place of TNF and/or the CD64 antibody or antibody fragment. The TNF conjugated with an moDC targeting moiety may include a TNF-Fc (IgG2A) or an anti-CD64 antibody conjugate (antibody-cytokine fusion protein such as anti-CD64-TNF). The CDN may include, but is not limited to, cyclic-di-GMP (CDG), cyclic-di-AMP (CD A), cyclic-di-IMP (CDI), cyclic-AMP-GMP (CDA/G), cyclic-AMP- IMP (CDA/I, and cyclic-GMP-IMP (CDG/I). In a specific embodiment, the CDN is CDG. In a specific embodiment, the CD64 antibody is monoclonal antibody.
[0029] According to another embodiment, provided is a method of eliciting an immune response in a subject that involves administering to the subject a vaccine composition as described herein. The vaccine composition is one that activates monocyte derived dendritic cells (moDCs), to induce lung mucosal antibody responses. [0030] One unique advantage of the compositions described herein is that allow for the generation of effective vaccine responses on mucosal surface. They also allow for the generation of long-termed memory responses that is critical for vaccine-induced protection.
[0031] According to another embodiment, provided is a method of eliciting an immune response that involves administering an antigen; and co-administering a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment. In this embodiment, the compositions are not necessarily administered in the same composition but may be administered separately, either by the same or different mode of administration.
[0032] Another embodiment pertains to a kit that includes an inhalative administration device and a therapeutically effective amount of a composition described herein. The kit may include a vaccine composition or a separate antigen composition and adjuvant composition. The one or more compositions may be loaded into the device or provided in a separate container for use in the device.
[0033] Another embodiment pertains to a method for treating, or preventing, a disease or condition in a subject. The method involves administering an antigen associated with the disease or condition; and co-administering a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment. Examples of the disease or condition include cancer or an infection. [0034] A further embodiment pertains to an adjuvant composition that includes a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment or, optionally, a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti- CD64 antibody conjugated-TNF) in addition to or in place of TNF or CD64 antibody or fragment, or both TNF and CD64 antibody or fragment. The adjuvant composition may optionally include a pharmaceutically acceptable carrier
Definitions
[0035] Unless otherwise defined, all technical and scientific terms used herein are intended to have the same meaning as commonly understood in the art to which this invention pertains and at the time of its filing. Although various methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. However, the skilled should understand that the methods and materials used and described are examples and may not be the only ones suitable for use in the invention. Moreover, it should also be understood that as measurements are subject to inherent variability, any temperature, weight, volume, time interval, pH, salinity, molarity or molality, range, concentration and any other measurements, quantities or numerical expressions given herein are intended to be approximate and not exact or critical figures unless expressly stated to the contrary. Hence, where appropriate to the invention and as understood by those of skill in the art, it is proper to describe the various aspects of the invention using approximate or relative terms and terms of degree commonly employed in patent applications, such as: so dimensioned, about, approximately, substantially, essentially, consisting essentially of, comprising, and effective amount. [0036] Generally, nomenclature used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics, protein, and nucleic acid chemistry and hybridization described herein are those well-known and commonly used in the art. The methods and techniques of the present invention generally are performed according to conventional methods well known in the art and as described in various general and more specific references, unless otherwise indicated. See, e.g., Sambrook et ah, Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989); Ausubel et ah, Current Protocols in Molecular Biology, Greene Publishing Associates (1992, and Supplements to 2002); Harlow and Lan, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1990); Kandel, Schwartz, and Jessell, eds., Principles of Neural Science, 4th ed., McGraw-Hill/ Appleton & Lange: New York, N.Y. (2000). Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art.
[0037] As used herein and in the appended claims, the singular forms“a,”“an,” and“the” include plural references unless the content clearly dictates otherwise.
[0038] The term“about” as used herein means approximately, roughly, around, or in the region of. When the term“about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20% up or down (higher or lower).
[0039] The term“TNF” as used herein refers to tumor necrosis factor. The TNF sequence used is typically that of the species being treated, that is, human TNF when used in inducing an immune response in humans. The term TNF, unless otherwise indicated, includes both soluble TNF and transmembrane TNF.
[0040] The terms“soluble TNF” or“sTNF” are used interchangeably herein to refer to the l7KDa trimeric soluble cytokine form of tumor necrosis factor that is found in blood plasma, i.e., the form that circulates throughout the body and confers TNF with its potent endocrine function.
[0041] The terms“transmembrane TNF” or“tmTNF” or“mTNF” are used interchangeably herein to refer to transmembrane TNF. tmTNF binds to only TNFR2.
[0042] The term“antibody(ies)” or antibody refers to an immunoglobulin molecule which specifically binds with an antigen. Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins. The term“antibody(ies)” includes whole antibodies of any isotype (e.g., IgG, IgA, IgM, IgE, etc.) or antibody fragments. Antibodies or antibody fragments include Fab fragments, a Fab' fragments, a heavy chain antibodies, single-domain antibodies (sdAb), variable domain of a heavy chain antibodies, VHH, Nanobodies, single-chain variable fragments (scFv), a tandem scFvs, a bispecific T-cell engagers (BITEs), a diabodies, single-chain diabodies, DARTs, triple bodies, or a nanoantibodies.
[0043] The term“moDC” targeting moiety, as used herein refers to a component associated with TNF that targets the TNF to moDC cells. In one example, the moDC targeting moiety is an anti- CD64 antibody or antibody fragment. In another example, the moDC targeting moiety is a fragment crystallizable region (Fc region) of an antibody. In a specific example, the Fc is an Fc of a mouse IgG2a isotype or a human IgGl, IgG3 isotype. [0044] A“disease” is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate. In contrast, a“disorder” in an animal is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.
[0045] The terms“patient,”“subject,”“individual,” and the like are used interchangeably herein, and refer to any animal, or cells thereof whether in vitro or in situ, amenable to the methods described herein. In certain non-limiting embodiments, the patient, subject or individual is a human.
[0046] The term“cancer” or“tumor” as used herein means is intended to include any neoplastic growth in a patient, including an initial tumor and any metastases. The cancer can be of the liquid or solid tumor type. Liquid tumors include tumors of hematological origin (hematological cancer), including, e.g., myelomas (e.g., multiple myeloma), leukemias (e.g., Waldenstrom's syndrome, chronic lymphocytic leukemia, other leukemias), and lymphomas (e g, B-cell lymphomas, non-Hodgkins lymphoma). Solid tumors can originate in organs, and include cancers such as lung, breast, prostate, ovary, colon, kidney, and liver.
[0047] The term“cyclic dinucleotide” or“CDN” refer to secondary signaling molecules including cyclic-di-GMP (CDG), cyclic-di-AMP (CDA), cyclic-di-IMP (CDI), cyclic-AMP- GMP (CDA/G), cyclic-AMP-IMP (CDA/I, and cyclic-GMP-IMP (CDG/I). Typically, CDNs trigger interferon in a STING-dependent manner.
[0048] The term“specifically binds,” is used herein in reference to the interaction of an antibody, a polypeptide, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant, binding domain, or epitope) on the chemical species. For example, a ligand recognizes and binds to a specific receptor structure rather than to proteins generally. For example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally.
[0049] The term“therapeutic” as used herein means a treatment and/or prophylaxis. A therapeutic effect is obtained by a diminution, suppression, remission, or eradication of a disease state.
[0050] The term“therapeutically effective amount” refers to the amount of the subject compound that will elicit the biological or clinical response of a tissue, system, or subject that is being sought by the researcher, veterinarian, medical doctor or other clinician. The term “therapeutically effective amount” includes that amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the signs or symptoms of the disorder or disease being treated. The therapeutically effective amount will vary depending on the compound, the disease and its severity and the age, weight, etc., of the subject to be treated.
[0051] To“treat” a disease as the term is used herein, means to reduce the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject.
[0052] In the context of the present invention, the term“vaccine” refers to a substance that induces anti-cancer or anti-pathogen immunity or suppresses the cancer or the pathogen upon later introduction of the cancer or pathogen into the subject. [0053] The term“antigen associated with a disease or condition” refers to an antigen that induces an immune response against abnormal cells (e.g. cancer cells or infected cells) or pathogen cells (e.g. microbes) in a subject.
[0054] The term“vaccine composition” refers to a composition that includes an antigen and one or more adjuvant component(s).
[0055] As used herein, the term“immune response” includes T cell mediated and/or B-cell mediated immune responses. Exemplary immune responses include T cell responses, e.g., cytokine production and cellular cytotoxicity, and B cell responses, e.g., antibody production. In addition, the term immune response includes immune responses that are indirectly affected by T cell activation, e.g., antibody production (humoral responses) and activation of cytokine responsive cells, e.g., macrophages Immune cells involved in the immune response include lymphocytes, such as B cells and T cells (CD4+, CD8+, Thl and Th2 cells); antigen presenting cells (e.g., professional antigen presenting cells such as dendritic cells, macrophages, B lymphocytes, Langerhans cells, and non-professional antigen presenting cells such as
keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes); natural killer cells; myeloid cells, such as macrophages, eosinophils, mast cells, basophils, and granulocytes. In a specific embodiment, an immune response involves activation of moDCs by a newly identified ( TNFR2 pRelB PD-L 1 CX3CRl+ cDC2) described herein.
[0056] The terms“immunogen,”“antigen” or“Ag,” as used herein, is defined as a molecule that induces an immune response. This immune response may involve either antibody production, or the activation of specific immunologically-competent cells, or both. The skilled artisan will understand that any macromolecule, including virtually all proteins or peptides, can serve as an immunogen. Furthermore, immunogens can be derived from recombinant or genomic DNA. A skilled artisan will understand that any DNA, which comprises a nucleotide sequences or a partial nucleotide sequence encoding a protein that elicits an immune response therefore encodes an“immunogen” as that term is used herein. Furthermore, one skilled in the art will understand that an immunogen need not be encoded solely by a full length nucleotide sequence of a gene. It is readily apparent that the present invention includes, but is not limited to, the use of partial nucleotide sequences of more than one gene and that these nucleotide sequences are arranged in various combinations to elicit the desired immune response. Moreover, a skilled artisan will understand that an immunogen need not be encoded by a“gene” at all. It is readily apparent that an immunogen can be generated, synthesized or can be derived from a biological sample.
[0057]‘‘Parenteral” administering or administration of a composition includes, e.g.,
subcutaneous (s.c.), intravenous (i.v.), intramuscular (i.m.), or intradermal (i.d.) injection or infusion techniques.
[0058]“Topical” administering or administration of a composition includes contacting a body surface of the subject, including the skin, the eye, or the mucosa, with the composition.
[0059]‘ Tnhalative” administering or administration refers to delivery to the upper respiratory tract or lungs. In one example inhalative administration includes delivery via inhaling of a composition through the mouth and/or nose. Intranasal administration includes inhalative administration through the nose.
[0060]The term“co-administration” or“co-administering” as used herein refers to the administration of an active agent before, concurrently, or after the administration of another active agent such that the biological effects of either agents overlap. The combination of agents as taught herein can act synergistically to treat or prevent the various diseases, disorders or conditions described herein. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects. Overview
[0061] Adjuvants improve vaccine safety profiles and enhance, and shape, antigen- specific immune responses. Understanding the mode of action of adjuvants is key for the development of rationally designed modern vaccines. Recently, the small molecule cyclic dinucleotides (CDNs) have emerged as a group of promising vaccine adjuvants in preclinical and clinical trials l. CDNs include the bacterial second messengers cyclic di-GMP (CDG), cyclic di-AMP, 3’3’-cyclic GMP-AMP and the mammalian second messenger 2’3’-cyclic GMP-AMP 1. CDG is the founding member and the most studied CDNs 2. As a mucosal adjuvant, CDG does not cause acute toxicity in mice l. Furthermore CDG is a more potent activator of Thl and Th2 immune response than LPS, CpG oligonucleotides (ODN) or aluminum salt based adjuvant in mice 3. Last, CDG adjuvanted vaccines protect mice from H5N 1 influenza 4, Acinetobacter baumannii 5, Staphylococcus aureus 6, Klebsiella pneumoniae 7 and Streptococcus pneumoniae 89. CDG also showed cancer vaccine adjuvant activity in animal models 10, u.
[0062] MPYS, also known as STING and MITA, is a receptor for CDNs 12 and a critical player in sensing cytosolic DNA 13, 14. MPYS 7 mice lose CDNs adjuvant activity 15. Additionally, TNF signaling, not type I IFN signaling, is essential for the adjuvant activity of CDG in vivo 15, 16. Notably, when administered intranasally, CDG not only induced lung production of TNF, IL- 1 b but also the anti-inflammatory cytokine IL-10 17. Consequently, CDG adjuvant does not induce over- inflammatory responses in the lung 17. The precise in vivo mechanism by which TNF mediates CDG adjuvant activity in vivo is unknown. [0063] DCs orchestrate vaccine adjuvant responses 18. They consist of developmentally and functionally distinct subsets that promote either immunogenic or tolerogenic immune responses 19 22. Lungs have three DC subsets: the CDl03+ conventional DC (cCDl), the
CDl lb+CD24+CD64 conventional DC (cDC2) and monocyte-derived CDl lb+CD24 CD64+ DC (moDC) 21. Using MPYSll/flCDl 1 CC C mice, we previously showed that CDG adjuvant activity depends on MPYS expression in DCs 17. The lung DC subset mediating CDG adjuvant activity is unknown.
[0064] As disclosed herein, identified is a new subset of lung DC (TNFR2 pRelB PD-Ll CX3CRl+ cDC2) mediating CDG adjuvant activity in vivo. Surprisingly, intranasally
administered CDG induces the activation, not maturation, of this new TNFR2 cDC2 subset. Activated TNFR2 cDC2 expresses mTNF, which interacts with TNFR2 on moDC to drive moDC maturation promoting CDG adjuvant responses.
Compositions and Modes of Administration
[0065] In various embodiments, the methods of the present invention comprise administering an antigen, or a polynucleotide encoding an immunogen, and co-administering one or more adjuvant components, directly to a subject. In a specific embodiment, the one or more adjuvant components include a CDN, sTNF and/or CD64 antibody or antibody fragment. In another embodiment, adjuvant components include a CDN and a TNF conjugated with an moDC targeting moiety (e.g. a TNF-Fc(IgG2A) or an anti-CD64 antibody conjugated-TNF).
Administration of the antigen and one or more adjuvant components can comprise, for example, inhalative, intramuscular, intravenous, peritoneal, subcutaneous, and intradermal administration.
In a specific example, the antigen and adjuvant component(s) are administered concurrently. In an even more specific example, the antigen and adjuvant component(s) are administered in the same composition.
[0066] Furthermore, the actual dose and schedule can vary depending on whether the antigen and adjuvant component(s) are administered in combination with other pharmaceutical compositions, or depending on inter-individual differences in pharmacokinetics, drug disposition, and metabolism. One skilled in the art can easily make any necessary adjustments in accordance with the exigencies of the particular situation.
[0067] These methods described herein are by no means all-inclusive, and further methods to suit the specific application will be apparent to the ordinary skilled artisan. Moreover, the effective amount of the antigen and adjuvant component(s) can be further approximated through analogy to compounds known to exert the desired effect.
[0068] Administration of the antigen and adjuvant component(s) may be continuous or intermittent, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners. The administration of the antigen and adjuvant component(s) may be essentially continuous over a preselected period of time or may be in a series of spaced doses. Both local and systemic administration is contemplated. The amount administered will vary depending on various factors including, but not limited to, the composition chosen, the particular disease, the weight, the physical condition, and the age of the subject, and whether prevention or treatment is to be achieved. Such factors can be readily determined by the clinician employing animal models or other test systems which are well known to the art.
[0069] When the antigen and adjuvant component(s) are prepared for administration, they may be combined with a pharmaceutically acceptable carrier, diluent or excipient to form a pharmaceutical formulation, or unit dosage form. The total active ingredients in such formulations include from 0.1 to 99.9% by weight of the formulation. A“pharmaceutically acceptable” carrier, diluent, excipient, and/or salt is one that is compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof. The active ingredient for administration may be present as a powder, as granules, as a solution, as a suspension or as an emulsion.
[0070] Pharmaceutical formulations containing the antigen and/or adjuvant component(s) can be prepared by procedures known in the art using well known and readily available ingredients. The antigen and adjuvant component(s) can also be formulated as solutions appropriate for inhalative administration or parenteral administration, for instance by intramuscular, subcutaneous, intradermal or intravenous routes.
[0071] The pharmaceutical formulations of antigen and adjuvant component(s) can also take the form of an aqueous or anhydrous solution or dispersion, or alternatively the form of an emulsion or suspension.
[0072] Thus, for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) the formulations may be presented in unit dose form in ampules, pre-filled syringes, small volume infusion containers or in multi-dose containers with an added
preservative. The active ingredients may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use. [0073] Devices for intranasal administration include spray devices. Suitable nasal spray devices are commercially available from Becton Dickinson, Pfeiffer GMBH and Valois. Spray devices for intranasal use do not depend for their performance on the pressure applied by the user.
Pressure threshold devices are particularly useful since liquid is released from the nozzle only when a threshold pressure is attained. These devices make it easier to achieve a spray with a regular droplet size. Pressure threshold devices suitable for use with the present invention are known in the art and are described for example in WO 91/13281 and EP 311 863 B. Such devices are currently available from Pfeiffer GmbH and are also described in Bommer, R.
Advances in Nasal drug delivery Technology, Pharmaceutical Technology Europe, September 1999, p26-33.
[0074] In one specific example, intranasal devices produce droplets (measured using water as the liquid) in the range 1 to 500 pm. Below 10 pm there is a risk of inhalation, therefore it is desirable to have no more than about 5% of droplets below 10 pm.
[0075] Bi-dose delivery may also be implemented for intranasal delivery. Bi-dose devices contain two subdoses of a single vaccine dose, one sub-dose for administration to each nostril.
[0076] The invention provides in a further aspect a pharmaceutical kit comprising
an intranasal administration device containing a vaccine formulation or comprising
an intranasal administration device and a separate vaccine formulation(s) for use with that device.
[0077] It will be appreciated that the unit content of active ingredient or ingredients contained in an individual dose of each dosage form need not in itself constitute an effective amount for treating the particular indication or disease since the necessary effective amount can be reached by administration of a plurality of dosage units. Moreover, the effective amount may be achieved using less than the dose in the dosage form, either individually, or in a series of administrations.
[0078] The pharmaceutical formulations may include, as optional ingredients, pharmaceutically acceptable carriers, diluents, solubilizing or emulsifying agents, and salts of the type that are well-known in the art. Specific non-limiting examples of the carriers and/or diluents that are useful in the pharmaceutical formulations of the present invention include water and
physiologically acceptable buffered saline solutions, such as phosphate buffered saline solutions pH 7.0-8.0.
Therapeutic Uses
[0079] Certain embodiment pertain to methods of preventing, inhibiting, and treating cancer or infection. In one embodiment, the vaccination methods of the invention induce protective immunity against cancer or a pathogen, by generating an immune response directed against the cancer or the pathogen. In one embodiment, the methods of the invention induce production of pathogen-specific antibodies. In another embodiment, the methods of the invention induce a pathogen-specific cell-mediated immune response. In another embodiment, the methods of the invention induce production of pathogen- specific antibodies and a pathogen-specific cell- mediated immune response. In one embodiment, the methods of the invention induce production of tumor- specific antibodies. In another embodiment, the methods of the invention induce a tumor- specific cell-mediated immune response. In another embodiment, the methods of the invention induce production of tumor- specific antibodies and a tumor- specific cell- mediated immune response.
[0080] Anti-cancer and anti-pathogen immunity can be induced by administering an antigen and adjuvant component(s) described herein, and the induction of anti-cancer or anti-pathogen immunity enables treatment and prevention of a disease associated with cancer or the presence of the pathogen. A decrease in mortality of individuals having a disease, a decrease of the disease markers in the blood, alleviation of detectable symptoms accompanying the disease and such are also included in the therapy or prevention of the disease associated with cancer or infection by the pathogen. Such therapeutic and preventive effects are preferably statistically significant, for example, observed at a significance level of 5% or less, wherein the therapeutic or preventive effect of a vaccine against a disease, is compared to a control without vaccine administration. For example, Student's t-test, the Mann-Whitney U-test or ANOVA may be used for determining statistical significance.
[0081] Certain embodiments provide a method for treating, or preventing, a disease or condition associated cancer or infection by a pathogen. The antigen and adjuvant(s) and methods of administering same may be administered prophylactic ally or therapeutically to subjects suffering from, or at risk of, or susceptible to, developing the disease or condition, including cancer or infection by a pathogen. Such subjects may be identified using standard clinical methods. In the context of the present invention, prophylactic administration occurs prior to the manifestation of overt clinical symptoms of disease, such that a disease or disorder is prevented or alternatively delayed in its progression. In the context of the field of medicine, the term“prevent”
encompasses any activity which reduces the burden of mortality or morbidity from disease. Prevention can occur at primary, secondary and tertiary prevention levels. While primary prevention avoids the development of a disease, secondary and tertiary levels of prevention encompass activities aimed at preventing the progression of a disease and the emergence of symptoms as well as reducing the negative impact of an already established disease by restoring function and reducing disease-related complications. [0082] The induction of anti-cancer or anti-pathogen immunity by composition embodiments described herein, can be further confirmed by observing the induction of antibody production against specific immunogens. For example, when antibodies against one or more antigens, are induced in a subject immunized with the antigen containing composition, and when pathology is suppressed by those antibodies, the one or more antigens, are determined to induce anti-cancer or anti-pathogen immunity.
Antigens
[0083] Those skilled in the art will appreciate that known antigens or developed antigens may be co-administered with adjuvant component(s) described herein. Examples of antigens include but are not limited to the following:
Figure imgf000028_0001
It is noted that the vaccine formulation can include multiple antigens. For example, one formulation can include CDG+PspA+H7/HA to protect infection from both Influenza and pneumococcal infection in one immunization. EXAMPLES
CDG directly targets lung cDCl and cDC2, not moDCs.
[0084] DCs mediate CDG adjuvant activity in vivo 17. There are three lung DC subsets: cDCl (CD 103+ CD24+ CD64-CDl lb-), cDC2 (CD103-CD24+CD64-CD1 lb+) and moDC (CD103- CD24-CD64+ CDl lb+) 21, 23-27 (Fig 10). All DC subsets express the CDG receptor MPYS, which is an ER-resident protein (Fig 1A). CDG has two phosphate groups preventing it from directly passing through the cell membrane. To determine which lung DC subset took up CDG, mice were intranasally administered with FITC-conjugated CDG and FITC+ lung cells were examined after 5hrs. Among lung DCs, cDCl and cDC2 had the highest percentage of CDG- FITC whereas moDCs had no CDG-FITC (Fig 1B). We concluded that intranasal administration of CDG directly targets lung cDCl and cDC2, not moDCs.
Alveolar macrophages (AM) take up CDG but are dispensable for CDG adjuvant activity.
[0085] AM (CD1 lc+MHC ITnt) took up most of the fluorescent CDG in vivo (Fig 11A). Unlike DCs, AM did not increase expression of CD86 following CDG treatment (Fig 11B). To determine whether AM are required for CDG responses in vivo, we used the MPYSfl/flLysMcre mice 17, which deleted MPYS expression in AM (Fig 11C). The activation of lung DCs by CDG was unaltered in the MPYSll/riLysMtrc mice (Fig 11D). Importantly, MPYSll/riLysMtrc mice produced similar anti-pneumococcal surface protein A (PspA) antibody as the WT upon CDG/PspA immunization (Fig 11E). We concluded that CDG targets AM but AM are dispensable for CDG adjuvant activity.
CDG differentially activates lung DC subsets in vivo. [0086] DCs subsets are functionally distinct. CDG increased CD86 and CCR7 expression in lung cDCl and cDC2 (Fig 1C & 1D) 17. In addition, both cDCs migrate, bearing processed antigen, to the lung draining lymph nodes (dLNs) (Fig 12A-B). However, we found that following intranasal CDG administration, cDCl activated RelA (Fig 1E) while cDC2 activated RelB (Fig 1F). Notably, some cDC2 have constitutively activated RelB (Fig 1F). To further demonstrate that CDG differentially activates lung DC subsets, we used the IRF4fl/flCDl lccre and Batf3_/ mice.
[0087] The development of cDCl and cDC2 are controlled by transcriptional factors Batf3 and IRF4 respectively. IRF4fl/flCDl lccre mice lack cDC2 in the lung (Fig 10C) 28-30 while Batf3-/- mice lack cDCl (Fig 10D) 31, 32. We found that CDG-induced lung production of TNF, IL- 1 b and IL-l2p70 were dramatically reduced in IRF4n/llCDl lccre mice but not in the Batf3-/- mice (Fig 12C). Conversely, CDG-induced MCP-l production was largely absent in the Batf3-/- mice but not in the IRFd^CDl lccre mice (Fig 12C). We concluded that intranasally administered CDG differentially activates lung cDCl and cDC2 in vivo.
CDG indirectly activates moDCs
[0088] Although moDCs did not take up CDG, they increased expression of CD86 in response to intranasal administration of CDG (Fig 1C). This was independent of MPYS expression in moDCs as moDCs in MPYS 11/11 LysMtrc mice had normal levels of CD86 expression (Fig 11D). Notably, activated moDCs did not increase CCR7 (Fig 1D) and did not migrate to dLNs (Fig 12B). Last, moDCs activated both RelA and RelB in response to CDG (Fig 1G). We concluded that CDG indirectly activate moDCs and activated lung moDCs were not migratory 25, 33. cDC2 play a central role in mediating CDG adjuvant activity [0089] We next asked which cDC subset mediates CDG adjuvant activity. IRF4fl/flCDl lccre mice lack cDC2 in the lung 28, 29(Fig 10C). The cDCl and moDCs were retained in the
IRF4fl/flCDl lccre mice (Fig 10C). We examined CDG adjuvant activity in the IRF4fl/flCDl lccre mice. Mice were intranasally administered with PspA alone or with CDG. PspA-specific Ab responses were examined in the blood and bronchoalveolar lavage fluid (BALF). Unlike the WT mice, CDG did not induce anti-PspA Abs in BALF and serum (Fig 1H, 12D-E) from immunized IRF4fl/flCDl lccre mice. Ex vivo recall assay in lung cells and splenocytes from immunized IRF4fl/flCDl lccre mice also did not show PspA-specific Thl, Th2 or Thl7 responses (Fig 12F-G).
[0090] To further demonstrate that lung cDC2 mediate the adjuvant activity of CDG, we adoptively transfer (i.n.) WT lung cDC2 into IRF4n/llCDl lccre mice. The recipient
IRF4n/llCDl lccre mice were then immunized with CDG/PspA. We found that adoptive transfer of WT cDC2 generated PspA-specific serum IgG and IgA in the IRF4fl/flCDl lccre mice similar to the WT mice (Fig II). We concluded that lung cDC2 are critical for CDG adjuvant activity.
[0091] In contrast to the IRFd^CDl lccre mice, Batf3_/ mice mounted antigen- specific IgG and IgA responses in a manner comparable to the WT following CDG/PspA immunization (Fig 1H, 12D-E). We concluded that cDC2 play a central role in mediating CDG adjuvant activity. Batf3_/ mice had impaired Thl responses following immunization (Fig 12F-G). Whether the defect is due to the lack of cDCl remains to be determined since T cells also express Batf3.
TNFR2 is essential for CDG adjuvant activity
[0092] TNF signaling is critical for CDG adjuvant activity in vivo 15, 16. TNF signals through two TNFR1 and TNFR2. TNFR1 binds transmembrane TNF (tmTNF) and soluble TNF (sTNF) while TNFR2 only binds to tmTNF 34-37. The lung DC compartment is not altered by the lack of either TNFR1 or TNFR2 (Fig. 13A-B). Consistent with the previous report, we found that CDG induced reduced humoral and cellular immune responses in TNFR1 7 mice (Fig 2A, 13C-F). Surprisingly, CDG completely lost its adjuvant activity in TNFR27 mice (Fig 2A, 13C-F). We concluded that TNFR2 expression is essential for CDG adjuvant activity.
TNFR2 expression on lung cDC2 is required for its maturation
[0093] cDC2 are critical for CDG adjuvant activity (Fig 1). We next examined lung cDC2 maturation in the TNFR2 7 mice. We found that CDG did not enhance CD86 or CCR7 expression in lung cDC2 of TNFR27 mice in vivo (Fig. 2B & 14A). In comparison, CDG induced CD86 and CCR7 expression on cDCl of TNFR2 7 mice in vivo (Fig 14A-B).
Furthermore, blocking TNFR2 by mAb inhibited CDG-induced CD86 expression on cDC2 in vivo (Fig. 14C). Last, adoptively transferred TNFR2-deficient cDC2 into IRF4fl/flCDl lccre recipient mice failed to upregulate CD86 expression in response to intranasal CDG
administration (Fig 2C). Collectively, we concluded that TNFR2 expression on lung cDC2 are required for their maturation in response to CDG in vivo.
TNFR2+ lung cDC2 have constitutively activated RelB
[0094] We next examine TNFR2 expression on lung cDC2. We found that a population of lung cDC2 constitutively express TNFR2 (Fig. 2D). In contrast, TNFR2 expression was not detected on steady-state lung cDCl though CDG dramatically increased TNFR2 expression (Fig 14D). Steady-state lung cDC2 have a pRelB+ population (Fig 1F). Interestingly, all the pRelB+ cDC2 are TNFR2+ (Fig 2E, left panel). CDG further activate RelB in lung cDC2 upon CDG treatment (Fig 1F). We found that all pRelB+ cells in activated cDC2 expressed TNFR2 (Fig 2E, right panel) and all the TNFR2+ cDC2 are pRelB+ (Fig 2F). Last, cDC2 in TNFR27 mice lack pRelB indicating that RelB activation requires TNFR2 signaling (Fig 2G). We concluded that the TNFR2+ and pRelB+ lung cDC2 are the same population. RelB in DCs is required for CDG-induced cDC2 maturation in vivo.
[0095] TNFR2 on lung cDC2 is required CDG-induced cDC2 maturation in vivo (Fig 2C). We reasoned that RelB was required for CDG-induced cDC2 maturation too. We used
RclBn/llCDl lccre mice to ablate RelB in DCs. RclBn/llCD l l ctrc mice have normal DC populations, with all subsets intact (Fig 14E). In the absence of RelB in DCs, cDC2 failed to upregulate CD86 in response to CDG (Fig 2H). cDCl, which do not activate RelB, upregulated CD86 (Fig 14F). We concluded that CDG-induced lung cDC2 maturation depends on the cell- intrinsic signal of TNFR2-RelB .
TNFR2+ cDC2 is required for CDG-induced Thl and TH17 responses but dispensable for the humoral responses
[0096] We next sought to determine if TNFR2 expression on cDC2 was needed for the adjuvant activity of CDG. Unexpectedly, even though adoptively transferred TNFR2-deficient cDC2 failed to upregulate CD86 in response to CDG (Fig 2C), they induced serum anti-PspA IgG and IgA when transferred into IRFd^CDl lccre mice (Fig 3A). In fact, the anti-PspA IgG and IgA in IRF4n/llCDl lccre mice receiving TNFR2-deficient cDC2 and WT cDC2 were comparable (Fig 3A).
[0097] We then examined CDG adjuvant activity in the RelB^CDl lccre mice, which also lack mature cDC2. Upon immunization with PspA and CDG, RclBn/llCD l l ctrc mice produced normal levels of IgG and IgA (Fig 3B). RelBfl/flCDl lccre mice failed to induce Thl and Thl7 responses in the lung (Fig 3C). Collectively, the data suggested that TNFR2+ cDC2 is important for CDG- induced maturation and generation of Thl/Thl7 responses, but is dispensable for humoral responses. TNFR2 defines two functionally distinct subsets of lung cDC2
[0098] cDC2 is a heterogeneous population 21·32·38· 39. We showed that lung TNFR2+pRelB+ cDC2 were mature and required for the Thl/Thl7 responses but not humoral responses while the TNFR2 cDC2 was not mature but mediates CDG-induced antibody response. We assessed whether TNFR2 expression could define functionally distinct lung cDC2 subsets.
[0099] Both cDC2 populations took up CDG in vivo (Fig 4A). When adoptively transferred into MPYS_/ mice, both produced lung TNF (Fig 4B). MPYS_/ mice themselves do not respond to CDG 15. Consistently, adoptively transferred TNFR2+ cDC2 upregulated CD86 and CCR7 in response to CDG whereas TNFR2 cDC2 failed to do so (Fig 4C). Importantly, adoptive transfer of TNFR2+ cDC2 into MPYS 7 (Fig 4D, 15A-B) and IRF4n/llCDl lctrc mice (Fig 4E) failed to rescue antibody production. Consistent with the RelB^CDl lccre results, TNFR2+ cDC2 were able to rescue Thl/Thl7 responses in the IRF4fl/flCDl lccre mice (Fig 4F). TNFR2+ cDC2 also rescued Th2 response, which is different from the RclBn/llCDl lccre mice (Fig 4F & 3C). We speculate that the ability of TNFR2+ cDC2 to mediate CDG-induced Th2 responses maybe redundant in vivo.
[0100] In contrast, adoptive transfer of TNFR2 cDC2 completely restored antibody, but not Th, responses in MPYS 7 (Fig 4D, 15A-B) and IRF4fl/flCDl lccre mice (Fig 4E-F). In fact, levels of anti-PspA IgG and IgA in IRF4n/llCDl lccre mice receiving TNFR2 cDC2 were similar to the WT (Fig 4E). We concluded that lung cDC2 can be divided into two functionally distinct subsets: TNFR2+ cDC2 and TNFR2 cDC2. The TNFR2+ cDC2 are important for CDG-induced cellular immunity, while TNFR2 cDC2 are responsible for CDG-induced humoral responses
(Fig 4G). TNFR2+ and TNFR2 lung cDC2 are derived from pre-cDC2 but not from each other
[0101] We further characterized these steady-state lung cDC2 populations. We found that the TNFR2+ cDC2 are positive for BTLA, PDL-l, arginase 1 (Argl). The TNFR2+ cDC2 also have mixed expression of PD-L2 and CD30lb (Fig 5A). The lung TNFR2 cDC2 expressed CX3CR1 (Fig 5A). Both populations express common cDC2 markers as SIRPa, CD26, IRF4 and Zbtb46 (Fig 5 A, 5B & 16A) 4041. Furthermore, both subsets of cDC2 were absent in Flt3 7 mice (Fig 16B-D). Last, TNFR2+ and TNFR2 cDC2 are negative for cDCl markers IRF8, XCR1 and not affected in Batf3_/ mice confirming their identity as cDC2 (Fig 5B, 16C-D).
[0102] To further establish that the lung TNFR2+ and TNFR2 cDC2 subset are cDC2, we adoptively transfer CD45.1 pre-cDC2 40, 42 into IRF4n/llCDl lctrc mice (Fig 17A-C). CD45.l+ cells were identified in lung five days after transfer and displayed a cDC2 phenotype (Fig 5D and 17C). Importantly, the CD45.l+ pre-cDC2 generated both TNFR2+ and TNFR2 lung cDC2 in the recipient mice (Fig 5D). The CD45.l+ TNFR2+ cDC2 expressed BTLA, PD-L1 and had mixed expression of PD-L2 and CD30lb (Fig 5E), similar to the resident TNFR2+ cDC2. The CD45.l+ TNFR2 cDC2 only expressed CX3CR1 (Fig 5E). We concluded that the lung TNFR2+ and TNFR2 cDC2 arise from the cDC2 lineage and express distinct surface markers.
[0103] We next asked if the TNFR2+ and TNFR2 cDC2 populations represent different developmental/active stage of cDC2. We adoptively transferred CD45.1 lung TNFR2+ and TNFR2 cDC2 into MPYS 7 mice (Fig 17D). The recipient mice were then activated by CDG (i.n.). Consistent with our previous observation, TNFR2 cDC2 did not upregulate TNFR2 while TNFR2+ cDC2 maintained their expression of TNFR2 (Fig 5F). CDG treatment did not affect PD-L1 expression either (Fig 5F). Collectively, we concluded that lung cDC2 consist of two functionally and developmentally distinct subsets, TNFR2+ and TNFR2 cDC2. CDG activates TNFR2-deficient cDC2 in vivo
[0104] Question remains how the immature TNFR2 cDC2 mediates CDG-induced antibody responses in vivo. We suspected that the TNFR2- cDC2, though not mature, may still be activated by CDG in vivo. CDG induces TNF production in vivo that is essential for its adjuvant activity 15 17. CDG-induced TNF production in vivo mainly depending on MPYS-expression in CDl lc+ cells (Fig 6A) 17 , specifically cDC2 as IRF4n/llCDl l ctrc mice had dramatically decreased lung TNF (Fig 12C). We found that CDG induced lung TNF in TNFR27 mice (Fig 6B).
Furthermore, cDC2 produced TNF in TNFR2 7 mice (Fig 6C). Deleting TBK1 in hematopoietic and endothelial lineages (TBK 111/11 VavCrc) dramatically reduced CDG-induced lung TNF production (Fig 6D) suggesting TBK1 is needed for TNF production by CDG. Indeed, TBK1 was activated in the TNFR27 cDC2 (Fig 6E). Last, adoptive transferred TNFR2 cDC2 produced lung TNF in MPYS 7 lung (Fig 4B). We concluded that although TNFR2 cDC2 fail to mature, they were activated by CDG in vivo.
Adoptive transfer of WT monocyte restored CDG adjuvant activity in TNFR2~/~ mice
[0105] We next investigated how the activated, but immature, TNFR2 cDC2 mediate CDG- induced antibody responses. First, we found that moDCs failed to upregulate CD86 in
IRF4n/llCDl lccre mice following CDG treatment (Fig 7A). The CD86 expression in Batf3_/ mice was unaltered (Fig 7B). Second, adoptively transferred TNFR2 7 cDC2 induced CD86 expression on moDCs in IRF4fl/flCDl lccre mice (Fig 7C). Third, CDG induced CD86 expression on moDCs in RclBn/llCDl l ctrc mice (Fig 7D). We concluded that cDC2, especially TNFR2 cDC2 promote CDG-induced moDCs maturation in vivo.
[0106] Adoptively transferred TNFR2 7 cDC2 restored CDG responses in IRF4n/llCD l l ctrc mice (Fig 3A). Yet, TNFR2 7 mice had no CDG responses (Fig 2A). We reasoned that TNFR2 expression on moDCs may be important for CDG responses in vivo. Indeed, we found that CDG induced TNFR2 on moDCs in WT mice (Fig 7E) and moDCs from TNFR27 mice did not upregulate CD86 in response to CDG in vivo (Fig 7F). Last, adoptive transfer of WT monocytes into TNFR27 mice restored CDG-induced IgG and IgA responses (Fig 7G). Notably, adoptive transfer WT cDC2 into TNFR2 7 mice did not restore CDG-induced antibody responses (Fig 7H). We concluded that moDCs expression of TNFR2 is critical for its maturation and subsequent induction of CDG adjuvant response.
CDG induces mTNF expression on TNFR2 cDC2 in vivo
[0107] Our data so far indicates that moDCs are matured by activated TNFR2 cDC2.
Furthermore, moDC maturation requires cell intrinsic TNFR2 expression. Only mTNF can efficiently engage TNFR2 36. Both TNFR2+ and TNFR2 cDC2 produced TNF upon intranasal CDG treatment (Fig 4B). We asked if the lung TNFR2 cDC2 specifically expressed mTNF.
[0108] We intranasally administered CDG to WT mice for 16 hours and detected cell surface mTNF expression using TNFR2-Fc recombinant protein (Fig 8A). We found that the majority of lung mTNF+ DCs were cDC2 (Fig 8B). Remarkably, TNFR2 cDC2 were the main mTNF+ cDC2 cells in vivo while TNFR2+ cDC2 expressed little mTNF (Fig 8C). moDCs, not TNFR2 cDC2, are very efficient in antigen processing in vivo
[0109] We next examined antigen processing in TNFR2 cDC2 in vivo. WT mice were intranasally administered with CDG/DQ-OVA 17. DQ+ cells were examined in lung cDC2 and moDCs (Fig 8D). DQ-OVA is a self-quenched conjugate of OVA exhibiting bright green fluorescence upon proteolytic degradation (DQ-Green). Furthermore, high concentration of digested fragments of DQ-OVA accumulating in organelles form excimers that exhibits bright red fluorescence (DQ-Red). We found that DQ+ moDCs are mostly DQ-Red indicating a high concentration of processed antigens in moDCs. Conversely, DQ+ cDC2 were DQ-Green (Fig 8D). Strikingly, comparing to the TNFR2+ cDC2 subset, very few TNFR2 cDC2 subset were DQ+ (Fig 8D) suggesting that the TNFR2 cDC2 either did not take up antigen or were not efficient at antigen processing. In comparison, all DQ+ moDCs were TNFR2+ cells (Fig 8D) indicating TNFR2+ moDCs were indeed mature DCs. moDCs promote CDG-induced Tfti and GC B cells generation in the lung
[0110] How does non-migratory moDCs (Fig 1D & 12B) 25, 33 promote CDG-induced antibody responses? moDCs were efficient at antigen processing (Fig 8D). We first asked if they presented antigen on cell surface. We intranasally administered C57BL/6 mice with CDG and Ea-OVA, and detected I-Ab/Ea+ cells with the YAE mAb. Indeed, CDG increased YAE+ moDCs in vivo (Fig 9A). Furthermore, the majority of YAE+ moDCs upregulated CD86 (Fig 9B), indicating their potential to activate CD4+ T cells.
[0111] Tfh cells and GC B cells play central roles in promoting humoral responses. We found that 14 days after CDG/PspA immunization (i.n.), lung from the WT mice had
PDl+CXCR5+Bcl6+ Tfh cells and Bcl6+ GC B cells (Fig 9C-9E). In contrast, TNFR2 7 mice were unable to generate lung Tfh or GC B cells (Fig 9C-9D). Importantly, adoptively transfer of WT monocytes into TNFR2 7 mice restored the generation of Tfh and GC B cells in the lung (Fig 9C-9D). We concluded that moDCs promote CDG-induced Tfh and GC B cells generation in the lung.
[0112] moDCs are activated by TNFR2 cDC2. Thus restoring TNFR2 cDC2 in the
IRF4n/llCDl lccre mice should restore Tfh cells. Indeed, we found that adoptive transfer of TNFR2 cDC2, but not TNFR2+ cDC2, into IRF4n/llCDl lccre mice generated Tfh cells (Fig 9E). Together, we propose that CDG activates TNFR2 cDC2 that matures moDCs to generate Tfh and GC B cells promoting CDG-induced antibody responses in vivo (Fig 9F).
Materials and Methods
Mice
[0113] Eight to sixteen-week old mice, both males, and females, were used for all experiments. MPYS ' mice (Trnernl73<tmlCamb>) have been described previously 3839. The following strains were obtained from The Jackson Laboratory: Irf fl (#009380)40, Batf3 (#Ol3755)30, TnfrP (#002818)41, Tnfr2- (#002620)42, Re IB1' (#0287l9)43, CDllCcre (#008068)44. Vavtrt'-TBK 111/11 mice were from Dr. Fitzgerald’s lab. All mice are on a C57BL/6 background. Mice were housed and bred in the Animal Research Facility at Albany Medical College and the University of Florida. All experiments with mice were performed by the regulations and approval of the Institutional Animal Care and Use Committee from Albany Medical College or the University of Florida.
Reagent
[0114] The following reagent was obtained through BEI Resources, NIAID, NIH: Streptococcus pneumoniae Family 1, Clade 2 Pneumococcal Surface Protein A (PspA UAB055) with C- Terminal Histidine Tag, Recombinant from Escherichia coli, NR-33178.
Intranasal CDG Immunization
[0115] Groups of mice (4 per group) were intranasally vaccinated with CDG (5pg, Invivogen, cat# vac-cdg) adjuvanted PspA (2pg, BEI Resources) or PspA alone 17. Mice were immunized twice at 14 days interval. For intranasal vaccination, animals were anesthetized using isoflurane in an E-Z Anesthesia system (Euthanex Corp, Palmer, PA). PspA, with or without CDG was administered in 20m1 saline. Sera were collected 14 days after the last immunization. The PspA- specific Abs were determined by ELISA. Secondary Abs used were anti-mouse IgGl-HRP (Southern Biotech, cat#l070-05), anti-mouse IgG2C-HRP (Southern Biotech, cat#l079-05), and anti-mouse IgA-HRP (Southern Biotech, cat#l040-05). To determine Ag-specific Th response, splenocytes from PspA or CDG + PspA immunized mice were stimulated with 5pg/ml PspA for four days in culture. Thl, Th2, and Thl7 cytokines were measured in the supernatant by ELISA.
Detection of Lung TNF Production
[0116] Mice were intranasally administered 5pg CDG, then sacrificed after 5hrs by C02 asphyxiation 17. Lungs were perfused with cold PBS. The harvested lungs were washed with PBS once, then stored in 0.7ml Tissue protein extraction reagent (T-PER) (Thermo Scientific, cat#785l0) containing protease inhibitors (Roche, cat#l 1836153001) at -80°C. Later, the lung was thawed on ice and homogenized with Minilys® (Precellys, 5,000 RPM for 30sec) using Precellys lysing kit (Precellys, cat# KT03961). Lung homogenates were transferred to a l.5ml tube and spun at l4,000g for 30min at 4°C. The supernatant was collected and analyzed for TNL production by ELISA (eBioscience, cat#88-7324).
In vivo TNFR2 neutralization
[0117] Mice were first given CDG (i.n.). Half an hour later, treated mice were administered (i.n.) with 50pg/50pl anti-TNLR2 Ab (BioLegend, TR75-32.4), or isotype control (BioLegend, HTK888). Lung was harvested l6hr later and analyzed by Llow cytometer.
Isolation of lung cells
[0118] Mice were intranasally administered with or without CDG (5pg, vaccine-grade). After 20hrs, the lungs were lavaged and perfused with ice-cold PBS. Excised lungs were digested in DMEM containing 200pg/ml DNase I (Roche, 10104159001), 25mg/ml Liberase TM (Roche, 05401119001) at 37°C for 3hrs. Red blood cells were then lysed and a single cell suspension was prepared and analyzed by BD™ LSR II and FACScan flow cytometry.
In vivo Ag Uptake and Processing
[0119] Mice were intranasally administered 20pg DQ™-Ovalbumin (DQ-OVA) (Life technologies, D12053) with, or without CDG (5pg, vaccine-grade). After 20hrs, the lungs were lavaged, perfused and harvested. Lung cells were analyzed by BD™ LSR II and FACScan flow cytometry.
Flow Cytometry and cell sorting
[0120] The following Abs from Biolegend were used in the flow cytometry: CD80 (16-10A1), CD86 (GL1), CD11B (Ml/70), CD11C (N418), FceRIa (MAR-1), MHC II (M5/114.15.2), CDl03(2E7), CD24 (ml/69), CD64 (x54-5/7.l), CCR7 (4B 12), TNF (MP6-XT22), TNFR2 (TR75-89), PD-L1 (10F.9G2), BTLA (8F4), PD-L2 (TY25), CD30lb (URA-l), CX3CR1 (SA011F11), CDl72a (P84), CD44 (IM7). The following Abs were from Cell Signaling: p- TBK1 (serl72, d52c2), p-RelB (ser552, d4lb9), p-RelA (ser536, cat#4887S). The following reagents were from Sino Biological: TNFR2 Ab (cat# 50128-R112-A), TNFR2-Fc ( mouse TNFR2 extracellular domain, Met l-Gly 258, was fused with the Fc region of human IgGl at the C-terminus, cat# 50128-M02H), human IgGl Fc (cat# 10702-HNAH). Alexa Fluor®488 mouse anti-human IgGl Fc was from Invitrogen (cat# A- 10631). FITC-CDG (2’-Fluo-AHC-CDG ) was from Biolog (cat# F009). Cell sorting was performed on the BD FACSArialll Flow Cytometer and Cell Sorter. After sorting, dendritic cells were CFSE labeled, according to the protocol from the manufacturer (Invitrogen). Intracellular staining
[0121] The intracellular cytokine staining was performed using the Cytofix/Cytoperm™ kit from BD Biosciences (cat#555028). Briefly, mice were intranasally administered saline or cyclic di- GMP (5pg, vaccine-grade). The lungs were lavaged, perfused, and harvested at 5hr post treatment. Excised lungs were washed in PBS and digested in DMEM containing 200pg/ml DNase I (Roche, 10104159001), 25pg/ml Liberase TM (Roche, 05401119001), and Golgi-plug at 37°C for 6hrs. The single lung cell suspension was fixed in Cytofix/perm buffer (BD
Biosciences) in the dark for 20min at RT. Fixed cells were then washed and kept in Perm/wash buffer at 4°C. Golgi-plug was present during every step before fixation.
Mouse cDC2 and monocyte purification
[0122] Primary mouse cDC2 (cat#l8970A, Stemcell Technologies; cat# 480097, Biolegend) were purified from lungs of naive mice following the protocol according to the manufacturer. Mouse monocytes (cat#l986l, Stemcell Technologies) were purified from the bone marrow of naive mice following the protocol according to the manufacturer.
Adoptive transfer
[0123] Pulmonary TNFR2+ and TNFR2 cDC2 were sorted from the lungs of naive donor mice with a FACSArialll flow cytometer. After sorting, dendritic cells were CFSE labeled, according to the protocol from the manufacturer (Invitrogen). Between 500,000 - 1,000,000 cells were administered intranasally into recipient mice. 24 hours later of transfer, recipient mice were intranasally vaccinated with CDG (5pg, Invivogen, cat# vac-cdg) adjuvanted PspA (2pg, BEI Resources) or PspA alone 17. Recipient mice received two doses of transferred cells and were immunized at 14 days interval. Statistical Analysis
[0124] All data are expressed as means ± SEM. Statistical significance was evaluated using Prism 5.0 software to perform a Student’s t-test (unpaired, two-tailed) for comparison between mean values. tm-TNF-Fc promotes moDCs expression of chemokine CXCL13 and TFH cells induction in the lung
[0125] C57BL/6J mice were immunized with Influenza nucleoprotein (NP)/TNF-Fc(IgG2A) or NP/TNFD22iN/A223R-Fc(IgG2A) as shown in FIG. 24. Fung moDCs (FIG. 24A) and TFH cells (FIG. 24B) were analyzed on day 14 by Flow cytometry. n=3. CXCF13 is a chemokine essential for the formation of germinal center. The data shows that transmembrane TNF-Fc induced CXCF13 expression in moDCs. This effect was greater than that observed by sTNF-Fc treatment. tmTNF-FC also induced TFH cell production from CD4+ Tcells. sTNF-Fc promotes moDCs expression of chemokine CCL20 and transcriptional factor Bcl6
[0126] C57BF/6J mice were immunized with Influenza nucleoprotein (NP)/TNF-Fc(IgG2A) or NP/TNFD22iN/A223R-Fc(IgG2A) as shown in FIG. 25. Fung cells were analyzed on day 14 by Flow cytometry. n=3. CCF20 is a T cells recruiting chemokine and Bcl6 is a transcriptional factor important for memory cells development. The data in FIG. 25A shows that soluble TNF- Fc induced CCF20 expression in moDCs, and that this effect was stronger than tmTNF-Fc treatment. In addition, FIG. 25B shows that sTNF-Fc induced Bcl6 expression in moDCs and that this effect was stronger than tmTNF-Fc treatment.
[0127] Those skilled in the art will appreciate that conjugations with TNF can be produced using conventional techniques and methods. Examples of IgG2a include, but are not limited to, accession nos AAN76044, AAN76043.1, AAN76042.1, AXN93670.2, and AXN93670.2 and
P01859-1 (UniParc). Examples of IgG3 include CAA67886.1, CAC10266.1, CAC10265.1, and CAC10264.1 Also see Human IgGl, IgG3, and IgG3 Hinge-Truncated Mutants Show Different Protection Capabilities against Meningococci Depending on the Target Antigen and Epitope Specificity S. Giuntini, D. M. Granoff, P. T. Beemink, O. Ihle, D. Bratlie, T. E. Michaelsen
Clinical and Vaccine Immunology Aug 2016, 23 (8) 698-706 and Dillon et al. J Biol Chem. 2008 Jun 6;283(23): 16206-15 for information on IgGl and IgG2 subclass. Also, see UniProt, P01857 for IgGl. Vidarsson et al. Front Immunol , 2014 5:520. Web materials for the different Fc regions can be found at invivogen.com/review-antibody-generation. All of the above are incorporated by reference.
References:
All publications mentioned herein are incorporated by reference in their entirety.
1. Libanova R, Becker PD, Guzman CA. Cyclic di-nucleotides: new era for small molecules as adjuvants. Microb Biotechnol 2012; 5(2): 168-176.
2. Ebensen T, Schulze K, Riese P, Morr M, Guzman CA. The bacterial second messenger cdiGMP exhibits promising activity as a mucosal adjuvant. Clin Vaccine Immunol 2007; 14(8): 952-958.
3. Gray PM, Forrest G, Wisniewski T, Porter G, Freed DC, DeMartino JA et al. Evidence for cyclic diguanylate as a vaccine adjuvant with novel immuno stimulatory activities. Cell Immunol 2012; 278(1-2): 113-119.
4. Madhun AS, Haaheim LR, Nostbakken JK, Ebensen T, Chichester J, Yusibov V et al. Intranasal c-di-GMP-adjuvanted plant-derived H5 influenza vaccine induces multifunctional Thl CD4+ cells and strong mucosal and systemic antibody responses in mice. Vaccine 2011; 29(31): 4973-4982.
5. Zhao L, KuoLee R, Harris G, Tram K, Yan H, Chen W. c-di-GMP protects against intranasal Acinetobacter baumannii infection in mice by chemokine induction and enhanced neutrophil recruitment. Int Immunopharmacol 2010; 11(9): 1378-1383. 6. Hu DL, Narita K, Hyodo M, Hayakawa Y, Nakane A, Karaolis DK. c-di-GMP as a vaccine adjuvant enhances protection against systemic methicillin-resistant Staphylococcus aureus (MRS A) infection. Vaccine 2009; 27(35): 4867-4873.
7. Karaolis DK, Newstead MW, Zeng X, Hyodo M, Hayakawa Y, Bhan U et al. Cyclic di- GMP stimulates protective innate immunity in bacterial pneumonia. Infect Immun 2007; 75(10): 4942-4950.
8. Ogunniyi AD, Paton JC, Kirby AC, McCullers JA, Cook J, Hyodo M et al. c-di-GMP is an effective immunomodulator and vaccine adjuvant against pneumococcal infection. Vaccine 2008; 26(36): 4676-4685.
9. Yan H, KuoLee R, Tram K, Qiu H, Zhang J, Patel GB et al. 3',5'-Cyclic diguanylic acid elicits mucosal immunity against bacterial infection. Biochem Biophys Res Commun 2009;
387(3): 581-584.
10. Smith TT, Moffett HF, Stephan SB, Opel CF, Dumigan AG, Jiang X et al. Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors. J Clin Invest 2017; 127(6): 2176-2191.
11. Wang Z, Celis E. STING activator c-di-GMP enhances the anti-tumor effects of peptide vaccines in melanoma-bearing mice. Cancer Immunol Immunother 2015; 64(8): 1057-1066.
12. Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature 2011; 478(7370): 515-518.
13. Dempsey A, Bowie AG. Innate immune recognition of DNA: A recent history. Virology 2015; 479-480: 146-152.
14. Wu J, Chen ZJ. Innate immune sensing and signaling of cytosolic nucleic acids. Annu Rev Immunol 2014; 32: 461-488.
15. Blaauboer SM, Gabrielle VD, Jin L. MPYS/STING-mediated TNF-alpha, not type I IFN, is essential for the mucosal adjuvant activity of (3'-5')-cyclic-di-guanosine-monophosphate in vivo. J Immunol 2014; 192(1): 492-502.
16. Hanson MC, Crespo MP, Abraham W, Moynihan KD, Szeto GL, Chen SH et al.
Nanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvants. J Clin Invest 2015; 125(6): 2532-2546. 17. Blaauboer SM, Mansouri S, Tucker HR, Wang HL, Gabrielle VD, Jin L. The mucosal adjuvant cyclic di-GMP enhances antigen uptake and selectively activates pinocytosis-efficient cells in vivo. Elife 2015; 4.
18. Steinman RM. Decisions about dendritic cells: past, present, and future. Annu Rev Immunol 2012; 30: 1-22.
19. Baratin M, Foray C, Demaria O, Habbeddine M, Pollet E, Maurizio J el al. Homeostatic NF-kappaB Signaling in Steady-State Migratory Dendritic Cells Regulates Immune Homeostasis and Tolerance. Immunity 2015; 42(4): 627-639.
20. Crowley M, Inaba K, Witmer-Pack M, Steinman RM. The cell surface of mouse dendritic cells: FACS analyses of dendritic cells from different tissues including thymus. Cell Immunol 1989; 118(1): 108-125.
21. Mildner A, Jung S. Development and function of dendritic cell subsets. Immunity 2014; 40(5): 642-656.
22. Vremec D, Zorbas M, Scollay R, Saunders DJ, Ardavin CF, Wu L et al. The surface phenotype of dendritic cells purified from mouse thymus and spleen: investigation of the CD8 expression by a subpopulation of dendritic cells. J Exp Med 1992; 176(1): 47-58.
23. Langlet C, Tamoutounour S, Henri S, Luche H, Ardouin L, Gregoire C et al. CD64 expression distinguishes monocyte-derived and conventional dendritic cells and reveals their distinct role during intramuscular immunization. J Immunol 2012; 188(4): 1751-1760.
24. Tamoutounour S, Henri S, Lelouard H, de Bovis B, de Haar C, van der Woude CJ et al. CD64 distinguishes macrophages from dendritic cells in the gut and reveals the Thl -inducing role of mesenteric lymph node macrophages during colitis. Eur J Immunol 2012; 42(12): 3150- 3166.
25. Plantinga M, Guilliams M, Vanheerswynghels M, Deswarte K, Branco-Madeira F, Toussaint W et al. Conventional and monocyte-derived CDl lb(+) dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 2013; 38(2): 322-335.
26. Holt PG, Schon-Hegrad MA, McMenamin PG. Dendritic cells in the respiratory tract. Int Rev Immunol 1990; 6(2-3): 139-149. 27. Schlitzer A, McGovern N, Teo P, Zelante T, Atarashi K, Low D el al. IRF4 transcription factor-dependent CDl lb-i- dendritic cells in human and mouse control mucosal IL-17 cytokine responses. Immunity 2013; 38(5): 970-983.
28. Suzuki S, Honma K, Matsuyama T, Suzuki K, Toriyama K, Akitoyo I et al. Critical roles of interferon regulatory factor 4 in CD1 lbhighCD8alpha- dendritic cell development. Proc Natl Acad Sci U SA 2004; 101(24): 8981-8986.
29. Vander Lugt B, Khan A A, Hackney JA, Agrawal S, Lesch J, Zhou M et al.
Transcriptional programming of dendritic cells for enhanced MHC class II antigen presentation. Nat Immunol 201· 4; 15(2): 161-167.
30. Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, Kohyama M et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 2008; 322(5904): 1097-1100.
31. Murphy KM. Transcriptional control of dendritic cell development. Adv Immunol 2013; 120: 239-267.
32. Wallach D, Varfolomeev EE, Malinin NL, Goltsev YV, Kovalenko AV, Boldin MP. Tumor necrosis factor receptor and Fas signaling mechanisms. Annu Rev Immunol 1999; 17: 331-367.
33. Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ 2003; 10(1): 45-65.
34. Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B et al. The
transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 1995; 83(5): 793-802.
35. Grell M, Wajant H, Zimmermann G, Scheurich P. The type 1 receptor (CDl20a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci U SA 1998; 95(2): 570-575.
36. Guilliams M, Dutertre CA, Scott CL, McGovern N, Sichien D, Chakarov S et al.
Unsupervised High-Dimensional Analysis Aligns Dendritic Cells across Tissues and Species. Immunity 2016; 45(3): 669-684. 37. Sichien D, Scott CL, Martens L, Vanderkerken M, Van Gassen S, Plantinga M et al.
IRF8 Transcription Factor Controls Survival and Function of Terminally Differentiated
Conventional and Plasmacytoid Dendritic Cells, Respectively. Immunity 2016; 45(3): 626-640.
38. Jin L, Getahun A, Knowles HM, Mogan J, Akerlund LJ, Packard TA et al.
STING/MPYS Mediates Host Defense against Listeria monocytogenes Infection by Regulating Ly6Chi Monocyte Migration. J Immunol 2013; 190(6): 2835-2843.
39. Jin L, Hill KK, Filak H, Mogan J, Knowles H, Zhang B et al. MPYS is required for IFN response factor 3 activation and type I IFN production in the response of cultured phagocytes to bacterial second messengers cyclic-di-AMP and cyclic-di-GMP. J Immunol 2011; 187(5): 2595- 2601.
40. Klein U, Casola S, Cattoretti G, Shen Q, Lia M, Mo T et al. Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol 2006; 7(7): 773-782.
41. Pfeffer K, Matsuyama T, Kundig TM, Wakeham A, Kishihara K, Shahinian A et al. Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection. Cell 1993; 73(3): 457-467.
42. Erickson SL, de Sauvage FJ, Kikly K, Carver-Moore K, Pitts-Meek S, Gillett N et al. Decreased sensitivity to tumour-necrosis factor but normal T-cell development in TNF receptor- 2-deficient mice. Nature 1994; 372(6506): 560-563.
43. De Silva NS, Silva K, Anderson MM, Bhagat G, Klein U. Impairment of Mature B Cell Maintenance upon Combined Deletion of the Alternative NF-kappaB Transcription Factors RELB and NF-kappaB 2 in B Cells. J Immunol 2016; 196(6): 2591-2601.
44. Caton ML, Smith-Raska MR, Reizis B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J Exp Med 2007; 204(7): 1653-1664.
[0128] Although the present invention has been described in considerable detail with reference to certain preferred versions thereof, other versions are possible. Therefore, the spirit and scope of the appended claims should not be limited to the description of the preferred versions contained herein. [0129] The reader’ s attention is directed to all papers and documents which are filed concurrently with this specification and which are open to public inspection with this specification, and the contents of all such papers and documents are incorporated herein by reference.
[0130] All the features disclosed in this specification (including any accompanying claims, abstract, and drawings) may be replaced by alternative features serving the same, equivalent or similar purpose, unless expressly stated otherwise. Thus, unless expressly stated otherwise, each feature disclosed is one example only of a generic series of equivalent or similar features.
[0131] Any element in a claim that does not explicitly state“means for” performing a specified function, or“step for” performing a specific function, is not to be interpreted as a“means” or “step” clause as specified in 35 U.S.C § 112, sixth paragraph. In particular, the use of‘step of in the claims herein is not intended to invoke the provisions of 35 U.S.C §112, sixth paragraph.

Claims

CLAIMS What is claimed is:
1. A vaccine composition comprising a therapeutically effective amount of an antigen; a cyclic dinucleotide; soluble tumor necrosis factor (TNF); or a CD64 antibody or antibody fragment; and wherein, optionally, the vaccine composition comprises a TNF conjugated with a moDC targeting moiety in addition to or in place of TNF or CD64 antibody or antibody fragment, or both TNF and CD64 antibody or antibody fragment.
2. The vaccine composition of claim 1, wherein the cyclic dinucleotide comprises cyclic-di- GMP (CDG), cyclic-di-AMP (CD A), cyclic-di-IMP (CDI), cyclic-AMP-GMP (CDA/G), cyclic- AMP-IMP (CD A/I, and cyclic-GMP-IMP (CDG/I).
3. The vaccine composition of claim 1, wherein the CD64 antibody or antibody fragment is a monoclonal antibody.
4. The vaccine composition of claim 1, wherein the antigen comprisesH7HA, H7N7, PspA, influenza nucleoprotein (NP), HIVenvVlV2 or HIV-lpr55Gag.
5. The vaccine composition of claim 1, wherein the vaccine composition further comprises a pharmaceutically acceptable carrier.
6. The vaccine composition of claim 1, wherein the TNF conjugated with a moDC targeting moiety comprises an anti-CD64 antibody conjugated-TNF and/or Fc conjugated TNF.
7. A method of eliciting an immune response in a subject comprising administering to the subject a vaccine composition of any of claims 1-6.
8. The method of claim 7, wherein the immune response comprises activation of monocyte derived dendritic cells (moDCs).
9. The method of claim 8, wherein the moDCs comprise (CD64+CD24 CDl lB+CDl lC+MHC class IIhl) DCs.
10. The method of any of claims 7-9, wherein the vaccine protection is persistent.
11. The method of any of claims 7-9 wherein the subject is human.
12. The method of claim 11, wherein the subject is 40 or older, 45 or older, 50 or older, 55 or older, or 60 or older.
13. A method of eliciting an immune response comprising administering an antigen; and co-administering a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment; and wherein, optionally, a TNF conjugated with a moDC targeting moiety in addition to or in place of TNF or CD64 antibody or antibody fragment, or both TNF and CD64 antibody or antibody fragment.
14. A kit comprising an inhalative administration device and the vaccine composition of claim
1.
15. The kit of claim 11, wherein the vaccine composition is provided in container separate to the inhalative administration device.
16. The kit of claim 11, wherein the vaccine composition is disposed within the inhalative administration device.
17. A method for treating, or preventing, a disease or condition in a subject, the method comprising administering an antigen associated with the disease or condition; and co-administering a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment; and, wherein, optionally a TNF conjugated with a moDC targeting moiety in addition to or in place of TNF or CD64 antibody or antibody fragment, or both TNF and CD64 antibody or antibody fragment.
18. The method of claim 17, wherein the disease or condition comprises cancer.
19. The method of claim 17, wherein the disease or condition comprises an infection.
20. The method of claim 19, wherein the infection by a virus, bacteria or fungi.
21. An adjuvant composition comprising a cyclic dinucleotide, soluble tumor necrosis factor (TNF); and a CD64 antibody or antibody fragment; or optionally, a TNF conjugated with a moDC targeting moiety in addition to or in place of TNF or CD64 antibody or antibody fragment, or both TNF and CD64 antibody or antibody fragment.
22. The adjuvant composition of claim 21, further comprising a pharmaceutically acceptable carrier.
23. An adjuvant composition comprising TNF conjugated with an moDC targeting moiety.
24. The adjuvant composition of claim 23, wherein the moDC targeting moiety comprises Fc of an IgG2a antibody isotype.
25. The adjuvant composition of claims 23 or 24, wherein TNF is sTNF or tmTNF.
26. The adjuvant composition of claim 25, wherein tmTNF binds only TNFR2.
27. A vaccine composition comprising a therapeutically effective amount of an antigen; a cyclic dinucleotide; and a TNF conjugated with a moDC targeting moiety.
28. The vaccine composition of claim 27, wherein the moDC targeting moiety comprises Fc from an IgG2A isotype.
29. The vaccine composition of claims 27 or 28, wherein TNF comprises sTNF or tmTNF.
30. The vaccine composition of claims 27 or 28, wherein TNF conjugated with a moDC targeting moiety comprises sTNF-Fc and tmTNF-Fc.
31. A method of eliciting an immune response in a subject comprising administering to the subject a vaccine composition of any of claims 27-30.
32. The method of claim 31, wherein the immune response comprises activation of monocyte derived dendritic cells (moDCs).
33. The method of claim 32, wherein the moDCs comprise (CD64+CD24 CDl lB+CDl lC+ MHC class IIhi) DCs.
34. The method of any of claims 31-33, wherein the vaccine protection is persistent.
35. The method of any of claims 31-30 wherein the subject is human.
36. The method of claim 35, wherein the subject is 40 or older, 45 or older, 50 or older, 55 or older, or 60 or older.
PCT/US2019/053548 2018-09-27 2019-09-27 TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE WO2020069375A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/280,986 US20210353748A1 (en) 2018-09-27 2019-09-27 TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE
EP19865436.0A EP3856199A4 (en) 2018-09-27 2019-09-27 Targeting modc to enhance vaccine efficacy on mucosal surface

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862737154P 2018-09-27 2018-09-27
US62/737,154 2018-09-27

Publications (1)

Publication Number Publication Date
WO2020069375A1 true WO2020069375A1 (en) 2020-04-02

Family

ID=69953063

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/053548 WO2020069375A1 (en) 2018-09-27 2019-09-27 TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE

Country Status (3)

Country Link
US (1) US20210353748A1 (en)
EP (1) EP3856199A4 (en)
WO (1) WO2020069375A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160175427A1 (en) * 2013-08-06 2016-06-23 David Weiner Influenza Nucleic Acid Molecules And Vaccines Made Therefrom
US20170095548A1 (en) * 2010-03-17 2017-04-06 Socpra - Sciences Et Génie, S.E.C. Bacterial vaccine components and uses thereof
WO2018053508A1 (en) * 2016-09-19 2018-03-22 The University Of North Carolina At Chapel Hill Methods and compositions for inducing an immune response

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1765870A2 (en) * 2004-06-03 2007-03-28 Medarex, Inc. Human monoclonal antibodies to fc gamma receptor i (cd64)

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170095548A1 (en) * 2010-03-17 2017-04-06 Socpra - Sciences Et Génie, S.E.C. Bacterial vaccine components and uses thereof
US20160175427A1 (en) * 2013-08-06 2016-06-23 David Weiner Influenza Nucleic Acid Molecules And Vaccines Made Therefrom
WO2018053508A1 (en) * 2016-09-19 2018-03-22 The University Of North Carolina At Chapel Hill Methods and compositions for inducing an immune response

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LANGLET ET AL.: "CD 64 Expression Distinguishes Monocyte-Derived and Conventional Dendritic Cells and Reveals Their Distinct Role During Intramuscular Immunization", J IMMUNOL, vol. 188, 2012, pages 1751 - 1760, XP055698261 *
See also references of EP3856199A4 *

Also Published As

Publication number Publication date
US20210353748A1 (en) 2021-11-18
EP3856199A1 (en) 2021-08-04
EP3856199A4 (en) 2022-09-07

Similar Documents

Publication Publication Date Title
US20200087370A1 (en) Cd40 agonist antibody /type 1 interferon synergistic adjuvant combination, conjugates containing and use thereof as a therapeutic to enhance cellular immunity
US20090004194A1 (en) Tlr agonist (flagellin)/cd40 agonist/antigen protein and dna conjugates and use thereof for inducing synergistic enhancement in immunity
JP2021191765A (en) Use of mva or mvaδe3l as immunotherapeutic agents against solid tumors
EP2170931A1 (en) Use of tlr agonists and/or type 1 interferons to alleviate toxicity of tnf-r agonist therapeutic regimens
JP5331100B2 (en) An adjuvant combination comprising an NKT activator, a CD40 agonist, and optionally an antigen, and its use to induce a synergistic enhancement in cellular immunity
JP5745577B2 (en) CD40 agonist antibody / type I interferon synergistic adjuvant conjugates, conjugates comprising the same, and their use as therapeutics to enhance cellular immunity
US20230080443A1 (en) Immunogenic compounds for cancer therapy
JP6273290B2 (en) Allogeneic autophagosome-enhancing composition for treatment of disease
US20210353748A1 (en) TARGETING moDC TO ENHANCE VACCINE EFFICACY ON MUCOSAL SURFACE
US20210401955A1 (en) CD4 T cells provide antibody access to immunoprivileged tissue
US20220008512A1 (en) Anti-cancer monotherapy using sa-4-1bbl
Cheng Anti-tumor responses of endosomal toll-like receptor-based immunotherapy in head and neck squamous cell carcinoma
AU2020398223A1 (en) A T cell-based immunotherapy for central nervous system viral infections and tumors

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: 19865436

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019865436

Country of ref document: EP

Effective date: 20210428