US20200282049A1 - Compositions and methods for activating "stimulator of interferon gene"-dependent signalling - Google Patents

Compositions and methods for activating "stimulator of interferon gene"-dependent signalling Download PDF

Info

Publication number
US20200282049A1
US20200282049A1 US16/877,218 US202016877218A US2020282049A1 US 20200282049 A1 US20200282049 A1 US 20200282049A1 US 202016877218 A US202016877218 A US 202016877218A US 2020282049 A1 US2020282049 A1 US 2020282049A1
Authority
US
United States
Prior art keywords
cyclic
cancer
cells
tumor
cell
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US16/877,218
Inventor
Thomas W. Dubensky, Jr.
David B. Kanne
Meredith Lai Ling Leong
Edward Emile Lemmens
Laura H. Glickman
Russell E. Vance
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Chinook Therapeutics Inc
Original Assignee
University of California
Aduro Biotech 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51934006&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20200282049(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by University of California, Aduro Biotech Inc filed Critical University of California
Priority to US16/877,218 priority Critical patent/US20200282049A1/en
Assigned to Aduro Biotech, Inc. reassignment Aduro Biotech, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DUBENSKY, THOMAS W., JR., GLICKMAN, LAURA H., KANNE, DAVID B., LEMMENS, EDWARD EMILE, LEONG, MEREDITH LAI LING
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VANCE, RUSSELL E.
Publication of US20200282049A1 publication Critical patent/US20200282049A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • 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
    • 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

Definitions

  • the human immune system may generally be divided into two arms, referred to as “innate immunity” and “adaptive immunity.”
  • the innate arm of the immune system is predominantly responsible for an initial inflammatory response via a number of soluble factors, including the complement system and the chemokine/cytokine system; and a number of specialized cell types including mast cells, macrophages, dendritic cells (DCs), and natural killer cells.
  • the adaptive immune arm involves a delayed and a longer lasting antibody response together with CD8+ and CD4+ T cell responses that play a critical role in immunological memory against an antigen.
  • a third arm of the immune system may be identified as involving ⁇ T cells and T cells with limited T cell receptor repertoires such as NKT cells and MAIT cells.
  • antigen presenting cells For an effective immune response to an antigen, antigen presenting cells (APCs) must process and display the antigen in a proper MHC context to a T cell, which then will result in either T cell stimulation of cytotoxic and helper T cells. Following antigen presentation successful interaction of co-stimulatory molecules on both APCs and T cells must occur or activation will be aborted.
  • GM-CSF and IL-12 serve as effective pro-inflammatory molecules in many tumor models.
  • GM-CSF induces myeloid precursor cells to proliferate and differentiate into dendritic cells (DCs) although additional signals are necessary to activate their maturation to effective antigen-presenting cells necessary for activation of T cells.
  • Barriers to effective immune therapies include tolerance to the targeted antigen that can limit induction of cytotoxic CD8 T cells of appropriate magnitude and function, poor trafficking of the generated T cells to sites of malignant cells, and poor persistence of the induced T cell response.
  • DCs that phagocytose tumor-cell debris process the material for major histocompatibility complex (MHC) presentation, upregulate expression of costimulatory molecules, and migrate to regional lymph nodes to stimulate tumor-specific lymphocytes.
  • MHC major histocompatibility complex
  • This pathway results in the proliferation and activation of CD4+ and CD8+ T cells that react to tumor-associated antigens. Indeed, such cells can be detected frequently in the blood, lymphoid tissues, and malignant lesions of patients.
  • STING is an adaptor protein in the cytoplasm of host mammalian cells which activates the TANK binding kinase (TBK1)-IRF3 signaling axis, resulting in the induction of IFN- ⁇ and other IRF-3 dependent gene products that strongly activate innate immunity. It is now recognized that STING is a component of the host cytosolic surveillance pathway, that senses infection with intracellular pathogens and in response induces the production of IFN- ⁇ , leading to the development of an adaptive protective pathogen-specific immune response consisting of both antigen-specific CD4 and CD8 T cells as well as pathogen-specific antibodies. Examples of cyclic purine dinucleotides are described in some detail in, e.g., U.S. Pat. Nos. 7,709,458 and 7,592,326; WO2007/054279; and Yan et al., Bioorg. Med. Chem Lett. 18: 5631 (2008), each of which is hereby incorporated by reference.
  • compositions comprising:
  • CDNs cyclic purine dinucleotides
  • STING INterferon Genes
  • Preferred cyclic purine dinucleotides include, but are not limited to, one or more of c-di-AMP, c-di-GMP, c-di-IMP, c-AMP-GMP, c-AMP-IMP, c-GMP-IMP, and analogs thereof. This list is not meant to be limiting.
  • c-di-GMP having 2′-5′ linkages refers to the molecule indicated above where each of R1 and R2 are guanine, and each phosphate linkage is 2′-to-5′.
  • this general structure is further modified to introduce substituents which confer the ability to bind to STING and induce a STING-dependent signaling cascade (and most preferably induce a human STING-dependent signaling cascade), and thereby induce STING-dependent type I interferon production and other co-regulated genes.
  • substituents which confer the ability to bind to STING and induce a STING-dependent signaling cascade (and most preferably induce a human STING-dependent signaling cascade), and thereby induce STING-dependent type I interferon production and other co-regulated genes.
  • each X is independently O or S
  • R3 and R4 are each independently H or an optionally substituted straight chain alkyl of from 1 to 18 carbons and from 0 to 3 heteroatoms, an optionally substituted alkenyl of from 1-9 carbons, an optionally substituted alkynyl of from 1-9 carbons, or an optionally substituted aryl, wherein substitution(s), when present, may be independently selected from the group consisting of C 1-6 alkyl straight or branched chain, benzyl, halogen, trihalomethyl, C 1-6 alkoxy, —NO 2 , —NH 2 , —OH, ⁇ O, —COOR′ where R′ is H or lower alkyl, —CH 2 OH, and —CONH 2 , wherein R3 and R4 are not both H.
  • one or both of R3 and R4 independently comprise a prodrug leaving group removed by cellular esterases.
  • one or both of R3 and R4 are a C6 to C18 fatty acid ester.
  • one or both of R3 and R4 are selected from the group consisting of myristoyl, pentanoyl, hexanoyl, heptanoyl, etc.
  • each X is S. In preferred embodiments when each X is S, the compositions comprise one or more substantially pure Sp,Sp, Rp,Rp, Sp,Rp, or Rp,Sp stereoisomers.
  • each of R1 and R2 are independently selected from the group consisting of adenine, guanine, inosine, and xanthine or analogs thereof.
  • each of R1 and R2 are independently adenine or guanine.
  • a cyclic purine dinucleotide composition according to the present invention can induce STING-dependent type I interferon production at least 2-fold, and more preferably 5-fold or 10-fold, or more, as compared to c-di-GMP having 3′-5′ linkages.
  • the STING is human STING.
  • a substantially pure cyclic purine dinucleotide composition according to the present invention activates human STING but the corresponding cyclic purine dinucleotidehaving only bis-(3′,5′) linkages does not.
  • the present compositions may be used as adjuvants in a therapeutic or prophylactic strategy employing vaccine(s).
  • the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof may be used together with one or more vaccines selected to stimulate an immune response to one or more predetermined antigens.
  • the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof may be provided together with, or in addition to, such vaccines.
  • Such vaccine(s) can comprise inactivated or attenuated bacteria or viruses comprising the antigens of interest, purified antigens, live viral or bacterial delivery vectors recombinantly engineered to express and/or secrete the antigens, antigen presenting cell (APC) vectors comprising cells that are loaded with the antigens or transfected with a composition comprising a nucleic acid encoding the antigens, liposomal antigen delivery vehicles, or naked nucleic acid vectors encoding the antigens.
  • APC antigen presenting cell
  • Such vaccine(s) may also comprise an inactivated tumor cell that expresses and secretes one or more of GM-CSF, CCL20, CCL3, IL-12p70, FLT-3 ligand.
  • the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, may be administered to individuals in need thereof by a variety of parenteral and nonparenteral routes in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles.
  • Preferred routes are parenteral, and include but, are not limited to, one or more of subcutaneous, intravenous, intramuscular, intraarterial, intradermal, intrathecal and epidural administrations. Intra-tumor routes are also preferred. Particularly preferred is administration by subcutaneous administration.
  • Preferred pharmaceutical compositions are formulated as aqueous or oil-in-water emulsions.
  • compositions of the present invention may comprise, or be administered together with, one or more additional pharmaceutically active components such as adjuvants, lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers, CTLA-4 and PD-1 pathway Antagonists, PD-1 pathway blocking agents, inactivated bacteria which induce innate immunity (e.g., inactivated or attenuated Listeria monocytogenes ), compositions which mediate innate immune activation via Toll-like Receptors (TLRs), (NOD)-like receptors (NLRs), Retinoic acid inducible gene-based (RIG)-I-like receptors (RLRs), C-type lectin receptors (CLRs), pathogen-associated molecular patterns (“PAMPs”), chemo
  • the present invention relates to methods of inducing, stimulating, or adjuvanting an immune response in an individual. These methods comprise administering the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, to the individual. Preferred routes of administration are parenteral. As noted above, particularly preferred are thiophosphate derivatives of such cyclic purine dinucleotides.
  • the method is a method of cancer treatment.
  • the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof may be provided alone, or together with or in addition to one or more cancer vaccine compositions that are known in the art.
  • the patient receiving such treatment may be suffering from a cancer selected from the group consisting of a colorectal cancer cell, an aero-digestive squamous cancer, a lung cancer, a brain cancer, a liver cancer, a stomach cancer, a sarcoma, a leukemia, a lymphoma, a multiple myeloma, an ovarian cancer, a uterine cancer, a breast cancer, a melanoma, a prostate cancer, a pancreatic carcinoma, and a renal carcinoma.
  • the method is a method of inducing, stimulating, or adjuvanting an immune response a pathogen.
  • the methods described herein can comprise administering to the mammal an effective amount of the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, optionally prior to or following a primary therapy administered to the mammal to remove or kill cancer cells expressing the cancer antigen.
  • the compositions of the present invention may be provided as a neoadjuvant therapy; however in preferred embodiments, the compositions of the present invention are administered following the primary therapy.
  • the primary therapy comprises surgery to remove the cancer cells from the mammal, radiation therapy to kill the cancer cells in the mammal, or both surgery and radiation therapy.
  • the methods described herein can comprise administering to the mammal an effective amount of the substantially pure CDNs of the present invention for the treatment of disorders in which shifting of Th1 to Th2 immunity confers clinical benefit.
  • CMI Cell-mediated immunity
  • IFN interferon
  • TNF tumor necrosis factor
  • humoral immunity is associated with TH2 CD4+ T lymphocytes producing IL-4, IL-6 and IL-10.
  • Immune deviation towards TH1 responses typically produces activation of cytotoxic T-cell lymphocytes (CTL), natural killer (NK) cells, macrophages and monocytes.
  • Th1 responses are more effective against intracellular pathogens (viruses and bacteria that are inside host cells) and tumors
  • Th2 responses are more effective against extracellular bacteria, parasites including helminths and toxins.
  • the activation of innate immunity is expected to normalize the T-helper type 1 and 2 (Th1/Th2) immune system balance and to suppress the excessive reaction of Th2 type responses that cause immunoglobulin (Ig) E-dependent allergies and allergic asthma.
  • FIG. 1 depicts cyclic purine dinucleotide (“CDN”)-mediated signaling.
  • CDN e.g., c-di-AMP or c-di-GMP
  • cytosolic receptor STING Stimulator of Interferon Genes
  • FIG. 2A depicts a synthesis scheme for c-[G(2′,5′)pG(3′,5′)p] and dithio derivatives.
  • FIG. 2B depicts a synthesis scheme for c-[A(2′,5′)pA(3′,5′)p] and dithio derivatives.
  • FIG. 2C depicts structures of compounds 10, 20, 21, 22, and 23.
  • FIG. 3A depicts 1 H-NMR results for compound 9a.
  • FIG. 3B depicts COSY (3.5-6.0 ppm 1 H-axis) results for compound 9a.
  • FIG. 3C depicts HMBC (3.0-5.5 ppm 1 H-axis) results for compound 9a.
  • FIG. 3D depicts 1 H-NMR results for compound 21.
  • FIG. 3E depicts COSY (3.5-6.0 ppm 1 H-axis) results for compound 21.
  • FIG. 3F depicts HMBC (0-9.5 ppm 1 H-axis) results for compound 21.
  • FIG. 3G depicts HMBC (3.5-5.5 ppm 1 H-axis)results for compound 21.
  • FIG. 3H depicts analytical HPLC (2-20% ACN/10 mM TEAA buffer—20 min) results for compound 19b.
  • FIG. 4 depicts c-[G(2′,5′)pG(3′,5′)p] and dithio ribose O-substitued derivatives.
  • FIG. 5 depicts c-[A(2′,5′)pA(3′,5′)p] and dithio ribose O-substitued derivatives.
  • FIG. 6 depicts c-[G(2′,5′)pA(3′,5′)p] and dithio ribose O-substitued derivatives.
  • FIG. 7 Depicts Type 1 interferon production in THP-1 cells following stimulation with various cyclic dinucleotide molecules
  • FIG. 8 depicts normalized RNA expression levels of Type 1 interferons and interferon gamma in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules
  • FIG. 9A depicts levels of Type 1 interferon alpha protein in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules.
  • FIG. 9B depicts levels of Type 1 interferon beta protein in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules.
  • FIG. 9C depicts levels of interferon gamma protein in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules.
  • FIG. 10 depicts IFN-3 induction in human cells as a signature of adjuvant potency following treatment with various cyclic dinucleotide molecules.
  • FIG. 11A depicts upregulation of surface CD69 expression on natural killer (NK) cells as a measure of immune activation following treatment with various cyclic dinucleotide molecules.
  • FIG. 11B depicts upregulation of surface CD69 expression on CD4 + cells as a measure of immune activation following treatment with various cyclic dinucleotide molecules.
  • FIG. 11C depicts upregulation of surface CD69 expression on CD8+ T cells as a measure of immune activation following treatment with various cyclic dinucleotide molecules.
  • FIG. 12 depicts resistance of various CDN derivatives to phosphodiesterase treatment.
  • FIG. 13 depicts various known STING variants.
  • FIG. 14 depicts stimulation of HEK293 cell lines encoding human STING variant alleles by measuring the fold induction of the IFN ⁇ -LUC reporter.
  • FIG. 15A depicts surface expression of MHC class I (HLA-ABC), CD80, CD83 and CD86 by stimulated human dendritic cells.
  • FIG. 15B representative histograms of CD80, CD86, CD83 and MHC Class I (HLA-ABC) expression in human DCs following LPS or CDN stimulation.
  • FIG. 16 depicts OVA-specific CD8 T cell immunity in PBMCs in C57BL/6 mice at 7 days post vaccination with cyclic dinucleotide adjuvanted OVA protein.
  • FIG. 17 depicts OVA-specific CD8 T cell immunity in PBMCs in C57BL/6 or goldentickt (STING ⁇ / ⁇ ) mice at 7 days post vaccination with cyclic dinucleotide adjuvanted OVA protein.
  • FIG. 18 depicts tumor volume in a B16 melanoma model following treatment with various cyclic dinucleotide molecules.
  • FIG. 19A depicts survival curves in a CT26 lung metastasis tumor model following treatment with various cyclic dinucleotide molecules.
  • FIG. 19B depicts survival curves in a CT26 colon carcinoma tumor model following treatment with various cyclic dinucleotide molecules.
  • FIG. 19C depicts survival curves in a 4T1 mammary carcinoma tumor model following treatment with various cyclic dinucleotide molecules.
  • FIG. 20A depicts tumor inhibition in wild-type C57BL/6 mice following ML RR-CDN administration as compared to control mice receiving HBSS and CpG dinculeotide.
  • FIG. 20B depicts results obtained in STING deficient mice.
  • FIG. 21A depicts rejection of established CT26 colon carcinomas following ML RR-CDN administration.
  • FIG. 21B depicts IFN- ⁇ induction from mice treated with ML RR-CDA.
  • FIG. 21C depicts survival in mice following re-challenge with CT26 tumor cells.
  • FIG. 22A depicts rejection of established 4T1 mammary carcinomas following ML RR-CDN administration.
  • FIG. 22B depicts protection from re-challenge with CT26 tumor cells.
  • FIG. 23A depicts inhibition of the treated primary tumor in CT26 tumor-bearing animals following ML RR-CDA administration, as compared to HBSS vehicle control.
  • FIG. 23B depicts inhibition of the treated primary tumor in 4T1 tumor-bearing animals following ML RR-CDA administration, as compared to HBSS vehicle control.
  • FIG. 24A depicts inhibition of the treated primary tumor in B16 melanoma following ML RR-CDA administration.
  • FIG. 24B depicts inhibition of growth of distal lung tumor nodules following ML RR-CDA administration, as compared to HBSS vehicle control in graphical form.
  • FIG. 24C depicts inhibition of growth of distal lung tumor nodules following ML RR-CDA administration, as compared to HBSS vehicle control in the lung tissue itself in photographic form.
  • the present invention relates to the use of novel and highly active cyclic-di-nucleotide (CDN) immune stimulators that activate DCs via a recently discovered cytoplasmic receptor known as STING (Stimulator of Interferon Genes).
  • CDNs of the present invention are provided in the form of a composition comprising one or more cyclic purine dinucleotides induce STING-dependent type I interferon production, wherein the cyclic purine dinuclotides present in the composition are substantially pure 2′,5′,2′,5′ and 2′,5′,3′,5′ CDNs.
  • PAMPs Pathogen-Associated Molecular Patterns
  • PRRs host cell Pattern Recognition Receptors
  • cytokines and chemokines a downstream signaling cascade resulting in the induction of cytokines and chemokines
  • An objective of adjuvant design is to select defined PAMPs or synthetic molecules specific for designated PRRs to initiate a desired response.
  • Adjuvants such as monophosphoryl lipid A (MPL) and CpG are PAMPs recognized by Toll-like receptors (TLRs), a class of transmembrane PRRs that signal through MyD88 and Trif adaptor molecules and mediate induction of NF-kB dependent proinflammatory cytokines.
  • MPL TLR-4 agonist
  • CpG TLR-9 agonist
  • TLRs present on the cell surface e.g., TLR-4
  • endosomes e.g., CpG
  • TLRs present on the cell surface e.g., TLR-4
  • CpG endosomes
  • the productive growth cycle of multiple pathogens including viruses and intracellular bacteria occurs in the cytosol.
  • the compartmentalization of extracellular, vacuolar, and cytosolic PRRs has led to the hypothesis that the innate immune system distinguishes between pathogenic and non-pathogenic microbes by monitoring the cytosol.
  • agonists specific for PRRs comprising the cytosolic surveillance pathway that initiate development of protective immunity against intracellular pathogens, and is relevant to vaccine design.
  • These same targeting ligands will also be essential in the development of effective vaccines targeting malignancies, know to require tumor-specific CD4+ and CD8+ T cells.
  • CSP Cytosolic Surveillance Pathway
  • LLO listeriolysin O
  • IFN- ⁇ in infected host cells is regulated by Lm multidrug efflux pumps (MDRs), which that secrete structurally unrelated small molecules, including antibiotics.
  • MDRs Lm multidrug efflux pumps
  • IFN- ⁇ is not induced in host cells infected with Lm LLO mutants that are confined to the phagolysosome.
  • Normal levels of IFN- ⁇ are induced in infected MyD88 ⁇ / ⁇ Trif ⁇ / ⁇ macrophages deficient in all TLR-mediated signaling.
  • Cyclic-di-Nucleotides activate the cytosolic surveillance pathway through direct binding of to the cytosolic PRR, STING.
  • the Type I interferon response to infection by Lm and other intracellular bacteria results from the secretion of c-di-AMP or its related cyclic dinucleotide (CDN), c-di-GMP, and its direct binding to DDX41 and DEAD (aspartate-glutamate-alanine-aspartate) box helicase and STING (Stimulator of Interferon Genes), a recently defined receptor of the cytosolic surveillance pathway.
  • CDNs are second messengers expressed by most bacteria and regulate diverse processes, including motility and formation of biofilms.
  • STING In addition to activating the TBK-1/IRF-3 signaling pathway, in response to binding CDNs STING also activates the IkB kinase, resulting in translocation of the NF-kB transcription factor to the nucleus, activating the expression of multiple pro-inflammatory genes.
  • STING senses cytoplasmic DNA remained elusive. Unlike AIM2 which directly binds dsDNA, STING lacks any obvious DNA-binding domains. Whether other candidate DNA sensors such as DDX41, DNA-PK and DAI kinase were essential mediators of dsDNA signaling through STING remained unclear.
  • cyclic GMP-AMP synthase a host cell nucleotidyl transferase that in response to binding dsDNA synthesizes a second messenger, cyclic di-GMP-AMP, which binds directly to STING and initiates a signaling cascade through the TBK-1/IRF-3 axis, resulting in the induction of IFNs.
  • cGAS cyclic GMP-AMP synthase
  • the cGAS innate immune DNA sensor produces a non-canonical cyclic di-nucleotide that activates STING signaling.
  • the internucleotide phosphate bridge in the cyclic-GMP-AMP synthesized by cGAS is joined by non-canonical 2′, 5′ and 3′,5′ linkages, represented c[G(2′,5′)pA(3′,5′)p].
  • STING Stimulator of Interferon Genes
  • CDNs cyclic dinucleotides
  • cGAS host cell cyclic GMP-AMP synthase
  • Native CDN molecules are sensitive to degradation by phosphodiesterases that are present in host cells, for example in antigen presenting cells, that take up vaccine formulations that contain said native CDN molecules.
  • the potency of a defined adjuvant may be diminished by such degradation, as the adjuvant would be unable to bind and activate its defined PRR target.
  • Lower adjuvant potency could be measured, for example by a lower amount of induced expression of a signature molecule of innate immunity (e.g., IFN- ⁇ ), correlated with weaker vaccine potency, as defined by the magnitude of a measured antigen-specific immune response.
  • substantially pure 2′,5′,2′,5′ and 2′,5′,3′,5′ CDNs, and particularly dithio-diphosphate derivatives of 2′,5′,2′,5′ and 2′,5′,3′,5′ c-di-AMP and c-di-GMP are provided.
  • the synthesis process for said dithio-diphosphate derivatives of c-di-AMP and c-di-GMP molecules results in a mixture of diastereomers, including Rp,Rp, Sp,Sp, SpRp, and Rp,Sp dithio-diphosphate derivatives of c-di-AMP and c-di-GMP molecules. These individual species may be separated, and exhibit substantial differences in their pharmaceutical characteristics.
  • administering refers without limitation to contact of an exogenous ligand, reagent, placebo, small molecule, pharmaceutical agent, therapeutic agent, diagnostic agent, or composition to the subject, cell, tissue, organ, or biological fluid, and the like.
  • administering can refer, e.g., to therapeutic, pharmacokinetic, diagnostic, research, placebo, and experimental methods. Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell.
  • administering also encompasses in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding composition, or by another cell.
  • administered together it is not meant to be implied that two or more agents be administered as a single composition.
  • administration as a single composition is contemplated by the present invention, such agents may be delivered to a single subject as separate administrations, which may be at the same or different time, and which may be by the same route or different routes of administration.
  • an “agonist,” as it relates to a ligand and receptor, comprises a molecule, combination of molecules, a complex, or a combination of reagents, that stimulates the receptor.
  • an agonist of granulocyte-macrophage colony stimulating factor (GM-CSF) can encompass GM-CSF, a mutein or derivative of GM-CSF, a peptide mimetic of GM-CSF, a small molecule that mimics the biological function of GM-CSF, or an antibody that stimulates GM-CSF receptor.
  • an “antagonist,” as it relates to a ligand and receptor, comprises a molecule, combination of molecules, or a complex, that inhibits, counteracts, downregulates, and/or desensitizes the receptor.
  • Antagonist encompasses any reagent that inhibits a constitutive activity of the receptor. A constitutive activity is one that is manifest in the absence of a ligand/receptor interaction.
  • Antagonist also encompasses any reagent that inhibits or prevents a stimulated (or regulated) activity of a receptor.
  • an antagonist of GM-CSF receptor includes, without implying any limitation, an antibody that binds to the ligand (GM-CSF) and prevents it from binding to the receptor, or an antibody that binds to the receptor and prevents the ligand from binding to the receptor, or where the antibody locks the receptor in an inactive conformation.
  • substantially purified with regard to CDNs of the invention is meant that a specified species accounts for at least 50%, more often accounts for at least 60%, typically accounts for at least 70%, more typically accounts for at least 75%, most typically accounts for at least 80%, usually accounts for at least 85%, more usually accounts for at least 90%, most usually accounts for at least 95%, and conventionally accounts for at least 98% by weight, or greater, of the CDN activity present in a composition.
  • the weights of water, buffers, salts, detergents, reductants, protease inhibitors, stabilizers (including an added protein such as albumin), and excipients are generally not used in the determination of purity.
  • nucleic acid/complementary nucleic acid e.g., a cytokine to a cytokine receptor
  • a target biomolecule e.g., a cytokine to a cytokine receptor
  • Specific binding can mean, e.g., that the binding compound, nucleic acid ligand, antibody, or binding composition derived from the antigen-binding site of an antibody, of the contemplated method binds to its target with an affinity that is often at least 25% greater, more often at least 50% greater, most often at least 100% (2-fold) greater, normally at least ten times greater, more normally at least 20-times greater, and most normally at least 100-times greater than the affinity with a non-target molecule.
  • Affinity can be determined at equilibrium by measuring the fraction bound (r) of labeled ligand at various concentrations (c).
  • r/c is plotted on the Y-axis versus r on the X-axis, thus producing a Scatchard plot.
  • Affinity measurement by Scatchard analysis is well known in the art. See, e.g., van Erp et al., J.
  • affinity can be measured by isothermal titration calorimetry (ITC).
  • ITC isothermal titration calorimetry
  • a binding curve is then obtained from a plot of the heats from each injection against the ratio of ligand and binding partner in the cell.
  • subject refers to a human or non-human organism.
  • the methods and compositions described herein are applicable to both human and veterinary disease.
  • subjects are “patients,” i.e., living humans that are receiving medical care for a disease or condition. This includes persons with no defined illness who are being investigated for signs of pathology.
  • subjects who have an existing diagnosis of a particular cancer which is being targeted by the compositions and methods of the present invention.
  • Preferred cancers for treatment with the compositions described herein include, but are not limited to prostate cancer, renal carcinoma, melanoma, pancreatic cancer, cervical cancer, ovarian cancer, colon cancer, head & neck cancer, lung cancer and breast cancer.
  • “Therapeutically effective amount” is defined as an amount of a reagent or pharmaceutical composition that is sufficient to show a patient benefit, i.e., to cause a decrease, prevention, or amelioration of the symptoms of the condition being treated.
  • a “diagnostically effective amount” is defined as an amount that is sufficient to produce a signal, image, or other diagnostic parameter. Effective amounts of the pharmaceutical formulation will vary according to factors such as the degree of susceptibility of the individual, the age, gender, and weight of the individual, and idiosyncratic responses of the individual.
  • Effective amount encompasses, without limitation, an amount that can ameliorate, reverse, mitigate, prevent, or diagnose a symptom or sign of a medical condition or disorder or a causative process thereof. Unless dictated otherwise, explicitly or by context, an “effective amount” is not limited to a minimal amount sufficient to ameliorate a condition.
  • Treatment or “treating” (with respect to a condition or a disease) is an approach for obtaining beneficial or desired results including and preferably clinical results.
  • beneficial or desired results with respect to a disease include, but are not limited to, one or more of the following: preventing a disease, improving a condition associated with a disease, curing a disease, lessening severity of a disease, delaying progression of a disease, alleviating one or more symptoms associated with a disease, increasing the quality of life of one suffering from a disease, and/or prolonging survival.
  • beneficial or desired results with respect to a condition include, but are not limited to, one or more of the following: preventing a condition, improving a condition, curing a condition, lessening severity of a condition, delaying progression of a condition, alleviating one or more symptoms associated with a condition, increasing the quality of life of one suffering from a condition, and/or prolonging survival.
  • the beneficial or desired results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cells, reducing metastasis of neoplastic cells found in cancers, shrinking the size of a tumor, decreasing symptoms resulting from the cancer, increasing the quality of life of those suffering from the cancer, decreasing the dose of other medications required to treat the disease, delaying the progression of the cancer, and/or prolonging survival of patients having cancer.
  • “treatment” of a subject can imply that the subject is in need of treatment, e.g., in the situation where the subject comprises a disorder expected to be ameliorated by administration of a reagent.
  • Vaccine encompasses preventative vaccines. Vaccine also encompasses therapeutic vaccines, e.g., a vaccine administered to a mammal that comprises a condition or disorder associated with the antigen or epitope provided by the vaccine.
  • Prokaryotic as well as eukaryotic cells use various small molecules for cell signaling and intra- and intercellular communication. Cyclic nucleotides like cGMP, cAMP, etc. are known to have regulatory and initiating activity in pro- and eukaryotic cells. Unlike eukaryotic cells, prokaryotic cells also use cyclic purine dinucleotides as regulatory molecules. In prokaryotes, the condensation of two GTP molecules is catalyst by the enzyme diguanylate cyclase (DGC) to give c-diGMP, which represents an important regulator in bacteria.
  • DGC diguanylate cyclase
  • cyclic diGMP or analogs thereof can also stimulate or enhance immune or inflammatory response in a patient or can enhance the immune response to a vaccine by serving as an adjuvant in mammals.
  • Cytosolic detection of pathogen-derived DNA requires signaling through TANK binding kinase 1 (TBK1) and its downstream transcription factor, IFN-regulatory factor 3 (IRF3).
  • TK1 TANK binding kinase 1
  • IRF3 IFN-regulatory factor 3
  • Cyclic purine dinucleotides for use as precursors to derive the CDNs of the present invention are described in some detail in, e.g., Gao et al., Cell (2013) 153: doi: 10.1016/j.cell.2013.04.046; U.S. Pat. Nos. 7,709,458 and 7,592,326; WO2007/054279; and Yan et al., Bioorg. Med. Chem Lett. 18: 5631 (2008), each of which is hereby incorporated by reference.
  • These CDNs may be modified using standard organic chemistry techniques in order to produce the CDNs of the present invention.
  • Preferred purines include, but are not limited to, adenine, guanine, inosine, hypoxanthine, xanthine, isoguanine, etc.
  • the CDNs of the present invention are preferably phosphorothioate analogues, and most preferably substantially pure Sp,Sp, Rp,Rp, SpRp, or Rp,Sp stereoisomers thereof.
  • each ribose comprises a 2′ or 3′ hydroxyl which may be substituted.
  • the CDNs of the present invention can comprise a substitution at one or both of these 2′ or 3′ hydroxyls (which is not part of the cyclic linkage) which provide a prodrug leaving group or other modification which affects activity, solubility, bioavailability, etc.
  • prodrug refers to a modification of contemplated compounds, wherein the modified compound exhibits less pharmacological activity (as compared to the modified compound) and wherein the modified compound is converted within the body (e.g., in a target cell or target organ) back into the unmodified form through enzymatic or non-enzymatic reactions.
  • the hydroxyl on one ribose comprises a prodrug leaving group.
  • Prodrugs can modify the physicochemical, biopharmaceutic, and pharmacokinetic properties of drugs. Traditional prodrugs are classified as drugs that are activated by undergoing transformation in vivo to form the active drug.
  • prodrug development is typically poor aqueous solubility, chemical instability, low oral bioavailability, lack of blood brain barrier penetration, and high first pass metabolism associated with the parent drug.
  • Suitable prodrug moieties are described in, for example, “Prodrugs and Targeted Delivery,” J. Rautico, Ed., John Wiley & Sons, 2011.
  • Preferred cyclic purine dinucleotides are phosphorothioate analogues, referred to herein as “thiophosphates”.
  • Phosphorothioates are a variant of normal nucleotides in which one of the nonbridging oxygens is replaced by a sulfur.
  • the sulfurization of the internucleotide bond dramatically reduces the action of endo- and exonucleases, including 5′ to 3′ and 3′ to 5′ DNA POL 1 exonuclease, nucleases S1 and P1, RNases, serum nucleases and snake venom phosphodiesterase.
  • the potential for crossing the lipid bilayer increases.
  • a phosphorothioate linkage in inherently chiral may each exist in R or S forms.
  • Rp,Rp, Sp,Sp, Sp,Rp, and Rp,Sp forms are possible.
  • cyclic purine dinucleotides of the present invention comprise 2′-O— and 3′-O— substituent forms of CDNs, and in particular CDN thiophosphates. Additional stability and bioavailability can be provided by the substitution of the 2′-OH of the ribose moiety.
  • each R aa , R bb and R cC is, independently, H, an optionally linked chemical functional group or a further substituent group with a preferred list including without limitation, H, alkyl, alkenyl, alkynyl, aliphatic, alkoxy, acyl, aryl, aralkyl, heteroaryl, alicyclic, heterocyclic and heteroarylalkyl. Selected substituents within the compounds described herein are present to a recursive degree.
  • alkyl refers to a saturated straight or branched hydrocarbon radical containing up to twenty four carbon atoms.
  • alkyl groups include without limitation, methyl, ethyl, propyl, butyl, isopropyl, n-hexyl, octyl, decyl, dodecyl and the like.
  • Alkyl groups typically include from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred.
  • the term “lower alkyl” as used herein includes from 1 to about 6 carbon atoms.
  • Alkyl groups as used herein may optionally include one or more further substituent groups.
  • alkenyl refers to a straight or branched hydrocarbon chain radical containing up to twenty four carbon atoms and having at least one carbon-carbon double bond.
  • alkenyl groups include without limitation, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, dienes such as 1,3-butadiene and the like.
  • Alkenyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred.
  • Alkenyl groups as used herein may optionally include one or more further substituent groups.
  • alkynyl refers to a straight or branched hydrocarbon radical containing up to twenty four carbon atoms and having at least one carbon-carbon triple bond.
  • alkynyl groups include, without limitation, ethynyl, 1-propynyl, 1-butynyl, and the like.
  • Alkynyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred.
  • Alkynyl groups as used herein may optionally include one or more further substituent groups.
  • acyl refers to a radical formed by removal of a hydroxyl group from an organic acid and has the general Formula —C(O)—X where X is typically aliphatic, alicyclic or aromatic. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates, aliphatic phosphates and the like. Acyl groups as used herein may optionally include further substituent groups.
  • alicyclic refers to a cyclic ring system wherein the ring is aliphatic.
  • the ring system can comprise one or more rings wherein at least one ring is aliphatic.
  • Preferred alicyclics include rings having from about 5 to about 9 carbon atoms in the ring.
  • Alicyclic as used herein may optionally include further substituent groups.
  • aliphatic refers to a straight or branched hydrocarbon radical containing up to twenty four carbon atoms wherein the saturation between any two carbon atoms is a single, double or triple bond.
  • An aliphatic group preferably contains from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred.
  • the straight or branched chain of an aliphatic group may be interrupted with one or more heteroatoms that include nitrogen, oxygen, sulfur and phosphorus.
  • Such aliphatic groups interrupted by heteroatoms include without limitation, polyalkoxys, such as polyalkylene glycols, polyamines, and polyimines. Aliphatic groups as used herein may optionally include further substituent groups.
  • alkoxy refers to a radical formed between an alkyl group and an oxygen atom wherein the oxygen atom is used to attach the alkoxy group to a parent molecule.
  • alkoxy groups include without limitation, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, neopentoxy, n-hexoxy and the like.
  • Alkoxy groups as used herein may optionally include further substituent groups.
  • aminoalkyl refers to an amino substituted C ⁇ -Cn alkyl radical.
  • the alkyl portion of the radical forms a covalent bond with a parent molecule.
  • the amino group can be located at any position and the aminoalkyl group can be substituted with a further substituent group at the alkyl and/or amino portions.
  • aralkyl and arylalkyl refer to an aromatic group that is covalently linked to a C ⁇ -Cn alkyl radical.
  • the alkyl radical portion of the resulting aralkyl (or arylalkyl) group forms a covalent bond with a parent molecule. Examples include without limitation, benzyl, phenethyl and the like.
  • Aralkyl groups as used herein may optionally include further substituent groups attached to the alkyl, the aryl or both groups that form the radical group.
  • aryl and aromatic refer to a mono- or polycyclic carbocyclic ring system radicals having one or more aromatic rings.
  • aryl groups include without limitation, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like.
  • Preferred aryl ring systems have from about 5 to about 20 carbon atoms in one or more rings.
  • Aryl groups as used herein may optionally include further substituent groups.
  • halo and “halogen,” as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.
  • heteroaryl refers to a radical comprising a mono- or poly-cyclic aromatic ring, ring system or fused ring system wherein at least one of the rings is aromatic and includes one or more heteroatoms. Heteroaryl is also meant to include fused ring systems including systems where one or more of the fused rings contain no heteroatoms. Heteroaryl groups typically include one ring atom selected from sulfur, nitrogen or oxygen.
  • heteroaryl groups include without limitation, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl and the like.
  • Heteroaryl radicals can be attached to a parent molecule directly or through a linking moiety such as an aliphatic group or hetero atom.
  • Heteroaryl groups as used herein may optionally include further substituent groups.
  • heteroarylalkyl refers to a heteroaryl group as previously defined that further includes a covalently attached C 1 -C 12 alkyl radical.
  • the alkyl radical portion of the resulting heteroarylalkyl group is capable of forming a covalent bond with a parent molecule. Examples include without limitation, pyridinylmethyl, pyrimidinylethyl, napthyridinylpropyl and the like.
  • Heteroarylalkyl groups as used herein may optionally include further substituent groups on one or both of the heteroaryl or alkyl portions.
  • preferred cyclic purine dinucleotides also include prodrug forms of CDNs, and in particular CDN thiophosphates.
  • Produrgs can modify the physicochemical, biopharmaceutic, and pharmacokinetic properties of drugs.
  • Traditional prodrugs are classified as drugs that are activated by undergoing transformation in vivo to form the active drug.
  • Reasons for prodrug development are typically poor aqueous solubility, chemical instability, low oral bioavailability, lack of blood brain barrier penetration, and high first pass metabolism associated with the parent drug. Suitable prodrug moieties are described in, for example, “Prodrugs and Targeted Delivery,” J. Rautico, Ed., John Wiley & Sons, 2011.
  • substantially pure refers to an Rp,Rp or Rp,Sp form which is at least 75% pure relative to other possible stereochemistries at the chiral centers indicated in the figure above.
  • a “substantially pure Rp,Rp c-di-GMP thiophosphate” would be at least 75% pure with regard to the Rp,Sp and Sp,Sp forms of c-di-GMP thiophosphate.
  • a substantially pure cyclic purine dinucleotide is at least 85% pure, at least 90% pure, at least 95% pure, at least 97% pure, and at least 99% pure.
  • a substantially pure cyclic purine dinucleotide preparation of the invention is “stereochemically pure,” this is not meant to indicate that all CDNs within the preparation having a particular stereochemistry at these chiral centers are otherwise identical.
  • a substantially pure cyclic purine dinucleotide preparation may contain a combination of Rp,Rp c-di-GMP thiophosphate and Rp,Rp c-di-AMP thiophosphate and still be a substantially pure cyclic purine dinucleotide preparation.
  • Such a preparation may also include other components as described hereinafter that are advantageous for patient treatment, provided that all CDNs within the preparation having a particular stereochemistry at these chiral centers.
  • the CDN compositions described herein can be administered to a host, either alone or in combination with a pharmaceutically acceptable excipient, in an amount sufficient to induce, modify, or stimulate an appropriate immune response.
  • the immune response can comprise, without limitation, specific immune response, non-specific immune response, both specific and non-specific response, innate response, primary immune response, adaptive immunity, secondary immune response, memory immune response, immune cell activation, immune cell proliferation, immune cell differentiation, and cytokine expression.
  • the CDN compositions are administered in conjunction with one or more additional compositions including vaccines intended to stimulate an immune response to one or more predetermined antigens; adjuvants; CTLA-4 and PD-1 pathway antagonists, lipids, liposomes, chemotherapeutic agents, immunomodulatory cell lines, etc.
  • the CDN compositions may be administered before, after, and/or together with an additional therapeutic or prophylactic composition or modality.
  • additional therapeutic or prophylactic composition or modality include, without limitation, B7 costimulatory molecule, interleukin-2, interferon- ⁇ , GM-CSF, CTLA-4 antagonists, OX-40/OX-40 ligand, CD40/CD40 ligand, sargramostim, levamisol, vaccinia virus, Bacille Calmette-Guerin (BCG), liposomes, alum, Freund's complete or incomplete adjuvant, detoxified endotoxins, mineral oils, surface active substances such as lipolecithin, pluronic polyols, polyanions, peptides, and oil or hydrocarbon emulsions.
  • BCG Bacille Calmette-Guerin
  • Carriers for inducing a T cell immune response which preferentially stimulate a cytolytic T cell response versus an antibody response are preferred, although those that stimulate both types of response can be used as well.
  • the agent is a polypeptide
  • the polypeptide itself or a polynucleotide encoding the polypeptide can be administered.
  • the carrier can be a cell, such as an antigen presenting cell (APC) or a dendritic cell.
  • APC antigen presenting cell
  • Antigen presenting cells include such cell types as macrophages, dendritic cells and ⁇ cells.
  • Other professional antigen-presenting cells include monocytes, marginal zone Kupffer cells, microglia, Langerhans' cells, interdigitating dendritic cells, follicular dendritic cells, and T cells.
  • Facultative antigen-presenting cells can also be used.
  • facultative antigen-presenting cells include astrocytes, follicular cells, endothelium and fibroblasts.
  • the carrier can be a bacterial cell that is transformed to express the polypeptide or to deliver a polynucleoteide which is subsequently expressed in cells of the vaccinated individual.
  • Adjuvants such as aluminum hydroxide or aluminum phosphate, can be added to increase the ability of the vaccine to trigger, enhance, or prolong an immune response.
  • Additional materials such as cytokines, chemokines, and bacterial nucleic acid sequences, like CpG, a toll-like receptor (TLR) 9 agonist as well as additional agonists for TLR 2, TLR 4, TLR 5, TLR 7, TLR 8, TLR9, including lipoprotein, LPS, monophosphoryl lipid A, lipoteichoic acid, imiquimod, resiquimod, and in addition retinoic acid-inducible gene I (RIG-I) agonists such as poly I:C, used separately or in combination with the described compositions are also potential adjuvants.
  • TLR toll-like receptor
  • adjuvants include the synthetic adjuvant QS-21 comprising a homogeneous saponin purified from the bark of Quillaja saponaria and Corynebacterium parvum (McCune et al., Cancer, 1979; 43:1619). It will be understood that the adjuvant is subject to optimization. In other words, the skilled artisan can engage in routine experimentation to determine the best adjuvant to use.
  • adjuvant properties of the compounds of the present invention may also combined with other therapeutic modalities including other vaccines, adjuvants, antigen, antibodies, and immune modulators. Examples are provided below.
  • compositions of the present invention may further comprise one or more additional substances which, because of their nature, can act to stimulate or otherwise utilize the immune system to respond to the cancer antigens present on the inactivated tumor cell(s).
  • Such adjuvants include, but are not limited to, lipids, liposomes, inactivated bacteria which induce innate immunity (e.g., inactivated or attenuated Listeria monocytogenes ), compositions which mediate innate immune activation via Toll-like Receptors (TLRs), (NOD)-like receptors (NLRs), Retinoic acid inducible gene-based (RIG)-I-like receptors (RLRs), and/or C-type lectin receptors (CLRs).
  • TLRs Toll-like Receptors
  • NLRs NOD-like receptors
  • RLRs Retinoic acid inducible gene-based
  • CLRs C-type lectin receptors
  • PAMPs examples include lipoproteins, lipopolypeptides, peptidoglycans, zymosan, lipopolysaccharide, neisserial porins, flagellin, profillin, galactoceramide, muramyl dipeptide.
  • Peptidoglycans, lipoproteins, and lipoteichoic acids are cell wall components of Gram-positive. Lipopolysaccharides are expressed by most bacteria, with MPL being one example.
  • Flagellin refers to the structural component of bacterial flagella that is secreted by pathogenic and commensal bacterial.
  • ⁇ -Galactosylceramide ( ⁇ -GalCer) is an activator of natural killer T (NKT) cells.
  • Muramyl dipeptide is a bioactive peptidoglycan motif common to all bacteria. This list is not meant to be limiting.
  • Preferred adjuvant compositions are described below.
  • CTLA-4 is thought to be an important negative regulator of the adaptive immune response. Activated T cells upregulate CTLA-4, which binds CD80 and CD86 on antigen-presenting cells with higher affinity than CD28, thus inhibiting T-cell stimulation, IL-2 gene expression and T-cell proliferation. Anti-tumor effects of CTLA4 blockade have been observed in murine models of colon carcinoma, metastatic prostate cancer, and metastatic melanoma.
  • Ipilimumab (YervoyTM) and tremelimumab are humanized monoclonal antibodies that bind to human CTLA4 and prevent its interaction with CD80 and CD86. Phase I and II studies using ipilimumab and tremelimumab have demonstrated clinical activity in cancer patients.
  • Other negative immune regulators which may be targeted by a similar strategy include programmed cell death 1, B and T lymphocyte attenuator, transforming growth factor beta ⁇ , interleukin-10, and vascular endothelial growth factor.
  • PD-1 is another negative regulator of adaptive immune response that is expressed on activated T-cells. PD-1 binds to B7-H1 and B7-DC, and the engagement of PD-1 suppresses T-cell activation. Anti-tumor effects have been demonstrated with PD-1 pathway blockade. BMS-936558, MK3475, CT-011, AMP-224 and MDX-1106 have been reported in the literature to be examples of PD-1 pathway blockers which may find use in the present invention.
  • TLR toll like receptor
  • DC dendritic cell
  • TLRs are a family of pattern recognition receptors that were initially identified as sensors of the innate immune system that recognize microbial pathogens.
  • TLRs comprise a family of conserved membrane spanning molecules containing an ectodomain of leucine-rich repeats, a transmembrane domain and an intracellular TIR (Toll/IL-1R) domain.
  • PAMPs pathogen associated molecular patterns.
  • Ligand binding to TLRs invokes a cascade of intra-cellular signaling pathways that induce the production of factors involved in inflammation and immunity.
  • TLRs that are expressed on the surface of cells include TLR-1,-2,-4,-5, and -6, while TLR-3, -7/8, and -9 are expressed with the ER compartment.
  • Human dendritic cell subsets can be identified on the basis of distinct TLR expression patterns.
  • the myeloid or “conventional” subset of DC expresses TLRs 1-8 when stimulated, and a cascade of activation markers (e.g. CD80, CD86, MHC class I and II, CCR7), pro-inflammatory cytokines, and chemokines are produced.
  • a cascade of activation markers e.g. CD80, CD86, MHC class I and II, CCR7
  • DCs acquire an enhanced capacity to take up antigens and present them in an appropriate form to T cells.
  • plasmacytoid subset of DC expresses only TLR7 and TLR9 upon activation, with a resulting activation of NK cells as well as T-cells.
  • activating DC with TLR agonists may be beneficial for priming anti-tumor immunity in an immunotherapy approach to the treatment of cancer. It has also been suggested that successful treatment of breast cancer using radiation and chemotherapy requires TLR4 activation.
  • TLR agonists known in the art and finding use in the present invention include, but are not limited to, the following:
  • TLR agonists are preferably used in combinations with other vaccines, adjuvants and/or immune modulators, and may be combined in various combinations.
  • the cyclic purine dinucleotides that bind to STING and induces STING-dependent TBK1 activation and an inactivated tumor cell which expresses and secretes one or more cytokines which stimulate dendritic cell induction, recruitment and/or maturation, as described herein can be administered together with one or more TLR agonists for therapeutic purposes.
  • ADCC Antibody-Dependent Cell-Mediated Cytotoxicity
  • Classical ADCC is mediated by natural killer (NK) cells; macrophages, neutrophils and eosinophils can also mediate ADCC.
  • NK natural killer
  • ADCC is an important mechanism of action of therapeutic monoclonal antibodies, including trastuzumab and rituximab, against tumors. Compounds of the present invention may act to potentiate ADCC.
  • Muromonab-CD3 Used to prevent acute rejection of organ, e.g., kidney, transplants.
  • organ e.g., kidney, transplants.
  • the humanized versions show promise in inhibiting the autoimmune destruction of beta cells in Type 1 diabetes mellitus.
  • Infliximab (Remicade®) and adalimumab (Humira®): Bind to tumor necrosis factor-alpha (TNF- ⁇ ). Used in some inflammatory diseases such as rheumatoid arthritis, psoriasis, Crohns disease.
  • Omalizumab (Xolair®). Binds to IgE thus preventing IgE from binding to mast cells. Used against allergic asthma.
  • Daclizumab (Zenapax®). Binds to part of the IL-2 receptor exposed at the surface of activated T cells. Used to prevent acute rejection of transplanted kidneys.
  • Tositumomab (Bexxar®). This is a conjugate of a monoclonal antibody against CD20 and the radioactive isotope iodine-131 (131I).
  • Cetuximab (Erbitux®). Blocks HER1, a receptor for epidermal growth factor (EGF) that is found on some tumor cells (some breast cancers, lymphomas).
  • EGF epidermal growth factor
  • trastuzumab (Herceptin®). Blocks HER2, a growth factor receptor over-expressed in some 20% of breast cancers.
  • Adcetris® A conjugate of a monoclonal antibody that binds CD30, a cell-surface molecule expressed by the cells of some lymphomas but not found on the normal stem cells needed to repopulate the bone marrow.
  • Alemtuzumab (Campath-1H®). Binds to CD52, a molecule found on lymphocytes and depletes both T cells and B cells. Has produced complete remission of chronic lymphocytic leukemia and shows promise in preventing rejection of kidney transplants.
  • Lym-1 (Oncolym®). Binds to the HLA-DR-encoded histocompatibility antigen that can be expressed at high levels on lymphoma cells.
  • Ipilimumab (Yervoy®) that acts to enhance the body's own immune response to tumors.
  • Vitaxin Binds to a vascular integrin (alpha-v/beta-3) found on the blood vessels of tumors but not on the blood vessels supplying normal tissues. In Phase II clinical trials, Vitaxin has shown some promise in shrinking solid tumors without harmful side effects.
  • Bevacizumab (Avastin®). Binds to vascular endothelial growth factor (VEGF) preventing it from binding to its receptor. Used for the treatment of colorectal cancers.
  • VEGF vascular endothelial growth factor
  • Abciximab (ReoPro®). Inhibits the clumping of platelets by binding the receptors on their surface that normally are linked by fibrinogen. Helpful in preventing reclogging of the coronary arteries in patients who have undergone angioplasty.
  • Liposomes are vesicles formed from one (“unilamellar”) or more (“multilamellar”) layers of phospholipid. Because of the amphipathic character of the phospholipid building blocks, liposomes typically comprise a hydrophilic layer presenting a hydrophilic external face and enclosing a hydrophilic core.
  • WO2010/104833 which is incorporated by reference herein in its entirety, describes suitable liposomal preparations.
  • Such liposomal formulations referred to herein as VesiVax® (Molecular Express, Inc.), with our without the “immunogenic polypeptide(s) or carbohydrate(s)” referred to above, can contain one or more additional components such as peptidoglycan, lipopeptide, lipopolysaccharide, monophosphoryl lipid A, lipoteichoic acid, resiquimod, imiquimod, flagellin, oligonucleotides containing unmethylated CpG motifs, beta-galactosylceramide, muramyl dipeptide, all-trans retinoic acid, double-stranded viral RNA, heat shock proteins, dioctadecyldimethylammonium bromide, cationic surfactants, toll-like receptor agonists, dimyristoyltrimethylammoniumpropane,
  • liposomal formulations discussed above employ a” steroid derivative” as an anchor for attaching an immunogenic polypeptide or carbohydrate to a liposome
  • the steroid may simply be provided as an unconjugated steroid such as cholesterol.
  • Suitable methods for preparing liposomes from lipid mixtures are well known in the art. See, e.g., Basu & Basu, Liposome Methods and Protocols (Methods in Molecular Biology), Humana Press, 2002; Gregoriadis, Liposome Technology, 3 rd Edition, Informa HealthCare, 2006. Preferred methods include extrusion, homogenization, and sonication methods described therein.
  • An exemplary method for preparing liposomes for use in the present invention which comprises drying a lipid mixture, followed by hydration in an aqueous vehicle and sonication to form liposomes, is described in WO2010/104833.
  • the liposomes are provided within a particular average size range.
  • Liposome size can be selected, for example, by extrusion of an aqueous vehicle comprising liposomes through membranes having a preselected pore size and collecting the material flowing through the membrane.
  • the liposomes are selected to be substantially between 50 and 500 nm in diameter, more preferably substantially between 50 and 200 nm in diameter, and most preferably substantially between 50 and 150 nm in diameter.
  • substantially as used herein in this context means that at least 75%, more preferably 80%, and most preferably at least 90% of the liposomes are within the designated range.
  • lipid and lipid-like adjuvants which may find use in the present invention include oil-in-water (o/w) emulsions (see, e.g., Muderhwa et al., J. Pharmaceut. Sci. 88: 1332-9, 1999)), VesiVax® TLR (Molecular Express, Inc.), digitonin (see, e.g., U.S. Pat. No. 5,698,432), and glucopyranosyl lipids (see, e.g., United States Patent Application 20100310602).
  • o/w oil-in-water
  • VesiVax® TLR Molecular Express, Inc.
  • digitonin see, e.g., U.S. Pat. No. 5,698,432
  • glucopyranosyl lipids see, e.g., United States Patent Application 20100310602.
  • Nanoparticles also represent drug delivery systems suitable for most administration routes. Over the years, a variety of natural and synthetic polymers have been explored for the preparation of nanoparticles, of which Poly(lactic acid) (PLA), Poly(glycolic acid) (PGA), and their copolymers (PLGA) have been extensively investigated because of their biocompatibility and biodegradability. Nanoparticles and other nanocarriers act as potential carries for several classes of drugs such as anticancer agents, antihypertensive agents, immunomodulators, and hormones; and macromolecules such as nucleic acids, proteins, peptides, and antibodies. See, e.g., Crit. Rev. Ther. Drug Carrier Syst. 21:387-422, 2004; Nanomedicine: Nanotechnology, Biology and Medicine 1:22-30, 2005.
  • the methods further involve administering to the subject an effective amount of one or more chemotherapeutics as an additional treatment.
  • the one or more chemotherapeutics is selected from abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly-1-Lproline-t-butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3′,4′-didehydro-4′-deoxy-8′-norvin-caleukoblastine, docetaxol, doxetaxel
  • inactivated tumor cell is meant a tumor cell (either “autologous” or “allogeneic” to the patient) which has which been treated to prevent division of the cells.
  • tumor cells preserve their immunogenicity and their metabolic activity.
  • Such tumor cells are genetically modified to express a transgene which is expressed within a patient as part of cancer therapy.
  • a composition or vaccine of the invention comprises neoplastic (e.g., tumor) cells that are autologous or allogeneic to the patient undergoing treatment and is most preferably the same general type of tumor cell as is afflicting the patient.
  • a patient suffering from melanoma will typically be administered a genetically modified cell derived from a melanoma.
  • the inactivated tumor cells of the present invention are administered to the patient together with one or more costimulatory molecules or agents.
  • a preferred costimulatory agent comprises one or more cytokines which stimulate dendritic cell induction, recruitment, and/or maturation. Methods for assessing such costimulatory agents are well known in the literature. Induction and maturation of DCs is typically assessed by increased expression of certain membrane molecules such as CD80 and CD86, and/or secretion of pro-inflammatory cytokines, such as IL-12 and type I interferons following stimulation.
  • the inactivated tumor cells themselves are modified to express and secrete one or more cytokines which stimulate dendritic cell induction, recruitment, and/or maturation.
  • cytokines which stimulate dendritic cell induction, recruitment, and/or maturation.
  • the present invention is described in exemplary terms with regard to the use of GM-CSF.
  • the tumor cell may express a transgene encoding GM-CSF as described in U.S. Pat. Nos. 5,637,483, 5,904,920, 6,277,368 and 6,350,445, as well as in US Patent Publication No. 20100150946, each of which is expressly incorporated by reference herein.
  • GM-CSF-expressing genetically modified cancer cells or a “cytokine-expressing cellular vaccine” for the treatment of pancreatic cancer is described in U.S. Pat. Nos. 6,033,674 and 5,985,290, both of which are expressly incorporated by reference herein.
  • cytokines which may be expressed by such inactivated tumor cells and/or bystander cells instead of, or together with, GM-CSF include, but are not limited to, one or more of CD40 ligand, IL-12, CCL3, CCL20, and CCL21. This list is not meant to be limiting.
  • the tumor cell line may be accompanied by an inactivated bystander cell line which expresses and secretes one or more cytokines which stimulate dendritic cell induction, recruitment, and/or maturation.
  • the bystander cell line may provide all of the cytokines which stimulate dendritic cell induction, recruitment, and/or maturation, or may supplement cytokines which stimulate dendritic cell induction, recruitment, and/or maturation expressed and secreted by the inactivated tumor cells.
  • immunomodulatory cytokine-expressing bystander cell lines are disclosed in U.S. Pat. Nos.
  • GM-CSF Granulocyte-macrophage colony stimulating factor
  • the CDN compositions are administered in conjunction with one or more vaccines intended to stimulate an immune response to one or more predetermined antigens.
  • target antigens examples include target antigens that may find use in the invention are listed in the following table.
  • the target antigen may also be a fragment or fusion polypeptide comprising an immunologically active portion of the antigens listed in the table. This list is not meant to be limiting.
  • Antigen Reference Tumor antigens Mesothelin GenBank Acc. No. NM_005823; U40434; NM_013404; BC003512 (see also, e.g., Hassan, et al. (2004) Clin. Cancer Res. 10:3937-3942; Muminova, et al. (2004) BMC Cancer 4:19; Iacobuzio-Donahue, et al. (2003) Cancer Res. 63:8614-8622).
  • Wilms' tumor-1 WT-1 isoform A GenBank Acc. associated protein Nos. NM_000378; NP_000369).
  • Wt-1 including WT-1 isoform B (GenBank Acc.
  • MHC class I See, e.g., Groh, et al. (2005) Proc. chain-related protein A Natl. Acad. Sci. USA 102:6461- (MICA); MHC class I 6466; GenBank Acc. Nos. NM_000247; chain-related protein A BC 016929; AY750850; (MICB). NM_005931. Gastrin and peptides Harris, et al. (2004) Cancer Res.
  • PAP phosphatase
  • PSA antigen
  • PSM PSM
  • Chan et al. (1999) Prostate 41:99- PSMA. 109; Ito, et al. (2005) Cancer 103:242-250; Schmittgen, et al. (2003) Int. J. Cancer 107:323-329; Millon, et al. (1999) Eur. Urol. 36:278-285.
  • Six-transmembrane See, e.g., Machlenkin, et al. (2005) epithelial antigen of Cancer Res. 65:6435-6442; prostate (STEAP).
  • Prostate carcinoma See, e.g., Machlenkin, et al. (2005) tumor antigen-1 Cancer Res. 65:6435-6442; (PCTA-1).
  • Prostate See, e.g., Machlenkin, et al. (2005) tumor-inducing gene-1 Cancer Res. 65:6435-6442).
  • PTI-1 Prostate-specific gene See, e.g., Machlenkin, et al. (2005) with homology to Cancer Res. 65:6435-6442).
  • G protein-coupled receptor G protein-coupled receptor.
  • Prostase an antrogen See, e.g., Machlenkin, et al. (2005) Cancer regulated serine Res. 65:6435-6442; GenBank Acc. No. protease). BC096178; BC096176; BC096175. Proteinase 3. GenBank Acc. No. X55668. Cancer-testis antigens, GenBank Acc. No. NM_001327 e.g., NY-ESO-1; SCP- (NY-ESO-1) (see also, e.g., Li, et al. 1; SSX-1; SSX-2; SSX- (2005) Clin. Cancer Res.
  • N-Acetylglucosaminyl- Dosaka-Akita, et al. (2004) Clin. tranferase V (GnT-V). Cancer Res. 10:1773-1779.
  • Biol. 12:1687-1697 Ras, e.g., wild type ras, GenBank Acc. Nos. P01112; ras with mutations at P01116; M54969; M54968; P01111; codon 12, 13, 59, or 61, P01112; K00654. See also, e.g., e.g., mutations G12C; GenBank Acc. Nos. M26261; G12D; G12R; G125; M34904; 1(01519; K01520; BC006499; G12V; G13D; A59T; NM_006270; NM_002890; Q61H.
  • BRAF an isoform of Tannapfel, et al. (2005) Am. J. Clin. RAF). Pathol. 123:256-2601; Tsao and Sober (2005) Dermatol. Clin. 23:323-333.
  • Melanoma antigens GenBank Acc. No. NM_206956; including HST-2 NM_206955; NM_206954; melanoma cell NM_206953; NM_006115; antigens.
  • GenBank Acc. No. AB028869; U75285 see also, e.g., Tsuruma, et al. (2004) J. Translational Med. 2:19 (11 pages); Pisarev, et al. (2003) Clin. Cancer Res. 9:6523-6533; Siegel, et al. (2003) Br. J. Haematol. 122:911-914; Andersen, et al. (2002) Histol. Histopathol.
  • MDM-2 NM_002392; NM_006878 see also, e.g., Mayo, et al. (1997) Cancer Res. 57:5013-5016; Demidenko and Blagosklonny (2004) Cancer Res. 64:3653-3660).
  • Methyl-CpG-binding Muller et al. (2003) Br. J. Cancer proteins (MeCP2; 89:1934-1939; Fang, et al. (2004) MBD2).
  • Histone deacetylases Waltregny, et al. (2004) Eur. J. (HDAC), e.g., HDAC5. Histochem. 48:273-290; Scanlan, et al. (2002) Cancer Res. 62:4041-4047. Cyclophilin B (Cyp-B). Tamura, et al. (2001) Jpn. J. Cancer Res. 92:762-767. CA 15-3; CA 27.29. Clinton, et al. (2003) Biomed. Sci. Instrum. 39:408-414. Heat shock protein Faure, et al. (2004) Int. J. Cancer Hsp70. 108:863-870. GAGE/PAGE family, Brinkmann, et al.
  • Cancer Res. 10:4307-4313 Carcinoembryonic GenBank Acc. No. M29540; E03352; antigen (CEA), X98311; M17303 (see also, CAP1-6D enhancer e.g., Zaremba (1997) Cancer Res. agonist peptide. 57:4570-4577; Sarobe, et al. (2004) Curr. Cancer Drug Targets 4:443-454; Tsang, et al. (1997) Clin. Cancer Res. 3:2439-2449; Fong, et al. (2001) Proc. Natl. Acad. Sci. USA 98:8809-8814).
  • Annexin e.g., Zimmerman, et al. (2004) Virchows Annexin II. Arch. 445:368-374.
  • AH003567; U31798; U31799;U31807; U31799 see also, e.g., Bronte, et al. (2000) Cancer Res. 60:253-258).
  • TARP See, e.g., Clifton, et al. (2004) Proc. Natl. Acad. Sci. USA 101:10166- 10171; Virok, et al. (2005) Infection Immunity 73:1939-1946.
  • Tyrosinase-related GenBank Acc. No. NM_001922. proteins 1 and 2 (TRP-1 see also, e.g., Bronte, et al. (2000) and TRP-2). Cancer Res. 60:253-258).
  • NM_010139 (Mouse); Genbank Accession No. AB038986 (Chicken, partial sequence); GenBank Accession Nos. NP_004422, AAH37166, and AAA53375 (human); GenBank Accession Nos. NP_034269 (mouse), AAH06954 (mouse), XP_345597 (rat), and BAB63910 (chicken).
  • EGFRvIII See, e.g., WO/2012/068360 Francisella tularensis antigens Francisella tularensis Complete genome of subspecies A and B. Schu S4 (GenBank Acc. No. AJ749949); of subspecies Schu 4 (GenBank Acc. No. NC_006570).
  • Antigenic components of F. tularensis include, e.g., 80 antigens, including 10 kDa and 60 kDa chaperonins (Havlasova, et al. (2002) Proteomics 2:857-86), nucleoside diphosphate kinase, isocitrate dehydrogenase, RNA-binding protein Hfq, the chaperone ClpB (Havlasova, et al.
  • MSP2 e.g., protein 1(MSP1); GenBank Acc. No. X96399; 195A; BVp42. X96397).
  • MSP1 see, e.g., GenBank Acc. No. X03371).
  • RESA see, e.g., GenBank Acc. No. X05181; X05182).
  • Apical membrane See, e.g. ,Gupta, et al. (2005) Protein antigen 1 (AMA1). Expr. Purif. 41:186-198.
  • AMA1 see, e.g., GenBank Acc. No. A ⁇ 13; AJ494905; AJ490565).
  • Viruses and viral antigens Hepatitis A GenBank Acc. Nos., e.g., NC_001489; AY644670; X83302; K02990; M14707.
  • Hepatitis B Complete genome see, e.g., GenBank Acc. Nos.
  • HTLV lymphotropic AIDS Res. Hum. Retrovirus 21:28- virus 42; Bhigjee, et al. (1999) AIDS Res. (HTLV) types Hum. Restrovirus 15:1229-1233; I and II, Vandamme, et al. (1998) J. Virol. including the 72:4327-4340; Vallejo, et al. (1996) HTLV type I J. Acquir. Immune Defic. Syndr. subtypes Hum. Retrovirol. 13:384-391. Cosmopolitan, HTLV type I (see, e.g., GenBank Acc. Central Nos. AY563954; AY563953.
  • African, and HTLV type II see, e.g., GenBank Austro-Melanesian, Acc. Nos. L03561; Y13051; and AF139382.
  • the HTLV type II subtypes Iia, Iib, Iic, and Iid. Coronaviridae See, e.g., Brian and Baric (2005) Curr. including Top. Microbiol. Immunol. Coronaviruses, 287:1-30; Gonzalez, et al. (2003) Arch. such as Virol. 148:2207-2235; Smits, SARS-coronavirus et al. (2003) J. Virol. 77:9567-9577; (SARS-CoV), and Jamieson, et al.
  • Coxsackie virus B See, e.g., Ahn, et al. (2005) J. Med. including Virol. 75:290-294; Patel, et al. subtypes 1-6. (2004) J. Virol. Methods 120:167-172; Rezig, et al. (2004) J. Med. Virol. 72:268-274.
  • GenBank Acc. No. X05690 GenBank Acc. No. X05690.
  • Human enteroviruses See, e.g., Oberste, et al. (2004) J. including, e.g., human Virol. 78:855-867.
  • Human enterovirus A enterovirus A GenBank Acc. (HEV-A, Nos. NC_001612); human CAV2 to CAV8, enterovirus B (NC_001472); human CAV10, CAV12, enterovirus C (NC_001428); CAV14, CAV16, and human enterovirus D (NC_001430).
  • EV71 and also Simian enterovirus A
  • GenBank including HEV-B Acc. No. NC_003988 Simian enterovirus A
  • Polioviruses See, e.g., He, et al. (2003) J. Virol. including 77:4827-4835; Hahsido, et al. PV1, PV2, and PV3. (1999) Microbiol. Immunol. 43:73-77.
  • GenBank Acc. No. AJ132961 type 1
  • AY278550 type 2
  • X04468 type 3
  • Viral encephalitides See, e.g., Hoke (2005) Mil. Med.
  • viruses including 170:92-105; Estrada-Franco, et al. equine encephalitis, (2004) Emerg. Infect. Dis. 10:2113-2121; Venezuelan equine Das, et al. (2004) Antiviral encephalitis (VEE) Res. 64:85-92; Aguilar, et al. (2004) (including Emerg. Infect. Dis. 10:880-888; subtypes IA, Weaver, et al. (2004) Arch. Virol. IB, IC, ID, IIIC, Suppl. 18:43-64; Weaver, et al. IIID), Eastern equine (2004) Annu. Rev. Entomol. 49:141-174.
  • EEE Eastern equine encephalitis Western equine
  • NC_003899 Eastern equine encephalitis Western equine
  • WEE encephalitis
  • NC_003908 Western equine St. Louis encephalitis, encephalitis
  • NC_003908 Western equine St. Louis encephalitis, encephalitis
  • NC_003908 Western equine St. Louis encephalitis, encephalitis
  • Murray Valley Australian
  • encephalitis Japanese encephalitis
  • tick-born encephalitis Human herpesviruses, See, e.g., Studahl, et al. (2000) including Scand. J. Infect. Dis. 32:237-248; cytomegalovirus Padilla, et al. (2003) J. Med. Virol. (CMV), Epstein-Barr 70 (Suppl.
  • herpesvirus-1 S103-S110; virus (EBV), human Jainkittivong and Lley (1998) herpesvirus-1 Oral Surg. Oral Med. 85:399-403.
  • HHV-1 GenBank Nos. NC_001806 HHV-2, HHV-3, (herpesvirus 1); NC_001798 HHV-4, HHV-5, (herpesvirus 2); X04370 and HHV-6, HHV-7, NC_001348 (herpesvirus 3); HHV-8, herpes NC_001345 (herpesvirus 4); NC_001347 B virus, (herpesvirus 5); X83413 herpes simplex virus and NC_000898 (herpesvirus 6); types 1 and 2 (HSV-1, NC_001716 (herpesvirus 7).
  • HSV-2 varicella Human herpesviruses types 6 and 7 zoster virus
  • VZV varicella Human herpesviruses types 6 and 7 zoster virus
  • HHV-6; HHV-7) are disclosed by, e.g., Padilla, et al. (2003) J. Med. Virol. 70 (Suppl. 1)S103-S110.
  • Human herpesvirus 8 (HHV-8), including subtypes A-E are disclosed in, e.g., Treurnicht, et al. (2002) J. Med. Virul. 66:235-240.
  • HIV-1 including See, e.g., Smith, et al. (1998) J. Med. group M Virol. 56:264-268. See also, (including subtypes e.g., GenBank Acc. Nos.
  • Epstein-Barr virus See, e.g., Peh, et al. (2002) Pathology (EBV), including 34:446-450.
  • strain B95-8 (GenBank Acc. No. V01555).
  • Reovirus including See, e.g., Barthold, et al. (1993) Lab. serotypes and Anim. Sci. 43:425-430; Roner, strains 1, et al. (1995) Proc. Natl. Acad. Sci. 2, and 3, type 1 Lang, USA 92:12362-12366; Kedl, et al. type 2 Jones, (1995) J. Virol. 69:552-559. GenBank and type 3 Acc. No. 1(02739 (sigma-3 Dearing. gene surface protein). Cytomegalovirus See, e.g., Chern, et al. (1998) J. Infect.
  • CMV CMV subtypes Dis. 178:1149-1153; Vilas include CMV Boas, et al. (2003) J. Med. Virol. subtypes 71:404-407; Trincado, et al. (2000) I-VII. J. Med. Virol. 61:481-487.
  • Rhinovirus, Human rhinovirus 2 GenBank including Acc. No. X02316); Human all serotypes.
  • rhinovirus B GenBank Acc. No. NC_001490
  • Human rhinovirus 89 GenBank Acc. No. NC_001617
  • Human rhinovirus 39 GenBank Acc. No. AY751783
  • Adenovirus AY803294; NC_004001; AC_000019; including AC_000018; AC_000017; all serotypes. AC_000015; AC_000008; AC_000007; AC_000006; AC_000005; AY737798; AY737797;NC_003266; NC_002067; AY594256; AY594254; AY875648; AJ854486; AY163756; AY594255; AY594253; NC_001460; NC_001405; AY598970; AY458656; AY487947; NC_001454; AF534906; AY45969; AY128640; L19443; AY339865; AF532578.
  • Arenaviruses Junin virus, segment S (GenBank including Acc. No. NC_005081); Junin virus, lymphocytic segment L (GenBank Acc. No. NC_005080). choriomeningitis (LCM) virus, Lassa virus, Junin virus, and Machupo virus. Rabies virus. See, e.g., GenBank Acc. Nos. NC_001542; AY956319; AY705373; AF499686; AB128149; AB085828; AB009663. Arboviruses, Dengue virus type 1 (see, e.g., including GenBank Acc. Nos. AB195673; West Nile virus, AY762084).
  • Dengue virus type 2 Dengue (see, e.g., GenBank Acc. Nos. viruses 1 to 4, NC_001474; AY702040; AY702039; Colorado AY702037).
  • Dengue virus type tick fever 3 (see, e.g., GenBank Acc. Nos. virus, Sindbis AY923865; AT858043).
  • Dengue virus, virus type 4 (see, e.g., GenBank Acc. Togaviraidae, Nos. AY947539; AY947539; Flaviviridae, AF326573).
  • Sindbis virus see, e.g., Bunyaviridae, GenBank Acc. Nos.
  • GenBank orthomyxoviridae Acc. Nos. NC_001563; AY603654
  • Poxvirus including Viriola virus (see, e.g., GenBank orthopoxvirus (variola Acc. Nos. NC_001611; Y16780; virus, monkeypox X72086; X69198). virus, vaccinia virus, cowpox virus), yatapoxvirus (tanapox virus, Yaba monkey tumor virus), parapoxvirus, and molluscipoxvirus.
  • Hantaviruses See, e.g., Elgh, et al. (1997) J. Clin. including Microbiol. 35:1122-1130; serotypes Hantaan Sjolander, et al. (2002) Epidemiol. (HTN), Infect. 128:99-103; Zeier, et al. Seoul (SEO), (2005) Virus Genes 30:157-180.
  • Flaviviruses See, e.g., Mukhopadhyay, et al. (2005) including Nature Rev. Microbiol. 3:13- Dengue virus, 22. GenBank Acc. Nos NC_001474 Japanese and AY702040 (Dengue). encephalitis virus, GenBank Acc. Nos. NC_001563 West Nile and AY603654. virus, and yellow fever virus. Measles virus. See, e.g., GenBank Acc. Nos. AB040874 and AY486084.
  • Human Human parainfluenza virus 2 (see, parainfluenzaviruses e.g., GenBank Acc. Nos. (HPV), AB176531; NC003443). Human including HPV parainfluenza virus 3 (see, e.g., types 1-56. GenBank Acc. No. NC_001796).
  • Influenza virus, Influenza nucleocapsid (see, e.g., including GenBank Acc. No. AY626145).
  • influenza Influenza hemagglutinin see, e.g., virus types GenBank Acc. Nos. AY627885; A, B, and C. AY555153).
  • Influenza neuraminidase see, e.g., GenBank Acc.
  • Influenza matrix protein 2 (see, e.g., GenBank Acc. Nos. AY626144(. Influenza basic protein 1 (see, e.g., GenBank Acc. No. AY627897). Influenza polymerase acid protein (see, e.g., GenBank Acc. No. AY627896). Influenza nucleoprotein (see, e.g., GenBank Acc. Nno. AY627895). Influenza A virus Hemagglutinin of H1N1 (GenBank Acc. subtypes, e.g., swine No. S67220).
  • Influenza A viruses SIV: H1N1 virus matrix protein (GenBank Acc. influenzaA and swine No. AY700216). Influenza virus influenza virus. A H5H1 nucleoprotein (GenBank Acc. No. AY646426). H1N1 haemagglutinin (GenBank Acc. No. D00837). See also, GenBank Acc. Nos. BD006058; BD006055; BD006052. See also, e.g., Wentworth, et al. (1994) J. Virol. 68:2051-2058; Wells, et al. (1991) J.A.M.A. 265:478-481.
  • Respiratory syncytial Respiratory syncytial virus (RSV) virus including (see, e.g., GenBank Acc. Nos. subgroup A and AY353550; NC_001803; NC001781).
  • human-RVV AY548957 human rotavirus A strain reassortments. major inner capsid protein (see, e.g., GenBank Acc. No. AY601554).
  • Polyomavirus See, e.g., Engels, et al. (2004) J. Infect. including simian Dis. 190:2065-2069; Vilchez virus 40 (SV40), JC and Butel (2004) Clin. Microbiol. virus (JCV) and BK Rev. 17:495-508; Shivapurkar, et virus (BKV). al. (2004) Cancer Res. 64:3757-3760; Carbone, et al. (2003) Oncogene 2:5173-5180; Barbanti-Brodano, et al.
  • Virology 318:1-9) (5V40 complete genome in, e.g., GenBank Acc. Nos. NC_001669; AF168994; AY271817; AY271816; AY120890; AF345344; AF332562).
  • Coltiviruses Attoui, et al. (1998) J. Gen. Virol. including 79:2481-2489. Segments of Eyach Colorado tick fever virus (see, e.g., GenBank Acc. virus, Eyach virus. Nos.
  • Calciviruses including Snow Mountain virus (see, e.g., GenBank the genogroups Acc. No. AY134748). Norwalk, Snow Mountain group (SMA), and Saaporo. Parvoviridae, See, e.g., Brown (2004) Dev. Biol.
  • Other organisms for which suitable antigens are known in the art include, but are not limited to, Chlamydia trachomatis, Streptococcus pyogenes (Group A Strep), Streptococcus agalactia (Group B Strep), Streptococcus pneumonia, Staphylococcus aureus, Escherichia coli, Haemophilus influenzae, Neisseria meningitidis, Neisseria gonorrheae, Vibrio cholerae, Salmonella species (including typhi, typhimurium ), enterica (including Helicobactor pylori Shigella flexneri and other Group D shigella species), Burkholderia mallei, Burkholderia pseudomallei, Klebsiella pneumonia, Clostridium species (including C. difficile ), Vibrio parahaemolyticus and V. vulnificus . This list is not meant to be limiting.
  • pharmaceutical refers to a chemical substance intended for use in the cure, treatment, or prevention of disease and which is subject to an approval process by the U.S. Food and Drug Administration (or a non-U.S. equivalent thereof) as a prescription or over-the-counter drug product. Details on techniques for formulation and administration of such compositions may be found in Remington, The Science and Practice of Pharmacy 21 st Edition (Mack Publishing Co., Easton, Pa.) and Nielloud and Marti-Mestres, Pharmaceutical Emulsions and Suspensions: 2 nd Edition (Marcel Dekker, Inc, New York).
  • the pharmaceutical compositions may be administered by a variety of means including orally, parenterally, by inhalation spray, topically, or rectally in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used here includes but is not limited to subcutaneous, intravenous, intramuscular, intraarterial, intradermal, intrathecal and epidural injections with a variety of infusion techniques.
  • Intraarterial and intravenous injection as used herein includes administration through catheters. Administration via intracoronary stents and intracoronary reservoirs is also contemplated.
  • Intra-tumoral administration of the compounds of the present invention may directly activate locally infiltrating DC, directly promote tumor cell apoptosis or sensitize tumor cells to cytotoxic agents.
  • oral as used herein includes, but is not limited to oral ingestion, or delivery by a sublingual or buccal route.
  • Oral administration includes fluid drinks, energy bars, as well as pill formulations.
  • compositions may be in any form suitable for the intended method of administration.
  • tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • Tablets containing a drug compound in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents; such as magnesium stearate, stearic acid or talc.
  • Tablets may be uncoated, or may be coated by known techniques including enteric coating, colonic coating, or microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and/or provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules where the drug compound is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example calcium phosphate or kaolin
  • an oil medium such as peanut oil, liquid paraffin or olive oil.
  • compositions may be formulated as aqueous suspensions in admixture with excipients suitable for the manufacture of aqueous-suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate).
  • a suspending agent
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or a mineral oil such as liquid paraffin.
  • the oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules of the disclosure suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives.
  • a dispersing or wetting agent and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
  • the pharmaceutical compositions of the disclosure may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these.
  • Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • sweetening agents such as glycerol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • compositions of the disclosure may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid
  • a time-release formulation intended for oral administration to humans may contain approximately 20 to 500 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions. It is preferred that the pharmaceutical composition be prepared which provides easily measurable amounts for administration.
  • an effective amount to be administered systemically is about 0.1 mg/kg to about 100 mg/kg and depends upon a number of factors including, for example, the age and weight of the subject (e.g., a mammal such as a human), the precise condition requiring treatment and its severity, the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs which have previously been administered; and the severity of the particular condition undergoing therapy, as is well understood by those skilled in the art.
  • formulations of the disclosure suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient, as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the pharmaceutical compositions may also be administered as a bolus, electuary or paste.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropyl ethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface active or dispersing agent.
  • Molded tablets may be made in a suitable machine using a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methylcellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric or colonic coating to provide release in parts of the gut other than the stomach. This is particularly advantageous with the compounds of formula 1 when such compounds are susceptible to acid hydrolysis.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • pharmaceutically acceptable salts include, but are not limited to: acetate, pyridine, ammonium, piperazine, diethylamine, nicotinamide, formic, urea, sodium, potassium, calcium, magnesium, zinc, lithium, cinnamic, methylamino, methanesulfonic, picric, tartaric, triethylamino, dimethylamino, and tris(hydoxymethyl)aminomethane. Additional pharmaceutically acceptable salts are known to those skilled in the art.
  • An effective amount for a particular patient may vary depending on factors such as the condition being treated, the overall health of the patient, the route and dose of administration and the severity of side effects.
  • Guidance for methods of treatment and diagnosis is available (see, e.g., Maynard, et al. (1996) A Handbook of SOPs for Good Clinical Practice, Interpharm Press, Boca Raton, Fla.; Dent (2001) Good Laboratory and Good Clinical Practice, Urch Publ., London, UK).
  • an effective amount may be given in one dose, but is not restricted to one dose.
  • the administration can be two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, or more, administrations of pharmaceutical composition.
  • the administrations can be spaced by time intervals of one minute, two minutes, three, four, five, six, seven, eight, nine, ten, or more minutes, by intervals of about one hour, two hours, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, and so on.
  • the term “about” means plus or minus any time interval within 30 minutes.
  • the administrations can also be spaced by time intervals of one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, and combinations thereof.
  • the invention is not limited to dosing intervals that are spaced equally in time, but encompass doses at non-equal intervals.
  • a dosing schedule of, for example, once/week, twice/week, three times/week, four times/week, five times/week, six times/week, seven times/week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, and the like, is available for the invention.
  • the dosing schedules encompass dosing for a total period of time of, for example, one week, two weeks, three weeks, four weeks, five weeks, six weeks, two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, and twelve months.
  • the cycle can be repeated about, e.g., every seven days; every 14 days; every 21 days; every 28 days; every 35 days; 42 days; every 49 days; every 56 days; every 63 days; every 70 days; and the like.
  • An interval of non dosing can occur between a cycle, where the interval can be about, e.g., seven days; 14 days; 21 days; 28 days; 35 days; 42 days; 49 days; 56 days; 63 days; 70 days; and the like.
  • the term “about” means plus or minus one day, plus or minus two days, plus or minus three days, plus or minus four days, plus or minus five days, plus or minus six days, or plus or minus seven days.
  • compositions of the present invention are preferably formulated as pharmaceutical compositions for parenteral or enteral delivery.
  • a typical pharmaceutical composition for administration to an animal comprises a pharmaceutically acceptable vehicle such as aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like. See, e.g., Remington's Pharmaceutical Sciences, 15th Ed., Easton ed., Mack Publishing Co., pp 1405-1412 and 1461-1487 (1975); The National Formulary XIV, 14th Ed., American Pharmaceutical Association, Washington, D.C. (1975).
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil and injectable organic esters such as ethyloleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles such as sodium chloride, Ringer's dextrose, etc.
  • Intravenous vehicles include fluid and nutrient replenishers.
  • Preservatives include antimicrobial agents, anti-oxidants, chelating agents and inert gases. The pH and exact concentration of the various components the pharmaceutical composition are adjusted according to routine skills in the art.
  • heterologous boosting Repeated administrations of a particular vaccine (homologous boosting) have proven effective for boosting humoral responses. Such an approach may not be effective at boosting cellular immunity because prior immunity to the vector tends to impair robust antigen presentation and the generation of appropriate inflammatory signals.
  • One approach to circumvent this problem has been the sequential administration of vaccines that use different antigen-delivery systems (heterologous boosting).
  • at least one prime or boost delivery comprises delivery of the inactivated tumor cell/cyclic purine dinucleotide compositions described herein.
  • the heterologous arm of the regimen may comprise delivery of antigen using one or more of the following strategies:
  • a prime vaccine and a boost vaccine can be administered by any one or combination of the following routes.
  • the prime vaccine and boost vaccine are administered by the same route.
  • the prime vaccine and boost vaccine are administered by different routes.
  • the term “different routes” encompasses, but is not limited to, different sites on the body, for example, a site that is oral, non-oral, enteral, parenteral, rectal, intranode (lymph node), intravenous, arterial, subcutaneous, intramuscular, intratumor, peritumor, intratumor, infusion, mucosal, nasal, in the cerebrospinal space or cerebrospinal fluid, and so on, as well as by different modes, for example, oral, intravenous, and intramuscular.
  • an effective amount of a prime or boost vaccine may be given in one dose, but is not restricted to one dose.
  • the administration can be two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, or more, administrations of the vaccine.
  • the administrations can be spaced by time intervals of one minute, two minutes, three, four, five, six, seven, eight, nine, ten, or more minutes, by intervals of about one hour, two hours, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, and so on.
  • the term “about” means plus or minus any time interval within 30 minutes.
  • the administrations can also be spaced by time intervals of one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, and combinations thereof.
  • the invention is not limited to dosing intervals that are spaced equally in time, but encompass doses at non-equal intervals, such as a priming schedule consisting of administration at 1 day, 4 days, 7 days, and 25 days, just to provide a non-limiting example.
  • Anhydrous solvents and reagents suitable for solution phase oligonucleotide synthesis were purchased and handled under dry argon or nitrogen using anhydrous technique.
  • Amidite coupling reactions and cyclizations were carried out in anhydrous acetonitrile or pyridine under dry argon or nitrogen.
  • the starting materials for all reactions in dry pyridine were dried by concentration (three times) from pyridine.
  • Preparative silica gel flash chromatography was carried out using Fluka 60A high-purity grade or Merck Grade 9385 silica using gradients of methanol in dichloromethane.
  • Analytical HPLC was carried out on a Varian ProStar 210 HPLC system with a ProStar 330 photodiode array detector monitoring at 254 nm using a either a Varian Microsorb 10 micron C18 250 ⁇ 4.6 mm or a Varian 3micronC18 100 ⁇ 4.6 mm column and gradients of 10 mM TEAA and acetonitrile.
  • Preparative HPLC was carried out on a Shimadzu preparative LC20-AP HPLC system, equipped with a SPD-20A UV/Vis detector monitoring at 254 nm on a Varian Microsorb 60-8 C-18 41.6 ⁇ 250 mm column using gradients of 10 mM TEAA and acetonitrile at a flow rate of 50 ml/min.
  • the 31P chemical shifts were referenced using the “unified scale” to the absolute 1H frequency of 0 ppm.
  • Some of the 1H and 31P spectra were acquired on a JEOL ECX-400 NMR spectrometer operating at 400 MHz for 1H and 162 MHz for 31P.
  • the gradient COSY spectrum was acquired in absolute value mode using 2048 data points in the direct dimension and 256 time points in the indirect dimension. Both dimensions were apodized using sinebell squared functions. The indirect dimension was zero filled to give a final matrix size of 2048 ⁇ 2048 points and a resolution of 3.91 Hz/data point in both dimensions.
  • cyclic[G(2′,5′)pA(3′,5′)p] ML-cGAMP.
  • ML-cGAMP (structure 23 in FIG. 2 c ) was prepared enzymatically from cellular cGAS and purified by prep HPLC.
  • reaction mix was then poured into 350 ml water containing 0.1 g NaHSO 3 and stirred for 5 minutes at room temperature. 2 g of NaHCO 3 was slowly added with stirring, then poured into a separatory funnel and extracted with 400 ml 1:1 ethyl acetate:diethylether. The aqueous layer was extracted again with 400 ml 1:1 ethyl acetate:diethylether, and the organic layers were combined, dried over sodium sulfate, and concentrated under reduced pressure to yield 0.75 g of a mixture containing 7a, the fully-protected cyclic-[G(2′,5′)pG(3′,5′)p].
  • the neutralized solution was desalted on a Waters C-18 Sep-Pak and the product eluted with CH3CN/10 mM aqueous triethylammonium acetate (1:1).
  • the CH 3 CN was evaporated under reduced pressure and the remaining aqueous solution was frozen and lyophilized overnight.
  • Multiple evaporations from methanol (3 ⁇ 3 ml) and a final evaporation from 50% acetonitrile in methanol (1 ⁇ 3 ml) gave 29.3 mg of cyclic-[G(2′,5′)pG(3′,5′)p] (9a) as the bis-triethylammonium salt.
  • the HPLC retention time of 9a is 7.25 min compared to 9.3 min for c-di-GMP using a gradient of 2 to 20% CH3CN in 10 mM triethylammonium acetate over 20 min on a C-18 column (3 micron, 100 ⁇ 4.6 mm, 0.6 ml/min.)
  • the HRMS (FT-ICR) confirmed the expected elemental formula: [M-H] ⁇ calcd for C 20 H 23 N 10 O 14 P 2 689.0876; found 689.0874.
  • the 31-P NMR of 9a showed two peaks (integrating 1:1) at 2.03 and -0.95 ppm consistent with a 2′,5′/3′,5′ mixed linkage (both c[G(3′,5′)pG(3′,5′)p] and c[G(2′,5′)pG(2′,5′)p], for example, would give only one 31-P NMR signal due to symmetry).
  • Direct evidence for the regiochemistry of the phosphodiester linkages was obtained by 1H-1H COSY in combination with phosphorous decoupling experiments, and by 1 H- 31 P HMBC two-dimensional NMR ( FIGS. 3 b and 3 c ).
  • the anomeric (H-1) protons appear as overlapping doublet of doublets (or triplet) at 5.82 ppm.
  • the “A” designation was given to the downfield anomeric (H-1) proton and “B” to the anomeric proton slightly upfield of that.
  • a 1H-1H COSY experiment ( FIG. 3 b ) allowed assignment of H-2A (4.96 ppm), H-3A (4.31 ppm), as well as H-2B (4.67 ppm) and H-3B (4.84 ppm).
  • N 6 -benzoyl-5′-O-(4,4′-dimethoxytrityl)-3′-O-tert-butyldimethylsilyl-2′-O-[(2-cyanoethyl)-N,N-diisopropylaminophinyl]adenosine (14, 6.4 g, 6.6 mmole) was dissolved in 40 ml anhydrous acetonitrile and dried by three co-evaporations with 40 ml anhydrous acetonitrile, the last time leaving 20 ml. Ten 3 ⁇ molecular sieves were added and the solution stored under argon until use.
  • reaction mix was then poured into 700 ml water containing 20 g NaHCO 3 and stirred for 5 minutes at room temperature, then poured into a separatory funnel and extracted with 800 ml 1:1 ethyl acetate:diethyl ether. The aqueous layer was extracted again with 600 ml 1:1 ethyl acetate:diethyl ether. The organic layers were combined and concentrated under reduced pressure to yield approximately 11 g of an oil containing diastereoisomers 17b and 17c.
  • the lyophilized crude mixture containing 18b and 18c was taken up in approximately 50 ml of CH3CN/10 mM aqueous triethylammonium acetate (60/40). After 0.45 micron PTFE filtration, 4-5 ml sample portions were applied to a C-18 Dynamax column (40 ⁇ 250 mm). Elution was performed with a gradient of acetonitrile and 10 mM aqueous triethylammonium acetate (30% to 50% CH 3 CN over 20 minutes at 50 ml/min flow).
  • the neutralized solution was desalted on a Waters C-18 Sep-Pak and the product eluted with CH3CN/10 mM aqueous triethylammonium acetate (1:4).
  • the CH 3 CN was evaporated under reduced pressure and the remaining aqueous solution was frozen and lyophilized. Multiple rounds of lyophilization from water gave 54 mg (76%) of dithio-(Rp,Rp)-[cyclic-A(2′,5′)pA(3′,5′)p] (19b) as the bis-triethylammonium salt.
  • TEA.3HF (1 mL, 6.1 mmol) was added to 18b (41 mg, 0.04 mmol) in a flask equipped with a vent needle and the mixture stirred at 45° C.
  • the reaction progress was monitored by LC and upon consumption of the starting material and mono-TBS analogs ( ⁇ 2 hr) the mixture was cooled to room temperature.
  • the mixture was slowly pipetted into a solution of 1 M TEAB (4.9 mL) and TEA (1.6 mL) at 0° C. and a slightly basic pH was confirmed by pH paper.
  • the ML-RR-CDA bis-TEA salt (19b) is readily converted to the pharmaceutically acceptable sodium salt (21) by ion exchange as described below.
  • ML-RR-CDA.2Na + (21).
  • BT AG® 50W-X2 Resin 100-200 Mesh, hydrogen form (100 mg) was slurry packed with DI water into a Bio-spin® column. The excess DI water was drained via gravity. 3 bed volumes of 1 M NaOH (1 mL) was passed through the column via gravity followed by 5 bed volumes of DI water (2 mL). After draining the excess DI water via gravity a solution of ML-RR-CDA.2TEA (19b, 10 mg) in DI water (1 mL) was loaded onto the column. The column was eluted with 5 bed volumes of DI water (2 mL), fractions were collected and checked for UV activity via TLC plate and UV lamp.
  • FIG. 2 c Compound 10 ( FIG. 2 c ) was synthesized analogously to ML-CDG following the procedures of ML-CDG series experimental with the following modifications ( FIG. 2 a ): n) myristic anhydride, DMF.
  • Compound 22 was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications ( FIG. 2 b ): n) BT AG® 50W-X2 Resin 100-200 Mesh, hydrogen form, 1 M NH 4 OH.
  • FIG. 2 c Compound 20 ( FIG. 2 c ) was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications ( FIG. 2 b ): d) pyr, 4; n) obtained as the TEA salt, no ion exchange was needed.
  • FIG. 4-6 Examples of derivatives finding use in the present invention are depicted in FIG. 4-6 .
  • THP1-BlueTM ISG cells a human monocyte cell line transfected with an IRF-inducible secreted embryonic alkaline phosphatase reporter gene (Invivogen) which express alkaline phosphatase under the control of a promoter comprised of five IFN-stimulated response elements
  • CDG cyclic [G(3′,5′)pG(3′,5′)p]
  • ML-CDG cyclic [G(2′,5′)pG(3′,5′)p]
  • cyclic [G(2′,5′)pG(3′,5′)p] (ML-CDG) induced significantly higher levels of IFN- ⁇ than cyclic [G(3′,5′)pG(3′,5′)p] across a broad range of time points.
  • ISD Interferon Stimulating DNA
  • TLR independent Stetston, D. B. et. al. Immunity 24, 93-103, January 2006
  • Poly I:C
  • IFNA2 interferon alpha 2
  • IFNB1 interferon beta 1
  • IFNG type II cytokine gene interferon gamma
  • gene expression of interferon alpha 2 was significantly higher for cyclic [G(2′,5′)pG(3′,5′)p] at 5 ⁇ M than for cyclic [G(3′,5′)pG(3′,5′)p] at 5 ⁇ M across all four donors.
  • gene expression of interferon beta 1 was significantly higher for cyclic [G(2′,5′)pG(3′,5′)p] at 5 ⁇ M than for cyclic [G(3′,5′)pG(3′,5′)p] at 5 ⁇ M in all four donors.
  • the levels of secreted IFN- ⁇ induced in primary human PBMCs by cyclic [G(2′,5′)pG(3′,5′)p] at 5 ⁇ M are higher than cyclic [G(3′,5′)pG(3′,5′)p] at the same or lower dose across all four donors.
  • cyclic [G(3′,5′)pG(3′,5′)p] at the same or lower dose across all four donors are higher than cyclic [G(3′,5′)pG(3′,5′)p] at the same or lower dose across all four donors.
  • FIG. 9( b ) levels of IFN-(3, as assessed by ELISA, for cyclic [G(2′,5′)pG(3′,5′)p] at 5 ⁇ M were also higher than with cyclic [G(3′,5′)pG(3′,5′)p]induced levels, as well as for the ISD and Poly I:C controls in all four donors.
  • FIG. 9( c ) demonstrates a similar finding for secretion of IFN- ⁇ , as assessed by CBA.
  • cyclic [G(2′,5′)pG(3′,5′)p] induced higher levels of IFN- ⁇ than cyclic [G(3′,5′)pG(3′,5′)p] at the same doses, and higher levels than the ISD and Poly I:C controls across all four donors.
  • These data demonstrate the increased potency of cyclic [G(2′,5′)pG(3′,5′)p] compared to cyclic [G(3′,5′)pG(3′,5′)p] to stimulate type I and II IFN production, critical to the induction of innate immunity across a broad sampling of human donors.
  • THP1-Blue cells a human monocyte cell line transfected with an IRF-inducible secreted embryonic alkaline phosphatase reporter gene (Invivogen) were incubated with 50 ⁇ M of cyclic [G(3′,5′)pG(3′,5′)p] (CDG), cyclic [G(2′,5′)pG(3′,5′)p] (mixed linkage, or ML-CDG), Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG), compared to [A(3′,5′)pA(3′,5′)p] (CDA), cyclic [A(2′,5′)pA(3′,5′)p] (mixed linkage, or
  • the Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) derivative induced significantly higher levels of IFN- ⁇ than the unmodified cyclic c-di-GMP (CDG) or modified CDG molecules.
  • the Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p](ML RR-CDA) molecule induced significantly higher IFN- ⁇ levels that either the unmodified CDA or ML CDA molecules.
  • CDN compounds were administered to 6-8 week old female BALB/c mice (in a total volume of 100 ⁇ L in HBSS) at doses of 50, 5 and 0.5 ⁇ M by subcutaneous injection into the base of the tail. Mice were assessed 24 hours later for lymphocyte immune cell activation by fluorescent activated cell sorting (FACS) for upregulation of surface CD69 expression on natural killer (NK) cells, CD4 + and CD8 + T cells, as compared to IgG1 isotype controls.
  • FACS fluorescent activated cell sorting
  • the Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) molecule induced potent immune activation of NK and T cells in a dose-dependent manner.
  • the Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) molecule also induced NK and T cell activation, although to a lesser extent than the ML RR-CDG molecule.
  • Both the ML RR-CDN molecules induced more immune cell activation that the ML CDN molecules at all doses.
  • each compound was diluted in either SVPD buffer (IX PBS and 0.6 mM MgCl 2 ), NP1 buffer (30 mM Na Acetate, pH 5.3, 2 mM ZnCl 2 ) or left untreated and then incubated for 2 hr at 37° C., followed by boiling for 10 min to inactivate the nucleases.
  • SVPD buffer IX PBS and 0.6 mM MgCl 2
  • NP1 buffer (30 mM Na Acetate, pH 5.3, 2 mM ZnCl 2 ) or left untreated and then incubated for 2 hr at 37° C., followed by boiling for 10 min to inactivate the nucleases.
  • THP1-BlueTM ISG cells a human monocyte cell line transfected with an IRF-inducible secreted embryonic alkaline phosphatase reporter gene (Invivogen) which express alkaline phosphatase under the control of a promoter comprised of five IFN-stimulated response elements
  • IRF-inducible secreted embryonic alkaline phosphatase reporter gene Invivogen which express alkaline phosphatase under the control of a promoter comprised of five IFN-stimulated response elements
  • the untreated Rp, Rp dithio compounds, Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA) and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) are more potent inducers of type I interferon than the oxo (cyclic[A(3′,5′)pA(3′,5′)p] (CDA) and cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA) and the Rp monothio.
  • Rp, Rp dithio compounds, Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA) and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) retain their potency after SVPD and NP1 treatment, whereas the oxo (cyclic[A(3′,5′)pA(3′,5′)p](CDA) and cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA) lost activity after digestion with both SVPD and NP1.
  • Rp The Rp monothio derivative (Rp, monothio cyclic [A(2′,5′)pA(3′,5′)p] (ML R-CDA) which contains a single thio substitution at the 3′-5 phosphodiester linkage retained activity after NP1 digestion, but was susceptible to SVPD treatment, which cleaves the 2′-5′ phosphodieseter linkage.
  • the differential susceptibility of the oxo, Rp monothio and Rp, Rp dithio derivatives to SVPD or NP1 digestion confirms the structure of the Rp monothio and Rp, Rp dithio derivatives.
  • HEK 293T cell lines that expressed the human STING alleles was generated.
  • the parental HEK 293T cell line does not express endogenous STING, so the responsiveness of exogenously expressed STING alleles can be evaluated.
  • MSCV2.2 plasmids encoding hSTING(REF)-GFP, hSTING(WT)-GFP, hSTING(HAQ)-GFP, hSTING(Q)-GFP and mSTING(WT)-GFP were obtained from the Vance Laboratory at UC Berkeley.
  • hSTING(AQ)-GFP was derived from hSTING(Q)-GFP using a QuickChange Site-Directed Mutagenesis kit (Stratagene).
  • the sequence of the hSTING(REF) allele is also known as the Barber allele (Ishikawa, H., and Barber, G. N. (2008). Nature 455, 674-678), and has the NCBI Reference Sequence NP_938023.1.
  • the amino acid difference between hSTING(REF) and the other WT, HAQ, AQ and Q human STING alleles are shown in FIG. 13 , which is adapted from Yi et al., Plos One 8: e77846 (2013).
  • Stable HEK 293T-derived cell lines expressing each of the individual human STING alleles were generated by FACS sorting of GFP positive cells using a Mo Flo cell sorter at the Cancer Research Laboratory Flow Cytometry Facility at UC Berkeley. 1 ⁇ 10 4 HEK293T STING cells were seeded in 96-well plates and transiently transfected (using Lipofectamine 2000) with 50 ng of a human IFN- ⁇ reporter plasmid (pLuc-IFN- ⁇ ) expressing the human IFN- ⁇ promoter upstream of a luciferase reporter and 10 ng of TK-renilla for normalization.
  • pLuc-IFN- ⁇ human IFN- ⁇ reporter plasmid
  • Each STING cell line was stimulated with 10 ⁇ M of cyclic [G(3′,5′)pA(3′,5′)p] (cGAMP), cyclic [G(2′,5′)pA(3′,5′)p](ML-cGAMP), Rp, Rp dithio cyclic [G(2′,5′)pA(3′,5′)p] (ML RR-cGAMP), cyclic[A(3′,5′)pA(3′,5′)p] (CDA), Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA), cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA), Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA), Rp, Rp dithio cyclic [
  • FIG. 14 depicts stimulation of HEK293 cell lines encoding human STING variant alleles by measuring the fold induction of the IFN ⁇ -LUC reporter (RLU plotted on y-axis).
  • hSTING The refractory human STING alleles, hSTING (REF) and hSTING (Q), responded poorly to stimulation with the native molecules with canonical internucleotide phosphate bridge linkages: cyclic [G(3′,5′)pA(3′,5′)p] (cGAMP), cyclic[A(3′,5′)pA(3′,5′)p] (CDA); and, cyclic [G(3′,5′)pG(3′,5′)p] (CDG).
  • cGAMP cyclic [G(3′,5′)pA(3′,5′)p]
  • CDA cyclic[A(3′,5′)pA(3′,5′)p]
  • CDG cyclic [G(3′,5′)pG(3′,5′)p]
  • cell lines expressing the refractory human STING alleles were responsive to stimulation with the synthetic Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA): ML RR-CDA; ML RR-CDG; and, ML RR-cGAMP.
  • ML RR-CDA synthetic Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p]
  • ML RR-CDA Rp dithio cyclic [A(2′,5′)pA(3′,5′)p]
  • FIG. 15A depicts bar graphs indicating the average of the mean fluorescence intensity (MFI) following stimulation with the CDN molecules indicated in the figure. Also shown in FIG.
  • 15B are representative histograms of CD80, CD86, CD83 and MHC Class I (HLA-ABC) expression in human DCs. Filled histograms correspond to unstimulated cells, the dotted line represents LPS stimulation, and the solid line represents Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) stimulation.
  • PBMCs 5 ⁇ 10 4 PBMCs were stimulated overnight in an IFN ⁇ ELISpot assay with media alone (unstimulated) or with 1 ⁇ M OVA 257-264 peptide in the presence of 1 ⁇ 10 5 na ⁇ ve splenocytes as feeder cells.
  • IFN ⁇ ELISpots were developed and quantified using a CTL plate reader and ImmunoSpot software.
  • both doses of cyclic [G(2′,5′)pG(3′,5′)p] induce OVA-specific CD8 immune responses in C57BL/6 mice. These responses are significantly higher than responses induced by unstimulated controls and by a no CDN control group.
  • ML-CDG c[G(2′,5′)pG(3′,5′)p]
  • PBMCs 5 ⁇ 10 4 PBMCs were stimulated overnight in an IFN ⁇ ELISpot assay with media alone (unstimulated) or with 1 ⁇ M OVA 257-264 peptide in the presence of 1 ⁇ 10 5 na ⁇ ve splenocytes as feeder cells.
  • IFN ⁇ ELISpots were developed and quantified using a CTL plate reader and ImmunoSpot software.
  • FIG. 17 shows that c[G(2′,5′)pG(3′,5′)p] (ML-CDG) induces an OVA-specific CD8 T cell response that is dependent on the presence of a functional STING molecule.
  • formulation of c[G(2′,5′)pG(3′,5′)p](ML-CDG) and ovalbumin protein induces significant OVA 257-264 immune responses compared to unstimulated control and a no CDN control.
  • B16 melanoma cells (5 ⁇ 10 4 cells in 100 ⁇ L PBS) were implanted subcutaneously on the lower back of 6-8 week old female C57BL/6 mice (8 mice per group). Treatments began when tumors reached a volume of approximately 75 mm 3 , on day 14 post tumor implantation.
  • the CDN compounds were administered (25 ⁇ g in a total volume of 40 ⁇ L HBSS) by subcutaneous injection into the center of the tumor using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • CDNs tested were cyclic [G(3′,5′)pG(3′,5′)p] (CDG); cyclic [G(2′,5′)pG(3′,5′)p] (mixed linkage, or ML CDG); Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG); cyclic [A(3′,5′)pA(3′,5′)p] (CDA); cyclic [A(2′,5′)pA(3′,5′)p] (mixed linkage, or ML CDA); and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA).
  • the ML RR-CDG and ML RR-CDA derivatives induced potent anti-tumor efficacy, as compared to the cyclic ML CDG and cyclic ML CDA cyclic dinucleotide molecules.
  • CT26 colon carcinoma cells (2 ⁇ 10 5 cells in 100 ⁇ L PBS) were implanted by intravenous injection into 6-8 week old female BALB/c mice and assessed for overall survival.
  • the CDN compounds (25 ⁇ g in a total volume of 100 ⁇ L HBSS) were administered one day post tumor implantation by subcutaneous injection into the base of the tail. Mice were boosted with an additional injection one week later for a total of two vaccinations.
  • the compounds Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) or Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) compounds 25 ⁇ g in a total volume of 40 L HBSS), or HBSS vehicle control
  • Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) 50 ⁇ g in a total volume of 40 ⁇ L HBSS) or HBSS vehicle control, were administered by subcutaneous injection into the center of the tumor using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • B16 melanoma cells (5 ⁇ 10 4 cells in 100 ⁇ L PBS) were implanted on the right flanks of 6-8 week old female goldenticket STING ⁇ / ⁇ mice, or wild-type C57BL/6 control mice (5 mice per group). Treatments began when tumors reached a volume of approximately 75 mm 3 , on day 14 post tumor implantation.
  • the compounds administered were Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) (25 ⁇ g in a total volume of 40 ⁇ L HBSS), Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) (50 ⁇ g in a total volume of 40 ⁇ L HBSS), the TLR9 agonist CpG 1668 (50 ⁇ g in a total volume of 40 ⁇ L HBSS), or HBSS vehicle control.
  • Mice were treated by subcutaneous injection into the center of tumor only using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • FIG. 20B tumor growth was not inhibited by either ML RR-CDG or ML RR-CDA, demonstrating that the anti-tumor efficacy observed in wild-type C57BL/6 mice ( FIG. 20A ) was entirely dependent on a functional STING signaling pathway.
  • mice 6-8 week old female BALB/c mice (8 mice per group) were implanted with CT26 colon carcinoma cells (1 ⁇ 10 5 cells in 100 ⁇ L PBS). Mice were treated with Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound (50 ⁇ g in a total volume of 40 ⁇ L HBSS) or HBSS vehicle control, and tumor growth was monitored as per previous example. Mice were bled on day 18 post tumor implantation and PBMCs were isolated by Ficoll gradient (Miltenyi Biotech).
  • mice and age-matched na ⁇ ve control mice were implanted on the contralateral flank with either CT26 or 4T1 (both 1 ⁇ 10 5 cells in 100 ⁇ L PBS) tumor cells (4 mice per group), and monitored for tumor growth.
  • mice treated with ML RR-CDA rejected the growth of established CT26 colon carcinomas.
  • PBMCs on day 18 post tumor induction were assessed for IFN- ⁇ production by ELISpot assay, in response to stimulation with the endogenous tumor antigen AH1.
  • CDN derivative molecules induce effective and durable anti-tumor immunity in an alternate tumor model
  • 6-8 week old female BALB/c mice 8 mice per group were implanted with 4T1 mammary carcinoma cells (1 ⁇ 10 5 cells in 100 ⁇ L PBS).
  • Mice were treated with Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound (50 ⁇ g in a total volume of 40 ⁇ L HBSS), or HBSS vehicle control, as per previous experiment.
  • mice and age-matched na ⁇ ve control mice were implanted on the contralateral flank with either CT26 or 4T1 (both 1 ⁇ 10 5 cells in 100 ⁇ L PBS) tumor cells (4 mice per group), and monitored for tumor growth.
  • CT26 colon carcinoma cells (1 ⁇ 10 5 cells in 100 ⁇ L PBS) and (B) 4T1 mammary carcinoma cells (1 ⁇ 10 5 cells in 100 ⁇ L PBS) were implanted subcutaneously on the contralateral flanks of 6-8 week old female BALB/c mice (8 mice per group). Treatments began when tumors reached a volume of approximately 75 mm 3 , on day 13 post tumor implantation.
  • the Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound (50 ⁇ g in a total volume of 40 ⁇ L HBSS), or HBSS vehicle control, was administered by subcutaneous injection into the center of the primary (right side) tumor only using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • mice 6-8 week old female C57BL/6 mice (8 mice per group) were implanted with B16 melanoma cells (5 ⁇ 10 4 cells in 100 ⁇ L PBS) in the right flank. One week later mice were implanted intravenously with 1 ⁇ 10 5 B16 melanoma cells to colonize the lung, along with a group of na ⁇ ve age-matched control mice.
  • mice When the subcutaneous flank tumor reached approximately 75 mm 3 on day 13, mice were treated intratumorally with Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) (50 ⁇ g in a total volume of 40 ⁇ L HBSS) or HBSS vehicle control, for three injections as per previous protocol.
  • ML RR-CDA Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p]
  • ML RR-CDA Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p]
  • HBSS HBSS vehicle control
  • treatment with ML RR-CDA significantly inhibited tumor growth of the primary flank tumor, as compared to the HBSS control group (P ⁇ 0.001, student's t-test).
  • treatment with the CDN derivative significantly inhibited the growth of distal lung tumor nodules, compared to the HBSS and na ⁇ ve (i.v. only) tumor groups.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Dermatology (AREA)
  • Saccharide Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention provides highly active cyclic-di-nucleotide (CDN) immune stimulators that activate DCs via a recently discovered cytoplasmic receptor known as STING (Stimulator of Interferon Genes). In particular, the CDNs of the present invention are provided in the form of a composition comprising one or more cyclic purine dinucleotides induce STING-dependent type I interferon production, wherein the cyclic purine dinuclotides present in the composition are substantially pure 2′,5′,2′,5′ and 2′,5′,3′,5′ CDNs, and prefereably Rp,Rp stereosiomers thereof.

Description

  • The present application is a continuation of U.S. patent application Ser. No. 15/665,378, filed Jul. 31, 2017, now U.S. Pat. No. 10,653,774, which is a continuation of U.S. patent application Ser. No. 14/280,667, filed May 18, 2014, now U.S. Pat. No. 9,724,408, which claims priority to U.S. Provisional Application No. 61/825,005 filed May 18, 2013, and to U.S. Provisional Application No. 61/902,125 filed Nov. 8, 2013, all of which are hereby incorporated by reference in their entirety.
  • BACKGROUND OF THE INVENTION
  • The following discussion of the background of the invention is merely provided to aid the reader in understanding the invention and is not admitted to describe or constitute prior art to the present invention.
  • The human immune system may generally be divided into two arms, referred to as “innate immunity” and “adaptive immunity.” The innate arm of the immune system is predominantly responsible for an initial inflammatory response via a number of soluble factors, including the complement system and the chemokine/cytokine system; and a number of specialized cell types including mast cells, macrophages, dendritic cells (DCs), and natural killer cells. In contrast, the adaptive immune arm involves a delayed and a longer lasting antibody response together with CD8+ and CD4+ T cell responses that play a critical role in immunological memory against an antigen. A third arm of the immune system may be identified as involving γδ T cells and T cells with limited T cell receptor repertoires such as NKT cells and MAIT cells.
  • For an effective immune response to an antigen, antigen presenting cells (APCs) must process and display the antigen in a proper MHC context to a T cell, which then will result in either T cell stimulation of cytotoxic and helper T cells. Following antigen presentation successful interaction of co-stimulatory molecules on both APCs and T cells must occur or activation will be aborted. GM-CSF and IL-12 serve as effective pro-inflammatory molecules in many tumor models. For example, GM-CSF induces myeloid precursor cells to proliferate and differentiate into dendritic cells (DCs) although additional signals are necessary to activate their maturation to effective antigen-presenting cells necessary for activation of T cells. Barriers to effective immune therapies include tolerance to the targeted antigen that can limit induction of cytotoxic CD8 T cells of appropriate magnitude and function, poor trafficking of the generated T cells to sites of malignant cells, and poor persistence of the induced T cell response.
  • DCs that phagocytose tumor-cell debris process the material for major histocompatibility complex (MHC) presentation, upregulate expression of costimulatory molecules, and migrate to regional lymph nodes to stimulate tumor-specific lymphocytes. This pathway results in the proliferation and activation of CD4+ and CD8+ T cells that react to tumor-associated antigens. Indeed, such cells can be detected frequently in the blood, lymphoid tissues, and malignant lesions of patients.
  • New insights into the mechanisms underlying immune-evasion, together with combination treatment regimens that potentiate the potency of therapeutic vaccination-either directly or indirectly-through combination with immune checkpoint inhibitors or other therapies, have served as a basis for the development of vaccines that induce effective antitumor immunity. The CDNs cyclic-di-AMP (produced by Listeria monocytogenes) and its analog cyclic-di-GMP (produced by Legionella pneumophila) are recognized by the host cell as a PAMP (Pathogen Associated Molecular Pattern), which bind to the PRR (Pathogen Recognition Receptor) known as STING. STING is an adaptor protein in the cytoplasm of host mammalian cells which activates the TANK binding kinase (TBK1)-IRF3 signaling axis, resulting in the induction of IFN-β and other IRF-3 dependent gene products that strongly activate innate immunity. It is now recognized that STING is a component of the host cytosolic surveillance pathway, that senses infection with intracellular pathogens and in response induces the production of IFN-β, leading to the development of an adaptive protective pathogen-specific immune response consisting of both antigen-specific CD4 and CD8 T cells as well as pathogen-specific antibodies. Examples of cyclic purine dinucleotides are described in some detail in, e.g., U.S. Pat. Nos. 7,709,458 and 7,592,326; WO2007/054279; and Yan et al., Bioorg. Med. Chem Lett. 18: 5631 (2008), each of which is hereby incorporated by reference.
  • There remains a need for improved compositions and methods for immunologic strategies to treating diseases such as cancer that can be refractory to traditional therapeutic approaches.
  • SUMMARY OF THE INVENTION
  • It is an object of the present invention to provide compositions which modulate immune responses to diseases.
  • In a first aspect, the present invention provides compositions comprising:
  • one or more cyclic purine dinucleotides (“CDNs”) which that induce STimulator of INterferon Genes (“STING”)-dependent type I interferon production. As described hereinafter, a number of CDNs find use in the present invention. Preferred cyclic purine dinucleotides include, but are not limited to, one or more of c-di-AMP, c-di-GMP, c-di-IMP, c-AMP-GMP, c-AMP-IMP, c-GMP-IMP, and analogs thereof. This list is not meant to be limiting.
  • The general structure of a cyclic purine dinucleotide according to the present invention is as follows:
  • Figure US20200282049A1-20200910-C00001
  • where each R1 and R2 is a purine, and the structure
  • Figure US20200282049A1-20200910-C00002
  • is intended to reflect that the phosphate linkage may be to either the 2′ or 3′ position on the ribose, and the other of the 2′ or 3′ position which is not participating in the cyclic linkage is an —OH. The present invention contemplates 2′,5′,2′,5′ CDNs and 2′,5′,3′,5′ CDNs. By way of example, c-di-GMP having 2′-5′ linkages refers to the molecule indicated above where each of R1 and R2 are guanine, and each phosphate linkage is 2′-to-5′.
  • For purposes of the present invention, this general structure is further modified to introduce substituents which confer the ability to bind to STING and induce a STING-dependent signaling cascade (and most preferably induce a human STING-dependent signaling cascade), and thereby induce STING-dependent type I interferon production and other co-regulated genes. By way of example, the present invention provides compositions comprising the following compounds:
  • Figure US20200282049A1-20200910-C00003
  • wherein each X is independently O or S, and R3 and R4 are each independently H or an optionally substituted straight chain alkyl of from 1 to 18 carbons and from 0 to 3 heteroatoms, an optionally substituted alkenyl of from 1-9 carbons, an optionally substituted alkynyl of from 1-9 carbons, or an optionally substituted aryl, wherein substitution(s), when present, may be independently selected from the group consisting of C1-6 alkyl straight or branched chain, benzyl, halogen, trihalomethyl, C1-6 alkoxy, —NO2, —NH2, —OH, ═O, —COOR′ where R′ is H or lower alkyl, —CH2OH, and —CONH2, wherein R3 and R4 are not both H.
  • In preferred embodiments, one or both of R3 and R4 independently comprise a prodrug leaving group removed by cellular esterases. In certain embodiments, one or both of R3 and R4 are a C6 to C18 fatty acid ester. In certain embodiments, one or both of R3 and R4 are selected from the group consisting of myristoyl, pentanoyl, hexanoyl, heptanoyl, etc.
  • In certain embodiments, each X is S. In preferred embodiments when each X is S, the compositions comprise one or more substantially pure Sp,Sp, Rp,Rp, Sp,Rp, or Rp,Sp stereoisomers.
  • In certain embodiments, each of R1 and R2 are independently selected from the group consisting of adenine, guanine, inosine, and xanthine or analogs thereof. Preferably, each of R1 and R2 are independently adenine or guanine.
  • As described hereinafter, a cyclic purine dinucleotide composition according to the present invention can induce STING-dependent type I interferon production at least 2-fold, and more preferably 5-fold or 10-fold, or more, as compared to c-di-GMP having 3′-5′ linkages. As noted herein, most preferably, the STING is human STING. In preferred embodiments, a substantially pure cyclic purine dinucleotide composition according to the present invention activates human STING but the corresponding cyclic purine dinucleotidehaving only bis-(3′,5′) linkages does not.
  • In their role as adjuvants, in certain embodiments the present compositions may be used as adjuvants in a therapeutic or prophylactic strategy employing vaccine(s). Thus, the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, may be used together with one or more vaccines selected to stimulate an immune response to one or more predetermined antigens. The substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, may be provided together with, or in addition to, such vaccines.
  • Such vaccine(s) can comprise inactivated or attenuated bacteria or viruses comprising the antigens of interest, purified antigens, live viral or bacterial delivery vectors recombinantly engineered to express and/or secrete the antigens, antigen presenting cell (APC) vectors comprising cells that are loaded with the antigens or transfected with a composition comprising a nucleic acid encoding the antigens, liposomal antigen delivery vehicles, or naked nucleic acid vectors encoding the antigens. This list is not meant to be limiting. By way of example, such vaccine(s) may also comprise an inactivated tumor cell that expresses and secretes one or more of GM-CSF, CCL20, CCL3, IL-12p70, FLT-3 ligand.
  • The substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, may be administered to individuals in need thereof by a variety of parenteral and nonparenteral routes in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles. Preferred routes are parenteral, and include but, are not limited to, one or more of subcutaneous, intravenous, intramuscular, intraarterial, intradermal, intrathecal and epidural administrations. Intra-tumor routes are also preferred. Particularly preferred is administration by subcutaneous administration. Preferred pharmaceutical compositions are formulated as aqueous or oil-in-water emulsions.
  • The compositions of the present invention may comprise, or be administered together with, one or more additional pharmaceutically active components such as adjuvants, lipids, interbilayer crosslinked multilamellar vesicles, biodegradeable poly(D,L-lactic-co-glycolic acid) [PLGA]-based or poly anhydride-based nanoparticles or microparticles, and nanoporous particle-supported lipid bilayers, CTLA-4 and PD-1 pathway Antagonists, PD-1 pathway blocking agents, inactivated bacteria which induce innate immunity (e.g., inactivated or attenuated Listeria monocytogenes), compositions which mediate innate immune activation via Toll-like Receptors (TLRs), (NOD)-like receptors (NLRs), Retinoic acid inducible gene-based (RIG)-I-like receptors (RLRs), C-type lectin receptors (CLRs), pathogen-associated molecular patterns (“PAMPs”), chemotherapeutic agents, etc.
  • In a related aspect, the present invention relates to methods of inducing, stimulating, or adjuvanting an immune response in an individual. These methods comprise administering the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, to the individual. Preferred routes of administration are parenteral. As noted above, particularly preferred are thiophosphate derivatives of such cyclic purine dinucleotides.
  • In certain embodiments, the method is a method of cancer treatment. By way of example, the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, may be provided alone, or together with or in addition to one or more cancer vaccine compositions that are known in the art. The patient receiving such treatment may be suffering from a cancer selected from the group consisting of a colorectal cancer cell, an aero-digestive squamous cancer, a lung cancer, a brain cancer, a liver cancer, a stomach cancer, a sarcoma, a leukemia, a lymphoma, a multiple myeloma, an ovarian cancer, a uterine cancer, a breast cancer, a melanoma, a prostate cancer, a pancreatic carcinoma, and a renal carcinoma. In other embodiments, the method is a method of inducing, stimulating, or adjuvanting an immune response a pathogen.
  • With regard to treatment of a mammal suffering from cancer, the methods described herein can comprise administering to the mammal an effective amount of the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, optionally prior to or following a primary therapy administered to the mammal to remove or kill cancer cells expressing the cancer antigen. The compositions of the present invention may be provided as a neoadjuvant therapy; however in preferred embodiments, the compositions of the present invention are administered following the primary therapy. In various embodiments, the primary therapy comprises surgery to remove the cancer cells from the mammal, radiation therapy to kill the cancer cells in the mammal, or both surgery and radiation therapy.
  • In other embodiments, the methods described herein can comprise administering to the mammal an effective amount of the substantially pure CDNs of the present invention for the treatment of disorders in which shifting of Th1 to Th2 immunity confers clinical benefit. Cell-mediated immunity (CMI) is associated with TH1 CD4+ T lymphocytes producing cytokines IL-2, interferon (IFN)-γ and tumor necrosis factor (TNF)-α. In contrast, humoral immunity is associated with TH2 CD4+ T lymphocytes producing IL-4, IL-6 and IL-10. Immune deviation towards TH1 responses typically produces activation of cytotoxic T-cell lymphocytes (CTL), natural killer (NK) cells, macrophages and monocytes. Generally, Th1 responses are more effective against intracellular pathogens (viruses and bacteria that are inside host cells) and tumors, while Th2 responses are more effective against extracellular bacteria, parasites including helminths and toxins. In addition, the activation of innate immunity is expected to normalize the T-helper type 1 and 2 (Th1/Th2) immune system balance and to suppress the excessive reaction of Th2 type responses that cause immunoglobulin (Ig) E-dependent allergies and allergic asthma.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 depicts cyclic purine dinucleotide (“CDN”)-mediated signaling. A CDN (e.g., c-di-AMP or c-di-GMP) induces production of IFN-β by binding to the cytosolic receptor STING (Stimulator of Interferon Genes), and inducing signaling through the TBK-1/IRF-3 pathway, resulting in both autocrine and paracrine activation of DCs through binding to the IFN receptor and subsequent signaling.
  • FIG. 2A depicts a synthesis scheme for c-[G(2′,5′)pG(3′,5′)p] and dithio derivatives.
  • FIG. 2B depicts a synthesis scheme for c-[A(2′,5′)pA(3′,5′)p] and dithio derivatives.
  • FIG. 2C depicts structures of compounds 10, 20, 21, 22, and 23.
  • FIG. 3A depicts 1H-NMR results for compound 9a.
  • FIG. 3B depicts COSY (3.5-6.0 ppm 1H-axis) results for compound 9a.
  • FIG. 3C depicts HMBC (3.0-5.5 ppm 1H-axis) results for compound 9a.
  • FIG. 3D depicts 1H-NMR results for compound 21.
  • FIG. 3E depicts COSY (3.5-6.0 ppm 1H-axis) results for compound 21.
  • FIG. 3F depicts HMBC (0-9.5 ppm 1H-axis) results for compound 21.
  • FIG. 3G depicts HMBC (3.5-5.5 ppm 1H-axis)results for compound 21.
  • FIG. 3H depicts analytical HPLC (2-20% ACN/10 mM TEAA buffer—20 min) results for compound 19b.
  • FIG. 4 depicts c-[G(2′,5′)pG(3′,5′)p] and dithio ribose O-substitued derivatives.
  • FIG. 5 depicts c-[A(2′,5′)pA(3′,5′)p] and dithio ribose O-substitued derivatives.
  • FIG. 6 depicts c-[G(2′,5′)pA(3′,5′)p] and dithio ribose O-substitued derivatives.
  • FIG. 7 Depicts Type 1 interferon production in THP-1 cells following stimulation with various cyclic dinucleotide molecules
  • FIG. 8 depicts normalized RNA expression levels of Type 1 interferons and interferon gamma in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules
  • FIG. 9A depicts levels of Type 1 interferon alpha protein in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules.
  • FIG. 9B depicts levels of Type 1 interferon beta protein in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules.
  • FIG. 9C depicts levels of interferon gamma protein in human PBMCs from independent donors following stimulation with various cyclic dinucleotide molecules.
  • FIG. 10 depicts IFN-3 induction in human cells as a signature of adjuvant potency following treatment with various cyclic dinucleotide molecules.
  • FIG. 11A depicts upregulation of surface CD69 expression on natural killer (NK) cells as a measure of immune activation following treatment with various cyclic dinucleotide molecules.
  • FIG. 11B depicts upregulation of surface CD69 expression on CD4+ cells as a measure of immune activation following treatment with various cyclic dinucleotide molecules.
  • FIG. 11C depicts upregulation of surface CD69 expression on CD8+ T cells as a measure of immune activation following treatment with various cyclic dinucleotide molecules.
  • FIG. 12 depicts resistance of various CDN derivatives to phosphodiesterase treatment.
  • FIG. 13 depicts various known STING variants.
  • FIG. 14 depicts stimulation of HEK293 cell lines encoding human STING variant alleles by measuring the fold induction of the IFNβ-LUC reporter.
  • FIG. 15A depicts surface expression of MHC class I (HLA-ABC), CD80, CD83 and CD86 by stimulated human dendritic cells.
  • FIG. 15B representative histograms of CD80, CD86, CD83 and MHC Class I (HLA-ABC) expression in human DCs following LPS or CDN stimulation.
  • FIG. 16 depicts OVA-specific CD8 T cell immunity in PBMCs in C57BL/6 mice at 7 days post vaccination with cyclic dinucleotide adjuvanted OVA protein.
  • FIG. 17 depicts OVA-specific CD8 T cell immunity in PBMCs in C57BL/6 or goldentickt (STING−/−) mice at 7 days post vaccination with cyclic dinucleotide adjuvanted OVA protein.
  • FIG. 18 depicts tumor volume in a B16 melanoma model following treatment with various cyclic dinucleotide molecules.
  • FIG. 19A depicts survival curves in a CT26 lung metastasis tumor model following treatment with various cyclic dinucleotide molecules.
  • FIG. 19B depicts survival curves in a CT26 colon carcinoma tumor model following treatment with various cyclic dinucleotide molecules.
  • FIG. 19C depicts survival curves in a 4T1 mammary carcinoma tumor model following treatment with various cyclic dinucleotide molecules.
  • FIG. 20A depicts tumor inhibition in wild-type C57BL/6 mice following ML RR-CDN administration as compared to control mice receiving HBSS and CpG dinculeotide.
  • FIG. 20B depicts results obtained in STING deficient mice.
  • FIG. 21A depicts rejection of established CT26 colon carcinomas following ML RR-CDN administration.
  • FIG. 21B depicts IFN-γ induction from mice treated with ML RR-CDA.
  • FIG. 21C depicts survival in mice following re-challenge with CT26 tumor cells.
  • FIG. 22A depicts rejection of established 4T1 mammary carcinomas following ML RR-CDN administration.
  • FIG. 22B depicts protection from re-challenge with CT26 tumor cells.
  • FIG. 23A depicts inhibition of the treated primary tumor in CT26 tumor-bearing animals following ML RR-CDA administration, as compared to HBSS vehicle control. FIG. 23B depicts inhibition of the treated primary tumor in 4T1 tumor-bearing animals following ML RR-CDA administration, as compared to HBSS vehicle control.
  • FIG. 24A depicts inhibition of the treated primary tumor in B16 melanoma following ML RR-CDA administration.
  • FIG. 24B depicts inhibition of growth of distal lung tumor nodules following ML RR-CDA administration, as compared to HBSS vehicle control in graphical form.
  • FIG. 24C depicts inhibition of growth of distal lung tumor nodules following ML RR-CDA administration, as compared to HBSS vehicle control in the lung tissue itself in photographic form.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to the use of novel and highly active cyclic-di-nucleotide (CDN) immune stimulators that activate DCs via a recently discovered cytoplasmic receptor known as STING (Stimulator of Interferon Genes). In particular, the CDNs of the present invention are provided in the form of a composition comprising one or more cyclic purine dinucleotides induce STING-dependent type I interferon production, wherein the cyclic purine dinuclotides present in the composition are substantially pure 2′,5′,2′,5′ and 2′,5′,3′,5′ CDNs.
  • Recent insights into the design and development of adjuvants are informed by a fundamental understanding that conserved microbial structures known as Pathogen-Associated Molecular Patterns (PAMPs) are sensed by host cell Pattern Recognition Receptors (PRRs), triggering a downstream signaling cascade resulting in the induction of cytokines and chemokines, and initiation of a specific adaptive immune response. How the innate immune system is engaged by the PAMP complement of a microbe shapes the development of an adaptive response that is appropriate to combat the invading pathogen from causing disease. An objective of adjuvant design is to select defined PAMPs or synthetic molecules specific for designated PRRs to initiate a desired response. Adjuvants such as monophosphoryl lipid A (MPL) and CpG are PAMPs recognized by Toll-like receptors (TLRs), a class of transmembrane PRRs that signal through MyD88 and Trif adaptor molecules and mediate induction of NF-kB dependent proinflammatory cytokines. MPL (TLR-4 agonist) and CpG (TLR-9 agonist) are clinically advanced adjuvants, and are components of vaccines that are approved or pending approval by the FDA. While TLRs present on the cell surface (e.g., TLR-4) and endosomes (e.g., CpG) sense extracellular and vacuolar pathogens, the productive growth cycle of multiple pathogens including viruses and intracellular bacteria occurs in the cytosol. The compartmentalization of extracellular, vacuolar, and cytosolic PRRs has led to the hypothesis that the innate immune system distinguishes between pathogenic and non-pathogenic microbes by monitoring the cytosol. It should be apparent to one skilled in the art that agonists specific for PRRs comprising the cytosolic surveillance pathway that initiate development of protective immunity against intracellular pathogens, and is relevant to vaccine design. These same targeting ligands will also be essential in the development of effective vaccines targeting malignancies, know to require tumor-specific CD4+ and CD8+ T cells.
  • Activation of the Cytosolic Surveillance Pathway (CSP) is Integral to Development of Protective Immunity to Intracellular Pathogens. The CSP detects bacterial, viral, and protozoan pathogens, leading to activation of the TANK binding kinase (TBK-1)/IRF-3 signaling axis and induction of IFN-β and other co-regulated genes. Both viral and bacterial nucleic acids activate this pathway, and induction of IFN-β is MyD88 and Trif independent. While Type I interferon is often thought of primarily as a host anti-viral response, induction of IFN-β is a signature of cytosolic growth in macrophages infected with the intracellular bacterium, Listeria monocytogenes (Lm). A well-known dichotomy in the mouse listeriosis model is that, whereas wild-type Lm primes potent CD4 and CD8 T-cell immunity that protects mice against bacterial challenge, vaccination with listeriolysin O (LLO)-deleted Lm does not elicit functional T cells or induce protective immunity. This difference is evidence of the requirement for expression of host cell genes and cytosolic access by Lm to elicit functional T-cell mediated protective immunity. The level of IFN-β in infected host cells is regulated by Lm multidrug efflux pumps (MDRs), which that secrete structurally unrelated small molecules, including antibiotics. IFN-β is not induced in host cells infected with Lm LLO mutants that are confined to the phagolysosome. Normal levels of IFN-β are induced in infected MyD88−/− Trif−/− macrophages deficient in all TLR-mediated signaling. These data demonstrate that although Lm engages TLRs, in response to infection with wild-type Lm, the host cell CSP is required for development of protective immunity, correlated with induction of IFN-β.
  • Cyclic-di-Nucleotides (CDNs) activate the cytosolic surveillance pathway through direct binding of to the cytosolic PRR, STING. The Type I interferon response to infection by Lm and other intracellular bacteria results from the secretion of c-di-AMP or its related cyclic dinucleotide (CDN), c-di-GMP, and its direct binding to DDX41 and DEAD (aspartate-glutamate-alanine-aspartate) box helicase and STING (Stimulator of Interferon Genes), a recently defined receptor of the cytosolic surveillance pathway. CDNs are second messengers expressed by most bacteria and regulate diverse processes, including motility and formation of biofilms. In addition to activating the TBK-1/IRF-3 signaling pathway, in response to binding CDNs STING also activates the IkB kinase, resulting in translocation of the NF-kB transcription factor to the nucleus, activating the expression of multiple pro-inflammatory genes.
  • Until recently, how STING senses cytoplasmic DNA remained elusive. Unlike AIM2 which directly binds dsDNA, STING lacks any obvious DNA-binding domains. Whether other candidate DNA sensors such as DDX41, DNA-PK and DAI kinase were essential mediators of dsDNA signaling through STING remained unclear. This conundrum was solved with the discovery of cyclic GMP-AMP synthase (cGAS), a host cell nucleotidyl transferase that in response to binding dsDNA synthesizes a second messenger, cyclic di-GMP-AMP, which binds directly to STING and initiates a signaling cascade through the TBK-1/IRF-3 axis, resulting in the induction of IFNs. Additionally, the cGAS innate immune DNA sensor produces a non-canonical cyclic di-nucleotide that activates STING signaling. Unlike the cyclic dinucleotide second messenger produced by bacteria, in which the internucleotide phosphate bridge is joined by bis-(3′, 5′) linkages, the internucleotide phosphate bridge in the cyclic-GMP-AMP synthesized by cGAS is joined by non-canonical 2′, 5′ and 3′,5′ linkages, represented c[G(2′,5′)pA(3′,5′)p]. Thus, STING (Stimulator of Interferon Genes) has emerged as a central pathway for sensing cytosolic pathogen nucleic acids, either through direct binding of cyclic dinucleotides (CDNs) secreted by intracellular bacterium6, or via binding of a c-GMP-AMP second messenger, synthesized by host cell cyclic GMP-AMP synthase (cGAS) in response to binding cytosolic pathogen nucleic acids.
  • Native CDN molecules are sensitive to degradation by phosphodiesterases that are present in host cells, for example in antigen presenting cells, that take up vaccine formulations that contain said native CDN molecules. The potency of a defined adjuvant may be diminished by such degradation, as the adjuvant would be unable to bind and activate its defined PRR target. Lower adjuvant potency could be measured, for example by a lower amount of induced expression of a signature molecule of innate immunity (e.g., IFN-β), correlated with weaker vaccine potency, as defined by the magnitude of a measured antigen-specific immune response.
  • In the present invention, substantially pure 2′,5′,2′,5′ and 2′,5′,3′,5′ CDNs, and particularly dithio-diphosphate derivatives of 2′,5′,2′,5′ and 2′,5′,3′,5′ c-di-AMP and c-di-GMP are provided. The synthesis process for said dithio-diphosphate derivatives of c-di-AMP and c-di-GMP molecules results in a mixture of diastereomers, including Rp,Rp, Sp,Sp, SpRp, and Rp,Sp dithio-diphosphate derivatives of c-di-AMP and c-di-GMP molecules. These individual species may be separated, and exhibit substantial differences in their pharmaceutical characteristics.
  • Definitions
  • “Administration” as it is used herein with regard to a human, mammal, mammalian subject, animal, veterinary subject, placebo subject, research subject, experimental subject, cell, tissue, organ, or biological fluid, refers without limitation to contact of an exogenous ligand, reagent, placebo, small molecule, pharmaceutical agent, therapeutic agent, diagnostic agent, or composition to the subject, cell, tissue, organ, or biological fluid, and the like. “Administration” can refer, e.g., to therapeutic, pharmacokinetic, diagnostic, research, placebo, and experimental methods. Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell. “Administration” also encompasses in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding composition, or by another cell. By “administered together” it is not meant to be implied that two or more agents be administered as a single composition. Although administration as a single composition is contemplated by the present invention, such agents may be delivered to a single subject as separate administrations, which may be at the same or different time, and which may be by the same route or different routes of administration.
  • An “agonist,” as it relates to a ligand and receptor, comprises a molecule, combination of molecules, a complex, or a combination of reagents, that stimulates the receptor. For example, an agonist of granulocyte-macrophage colony stimulating factor (GM-CSF) can encompass GM-CSF, a mutein or derivative of GM-CSF, a peptide mimetic of GM-CSF, a small molecule that mimics the biological function of GM-CSF, or an antibody that stimulates GM-CSF receptor.
  • An “antagonist,” as it relates to a ligand and receptor, comprises a molecule, combination of molecules, or a complex, that inhibits, counteracts, downregulates, and/or desensitizes the receptor. “Antagonist” encompasses any reagent that inhibits a constitutive activity of the receptor. A constitutive activity is one that is manifest in the absence of a ligand/receptor interaction. “Antagonist” also encompasses any reagent that inhibits or prevents a stimulated (or regulated) activity of a receptor. By way of example, an antagonist of GM-CSF receptor includes, without implying any limitation, an antibody that binds to the ligand (GM-CSF) and prevents it from binding to the receptor, or an antibody that binds to the receptor and prevents the ligand from binding to the receptor, or where the antibody locks the receptor in an inactive conformation.
  • By “substantially purified” with regard to CDNs of the invention is meant that a specified species accounts for at least 50%, more often accounts for at least 60%, typically accounts for at least 70%, more typically accounts for at least 75%, most typically accounts for at least 80%, usually accounts for at least 85%, more usually accounts for at least 90%, most usually accounts for at least 95%, and conventionally accounts for at least 98% by weight, or greater, of the CDN activity present in a composition. The weights of water, buffers, salts, detergents, reductants, protease inhibitors, stabilizers (including an added protein such as albumin), and excipients are generally not used in the determination of purity.
  • “Specifically” or “selectively” binds, when referring to a ligand/receptor, nucleic acid/complementary nucleic acid, antibody/antigen, or other binding pair (e.g., a cytokine to a cytokine receptor) (each generally referred to herein as a “target biomolecule” or a “target”) indicates a binding reaction which is related to the presence of the target in a heterogeneous population of proteins and other biologics. Specific binding can mean, e.g., that the binding compound, nucleic acid ligand, antibody, or binding composition derived from the antigen-binding site of an antibody, of the contemplated method binds to its target with an affinity that is often at least 25% greater, more often at least 50% greater, most often at least 100% (2-fold) greater, normally at least ten times greater, more normally at least 20-times greater, and most normally at least 100-times greater than the affinity with a non-target molecule.
  • “Ligand” refers to a small molecule, nucleic acid, peptide, polypeptide, saccharide, polysaccharide, glycan, glycoprotein, glycolipid, or combinations thereof that binds to a target biomolecule. While such ligands may be agonists or antagonists of a receptor, a ligand also encompasses a binding agent that is not an agonist or antagonist, and has no agonist or antagonist properties. Specific binding of a ligand for its cognate target is often expressed in terms of an “Affinity.” In preferred embodiments, the ligands of the present invention bind with affinities of between about 104 M−1 and about 108 M−1. Affinity is calculated as Kd=koff/kon(koff is the dissociation rate constant, Kon is the association rate constant and Kd is the equilibrium constant).
  • Affinity can be determined at equilibrium by measuring the fraction bound (r) of labeled ligand at various concentrations (c). The data are graphed using the Scatchard equation: r/c=K(n−r): where r=moles of bound ligand/mole of receptor at equilibrium; c=free ligand concentration at equilibrium; K=equilibrium association constant; and n=number of ligand binding sites per receptor molecule. By graphical analysis, r/c is plotted on the Y-axis versus r on the X-axis, thus producing a Scatchard plot. Affinity measurement by Scatchard analysis is well known in the art. See, e.g., van Erp et al., J. Immunoassay 12: 425-43, 1991; Nelson and Griswold, Comput. Methods Programs Biomed. 27: 65-8, 1988. In an alternative, affinity can be measured by isothermal titration calorimetry (ITC). In a typical ITC experiment, a solution of ligand is titrated into a solution of its cognate target. The heat released upon their interaction (ΔH) is monitored over time. As successive amounts of the ligand are titrated into the ITC cell, the quantity of heat absorbed or released is in direct proportion to the amount of binding. As the system reaches saturation, the heat signal diminishes until only heats of dilution are observed. A binding curve is then obtained from a plot of the heats from each injection against the ratio of ligand and binding partner in the cell. The binding curve is analyzed with the appropriate binding model to determine KB, n and ΔH. Note that KB=1/Kd.
  • The term “subject” as used herein refers to a human or non-human organism. Thus, the methods and compositions described herein are applicable to both human and veterinary disease. In certain embodiments, subjects are “patients,” i.e., living humans that are receiving medical care for a disease or condition. This includes persons with no defined illness who are being investigated for signs of pathology. Preferred are subjects who have an existing diagnosis of a particular cancer which is being targeted by the compositions and methods of the present invention. Preferred cancers for treatment with the compositions described herein include, but are not limited to prostate cancer, renal carcinoma, melanoma, pancreatic cancer, cervical cancer, ovarian cancer, colon cancer, head & neck cancer, lung cancer and breast cancer.
  • “Therapeutically effective amount” is defined as an amount of a reagent or pharmaceutical composition that is sufficient to show a patient benefit, i.e., to cause a decrease, prevention, or amelioration of the symptoms of the condition being treated. When the agent or pharmaceutical composition comprises a diagnostic agent, a “diagnostically effective amount” is defined as an amount that is sufficient to produce a signal, image, or other diagnostic parameter. Effective amounts of the pharmaceutical formulation will vary according to factors such as the degree of susceptibility of the individual, the age, gender, and weight of the individual, and idiosyncratic responses of the individual. “Effective amount” encompasses, without limitation, an amount that can ameliorate, reverse, mitigate, prevent, or diagnose a symptom or sign of a medical condition or disorder or a causative process thereof. Unless dictated otherwise, explicitly or by context, an “effective amount” is not limited to a minimal amount sufficient to ameliorate a condition.
  • “Treatment” or “treating” (with respect to a condition or a disease) is an approach for obtaining beneficial or desired results including and preferably clinical results. For purposes of this invention, beneficial or desired results with respect to a disease include, but are not limited to, one or more of the following: preventing a disease, improving a condition associated with a disease, curing a disease, lessening severity of a disease, delaying progression of a disease, alleviating one or more symptoms associated with a disease, increasing the quality of life of one suffering from a disease, and/or prolonging survival. Likewise, for purposes of this invention, beneficial or desired results with respect to a condition include, but are not limited to, one or more of the following: preventing a condition, improving a condition, curing a condition, lessening severity of a condition, delaying progression of a condition, alleviating one or more symptoms associated with a condition, increasing the quality of life of one suffering from a condition, and/or prolonging survival. For instance, in embodiments where the compositions described herein are used for treatment of cancer, the beneficial or desired results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cells, reducing metastasis of neoplastic cells found in cancers, shrinking the size of a tumor, decreasing symptoms resulting from the cancer, increasing the quality of life of those suffering from the cancer, decreasing the dose of other medications required to treat the disease, delaying the progression of the cancer, and/or prolonging survival of patients having cancer. Depending on the context, “treatment” of a subject can imply that the subject is in need of treatment, e.g., in the situation where the subject comprises a disorder expected to be ameliorated by administration of a reagent.
  • “Vaccine” encompasses preventative vaccines. Vaccine also encompasses therapeutic vaccines, e.g., a vaccine administered to a mammal that comprises a condition or disorder associated with the antigen or epitope provided by the vaccine.
  • Cyclic Purine Dinucleotides
  • Prokaryotic as well as eukaryotic cells use various small molecules for cell signaling and intra- and intercellular communication. Cyclic nucleotides like cGMP, cAMP, etc. are known to have regulatory and initiating activity in pro- and eukaryotic cells. Unlike eukaryotic cells, prokaryotic cells also use cyclic purine dinucleotides as regulatory molecules. In prokaryotes, the condensation of two GTP molecules is catalyst by the enzyme diguanylate cyclase (DGC) to give c-diGMP, which represents an important regulator in bacteria.
  • Recent work suggests that cyclic diGMP or analogs thereof can also stimulate or enhance immune or inflammatory response in a patient or can enhance the immune response to a vaccine by serving as an adjuvant in mammals. Cytosolic detection of pathogen-derived DNA requires signaling through TANK binding kinase 1 (TBK1) and its downstream transcription factor, IFN-regulatory factor 3 (IRF3). A transmembrane protein called STING (stimulator of IFN genes; also known as MITA, ERIS, MPYS and TMEM173) functions as the signaling receptor for these cyclic purine dinucleotides, causing stimulation of the TBK1-IRF3 signalling axis and a STING-dependent type I interferon response. See, e.g., FIG. 1. Burdette et al., Nature 478: 515-18, 2011 demonstrated that STING binds directly to cyclic diguanylate monophosphate, but not to other unrelated nucleotides or nucleic acids.
  • Cyclic purine dinucleotides for use as precursors to derive the CDNs of the present invention are described in some detail in, e.g., Gao et al., Cell (2013) 153: doi: 10.1016/j.cell.2013.04.046; U.S. Pat. Nos. 7,709,458 and 7,592,326; WO2007/054279; and Yan et al., Bioorg. Med. Chem Lett. 18: 5631 (2008), each of which is hereby incorporated by reference. These CDNs may be modified using standard organic chemistry techniques in order to produce the CDNs of the present invention.
  • Preferred purines include, but are not limited to, adenine, guanine, inosine, hypoxanthine, xanthine, isoguanine, etc. The CDNs of the present invention are preferably phosphorothioate analogues, and most preferably substantially pure Sp,Sp, Rp,Rp, SpRp, or Rp,Sp stereoisomers thereof.
  • As denoted in the structures, each ribose comprises a 2′ or 3′ hydroxyl which may be substituted. As described hereinafter, the CDNs of the present invention can comprise a substitution at one or both of these 2′ or 3′ hydroxyls (which is not part of the cyclic linkage) which provide a prodrug leaving group or other modification which affects activity, solubility, bioavailability, etc. The term “prodrug” as used herein refers to a modification of contemplated compounds, wherein the modified compound exhibits less pharmacological activity (as compared to the modified compound) and wherein the modified compound is converted within the body (e.g., in a target cell or target organ) back into the unmodified form through enzymatic or non-enzymatic reactions. In certain embodiments, the hydroxyl on one ribose comprises a prodrug leaving group. Prodrugs can modify the physicochemical, biopharmaceutic, and pharmacokinetic properties of drugs. Traditional prodrugs are classified as drugs that are activated by undergoing transformation in vivo to form the active drug. Reasons for prodrug development are typically poor aqueous solubility, chemical instability, low oral bioavailability, lack of blood brain barrier penetration, and high first pass metabolism associated with the parent drug. Suitable prodrug moieties are described in, for example, “Prodrugs and Targeted Delivery,” J. Rautico, Ed., John Wiley & Sons, 2011.
  • Preferred cyclic purine dinucleotides are phosphorothioate analogues, referred to herein as “thiophosphates”. Phosphorothioates are a variant of normal nucleotides in which one of the nonbridging oxygens is replaced by a sulfur. The sulfurization of the internucleotide bond dramatically reduces the action of endo- and exonucleases, including 5′ to 3′ and 3′ to 5′ DNA POL 1 exonuclease, nucleases S1 and P1, RNases, serum nucleases and snake venom phosphodiesterase. In addition, the potential for crossing the lipid bilayer increases.
  • A phosphorothioate linkage in inherently chiral. The skilled artisan will recognize that the phosphates in this structure may each exist in R or S forms. Thus, Rp,Rp, Sp,Sp, Sp,Rp, and Rp,Sp forms are possible.
  • As noted above, cyclic purine dinucleotides of the present invention comprise 2′-O— and 3′-O— substituent forms of CDNs, and in particular CDN thiophosphates. Additional stability and bioavailability can be provided by the substitution of the 2′-OH of the ribose moiety. Substituent groups amenable herein include without limitation, halogen, hydroxyl, alkyl, alkenyl, alkynyl, acyl (—C(O)Raa), carboxyl (—C(O)O—Raa), aliphatic groups, alicyclic groups, alkoxy, substituted oxy (—O—Raa), aryl, aralkyl, heterocyclic radical, heteroaryl, heteroarylalkyl, amino (—N(Rbb)(Rcc)), imino(=NRbb), amido (—C(O)N(Rbb)(RcC) or —N(Rbb)C(O)Raa), azido (—N3), nitro (—NO2), cyano (—CN), carbamido (—OC(O)N(Rbb)(Rcc) or —N(Rbb)C(O)ORaa), ureido (—N(Rbb)C(O)—N(Rbb)(Rcc)), thioureido (—N(Rbb)C(S)N(Rbb)(Rcc)), guanidinyl (—N(Rbb)C(═NRbb)N(Rbb)(Rcc)), amidinyl (—C(═NRbb)N(Rbb)(RcC) or —N(Rbb)C(═NRbb)(Raa)), thiol (—SRbb), sulfinyl (—S(O)Rbb), sulfonyl (—S(O)2Rb) and sulfonamidyl (—S(O)2N(Rbb)(RcC) or —N(Rbb)S(O)2Rbb). Wherein each Raa, Rbb and RcC is, independently, H, an optionally linked chemical functional group or a further substituent group with a preferred list including without limitation, H, alkyl, alkenyl, alkynyl, aliphatic, alkoxy, acyl, aryl, aralkyl, heteroaryl, alicyclic, heterocyclic and heteroarylalkyl. Selected substituents within the compounds described herein are present to a recursive degree.
  • The term “alkyl,” as used herein, refers to a saturated straight or branched hydrocarbon radical containing up to twenty four carbon atoms. Examples of alkyl groups include without limitation, methyl, ethyl, propyl, butyl, isopropyl, n-hexyl, octyl, decyl, dodecyl and the like. Alkyl groups typically include from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred. The term “lower alkyl” as used herein includes from 1 to about 6 carbon atoms. Alkyl groups as used herein may optionally include one or more further substituent groups.
  • The term “alkenyl,” as used herein, refers to a straight or branched hydrocarbon chain radical containing up to twenty four carbon atoms and having at least one carbon-carbon double bond. Examples of alkenyl groups include without limitation, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, dienes such as 1,3-butadiene and the like. Alkenyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred. Alkenyl groups as used herein may optionally include one or more further substituent groups.
  • The term “alkynyl,” as used herein, refers to a straight or branched hydrocarbon radical containing up to twenty four carbon atoms and having at least one carbon-carbon triple bond. Examples of alkynyl groups include, without limitation, ethynyl, 1-propynyl, 1-butynyl, and the like. Alkynyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred. Alkynyl groups as used herein may optionally include one or more further substituent groups.
  • The term “acyl,” as used herein, refers to a radical formed by removal of a hydroxyl group from an organic acid and has the general Formula —C(O)—X where X is typically aliphatic, alicyclic or aromatic. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates, aliphatic phosphates and the like. Acyl groups as used herein may optionally include further substituent groups.
  • The term “alicyclic” refers to a cyclic ring system wherein the ring is aliphatic. The ring system can comprise one or more rings wherein at least one ring is aliphatic. Preferred alicyclics include rings having from about 5 to about 9 carbon atoms in the ring. Alicyclic as used herein may optionally include further substituent groups.
  • The term “aliphatic,” as used herein, refers to a straight or branched hydrocarbon radical containing up to twenty four carbon atoms wherein the saturation between any two carbon atoms is a single, double or triple bond. An aliphatic group preferably contains from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred. The straight or branched chain of an aliphatic group may be interrupted with one or more heteroatoms that include nitrogen, oxygen, sulfur and phosphorus. Such aliphatic groups interrupted by heteroatoms include without limitation, polyalkoxys, such as polyalkylene glycols, polyamines, and polyimines. Aliphatic groups as used herein may optionally include further substituent groups.
  • The term “alkoxy,” as used herein, refers to a radical formed between an alkyl group and an oxygen atom wherein the oxygen atom is used to attach the alkoxy group to a parent molecule. Examples of alkoxy groups include without limitation, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, neopentoxy, n-hexoxy and the like. Alkoxy groups as used herein may optionally include further substituent groups.
  • The term “aminoalkyl” as used herein, refers to an amino substituted C\-Cn alkyl radical. The alkyl portion of the radical forms a covalent bond with a parent molecule. The amino group can be located at any position and the aminoalkyl group can be substituted with a further substituent group at the alkyl and/or amino portions.
  • The terms “aralkyl” and “arylalkyl,” as used herein, refer to an aromatic group that is covalently linked to a C\-Cn alkyl radical. The alkyl radical portion of the resulting aralkyl (or arylalkyl) group forms a covalent bond with a parent molecule. Examples include without limitation, benzyl, phenethyl and the like. Aralkyl groups as used herein may optionally include further substituent groups attached to the alkyl, the aryl or both groups that form the radical group.
  • The terms “aryl” and “aromatic,” as used herein, refer to a mono- or polycyclic carbocyclic ring system radicals having one or more aromatic rings. Examples of aryl groups include without limitation, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like. Preferred aryl ring systems have from about 5 to about 20 carbon atoms in one or more rings. Aryl groups as used herein may optionally include further substituent groups.
  • The terms “halo” and “halogen,” as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.
  • The terms “heteroaryl,” and “heteroaromatic,” as used herein, refer to a radical comprising a mono- or poly-cyclic aromatic ring, ring system or fused ring system wherein at least one of the rings is aromatic and includes one or more heteroatoms. Heteroaryl is also meant to include fused ring systems including systems where one or more of the fused rings contain no heteroatoms. Heteroaryl groups typically include one ring atom selected from sulfur, nitrogen or oxygen. Examples of heteroaryl groups include without limitation, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl and the like. Heteroaryl radicals can be attached to a parent molecule directly or through a linking moiety such as an aliphatic group or hetero atom. Heteroaryl groups as used herein may optionally include further substituent groups.
  • The term “heteroarylalkyl,” as used herein, refers to a heteroaryl group as previously defined that further includes a covalently attached C1-C12 alkyl radical. The alkyl radical portion of the resulting heteroarylalkyl group is capable of forming a covalent bond with a parent molecule. Examples include without limitation, pyridinylmethyl, pyrimidinylethyl, napthyridinylpropyl and the like. Heteroarylalkyl groups as used herein may optionally include further substituent groups on one or both of the heteroaryl or alkyl portions.
  • As noted above, preferred cyclic purine dinucleotides also include prodrug forms of CDNs, and in particular CDN thiophosphates. Produrgs can modify the physicochemical, biopharmaceutic, and pharmacokinetic properties of drugs. Traditional prodrugs are classified as drugs that are activated by undergoing transformation in vivo to form the active drug. Reasons for prodrug development are typically poor aqueous solubility, chemical instability, low oral bioavailability, lack of blood brain barrier penetration, and high first pass metabolism associated with the parent drug. Suitable prodrug moieties are described in, for example, “Prodrugs and Targeted Delivery,” J. Rautico, Ed., John Wiley & Sons, 2011.
  • The term “substantially pure” as used herein with regard to cyclic purine dinucleotides refers to an Rp,Rp or Rp,Sp form which is at least 75% pure relative to other possible stereochemistries at the chiral centers indicated in the figure above. By way of example, a “substantially pure Rp,Rp c-di-GMP thiophosphate” would be at least 75% pure with regard to the Rp,Sp and Sp,Sp forms of c-di-GMP thiophosphate. In preferred embodiments, a substantially pure cyclic purine dinucleotide is at least 85% pure, at least 90% pure, at least 95% pure, at least 97% pure, and at least 99% pure. While a substantially pure cyclic purine dinucleotide preparation of the invention is “stereochemically pure,” this is not meant to indicate that all CDNs within the preparation having a particular stereochemistry at these chiral centers are otherwise identical. For example, a substantially pure cyclic purine dinucleotide preparation may contain a combination of Rp,Rp c-di-GMP thiophosphate and Rp,Rp c-di-AMP thiophosphate and still be a substantially pure cyclic purine dinucleotide preparation. Such a preparation may also include other components as described hereinafter that are advantageous for patient treatment, provided that all CDNs within the preparation having a particular stereochemistry at these chiral centers.
  • The CDN compositions described herein can be administered to a host, either alone or in combination with a pharmaceutically acceptable excipient, in an amount sufficient to induce, modify, or stimulate an appropriate immune response. The immune response can comprise, without limitation, specific immune response, non-specific immune response, both specific and non-specific response, innate response, primary immune response, adaptive immunity, secondary immune response, memory immune response, immune cell activation, immune cell proliferation, immune cell differentiation, and cytokine expression. In certain embodiments, the CDN compositions are administered in conjunction with one or more additional compositions including vaccines intended to stimulate an immune response to one or more predetermined antigens; adjuvants; CTLA-4 and PD-1 pathway antagonists, lipids, liposomes, chemotherapeutic agents, immunomodulatory cell lines, etc.
  • The CDN compositions may be administered before, after, and/or together with an additional therapeutic or prophylactic composition or modality. These include, without limitation, B7 costimulatory molecule, interleukin-2, interferon-γ, GM-CSF, CTLA-4 antagonists, OX-40/OX-40 ligand, CD40/CD40 ligand, sargramostim, levamisol, vaccinia virus, Bacille Calmette-Guerin (BCG), liposomes, alum, Freund's complete or incomplete adjuvant, detoxified endotoxins, mineral oils, surface active substances such as lipolecithin, pluronic polyols, polyanions, peptides, and oil or hydrocarbon emulsions. Carriers for inducing a T cell immune response which preferentially stimulate a cytolytic T cell response versus an antibody response are preferred, although those that stimulate both types of response can be used as well. In cases where the agent is a polypeptide, the polypeptide itself or a polynucleotide encoding the polypeptide can be administered. The carrier can be a cell, such as an antigen presenting cell (APC) or a dendritic cell. Antigen presenting cells include such cell types as macrophages, dendritic cells and κ cells. Other professional antigen-presenting cells include monocytes, marginal zone Kupffer cells, microglia, Langerhans' cells, interdigitating dendritic cells, follicular dendritic cells, and T cells. Facultative antigen-presenting cells can also be used. Examples of facultative antigen-presenting cells include astrocytes, follicular cells, endothelium and fibroblasts. The carrier can be a bacterial cell that is transformed to express the polypeptide or to deliver a polynucleoteide which is subsequently expressed in cells of the vaccinated individual. Adjuvants, such as aluminum hydroxide or aluminum phosphate, can be added to increase the ability of the vaccine to trigger, enhance, or prolong an immune response. Additional materials, such as cytokines, chemokines, and bacterial nucleic acid sequences, like CpG, a toll-like receptor (TLR) 9 agonist as well as additional agonists for TLR 2, TLR 4, TLR 5, TLR 7, TLR 8, TLR9, including lipoprotein, LPS, monophosphoryl lipid A, lipoteichoic acid, imiquimod, resiquimod, and in addition retinoic acid-inducible gene I (RIG-I) agonists such as poly I:C, used separately or in combination with the described compositions are also potential adjuvants. Other representative examples of adjuvants include the synthetic adjuvant QS-21 comprising a homogeneous saponin purified from the bark of Quillaja saponaria and Corynebacterium parvum (McCune et al., Cancer, 1979; 43:1619). It will be understood that the adjuvant is subject to optimization. In other words, the skilled artisan can engage in routine experimentation to determine the best adjuvant to use.
  • Methods for co-administration with an additional therapeutic agent are well known in the art (Hardman, et al. (eds.) (2001) Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed., McGraw-Hill, New York, N.Y.; Poole and Peterson (eds.) (2001) Pharmacotherapeutics for Advanced Practice:A Practical Approach, Lippincott, Williams & Wilkins, Phila., Pa.; Chabner and Longo (eds.) (2001) Cancer Chemotherapy and Biotherapy, Lippincott, Williams & Wilkins, Phila., Pa.).
  • Because of the adjuvant properties of the compounds of the present invention, their use may also combined with other therapeutic modalities including other vaccines, adjuvants, antigen, antibodies, and immune modulators. Examples are provided below.
  • Adjuvants
  • In addition to the cyclic purine dinuclotide(s) described above, the compositions of the present invention may further comprise one or more additional substances which, because of their nature, can act to stimulate or otherwise utilize the immune system to respond to the cancer antigens present on the inactivated tumor cell(s). Such adjuvants include, but are not limited to, lipids, liposomes, inactivated bacteria which induce innate immunity (e.g., inactivated or attenuated Listeria monocytogenes), compositions which mediate innate immune activation via Toll-like Receptors (TLRs), (NOD)-like receptors (NLRs), Retinoic acid inducible gene-based (RIG)-I-like receptors (RLRs), and/or C-type lectin receptors (CLRs). Examples of PAMPs include lipoproteins, lipopolypeptides, peptidoglycans, zymosan, lipopolysaccharide, neisserial porins, flagellin, profillin, galactoceramide, muramyl dipeptide. Peptidoglycans, lipoproteins, and lipoteichoic acids are cell wall components of Gram-positive. Lipopolysaccharides are expressed by most bacteria, with MPL being one example. Flagellin refers to the structural component of bacterial flagella that is secreted by pathogenic and commensal bacterial. α-Galactosylceramide (α-GalCer) is an activator of natural killer T (NKT) cells. Muramyl dipeptide is a bioactive peptidoglycan motif common to all bacteria. This list is not meant to be limiting. Preferred adjuvant compositions are described below.
  • CTLA-4 and PD-1 Pathway Antagonists
  • CTLA-4 is thought to be an important negative regulator of the adaptive immune response. Activated T cells upregulate CTLA-4, which binds CD80 and CD86 on antigen-presenting cells with higher affinity than CD28, thus inhibiting T-cell stimulation, IL-2 gene expression and T-cell proliferation. Anti-tumor effects of CTLA4 blockade have been observed in murine models of colon carcinoma, metastatic prostate cancer, and metastatic melanoma.
  • Ipilimumab (Yervoy™) and tremelimumab are humanized monoclonal antibodies that bind to human CTLA4 and prevent its interaction with CD80 and CD86. Phase I and II studies using ipilimumab and tremelimumab have demonstrated clinical activity in cancer patients. Other negative immune regulators which may be targeted by a similar strategy include programmed cell death 1, B and T lymphocyte attenuator, transforming growth factor beta β, interleukin-10, and vascular endothelial growth factor.
  • PD-1 is another negative regulator of adaptive immune response that is expressed on activated T-cells. PD-1 binds to B7-H1 and B7-DC, and the engagement of PD-1 suppresses T-cell activation. Anti-tumor effects have been demonstrated with PD-1 pathway blockade. BMS-936558, MK3475, CT-011, AMP-224 and MDX-1106 have been reported in the literature to be examples of PD-1 pathway blockers which may find use in the present invention.
  • TLR Agonists
  • The term “Toll like receptor” (or “TLR”) as used herein refers to a member of the Toll-like receptor family of proteins or a fragment thereof that senses a microbial product and/or initiates an adaptive immune response. In one embodiment, a TLR activates a dendritic cell (DC). Toll like receptors (TLRs) are a family of pattern recognition receptors that were initially identified as sensors of the innate immune system that recognize microbial pathogens. TLRs comprise a family of conserved membrane spanning molecules containing an ectodomain of leucine-rich repeats, a transmembrane domain and an intracellular TIR (Toll/IL-1R) domain. TLRs recognize distinct structures in microbes, often referred to as “PAMPs” (pathogen associated molecular patterns). Ligand binding to TLRs invokes a cascade of intra-cellular signaling pathways that induce the production of factors involved in inflammation and immunity.
  • In humans, ten TLR have been identified. TLRs that are expressed on the surface of cells include TLR-1,-2,-4,-5, and -6, while TLR-3, -7/8, and -9 are expressed with the ER compartment. Human dendritic cell subsets can be identified on the basis of distinct TLR expression patterns. By way of example, the myeloid or “conventional” subset of DC (mDC) expresses TLRs 1-8 when stimulated, and a cascade of activation markers (e.g. CD80, CD86, MHC class I and II, CCR7), pro-inflammatory cytokines, and chemokines are produced. A result of this stimulation and resulting expression is antigen-specific CD4+ and CD8+ T cell priming. These DCs acquire an enhanced capacity to take up antigens and present them in an appropriate form to T cells. In contrast, the plasmacytoid subset of DC (pDC) expresses only TLR7 and TLR9 upon activation, with a resulting activation of NK cells as well as T-cells. As dying tumor cells may adversely affect DC function, it has been suggested that activating DC with TLR agonists may be beneficial for priming anti-tumor immunity in an immunotherapy approach to the treatment of cancer. It has also been suggested that successful treatment of breast cancer using radiation and chemotherapy requires TLR4 activation.
  • TLR agonists known in the art and finding use in the present invention include, but are not limited to, the following:
  • Pam3Cys, a TLR-1/2 agonist;
    CFA, a TLR-2 agonist;
    MALP2, a TLR-2 agonist;
    Pam2Cys, a TLR-2 agonist;
    FSL-1, a TLR-2 agonist;
    Hib-OMPC, a TLR-2 agonist;
    polyribosinic:polyribocytidic acid (Poly I:C), a TLR-3 agonist;
    polyadenosine-polyuridylic acid (poly AU), a TLR-3 agonist;
    Polyinosinic-Polycytidylic acid stabilized with poly-L-lysine and carboxymethylcellulose (Hiltonol®), a TLR-3 agonist;
    monophosphoryl lipid A (MPL), a TLR-4 agonist; LPS, a TLR-4 agonist;
    bacterial flagellin, a TLR-5 agonist;
    sialyl-Tn (STn), a carbohydrate associated with the MUC1 mucin on a number of human cancer cells and a TLR-4 agonist;
    imiquimod, a TLR-7 agonist;
    resiquimod, a TLR-7/8 agonist;
    loxoribine, a TLR-7/8 agonist; and
    unmethylated CpG dinucleotide (CpG-ODN), a TLR-9 agonist.
  • Because of their adjuvant qualities, TLR agonists are preferably used in combinations with other vaccines, adjuvants and/or immune modulators, and may be combined in various combinations. Thus, in certain embodiments, the cyclic purine dinucleotides that bind to STING and induces STING-dependent TBK1 activation and an inactivated tumor cell which expresses and secretes one or more cytokines which stimulate dendritic cell induction, recruitment and/or maturation, as described herein can be administered together with one or more TLR agonists for therapeutic purposes.
  • Antibody Therapeutics
  • Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) is a mechanism of cell-mediated immune defense whereby an effector cell of the immune system actively lyses a target cell, whose membrane-surface antigens have been bound by specific antibodies. It is one of the mechanisms through which antibodies, as part of the humoral immune response, can act to limit and contain infection. Classical ADCC is mediated by natural killer (NK) cells; macrophages, neutrophils and eosinophils can also mediate ADCC. ADCC is an important mechanism of action of therapeutic monoclonal antibodies, including trastuzumab and rituximab, against tumors. Compounds of the present invention may act to potentiate ADCC.
  • The following are an exemplary list of antibodies which may be used together with the compounds of the present invention.
  • Muromonab-CD3: Used to prevent acute rejection of organ, e.g., kidney, transplants. The humanized versions show promise in inhibiting the autoimmune destruction of beta cells in Type 1 diabetes mellitus.
  • Infliximab (Remicade®) and adalimumab (Humira®): Bind to tumor necrosis factor-alpha (TNF-α). Used in some inflammatory diseases such as rheumatoid arthritis, psoriasis, Crohns disease.
  • Omalizumab (Xolair®). Binds to IgE thus preventing IgE from binding to mast cells. Used against allergic asthma.
  • Daclizumab (Zenapax®). Binds to part of the IL-2 receptor exposed at the surface of activated T cells. Used to prevent acute rejection of transplanted kidneys.
  • Rituximab (trade name=Rituxan®). Binds to the CD20 molecule found on most B-cells and is used to treat B-cell lymphomas.
  • Ibritumomab (trade name=Zevalin®). This is a monoclonal antibody against the CD20 molecule on B cells (and lymphomas) conjugated to isotopes. Given to the lymphoma patient supplemented with Rituxan.
  • Tositumomab (Bexxar®). This is a conjugate of a monoclonal antibody against CD20 and the radioactive isotope iodine-131 (131I).
  • Cetuximab (Erbitux®). Blocks HER1, a receptor for epidermal growth factor (EGF) that is found on some tumor cells (some breast cancers, lymphomas).
  • Trastuzumab (Herceptin®). Blocks HER2, a growth factor receptor over-expressed in some 20% of breast cancers.
  • Adcetris®. A conjugate of a monoclonal antibody that binds CD30, a cell-surface molecule expressed by the cells of some lymphomas but not found on the normal stem cells needed to repopulate the bone marrow.
  • Alemtuzumab (Campath-1H®). Binds to CD52, a molecule found on lymphocytes and depletes both T cells and B cells. Has produced complete remission of chronic lymphocytic leukemia and shows promise in preventing rejection of kidney transplants.
  • Lym-1 (Oncolym®). Binds to the HLA-DR-encoded histocompatibility antigen that can be expressed at high levels on lymphoma cells.
  • Ipilimumab (Yervoy®) that acts to enhance the body's own immune response to tumors.
  • Vitaxin. Binds to a vascular integrin (alpha-v/beta-3) found on the blood vessels of tumors but not on the blood vessels supplying normal tissues. In Phase II clinical trials, Vitaxin has shown some promise in shrinking solid tumors without harmful side effects.
  • Bevacizumab (Avastin®). Binds to vascular endothelial growth factor (VEGF) preventing it from binding to its receptor. Used for the treatment of colorectal cancers.
  • Abciximab (ReoPro®). Inhibits the clumping of platelets by binding the receptors on their surface that normally are linked by fibrinogen. Helpful in preventing reclogging of the coronary arteries in patients who have undergone angioplasty.
  • Delivery Agents
  • Liposomes are vesicles formed from one (“unilamellar”) or more (“multilamellar”) layers of phospholipid. Because of the amphipathic character of the phospholipid building blocks, liposomes typically comprise a hydrophilic layer presenting a hydrophilic external face and enclosing a hydrophilic core. The versatility of liposomes in the incorporation of hydrophilic/hydrophobic components, their non-toxic nature, biodegradability, biocompatibility, adjuvanticity, induction of cellular immunity, property of sustained release and prompt uptake by macrophages, makes them attractive candidates for the delivery of antigens.
  • WO2010/104833, which is incorporated by reference herein in its entirety, describes suitable liposomal preparations. Such liposomal formulations, referred to herein as VesiVax® (Molecular Express, Inc.), with our without the “immunogenic polypeptide(s) or carbohydrate(s)” referred to above, can contain one or more additional components such as peptidoglycan, lipopeptide, lipopolysaccharide, monophosphoryl lipid A, lipoteichoic acid, resiquimod, imiquimod, flagellin, oligonucleotides containing unmethylated CpG motifs, beta-galactosylceramide, muramyl dipeptide, all-trans retinoic acid, double-stranded viral RNA, heat shock proteins, dioctadecyldimethylammonium bromide, cationic surfactants, toll-like receptor agonists, dimyristoyltrimethylammoniumpropane, and nod-like receptor agonists. Advantageously, these liposomal formulations can be used to deliver one or more cyclic purine dinucleotides in accordance with the present invention.
  • Moreover, while the liposomal formulations discussed above employ a” steroid derivative” as an anchor for attaching an immunogenic polypeptide or carbohydrate to a liposome, the steroid may simply be provided as an unconjugated steroid such as cholesterol.
  • Suitable methods for preparing liposomes from lipid mixtures are well known in the art. See, e.g., Basu & Basu, Liposome Methods and Protocols (Methods in Molecular Biology), Humana Press, 2002; Gregoriadis, Liposome Technology, 3rd Edition, Informa HealthCare, 2006. Preferred methods include extrusion, homogenization, and sonication methods described therein. An exemplary method for preparing liposomes for use in the present invention, which comprises drying a lipid mixture, followed by hydration in an aqueous vehicle and sonication to form liposomes, is described in WO2010/104833.
  • In certain embodiments, the liposomes are provided within a particular average size range. Liposome size can be selected, for example, by extrusion of an aqueous vehicle comprising liposomes through membranes having a preselected pore size and collecting the material flowing through the membrane. In preferred embodiments, the liposomes are selected to be substantially between 50 and 500 nm in diameter, more preferably substantially between 50 and 200 nm in diameter, and most preferably substantially between 50 and 150 nm in diameter. The term “substantially” as used herein in this context means that at least 75%, more preferably 80%, and most preferably at least 90% of the liposomes are within the designated range.
  • Other lipid and lipid-like adjuvants which may find use in the present invention include oil-in-water (o/w) emulsions (see, e.g., Muderhwa et al., J. Pharmaceut. Sci. 88: 1332-9, 1999)), VesiVax® TLR (Molecular Express, Inc.), digitonin (see, e.g., U.S. Pat. No. 5,698,432), and glucopyranosyl lipids (see, e.g., United States Patent Application 20100310602).
  • Nanoparticles also represent drug delivery systems suitable for most administration routes. Over the years, a variety of natural and synthetic polymers have been explored for the preparation of nanoparticles, of which Poly(lactic acid) (PLA), Poly(glycolic acid) (PGA), and their copolymers (PLGA) have been extensively investigated because of their biocompatibility and biodegradability. Nanoparticles and other nanocarriers act as potential carries for several classes of drugs such as anticancer agents, antihypertensive agents, immunomodulators, and hormones; and macromolecules such as nucleic acids, proteins, peptides, and antibodies. See, e.g., Crit. Rev. Ther. Drug Carrier Syst. 21:387-422, 2004; Nanomedicine: Nanotechnology, Biology and Medicine 1:22-30, 2005.
  • Chemotherapeutic Agents
  • In additional embodiments the methods further involve administering to the subject an effective amount of one or more chemotherapeutics as an additional treatment. In certain embodiments the one or more chemotherapeutics is selected from abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly-1-Lproline-t-butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3′,4′-didehydro-4′-deoxy-8′-norvin-caleukoblastine, docetaxol, doxetaxel, cyclophosphamide, carboplatin, carmustine, cisplatin, cryptophycin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, daunorubicin, decitabine dolastatin, doxorubicin (adriamycin), etoposide, 5-fluorouracil, finasteride, flutamide, hydroxyurea and hydroxyureataxanes, ifosfamide, liarozole, lonidamine, lomustine (CCNU), MDV3100, mechlorethamine (nitrogen mustard), melphalan, mivobulin isethionate, rhizoxin, sertenef, streptozocin, mitomycin, methotrexate, taxanes, nilutamide, onapristone, paclitaxel, prednimustine, procarbazine, RPR109881, stramustine phosphate, tamoxifen, tasonermin, taxol, tretinoin, vinblastine, vincristine, vindesine sulfate, and vinflunine.
  • Immunomodulatory Cell Lines
  • By “inactivated tumor cell” is meant a tumor cell (either “autologous” or “allogeneic” to the patient) which has which been treated to prevent division of the cells. For purposes of the present invention, such cells preserve their immunogenicity and their metabolic activity. Such tumor cells are genetically modified to express a transgene which is expressed within a patient as part of cancer therapy. Thus, a composition or vaccine of the invention comprises neoplastic (e.g., tumor) cells that are autologous or allogeneic to the patient undergoing treatment and is most preferably the same general type of tumor cell as is afflicting the patient. For example, a patient suffering from melanoma will typically be administered a genetically modified cell derived from a melanoma. Methods for inactivating tumor cells for use in the present invention, such as the use of irradiation, are well known in the art.
  • The inactivated tumor cells of the present invention are administered to the patient together with one or more costimulatory molecules or agents. A preferred costimulatory agent comprises one or more cytokines which stimulate dendritic cell induction, recruitment, and/or maturation. Methods for assessing such costimulatory agents are well known in the literature. Induction and maturation of DCs is typically assessed by increased expression of certain membrane molecules such as CD80 and CD86, and/or secretion of pro-inflammatory cytokines, such as IL-12 and type I interferons following stimulation.
  • In preferred embodiments, the inactivated tumor cells themselves are modified to express and secrete one or more cytokines which stimulate dendritic cell induction, recruitment, and/or maturation. The present invention is described in exemplary terms with regard to the use of GM-CSF. Thus, by way of example, the tumor cell may express a transgene encoding GM-CSF as described in U.S. Pat. Nos. 5,637,483, 5,904,920, 6,277,368 and 6,350,445, as well as in US Patent Publication No. 20100150946, each of which is expressly incorporated by reference herein. A form of GM-CSF-expressing genetically modified cancer cells or a “cytokine-expressing cellular vaccine” for the treatment of pancreatic cancer is described in U.S. Pat. Nos. 6,033,674 and 5,985,290, both of which are expressly incorporated by reference herein.
  • Other suitable cytokines which may be expressed by such inactivated tumor cells and/or bystander cells instead of, or together with, GM-CSF include, but are not limited to, one or more of CD40 ligand, IL-12, CCL3, CCL20, and CCL21. This list is not meant to be limiting.
  • While it is preferred that the inactivated tumor cells administered to the subject express one or more cytokines of interest, the tumor cell line may be accompanied by an inactivated bystander cell line which expresses and secretes one or more cytokines which stimulate dendritic cell induction, recruitment, and/or maturation. The bystander cell line may provide all of the cytokines which stimulate dendritic cell induction, recruitment, and/or maturation, or may supplement cytokines which stimulate dendritic cell induction, recruitment, and/or maturation expressed and secreted by the inactivated tumor cells. By way of example, immunomodulatory cytokine-expressing bystander cell lines are disclosed in U.S. Pat. Nos. 6,464,973, and 8,012,469, Dessureault et al., Ann. Surg. Oncol. 14: 869-84, 2007, and Eager and Nemunaitis, Mol. Ther. 12: 18-27, 2005, each of which is expressly incorporated by reference herein.
  • By “Granulocyte-macrophage colony stimulating factor (GM-CSF) polypeptide” is meant a cytokine or fragment thereof having immunomodulatory activity and having at least about 85% amino acid sequence identity to GenBank Accession No. AAA52122.1.
  • Vaccines
  • In certain embodiments, the CDN compositions are administered in conjunction with one or more vaccines intended to stimulate an immune response to one or more predetermined antigens. Examples of target antigens that may find use in the invention are listed in the following table. The target antigen may also be a fragment or fusion polypeptide comprising an immunologically active portion of the antigens listed in the table. This list is not meant to be limiting.
  • TABLE 1
    Antigens.
    Antigen Reference
    Tumor antigens
    Mesothelin GenBank Acc. No. NM_005823;
    U40434; NM_013404; BC003512
    (see also, e.g., Hassan, et al. (2004)
    Clin. Cancer Res. 10:3937-3942;
    Muminova, et al. (2004) BMC
    Cancer 4:19; Iacobuzio-Donahue, et
    al. (2003) Cancer Res. 63:8614-8622).
    Wilms' tumor-1 WT-1 isoform A (GenBank Acc.
    associated protein Nos. NM_000378; NP_000369).
    (Wt-1), including WT-1 isoform B (GenBank Acc.
    isoform A; isoform B; Nos. NM_024424; NP_077742).
    isoform C; isoform D. WT-1 isoform C (GenBank Acc.
    Nos. NM_024425; NP_077743).
    WT-1 isoform D (GenBank Acc.
    Nos. NM_024426; NP_077744).
    Stratum corneum GenBank Acc. No. NM_005046;
    chymotryptic enzyme NM_139277; AF332583. See also,
    (SCCE), and variants e.g., Bondurant, et al. (2005) Clin.
    thereof. Cancer Res. 11:3446-3454; Santin,
    et al. (2004) Gynecol. Oncol.
    94:283-288; Shigemasa, et al. (2001)
    Int. J. Gynecol. Cancer 11:454-461;
    Sepehr, et al. (2001) Oncogene
    20:7368-7374.
    MHC class I See, e.g., Groh, et al. (2005) Proc.
    chain-related protein A Natl. Acad. Sci. USA 102:6461-
    (MICA); MHC class I 6466; GenBank Acc. Nos. NM_000247;
    chain-related protein A BC 016929; AY750850;
    (MICB). NM_005931.
    Gastrin and peptides Harris, et al. (2004) Cancer Res.
    derived from gastrin; 64:5624-5631; Gilliam, et al. (2004)
    gastrin/CCK-2 receptor Eur. J. Surg. Oncol. 30:536-543;
    (also known as Laheru and Jaffee (2005) Nature
    CCK-B). Reviews Cancer 5:459-467.
    Glypican-3 (an antigen GenBank Acc. No. NM_004484.
    of, e.g., hepatocellular Nakatsura, et al. (2003) Biochem.
    carcinoma and Biophys. Res. Commun. 306:16-25;
    melanoma). Capurro, et al. (2003)
    Gasteroenterol. 125:89-97; Nakatsura,
    et al. (2004) Clin. Cancer Res.
    10:6612-6621).
    Coactosin-like protein. Nakatsura, et al. (2002) Eur. J. Immunol.
    32:826-836; Laheru and Jaffee (2005)
    Nature Reviews Cancer 5:459-467.
    Prostate stem cell GenBank Acc. No. AF043498;
    antigen (PSCA). AR026974; AR302232 (see also, e.g.,
    Argani, et al. (2001) Cancer Res.
    61:4320-4324; Christiansen, et al.
    (2003) Prostate 55:9-19; Fuessel,
    et al. (2003) 23:221-228).
    Prostate acid Small, et al. (2000) J. Clin. Oncol.
    phosphatase (PAP); 18:3894-3903; Altwein and
    prostate-specific Luboldt (1999) Urol. Int. 63:62-71;
    antigen (PSA); PSM; Chan, et al. (1999) Prostate 41:99-
    PSMA. 109; Ito, et al. (2005) Cancer
    103:242-250; Schmittgen, et al. (2003)
    Int. J. Cancer 107:323-329; Millon,
    et al. (1999) Eur. Urol. 36:278-285.
    Six-transmembrane See, e.g., Machlenkin, et al. (2005)
    epithelial antigen of Cancer Res. 65:6435-6442;
    prostate (STEAP). GenBank Acc. No. NM_018234;
    NM_001008410; NM_182915;
    NM_024636; NM_012449; BC011802.
    Prostate carcinoma See, e.g., Machlenkin, et al. (2005)
    tumor antigen-1 Cancer Res. 65:6435-6442;
    (PCTA-1). GenBank Acc. No. L78132.
    Prostate See, e.g., Machlenkin, et al. (2005)
    tumor-inducing gene-1 Cancer Res. 65:6435-6442).
    (PTI-1).
    Prostate-specific gene See, e.g., Machlenkin, et al. (2005)
    with homology to Cancer Res. 65:6435-6442).
    G protein-coupled
    receptor.
    Prostase (an antrogen See, e.g., Machlenkin, et al. (2005) Cancer
    regulated serine Res. 65:6435-6442; GenBank Acc. No.
    protease). BC096178; BC096176; BC096175.
    Proteinase 3. GenBank Acc. No. X55668.
    Cancer-testis antigens, GenBank Acc. No. NM_001327
    e.g., NY-ESO-1; SCP- (NY-ESO-1) (see also, e.g., Li, et al.
    1; SSX-1; SSX-2; SSX- (2005) Clin. Cancer Res. 11:1809-1814;
    4; GAGE, CT7; CT8; Chen, et al. (2004) Proc.
    CT10; MAGE-1; Natl. Acad. Sci. USA. 101(25):9363-9368;
    MAGE-2; MAGE-3; Kubuschok, et al. (2004)
    MAGE-4; MAGE-6; Int. J. Cancer. 109:568-575; Scanlan,
    LAGE-1. et al. (2004) Cancer Immun.
    4:1; Scanlan, et al. (2002) Cancer Res.
    62:4041-4047; Scanlan, et al.
    (2000) Cancer Lett. 150:155-164; Dalerba,
    et al. (2001) Int. J. Cancer
    93:85-90; Ries, et al. (2005) Int. J.
    Oncol. 26:817-824.
    MAGE-A1, Otte, et al. (2001) Cancer Res. 61:6682-6687;
    MAGE-A2; Lee, et al. (2003) Proc.
    MAGE-A3; Natl. Acad. Sci. USA 100:2651-2656;
    MAGE-A4; Sarcevic, et al. (2003)
    MAGE-A6; Oncology 64:443-449; Lin, et al. (2004)
    MAGE-A9; Clin. Cancer Res. 10:5708-5716.
    MAGE-A10;
    MAGE-A12;
    GAGE-3/6;
    NT-SAR-35; BAGE;
    CA125.
    GAGE-1; GAGE-2; De Backer, et al. (1999) Cancer Res.
    GAGE-3; GAGE-4; 59:3157-3165; Scarcella, et al.
    GAGE-5; GAGE-6; (1999) Clin. Cancer Res. 5:335-341.
    GAGE-7; GAGE-8;
    GAGE-65; GAGE-11;
    GAGE-13; GAGE-7B.
    HIP1R; LMNA; Scanlan, et al. (2002) Cancer
    KIAA1416; Seb4D; Res. 62:4041-4047.
    KNSL6; TRIP4;
    MBD2; HCAC5;
    MAGEA3.
    DAM family of genes, Fleishhauer, et al. (1998) Cancer
    e.g., DAM-1; DAM-6. Res. 58:2969-2972.
    RCAS1. Enjoji, et al. (2004) Dig. Dis. Sci.
    49:1654-1656.
    RU2. Van Den Eynde, et al. (1999) J. Exp.
    Med. 190:1793-1800.
    CAMEL. Slager, et al. (2004) J. Immunol.
    172:5095-5102; Slager, et al. (2004)
    Cancer Gene Ther. 11:227-236.
    Colon cancer associated Scanlan, et al. (2002) Cancer
    antigens, e.g., Res. 62:4041-4047.
    NY-CO-8; NY-CO-9;
    NY-CO-13;
    NY-CO-16;
    NY-CO-20;
    NY-CO-38;
    NY-CO-45;
    NY-CO-9/HDAC5;
    NY-CO-41/MBD2;
    NY-CO-42/TRIP4;
    NY-CO-95/KIAA1416;
    KNSL6; seb4D.
    N-Acetylglucosaminyl- Dosaka-Akita, et al. (2004) Clin.
    tranferase V (GnT-V). Cancer Res. 10:1773-1779.
    Elongation factor 2 Renkvist, et al. (2001) Cancer
    mutated (ELF2M). Immunol Immunother. 50:3-15.
    HOM-MEL-40/SSX2 Neumann, et al. (2004) Int. J.
    Cancer 112:661-668; Scanlan, et al.
    (2000) Cancer Lett. 150:155-164.
    BRDT. Scanlan, et al. (2000) Cancer
    Lett. 150:155-164.
    SAGE; HAGE. Sasaki, et al. (2003) Eur. J. Surg.
    Oncol. 29:900-903.
    RAGE. See, e.g., Li, et al. (2004) Am. J.
    Pathol. 164:1389-1397; Shirasawa,
    et al. (2004) Genes to Cells 9:165-174.
    MUM-1 (melanoma Gueguen, et al. (1998) J. Immunol.
    ubiquitous mutated); 160:6188-6194; Hirose, et al.
    MUM-2; MUM-2 Arg- (2005) Int. J. Hematol. 81:48-57;
    Gly mutation; MUM-3. Baurain, et al. (2000) J. Immunol.
    164:6057-6066; Chiari, et al. (1999)
    Cancer Res. 59:5785-5792.
    LDLR/FUT fusion Wang, et al. (1999) J. Exp. Med.
    protein antigen of 189:1659-1667.
    melanoma.
    NY-REN series of Scanlan, et al. (2002) Cancer Res.
    renal cancer antigens. 62:4041-4047; Scanlan, et al.
    (1999) Cancer Res. 83:456-464.
    NY-BR series of breast Scanlan, et al. (2002) Cancer Res.
    cancer antigens, e.g., 62:4041-4047; Scanlan, et al.
    NY-BR-62; NY- (2001) Cancer Immunity 1:4.
    BR-75; NY-BR-85;
    NY-BR-62; NY-BR-85.
    BRCA-1; BRCA-2. Stolier, et al. (2004) Breast J.
    10:475-480; Nicoletto, et al. (2001)
    Cancer Treat Rev. 27:295-304.
    DEK/CAN fusion Von Lindern, et al. (1992) Mol. Cell.
    protein. Biol. 12:1687-1697.
    Ras, e.g., wild type ras, GenBank Acc. Nos. P01112;
    ras with mutations at P01116; M54969; M54968; P01111;
    codon 12, 13, 59, or 61, P01112; K00654. See also, e.g.,
    e.g., mutations G12C; GenBank Acc. Nos. M26261;
    G12D; G12R; G125; M34904; 1(01519; K01520; BC006499;
    G12V; G13D; A59T; NM_006270; NM_002890;
    Q61H. K-RAS; NM_004985; NM_033360;
    H-RAS; N-RAS. NM_176795; NM_005343.
    BRAF (an isoform of Tannapfel, et al. (2005) Am. J. Clin.
    RAF). Pathol. 123:256-2601; Tsao and Sober
    (2005) Dermatol. Clin. 23:323-333.
    Melanoma antigens, GenBank Acc. No. NM_206956;
    including HST-2 NM_206955; NM_206954;
    melanoma cell NM_206953; NM_006115;
    antigens. NM_005367; NM_004988; AY148486;
    U10340; U10339; M77481. See,
    e g., Suzuki, et al. (1999) J.
    Immunol. 163:2783-2791.
    Survivin GenBank Acc. No. AB028869; U75285
    (see also, e.g., Tsuruma, et al.
    (2004) J. Translational Med. 2:19
    (11 pages); Pisarev, et al. (2003)
    Clin. Cancer Res. 9:6523-6533;
    Siegel, et al. (2003) Br. J. Haematol.
    122:911-914; Andersen, et al. (2002)
    Histol. Histopathol. 17:669-675).
    MDM-2 NM_002392; NM_006878 (see also,
    e.g., Mayo, et al. (1997) Cancer
    Res. 57:5013-5016; Demidenko and
    Blagosklonny (2004) Cancer
    Res. 64:3653-3660).
    Methyl-CpG-binding Muller, et al. (2003) Br. J. Cancer
    proteins (MeCP2; 89:1934-1939; Fang, et al. (2004)
    MBD2). World J. Gastreenterol. 10:3394-3398.
    NA88-A. Moreau-Aubry, et al. (2000) J. Exp.
    Med. 191:1617-1624.
    Histone deacetylases Waltregny, et al. (2004) Eur. J.
    (HDAC), e.g., HDAC5. Histochem. 48:273-290; Scanlan, et
    al. (2002) Cancer Res. 62:4041-4047.
    Cyclophilin B (Cyp-B). Tamura, et al. (2001) Jpn. J. Cancer
    Res. 92:762-767.
    CA 15-3; CA 27.29. Clinton, et al. (2003) Biomed.
    Sci. Instrum. 39:408-414.
    Heat shock protein Faure, et al. (2004) Int. J. Cancer
    Hsp70. 108:863-870.
    GAGE/PAGE family, Brinkmann, et al. (1999) Cancer
    e.g., PAGE-1; PAGE-2; Res. 59:1445-1448.
    PAGE-3; PAGE-4;
    XAGE-1; XAGE-2;
    XAGE-3.
    MAGE-A, B, C, and D Lucas, et al. (2000) Int. J. Cancer
    families. MAGE-B5; 87:55-60; Scanlan, et al. (2001)
    MAGE-B6; Cancer Immun. 1:4.
    MAGE-C2;
    MAGE-C3; MAGE-3;
    MAGE-6.
    Kinesin 2; TATA Scanlan, et al. (2001) Cancer
    element modulatory Immun. 30:1-4.
    factor 1; tumor protein
    D53; NY
    Alpha-fetoprotein Grimm, et al. (2000) Gastroenterol.
    (AFP) 119:1104-1112.
    SART1; SART2; Kumamuru, et al. (2004) Int. J. Cancer
    SART3; ART4. 108:686-695; Sasatomi, et al.
    (2002) Cancer 94:1636-1641; Matsumoto,
    et al. (1998) Jpn. J. Cancer
    Res. 89:1292-1295; Tanaka, et al. (2000)
    Jpn. J. Cancer Res. 91:1177-
    1184.
    Preferentially expressed Matsushita, et al. (2003) Leuk.
    antigen of melanoma Lymphoma 44:439-444; Oberthuer, et
    (PRAME). al. (2004) Clin. Cancer Res. 10:4307-4313.
    Carcinoembryonic GenBank Acc. No. M29540; E03352;
    antigen (CEA), X98311; M17303 (see also,
    CAP1-6D enhancer e.g., Zaremba (1997) Cancer Res.
    agonist peptide. 57:4570-4577; Sarobe, et al. (2004)
    Curr. Cancer Drug Targets 4:443-454;
    Tsang, et al. (1997) Clin.
    Cancer Res. 3:2439-2449; Fong, et al.
    (2001) Proc. Natl. Acad. Sci.
    USA 98:8809-8814).
    HER-2/neu. Disis, et al. (2004) J. Clin. Immunol.
    24:571-578; Disis and Cheever
    (1997) Adv. Cancer Res. 71:343-371.
    Cdk4; cdk6; p16 Ghazizadeh, et al. (2005) Respiration
    (INK4); Rb protein. 72:68-73; Ericson, et al. (2003)
    Mol. Cancer Res. 1:654-664.
    TEL; AML1; Stams, et al. (2005) Clin. Cancer
    TEL/AML1. Res. 11:2974-2980.
    Telomerase (TERT). Nair, et al. (2000) Nat. Med. 6:1011-1017.
    707-AP. Takahashi, et al. (1997) Clin. Cancer
    Res. 3:1363-1370.
    Annexin, e.g., Zimmerman, et al. (2004) Virchows
    Annexin II. Arch. 445:368-374.
    BCR/ABL; BCR/ABL Cobaldda, et al. (2000) Blood
    p210; BCR/ABL p190; 95:1007-1013; Hakansson, et al. (2004)
    CML-66; CML-28. Leukemia 18:538-547; Schwartz,
    et al. (2003) Semin. Hematol.
    40:87-96; Lim, et al. (1999) Int.
    J. Mol. Med. 4:665-667.
    BCL2; BLC6; Iqbal, et al. (2004) Am. J. Pathol.
    CD10 protein. 165:159-166.
    CDC27 (this is a Wang, et al. (1999) Science
    melanoma antigen). 284:1351-1354.
    Sperm protein 17 Arora, et al. (2005) Mol. Carcinog.
    (SP17); 14-3-3-zeta; 42:97-108.
    MEMD; KIAA0471;
    TC21.
    Tyrosinase-related GenBank Acc. No. NM_001922.
    proteins 1 and 2 (TRP-1 (see also, e.g., Bronte, et al. (2000)
    and TRP-2). Cancer Res. 60:253-258).
    Gp100/pme1-17. GenBank Acc. Nos. AH003567;
    U31798; U31799;U31807; U31799
    (see also, e.g., Bronte, et al. (2000)
    Cancer Res. 60:253-258).
    TARP. See, e.g., Clifton, et al. (2004) Proc.
    Natl. Acad. Sci. USA 101:10166-
    10171; Virok, et al. (2005) Infection
    Immunity 73:1939-1946.
    Tyrosinase-related GenBank Acc. No. NM_001922.
    proteins 1 and 2 (TRP-1 (see also, e.g., Bronte, et al. (2000)
    and TRP-2). Cancer Res. 60:253-258).
    Melanocortin 1 receptor Salazar-Onfray, et al. (1997) Cancer
    (MC1R); MAGE-3; Res. 57:4348-4355; Reynolds, et
    gp100; tyrosinase; al. (1998) J. Immunol. 161:6970-6976;
    dopachrome Chang, et al. (2002) Clin.
    tautomerase (TRP-2); Cancer Res. 8:1021-1032.
    MART-1.
    MUC-1; MUC-2. See, e.g., Davies, et al. (1994) Cancer
    Lett. 82:179-184; Gambus, et
    al. (1995) Int. J. Cancer 60:146-148;
    McCool, et al. (1999) Biochem.
    J. 341:593-600.
    Spas-1. U.S. Published Pat. Appl. No. 20020150588
    of Allison, et al.
    CASP-8; FLICE; Mandruzzato, et al. (1997) J. Exp. Med.
    MACH. 186:785-793.
    CEACAM6; CAP-1. Duxbury, et al. (2004) Biochem.
    Biophys. Res. Commun. 317:837-
    843; Morse, et al. (1999) Clin.
    Cancer Res. 5:1331-1338.
    HMGB1 (a DNA Brezniceanu, et al. (2003) FASEB
    binding protein and J. 17:1295-1297.
    cytokine).
    ETV6/AML1. Codrington, et al. (2000) Br. J. Haematol.
    111:1071-1079.
    Mutant and wild type Clements, et al. (2003) Clin. Colorectal
    forms of adenomatous Cancer 3:113-120; Gulmann,
    polyposis coli (APC); et al. (2003) Appl. Immunohistochem.
    beta-catenin; c-met; Mol. Morphol. 11:230-237;
    p53; E-cadherin; Jungck, et al. (2004) Int. J. Colorectal.
    cyclooxygenase-2 Dis. 19:438-445; Wang, et al.
    (COX-2). (2004) J. Surg. Res. 120:242-248;
    Abutaily, et al. (2003) J. Pathol.
    201:355-362; Liang, et al. (2004) Br.
    J. Surg. 91:355-361; Shirakawa,
    et al. (2004) Clin. Cancer Res.
    10:4342-4348.
    Renal cell carcinoma Mulders, et al. (2003) Urol. Clin.
    antigen bound by mAB North Am. 30:455-465; Steffens, et
    G250. al. (1999) Anticancer Res. 19:1197-1200.
    EphA2 See, e.g., U.S. Patent Publication
    No. 2005/0281783 A1; Genbank
    Accession No. NM_004431 (human);
    Genbank Accession No.
    NM_010139 (Mouse); Genbank
    Accession No. AB038986 (Chicken,
    partial sequence); GenBank Accession
    Nos. NP_004422, AAH37166,
    and AAA53375 (human); GenBank
    Accession Nos. NP_034269
    (mouse), AAH06954 (mouse), XP_345597
    (rat), and BAB63910
    (chicken).
    EGFRvIII See, e.g., WO/2012/068360
    Francisella tularensis antigens
    Francisella tularensis Complete genome of subspecies
    A and B. Schu S4 (GenBank Acc. No.
    AJ749949); of subspecies Schu 4
    (GenBank Acc. No. NC_006570).
    Outer membrane protein (43 kDa)
    Bevanger, et al. (1988) J. Clin. Microbiol.
    27:922-926; Porsch-Ozcurumez,
    et al. (2004) Clin. Diagnostic.
    Lab. Immunol. 11:1008-1015).
    Antigenic components of
    F. tularensis include, e.g., 80 antigens,
    including 10 kDa and 60 kDa
    chaperonins (Havlasova, et al. (2002)
    Proteomics 2:857-86),
    nucleoside diphosphate kinase,
    isocitrate dehydrogenase,
    RNA-binding protein Hfq, the
    chaperone ClpB (Havlasova, et al.
    (2005) Proteomics 5:2090-2103).
    See also, e.g., Oyston and Quarry
    (2005) Antonie Van Leeuwenhoek
    87:277-281; Isherwood, et al.
    (2005) Adv. Drug Deliv. Rev.
    57:1403-1414; Biagini, et al. (2005)
    Anal. Bioanal. Chem. 382:1027-1034.
    Malarial antigens
    Circumsporozoite See, e.g., Haddad, et al. (2004)
    protein (CSP); SSP2; Infection Immunity 72:1594-1602;
    HEP17; Exp-1 Hoffman, et al. (1997) Vaccine
    orthologs found in 15:842-845; Oliveira-Ferreira and
    P. falciparum; and Daniel-Ribeiro (2001) Mem. Inst.
    LSA-1. Oswaldo Cruz, Rio de Janeiro
    96:221-227. CSP (see, e.g., GenBank
    Acc. No. AB121024). SSP2
    (see, e.g., GenBank Acc. No.
    AF249739). LSA-1 (see, e.g., GenBank
    Acc. No. Z30319).
    Ring-infected See, e.g., Stirnadel, et al. (2000) Int.
    erythrocyte survace J. Epidemiol. 29:579-586;
    protein (RESA); Krzych, et al. (1995) J. Immunol.
    merozoite surface 155:4072-4077. See also, Good, et
    protein 2 (MSP2); al. (2004) Immunol. Rev. 201:254-267;
    Spf66; merozoite Good, et al. (2004) Ann. Rev.
    surface Immunol. 23:69-99. MSP2 (see, e.g.,
    protein 1(MSP1); GenBank Acc. No. X96399;
    195A; BVp42. X96397). MSP1 (see, e.g., GenBank
    Acc. No. X03371). RESA (see,
    e.g., GenBank Acc. No. X05181;
    X05182).
    Apical membrane See, e.g. ,Gupta, et al. (2005) Protein
    antigen 1 (AMA1). Expr. Purif. 41:186-198. AMA1
    (see, e.g., GenBank Acc. No. A`13;
    AJ494905; AJ490565).
    Viruses and viral antigens
    Hepatitis A GenBank Acc. Nos., e.g., NC_001489;
    AY644670; X83302; K02990; M14707.
    Hepatitis B Complete genome (see, e.g.,
    GenBank Acc. Nos. AB214516;
    NC_003977; AB205192; AB205191;
    AB205190; AJ748098;
    AB198079; AB198078; AB198076;
    AB074756).
    Hepatitis C Complete genome (see, e.g., GenBank
    Acc. Nos. NC_004102;
    AJ238800; AJ238799; AJ132997;
    AJ132996; AJ000009; D84263).
    Hepatitis D GenBank Acc. Nos, e.g. NC_001653;
    AB118847; AY261457.
    Human See, e.g., Trimble, et al. (2003)
    papillomavirus, Vaccine 21:4036-4042; Kim, et al.
    including all (2004) Gene Ther. 11:1011-1018;
    200+ Simon, et al. (2003) Eur. J. Obstet.
    subtypes Gynecol. Reprod. Biol. 109:219-223;
    (classed in Jung, et al. (2004) J. Microbiol.
    16 groups), 42:255-266; Damasus-Awatai and
    such as the Freeman-Wang (2003) Curr. Opin.
    high risk Obstet. Gynecol. 15:473-477; Jansen
    subtypes 16, and Shaw (2004) Annu. Rev.
    18, 30, 31, 33, 45. Med. 55:319-331; Roden and Wu
    (2003) Expert Rev. Vaccines 2:495-
    516; de Villiers, et al. (2004)
    Virology 324:17-24; Hussain and
    Paterson (2005) Cancer Immunol.
    Immunother. 54:577-586; Molijn,
    et al. (2005) J. Clin. Virol. 32
    (Suppl. 1) S43-S51. GenBank Acc.
    Nos. AY686584; AY686583;
    AY686582; NC_006169; NC_006168;
    NC_006164; NC_001355; NC_001349;
    NC_005351; NC_001596).
    Human T-cell See, e.g., Capdepont, et al. (2005)
    lymphotropic AIDS Res. Hum. Retrovirus 21:28-
    virus 42; Bhigjee, et al. (1999) AIDS Res.
    (HTLV) types Hum. Restrovirus 15:1229-1233;
    I and II, Vandamme, et al. (1998) J. Virol.
    including the 72:4327-4340; Vallejo, et al. (1996)
    HTLV type I J. Acquir. Immune Defic. Syndr.
    subtypes Hum. Retrovirol. 13:384-391.
    Cosmopolitan, HTLV type I (see, e.g., GenBank Acc.
    Central Nos. AY563954; AY563953.
    African, and HTLV type II (see, e.g., GenBank
    Austro-Melanesian, Acc. Nos. L03561; Y13051;
    and AF139382).
    the HTLV type II
    subtypes Iia, Iib, Iic,
    and Iid.
    Coronaviridae, See, e.g., Brian and Baric (2005) Curr.
    including Top. Microbiol. Immunol.
    Coronaviruses, 287:1-30; Gonzalez, et al. (2003) Arch.
    such as Virol. 148:2207-2235; Smits,
    SARS-coronavirus et al. (2003) J. Virol. 77:9567-9577;
    (SARS-CoV), and Jamieson, et al. (1998) J. Infect.
    Toroviruses. Dis. 178:1263-1269 (GenBank Acc.
    Nos. AY348314; NC_004718;
    AY394850).
    Rubella virus. GenBank Acc. Nos. NC_001545;
    AF435866.
    Mumps virus, See, e.g., Orvell, eta 1. (2002) J. Gen.
    including Virol. 83:2489-2496. See, e.g.,
    the genotypes GenBank Acc. Nos. AY681495;
    A, C, D, NC_002200; AY685921; AF201473.
    G, H, and I.
    Coxsackie virus A See, e.g., Brown, et al. (2003) J.
    including the Virol. 77:8973-8984. GenBank Acc.
    serotypes Nos. AY421768; AY790926: X67706.
    1, 11, 13, 15, 17, 18,
    19, 20, 21, 22, and 24
    (also known as
    Human enterovirus
    C; HEV-C).
    Coxsackie virus B, See, e.g., Ahn, et al. (2005) J. Med.
    including Virol. 75:290-294; Patel, et al.
    subtypes 1-6. (2004) J. Virol. Methods 120:167-172;
    Rezig, et al. (2004) J. Med.
    Virol. 72:268-274. GenBank
    Acc. No. X05690.
    Human enteroviruses See, e.g., Oberste, et al. (2004) J.
    including, e.g., human Virol. 78:855-867. Human
    enterovirus A enterovirus A (GenBank Acc.
    (HEV-A, Nos. NC_001612); human
    CAV2 to CAV8, enterovirus B (NC_001472); human
    CAV10, CAV12, enterovirus C (NC_001428);
    CAV14, CAV16, and human enterovirus D (NC_001430).
    EV71) and also Simian enterovirus A (GenBank
    including HEV-B Acc. No. NC_003988).
    (CAV9, CBV1 to
    CBV6, E1 to E7, E9,
    E11 to E21, E24 to
    E27, E29 to E33, and
    EV69 and E73),
    as well
    as HEV.
    Polioviruses See, e.g., He, et al. (2003) J. Virol.
    including 77:4827-4835; Hahsido, et al.
    PV1, PV2, and PV3. (1999) Microbiol. Immunol. 43:73-77.
    GenBank Acc. No. AJ132961
    (type 1); AY278550 (type 2);
    X04468 (type 3).
    Viral encephalitides See, e.g., Hoke (2005) Mil. Med.
    viruses, including 170:92-105; Estrada-Franco, et al.
    equine encephalitis, (2004) Emerg. Infect. Dis. 10:2113-2121;
    Venezuelan equine Das, et al. (2004) Antiviral
    encephalitis (VEE) Res. 64:85-92; Aguilar, et al. (2004)
    (including Emerg. Infect. Dis. 10:880-888;
    subtypes IA, Weaver, et al. (2004) Arch. Virol.
    IB, IC, ID, IIIC, Suppl. 18:43-64; Weaver, et al.
    IIID), Eastern equine (2004) Annu. Rev. Entomol. 49:141-174.
    encephalitis (EEE), Eastern equine encephalitis
    Western equine (GenBank Acc. No. NC_003899;
    encephalitis (WEE), AY722102); Western equine
    St. Louis encephalitis, encephalitis (NC_003908).
    Murray Valley
    (Australian)
    encephalitis, Japanese
    encephalitis, and
    tick-born encephalitis.
    Human herpesviruses, See, e.g., Studahl, et al. (2000)
    including Scand. J. Infect. Dis. 32:237-248;
    cytomegalovirus Padilla, et al. (2003) J. Med. Virol.
    (CMV), Epstein-Barr 70 (Suppl. 1) S103-S110;
    virus (EBV), human Jainkittivong and Langlais (1998)
    herpesvirus-1 Oral Surg. Oral Med. 85:399-403.
    (HHV-1), GenBank Nos. NC_001806
    HHV-2, HHV-3, (herpesvirus 1); NC_001798
    HHV-4, HHV-5, (herpesvirus 2); X04370 and
    HHV-6, HHV-7, NC_001348 (herpesvirus 3);
    HHV-8, herpes NC_001345 (herpesvirus 4); NC_001347
    B virus, (herpesvirus 5); X83413
    herpes simplex virus and NC_000898 (herpesvirus 6);
    types 1 and 2 (HSV-1, NC_001716 (herpesvirus 7).
    HSV-2), and varicella Human herpesviruses types 6 and 7
    zoster virus (VZV). (HHV-6; HHV-7) are disclosed
    by, e.g., Padilla, et al. (2003) J. Med.
    Virol. 70 (Suppl. 1)S103-S110.
    Human herpesvirus 8 (HHV-8),
    including subtypes A-E, are disclosed
    in, e.g., Treurnicht, et al. (2002)
    J. Med. Virul. 66:235-240.
    HIV-1 including See, e.g., Smith, et al. (1998) J. Med.
    group M Virol. 56:264-268. See also,
    (including subtypes e.g., GenBank Acc. Nos. DQ054367;
    A to J) and group O NC_001802; AY968312;
    (including any DQ011180; DQ011179; DQ011178;
    distinguishable DQ011177; AY588971;
    subtypes) (HIV-2, AY588970; AY781127; AY781126;
    including subtypes AY970950; AY970949;
    A-E. AY970948; X61240; AJ006287;
    AJ508597; and AJ508596.
    Epstein-Barr virus See, e.g., Peh, et al. (2002) Pathology
    (EBV), including 34:446-450. Epstein-Barr virus
    subtypes A and B. strain B95-8 (GenBank Acc. No. V01555).
    Reovirus, including See, e.g., Barthold, et al. (1993) Lab.
    serotypes and Anim. Sci. 43:425-430; Roner,
    strains 1, et al. (1995) Proc. Natl. Acad. Sci.
    2, and 3, type 1 Lang, USA 92:12362-12366; Kedl, et al.
    type 2 Jones, (1995) J. Virol. 69:552-559. GenBank
    and type 3 Acc. No. 1(02739 (sigma-3
    Dearing. gene surface protein).
    Cytomegalovirus See, e.g., Chern, et al. (1998) J. Infect.
    (CMV) subtypes Dis. 178:1149-1153; Vilas
    include CMV Boas, et al. (2003) J. Med. Virol.
    subtypes 71:404-407; Trincado, et al. (2000)
    I-VII. J. Med. Virol. 61:481-487.
    GenBank Acc. No. X17403.
    Rhinovirus, Human rhinovirus 2 (GenBank
    including Acc. No. X02316); Human
    all serotypes. rhinovirus B (GenBank Acc. No.
    NC_001490); Human rhinovirus 89
    (GenBank Acc. No. NC_001617);
    Human rhinovirus 39 (GenBank
    Acc. No. AY751783).
    Adenovirus, AY803294; NC_004001; AC_000019;
    including AC_000018; AC_000017;
    all serotypes. AC_000015; AC_000008; AC_000007;
    AC_000006; AC_000005;
    AY737798; AY737797;NC_003266;
    NC_002067; AY594256;
    AY594254; AY875648; AJ854486;
    AY163756; AY594255;
    AY594253; NC_001460; NC_001405;
    AY598970; AY458656;
    AY487947; NC_001454; AF534906;
    AY45969; AY128640; L19443;
    AY339865; AF532578.
    Filoviruses, See, e.g., Geisbert and Jahrling
    including (1995) Virus Res. 39:129-150;
    Marburg virus and Hutchinson, et al. (2001) J. Med.
    Ebola virus, Virol. 65:561-566. Marburg virus
    and strains (see, e.g., GenBank Acc. No.
    such as NC_001608). Ebola virus (see, e.g.,
    Ebola-Sudan GenBank Acc. Nos. NC_006432;
    (EBO-S), AY769362; NC_002549;
    Ebola-Zaire AF272001; AF086833).
    (EBO-Z), and
    Ebola-Reston
    (EBO-R).
    Arenaviruses, Junin virus, segment S (GenBank
    including Acc. No. NC_005081); Junin virus,
    lymphocytic segment L (GenBank Acc. No. NC_005080).
    choriomeningitis
    (LCM) virus, Lassa
    virus, Junin virus, and
    Machupo virus.
    Rabies virus. See, e.g., GenBank Acc. Nos.
    NC_001542; AY956319; AY705373;
    AF499686; AB128149; AB085828;
    AB009663.
    Arboviruses, Dengue virus type 1 (see, e.g.,
    including GenBank Acc. Nos. AB195673;
    West Nile virus, AY762084). Dengue virus type 2
    Dengue (see, e.g., GenBank Acc. Nos.
    viruses 1 to 4, NC_001474; AY702040; AY702039;
    Colorado AY702037). Dengue virus type
    tick fever 3 (see, e.g., GenBank Acc. Nos.
    virus, Sindbis AY923865; AT858043). Dengue
    virus, virus type 4 (see, e.g., GenBank Acc.
    Togaviraidae, Nos. AY947539; AY947539;
    Flaviviridae, AF326573). Sindbis virus (see, e.g.,
    Bunyaviridae, GenBank Acc. Nos. NC_001547;
    Reoviridae, AF429428; J02363; AF103728). West
    Rhabdoviridae, Nile virus (see, e.g., GenBank
    Orthomyxoviridae, Acc. Nos. NC_001563; AY603654).
    and
    the like.
    Poxvirus including Viriola virus (see, e.g., GenBank
    orthopoxvirus (variola Acc. Nos. NC_001611; Y16780;
    virus, monkeypox X72086; X69198).
    virus, vaccinia virus,
    cowpox virus),
    yatapoxvirus (tanapox
    virus, Yaba monkey
    tumor virus),
    parapoxvirus, and
    molluscipoxvirus.
    Yellow fever. See, e.g., GenBank Acc. No. NC_002031;
    AY640589; X03700.
    Hantaviruses, See, e.g., Elgh, et al. (1997) J. Clin.
    including Microbiol. 35:1122-1130;
    serotypes Hantaan Sjolander, et al. (2002) Epidemiol.
    (HTN), Infect. 128:99-103; Zeier, et al.
    Seoul (SEO), (2005) Virus Genes 30:157-180. GenBank
    Dobrava Acc. No. NC_005222 and
    (DOB), Sin NC_005219 (Hantavirus). See also,
    Nombre (SN), e.g., GenBank Acc. Nos.
    Puumala NC_005218; NC_005222; NC_005219.
    (PUU), and
    Dobrava-like
    Saaremaa
    (SAAV).
    Flaviviruses, See, e.g., Mukhopadhyay, et al. (2005)
    including Nature Rev. Microbiol. 3:13-
    Dengue virus, 22. GenBank Acc. Nos NC_001474
    Japanese and AY702040 (Dengue).
    encephalitis virus, GenBank Acc. Nos. NC_001563
    West Nile and AY603654.
    virus, and yellow
    fever virus.
    Measles virus. See, e.g., GenBank Acc. Nos.
    AB040874 and AY486084.
    Human Human parainfluenza virus 2 (see,
    parainfluenzaviruses e.g., GenBank Acc. Nos.
    (HPV), AB176531; NC003443). Human
    including HPV parainfluenza virus 3 (see, e.g.,
    types 1-56. GenBank Acc. No. NC_001796).
    Influenza virus, Influenza nucleocapsid (see, e.g.,
    including GenBank Acc. No. AY626145).
    influenza Influenza hemagglutinin (see, e.g.,
    virus types GenBank Acc. Nos. AY627885;
    A, B, and C. AY555153). Influenza neuraminidase
    (see, e.g., GenBank Acc. Nos.
    AY555151; AY577316). Influenza
    matrix protein 2 (see, e.g.,
    GenBank Acc. Nos. AY626144(. Influenza
    basic protein 1 (see, e.g.,
    GenBank Acc. No. AY627897).
    Influenza polymerase acid protein
    (see, e.g., GenBank Acc. No. AY627896).
    Influenza nucleoprotein
    (see, e.g., GenBank Acc. Nno. AY627895).
    Influenza A virus Hemagglutinin of H1N1 (GenBank Acc.
    subtypes, e.g., swine No. S67220). Influenza A
    viruses (SIV): H1N1 virus matrix protein (GenBank Acc.
    influenzaA and swine No. AY700216). Influenza virus
    influenza virus. A H5H1 nucleoprotein (GenBank
    Acc. No. AY646426). H1N1
    haemagglutinin (GenBank Acc. No.
    D00837). See also, GenBank
    Acc. Nos. BD006058; BD006055;
    BD006052. See also, e.g.,
    Wentworth, et al. (1994) J. Virol.
    68:2051-2058; Wells, et al. (1991)
    J.A.M.A. 265:478-481.
    Respiratory syncytial Respiratory syncytial virus (RSV)
    virus (RSV), including (see, e.g., GenBank Acc. Nos.
    subgroup A and AY353550; NC_001803; NC001781).
    subgroup B.
    Rotaviruses, Human rotavirus C segment 8
    including (GenBank Acc. No. AJ549087);
    human rotaviruses Human rotavirus G9 strain outer
    A to capsid protein (see, e.g., GenBank
    E, bovine rotavirus, Acc. No. DQ056300); Human rotavirus
    rhesus monkey B strain non-structural protein
    rotavirus, and 4 (see, e.g., GenBank Acc. No.
    human-RVV AY548957); human rotavirus A strain
    reassortments. major inner capsid protein (see, e.g.,
    GenBank Acc. No. AY601554).
    Polyomavirus, See, e.g., Engels, et al. (2004) J. Infect.
    including simian Dis. 190:2065-2069; Vilchez
    virus 40 (SV40), JC and Butel (2004) Clin. Microbiol.
    virus (JCV) and BK Rev. 17:495-508; Shivapurkar, et
    virus (BKV). al. (2004) Cancer Res. 64:3757-3760;
    Carbone, et al. (2003)
    Oncogene 2:5173-5180;
    Barbanti-Brodano, et al. (2004) Virology
    318:1-9) (5V40 complete genome
    in, e.g., GenBank Acc. Nos.
    NC_001669; AF168994; AY271817;
    AY271816; AY120890;
    AF345344; AF332562).
    Coltiviruses, Attoui, et al. (1998) J. Gen. Virol.
    including 79:2481-2489. Segments of Eyach
    Colorado tick fever virus (see, e.g., GenBank Acc.
    virus, Eyach virus. Nos. AF282475; AF282472;
    AF282473; AF282478; AF282476;
    NC_003707; NC_003702;
    NC_003703; NC_003704; NC_003705;
    NC_003696; NC_003697;
    NC_003698; NC_003699; NC_003701;
    NC_003706; NC_003700;
    AF282471; AF282477).
    Calciviruses, including Snow Mountain virus (see, e.g., GenBank
    the genogroups Acc. No. AY134748).
    Norwalk, Snow
    Mountain group
    (SMA), and Saaporo.
    Parvoviridae, See, e.g., Brown (2004) Dev. Biol.
    including (Basel) 118:71-77; Alvarez-
    dependovirus, Lafuente, et al. (2005) Ann. Rheum.
    parvovirus Dis. 64:780-782; Ziyaeyan, et al.
    (including (2005) Jpn. J. Infect. Dis. 58:95-97;
    parvovirus Kaufman, et al. (2005) Virology
    B19), and 332:189-198.
    erythrovirus.
  • Other organisms for which suitable antigens are known in the art include, but are not limited to, Chlamydia trachomatis, Streptococcus pyogenes (Group A Strep), Streptococcus agalactia (Group B Strep), Streptococcus pneumonia, Staphylococcus aureus, Escherichia coli, Haemophilus influenzae, Neisseria meningitidis, Neisseria gonorrheae, Vibrio cholerae, Salmonella species (including typhi, typhimurium), enterica (including Helicobactor pylori Shigella flexneri and other Group D shigella species), Burkholderia mallei, Burkholderia pseudomallei, Klebsiella pneumonia, Clostridium species (including C. difficile), Vibrio parahaemolyticus and V. vulnificus. This list is not meant to be limiting.
  • Pharmaceutical Compositions
  • The term “pharmaceutical” as used herein refers to a chemical substance intended for use in the cure, treatment, or prevention of disease and which is subject to an approval process by the U.S. Food and Drug Administration (or a non-U.S. equivalent thereof) as a prescription or over-the-counter drug product. Details on techniques for formulation and administration of such compositions may be found in Remington, The Science and Practice of Pharmacy 21st Edition (Mack Publishing Co., Easton, Pa.) and Nielloud and Marti-Mestres, Pharmaceutical Emulsions and Suspensions: 2nd Edition (Marcel Dekker, Inc, New York).
  • For the purposes of this disclosure, the pharmaceutical compositions may be administered by a variety of means including orally, parenterally, by inhalation spray, topically, or rectally in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used here includes but is not limited to subcutaneous, intravenous, intramuscular, intraarterial, intradermal, intrathecal and epidural injections with a variety of infusion techniques. Intraarterial and intravenous injection as used herein includes administration through catheters. Administration via intracoronary stents and intracoronary reservoirs is also contemplated. Intra-tumoral administration of the compounds of the present invention may directly activate locally infiltrating DC, directly promote tumor cell apoptosis or sensitize tumor cells to cytotoxic agents. The term oral as used herein includes, but is not limited to oral ingestion, or delivery by a sublingual or buccal route. Oral administration includes fluid drinks, energy bars, as well as pill formulations.
  • Pharmaceutical compositions may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing a drug compound in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents; such as magnesium stearate, stearic acid or talc. Tablets may be uncoated, or may be coated by known techniques including enteric coating, colonic coating, or microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and/or provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • Formulations for oral use may be also presented as hard gelatin capsules where the drug compound is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
  • Pharmaceutical compositions may be formulated as aqueous suspensions in admixture with excipients suitable for the manufacture of aqueous-suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or a mineral oil such as liquid paraffin. The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules of the disclosure suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
  • The pharmaceutical compositions of the disclosure may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally-occurring gums, such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
  • The pharmaceutical compositions of the disclosure may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 20 to 500 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions. It is preferred that the pharmaceutical composition be prepared which provides easily measurable amounts for administration. Typically, an effective amount to be administered systemically is about 0.1 mg/kg to about 100 mg/kg and depends upon a number of factors including, for example, the age and weight of the subject (e.g., a mammal such as a human), the precise condition requiring treatment and its severity, the route of administration, and will ultimately be at the discretion of the attendant physician or veterinarian. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs which have previously been administered; and the severity of the particular condition undergoing therapy, as is well understood by those skilled in the art.
  • As noted above, formulations of the disclosure suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient, as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The pharmaceutical compositions may also be administered as a bolus, electuary or paste.
  • A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free flowing form such as a powder or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropyl ethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) surface active or dispersing agent. Molded tablets may be made in a suitable machine using a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methylcellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric or colonic coating to provide release in parts of the gut other than the stomach. This is particularly advantageous with the compounds of formula 1 when such compounds are susceptible to acid hydrolysis.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • As used herein, pharmaceutically acceptable salts include, but are not limited to: acetate, pyridine, ammonium, piperazine, diethylamine, nicotinamide, formic, urea, sodium, potassium, calcium, magnesium, zinc, lithium, cinnamic, methylamino, methanesulfonic, picric, tartaric, triethylamino, dimethylamino, and tris(hydoxymethyl)aminomethane. Additional pharmaceutically acceptable salts are known to those skilled in the art.
  • An effective amount for a particular patient may vary depending on factors such as the condition being treated, the overall health of the patient, the route and dose of administration and the severity of side effects. Guidance for methods of treatment and diagnosis is available (see, e.g., Maynard, et al. (1996) A Handbook of SOPs for Good Clinical Practice, Interpharm Press, Boca Raton, Fla.; Dent (2001) Good Laboratory and Good Clinical Practice, Urch Publ., London, UK).
  • An effective amount may be given in one dose, but is not restricted to one dose. Thus, the administration can be two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, or more, administrations of pharmaceutical composition. Where there is more than one administration of a pharmaceutical composition in the present methods, the administrations can be spaced by time intervals of one minute, two minutes, three, four, five, six, seven, eight, nine, ten, or more minutes, by intervals of about one hour, two hours, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, and so on. In the context of hours, the term “about” means plus or minus any time interval within 30 minutes. The administrations can also be spaced by time intervals of one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, and combinations thereof. The invention is not limited to dosing intervals that are spaced equally in time, but encompass doses at non-equal intervals.
  • A dosing schedule of, for example, once/week, twice/week, three times/week, four times/week, five times/week, six times/week, seven times/week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, and the like, is available for the invention. The dosing schedules encompass dosing for a total period of time of, for example, one week, two weeks, three weeks, four weeks, five weeks, six weeks, two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, and twelve months.
  • Provided are cycles of the above dosing schedules. The cycle can be repeated about, e.g., every seven days; every 14 days; every 21 days; every 28 days; every 35 days; 42 days; every 49 days; every 56 days; every 63 days; every 70 days; and the like. An interval of non dosing can occur between a cycle, where the interval can be about, e.g., seven days; 14 days; 21 days; 28 days; 35 days; 42 days; 49 days; 56 days; 63 days; 70 days; and the like. In this context, the term “about” means plus or minus one day, plus or minus two days, plus or minus three days, plus or minus four days, plus or minus five days, plus or minus six days, or plus or minus seven days.
  • Methods for co-administration with an additional therapeutic agent are well known in the art (Hardman, et al. (eds.) (2001) Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed., McGraw-Hill, New York, N.Y.; Poole and Peterson (eds.) (2001) Pharmacotherapeutics for Advanced Practice:A Practical Approach, Lippincott, Williams & Wilkins, Phila., Pa.; Chabner and Longo (eds.) (2001) Cancer Chemotherapy and Biotherapy, Lippincott, Williams & Wilkins, Phila., Pa.).
  • As noted, the compositions of the present invention are preferably formulated as pharmaceutical compositions for parenteral or enteral delivery. A typical pharmaceutical composition for administration to an animal comprises a pharmaceutically acceptable vehicle such as aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like. See, e.g., Remington's Pharmaceutical Sciences, 15th Ed., Easton ed., Mack Publishing Co., pp 1405-1412 and 1461-1487 (1975); The National Formulary XIV, 14th Ed., American Pharmaceutical Association, Washington, D.C. (1975). Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oil and injectable organic esters such as ethyloleate. Aqueous carriers include water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles such as sodium chloride, Ringer's dextrose, etc. Intravenous vehicles include fluid and nutrient replenishers. Preservatives include antimicrobial agents, anti-oxidants, chelating agents and inert gases. The pH and exact concentration of the various components the pharmaceutical composition are adjusted according to routine skills in the art.
  • Repeated administrations of a particular vaccine (homologous boosting) have proven effective for boosting humoral responses. Such an approach may not be effective at boosting cellular immunity because prior immunity to the vector tends to impair robust antigen presentation and the generation of appropriate inflammatory signals. One approach to circumvent this problem has been the sequential administration of vaccines that use different antigen-delivery systems (heterologous boosting). In a heterologous boosting regimen, at least one prime or boost delivery comprises delivery of the inactivated tumor cell/cyclic purine dinucleotide compositions described herein. The heterologous arm of the regimen may comprise delivery of antigen using one or more of the following strategies:
      • inactivated or attenuated bacteria or viruses comprising the antigen of interest, which are particles that have been treated with some denaturing condition to render them ineffective or inefficient in mounting a pathogenic invasion;
      • purified antigens, which are typically naturally-produced antigens purified from a cell culture of the pathogen or a tissue sample containing the pathogen, or a recombinant version thereof;
      • live viral or bacterial delivery vectors recombinantly engineered to express and/or secrete antigens in the host cells of the subject. These strategies rely on attenuating (e.g., via genetic engineering) the viral or bacterial vectors to be non-pathogenic and non-toxic;
      • antigen presenting cell (APC) vectors, such as a dendritic cell (DC) vector, which comprise cells that are loaded with an antigen, or transfected with a composition comprising a nucleic acid encoding the antigen (e.g., Provenge® (Dendreon Corporation) for the treatment of castration-resistant metastatic prostate cancer);
      • liposomal antigen delivery vehicles; and
      • naked DNA vectors and naked RNA vectors which may be administered by a gene gun, electroporation, bacterial ghosts, microspheres, microparticles, liposomes, polycationic nanoparticles, and the like.
  • A prime vaccine and a boost vaccine can be administered by any one or combination of the following routes. In one aspect, the prime vaccine and boost vaccine are administered by the same route. In another aspect, the prime vaccine and boost vaccine are administered by different routes. The term “different routes” encompasses, but is not limited to, different sites on the body, for example, a site that is oral, non-oral, enteral, parenteral, rectal, intranode (lymph node), intravenous, arterial, subcutaneous, intramuscular, intratumor, peritumor, intratumor, infusion, mucosal, nasal, in the cerebrospinal space or cerebrospinal fluid, and so on, as well as by different modes, for example, oral, intravenous, and intramuscular.
  • An effective amount of a prime or boost vaccine may be given in one dose, but is not restricted to one dose. Thus, the administration can be two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty, or more, administrations of the vaccine. Where there is more than one administration of a vaccine the administrations can be spaced by time intervals of one minute, two minutes, three, four, five, six, seven, eight, nine, ten, or more minutes, by intervals of about one hour, two hours, three, four, five, six, seven, eight, nine, ten, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, and so on. In the context of hours, the term “about” means plus or minus any time interval within 30 minutes. The administrations can also be spaced by time intervals of one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, and combinations thereof. The invention is not limited to dosing intervals that are spaced equally in time, but encompass doses at non-equal intervals, such as a priming schedule consisting of administration at 1 day, 4 days, 7 days, and 25 days, just to provide a non-limiting example.
  • EXAMPLES
  • The following examples serve to illustrate the present invention. These examples are in no way intended to limit the scope of the invention.
  • Example 1. General Methods
  • Anhydrous solvents and reagents suitable for solution phase oligonucleotide synthesis were purchased and handled under dry argon or nitrogen using anhydrous technique. Amidite coupling reactions and cyclizations were carried out in anhydrous acetonitrile or pyridine under dry argon or nitrogen. The starting materials for all reactions in dry pyridine were dried by concentration (three times) from pyridine. Preparative silica gel flash chromatography was carried out using Fluka 60A high-purity grade or Merck Grade 9385 silica using gradients of methanol in dichloromethane. Analytical HPLC was carried out on a Varian ProStar 210 HPLC system with a ProStar 330 photodiode array detector monitoring at 254 nm using a either a Varian Microsorb 10 micron C18 250×4.6 mm or a Varian 3micronC18 100×4.6 mm column and gradients of 10 mM TEAA and acetonitrile. Preparative HPLC was carried out on a Shimadzu preparative LC20-AP HPLC system, equipped with a SPD-20A UV/Vis detector monitoring at 254 nm on a Varian Microsorb 60-8 C-18 41.6×250 mm column using gradients of 10 mM TEAA and acetonitrile at a flow rate of 50 ml/min. Solid phase extractions using C-18 Sep-Pak (Waters) were carried out at loadings of 3% (wt/wt). LC/MS (ESI/APCI) was obtained on a single quadrapole Shimadzu 2010EV instrument with PDA, MS, and ELSD detection using a Shimadzu LC20D analytical HPLC. High resolution FT-ICR mass spec was obtained from both WM Keck Foundation Biotechnology Resource Laboratory at Yale University in New Haven, Conn., and the QB3/Chemistry Mass Spect Lab at UC Berkeley.
  • 1H, 31p, 1H-1H COSY (2D NMR correlation spectroscopy), 1H-31P HMBC (heteronuclear multiple-bond correlation spectroscopy) spectra were acquired in d6-DMSO with 10 uL D2O (16 hr delay after D2O addition) at 45° C. on a Varian INOVA-500 NMR spectrometer operating at 500 MHz for 1H and 202 MHz for 31P. The resulting FIDs were transferred to a PC and processed using NUTS NMR processing software from Acorn NMR Inc. The chemical shifts were referenced to the DMSO solvent, 2.50 ppm for 1H. Per IUPAC recommendations for referencing of NMR spectral, the 31P chemical shifts were referenced using the “unified scale” to the absolute 1H frequency of 0 ppm. Some of the 1H and 31P spectra were acquired on a JEOL ECX-400 NMR spectrometer operating at 400 MHz for 1H and 162 MHz for 31P.
  • The gradient COSY spectrum was acquired in absolute value mode using 2048 data points in the direct dimension and 256 time points in the indirect dimension. Both dimensions were apodized using sinebell squared functions. The indirect dimension was zero filled to give a final matrix size of 2048×2048 points and a resolution of 3.91 Hz/data point in both dimensions.
  • Assignment of regiochemistry at phosphodiester linkage: 1H-1H COSY in combination with 1H-31P HMBC (and in some cases phosphorous decoupling) experiments were used to provide direct evidence that the regiochemistry of the phosphodiester linkages are 2′, 5′-3′, 5′ (see discussion in experimental for 9a and FIG. 3A-G). Similar 1H-31P HMBC experiments confirmed the non-canonical regiochemistry (2′, 5′-3′, 5′) at the phoshodiester linkage of all the synthesized cyclic di-nucleotides after final silyl deprotection or ion exchange
  • Assignment of the RR- and RS-diastereomers (main CDN products of the synthetic sequence) followed literature methods (Zhao et al. Nucleosides, Nucleotides and Nucleic Acids 289:352-378, 2009).
  • All CDN products (FIG. 2A-2C) were ≥95% pure as indicated by C18 reverse phase HPLC analysis (UV detection at 254 nm)
  • Abbreviations and Acronyms: Guanine=G. isobutyryl guanine=Gib. 4,4-dimethoxytrityl=DMT. OCH2CH2CN=CEO. tert-butyldimethylsilyl=TBS. adenine=A. benzoyl adenine=ABz. cyclic-[A(2′,5′)pA(3′,5′)p]=ML-CDA=19a(TEA salt). dithio-[RP, RP]-cyclic-[A(2′,5′)pA(3′,5′)p]=ML-RR-CDA=19b (TEA salt); 21 (sodium salt); 22 (ammonium salt). dithio-[RP, SP]-cyclic-[A(2′,5′)pA(3′,5′)p]=ML-RS-CDA=19c(TEA salt). cyclic-[G(2′,5′)pG(3′,5′)p]=ML-CDG=9a(TEA salt). dithio-[RP, RP]-cyclic-[G(2′,5′)pG(3′,5′)p]=ML-RR-CDG=9b (TEA salt). dithio-[RP, SP]-cyclic-[G(2′,5′)pG(3′,5′)p]=ML-RS-CDG=9c (TEA salt). cyclic[G(2′,5′)pA(3′,5′)p]=ML-cGAMP. dithio-[RP, RP]-cyclic-[G(2′,5′)pA(3′,5′)p]=ML-RR-cGAMP=20 (TEA salt). monothio-cyclic-[A(2′,5′)pA(3′,5′)Rp]=ML-3′,5′-R-CDA=19d (TEA salt). 2′-O-myristoyl-cyclic-[G(2′,5′)pG(3′,5′)p]=C14-ML-CDG=10 (TEA salt). ML-cGAMP=2′,3′-cGAMP=cyclic-[G(2′,5′)pA(3′,5′)p]=23 (TEA salt)
  • ML-cGAMP (structure 23 in FIG. 2c ) was prepared enzymatically from cellular cGAS and purified by prep HPLC.
  • Example 2. General Experimental for the ML-CDG Series (FIG. 2 a): Synthesis of cyclic [G(2′,5′)pG(3′,5′)p] 9 a
  • 1) Preparation of 3. To a solution of 4.87 g (5.0 mmol) N2-isobutyryl-5′-O-(4,4′-dimethoxytrityl)-2′-O-tert-butyldimethylsilyl-3′-O-[(2-cyanoethyl)-N,N-diisopropylaminophinyl]guanosine (1) in 25 ml acetonitrile was added 0.18 ml (10 mmole) water and 1.23 g (6 mmole) pyridinium trifluroacetate. After 5 minutes stirring at room temperature 25 ml t-butylamine was added and the reaction stirred for 15 minutes at room temperature. The solvents were removed under reduced pressure to give 2 as a foam which was then co-evaporated with acetonitrile (2×50 ml). To a solution of 2 in 60 ml dichloromethane was added 0.9 ml (50 mmole) water and 60 ml 6% (v/v) dichloroacetic acid in dichloromethane (44 mmol). After 10 minutes at room temperature the reaction was quenched by the addition of pyridine (7.0 ml, 87 mmol). The reaction mixture was concentrated to an oil which was dried by three co-evaporations with 40 ml anhydrous acetonitrile, the last time leaving 3 in a volume of 12 ml.
  • 2) Preparation of a dry solution of 4. N2-isobutyryl-5′-O-(4,4′-dimethoxytrityl)-3′-O-tert-butyldimethylsilyl-2′-O-[(2-cyanoethyl)-N,N-diisopropylaminophinyl]guanosine (4, 6.33 g, 6.5 mmole) was dissolved in 40 ml anhydrous acetonitrile and dried by three co-evaporations with 40 ml anhydrous acetonitrile, the last time leaving 20 ml. Ten 3 Å molecular sieves were added and the solution stored under argon until use.
  • 3) Coupling of 3 and 4 to give after oxidation and detritylation the 2′,5′linear dimer 6a. Azeo dried 4 (6.5 mmole) in 20 ml acetonitrile was added via syringe to 3 (5.0 mmole). After 5 minutes stirring at room temperature, 2.37 ml (15 mmole) of 5.5 M t-butylhydroperoxide in decane was added and the reaction stirred for 30 minutes at room temperature. The reaction was then cooled to 0° C., and 1.25 g NaHSO3 in 2.5 ml water was added, the ice bath removed, and the reaction stirred for 5 minutes. The reaction was concentrated to a foam, which was then taken up in 80 ml dichloromethane. 0.9 ml water and 80 ml 6% (v/v) dichloroacetic acid in dichloromethane was added, and the reaction stirred for 10 minutes at room temperature. 50 ml pyridine was added to quench the dichloroacetic acid. The solvents were removed under reduced pressure to give crude 6a as a solid.
  • 4) Cyclization of 6a to give 7a. 6a was dissolved in 50 ml dry pyridine and 5 ml ( 1/10th of total reaction, approximately 0.5 mmole) was transferred via syringe to 150 ml dry pyridine. This was concentrated to a volume of approximately 100 ml. 2-chloro-5,5-dimethyl-1,3,2-dioxaphosphorinane-2-oxide (DMOCP, 0.35 g, 1.8 mmole) was then added and the reaction stirred for 30 minutes at room temperature. 0.32 ml water was added immediately followed by addition of 0.16 g iodine, and the reaction stirred for 5 minutes at room temperature. The reaction mix was then poured into 350 ml water containing 0.1 g NaHSO3 and stirred for 5 minutes at room temperature. 2 g of NaHCO3 was slowly added with stirring, then poured into a separatory funnel and extracted with 400 ml 1:1 ethyl acetate:diethylether. The aqueous layer was extracted again with 400 ml 1:1 ethyl acetate:diethylether, and the organic layers were combined, dried over sodium sulfate, and concentrated under reduced pressure to yield 0.75 g of a mixture containing 7a, the fully-protected cyclic-[G(2′,5′)pG(3′,5′)p].
  • 5) Deprotection of crude 7a with methylamine to give crude 8a. To 750 mg of 7a was added 18 ml of methylamine in anhydrous ethanol (33% by weight) and the mixture was stirred for 90 min at which point analysis by HPLC indicated the reaction was complete. The reaction mixture was concentrated to give an oil which upon treatment with 10 ml of hexane/ethyl acetate (50:50) produced an off-white solid. The trituration/wash solvent was decanted and residual solvent was removed under reduced pressure to give 240 mg of an off-white solid.
  • 6) Preparative HPLC of crude 8a. A 120 mg portion of crude 8a was taken up in 5 ml of CH3CN/10 mM aqueous triethylammonium acetate (20/80). After 0.45 micron PTFE filtration the injection sample was applied to a C-18 Dynamax column (40×250 mm). Elution was performed with a gradient of acetonitrile and 10 mM aqueous triethylammonium acetate (20% to 50% CH3CN over 20 minutes at 50 ml/min flow). HPLC fractions from the two HPLC runs containing pure 8a were pooled, evaporated to remove CH3CN and lyophilized to remove most of remaining water and volatile buffer to give after azeotropic drying with acetonitrile (3×4 ml) 42 mg of pure 8a as the bis-triethylammonium salt. (It is also possible to defer the prep HPLC purification until after the last step). HRMS (FT-ICR) m/z: [M-H] calcd for C32H51N10O14P2Si2 917.2606; found 917.2622. 1H NMR (DMSO-d6+trace D2O) 45° C. δ 8.22 (1H, s), 7.85 (1H, s), 5.76-5.79 (2H, dd), 5.21 (1H, m), 4.85 (1H, m), 4.58 (1H, t), 4.49 (1H, d), 4.31 (1H, m), 4.21 (1H, m), 3.97 (1H, d), 3.83 (3H, m), 2.94 (12H, m), 1.12 (18H, t), 0.90 (9H, s), 0.72 (9H, s), 0.14 (6H, d), 0.09 (3H, s), −0.02 (3H, s). 31P NMR (DMSO-d6+trace D2O) 45° C. δ−1.26, −2.02 (FIG. 3a-3c ).
  • 7) Deprotection of TBS groups of 8a with triethylamine trihydrofluoride, neutralization with TEAB, and solid phase extraction with a C-18 Sep-Pak to give pure 9a as the bis-triethylammonium salt. To 40 mg of 8a was added 1.0 ml of triethylamine trihydrofluoride. The mixture was stirred at room temperature for 30 h. After confirming completion of reaction by analytical HPLC, the sample was neutralized by dropwise addition into 12 ml of chilled 1M triethylammonium bicarbonate. The neutralized solution was desalted on a Waters C-18 Sep-Pak and the product eluted with CH3CN/10 mM aqueous triethylammonium acetate (1:1). The CH3CN was evaporated under reduced pressure and the remaining aqueous solution was frozen and lyophilized overnight. Multiple evaporations from methanol (3×3 ml) and a final evaporation from 50% acetonitrile in methanol (1×3 ml) gave 29.3 mg of cyclic-[G(2′,5′)pG(3′,5′)p] (9a) as the bis-triethylammonium salt. HRMS (FT-ICR) m/z: [M-H] calcd for C20H23N10O14P2 689.0876; found 689.0874. 1H NMR (DMSO-d6+trace D2O) 45° C. δ 7.92 (1H, s), 7.90 (1H, s), 5.82 (1H, d), 5.80 (1H, d), 4.97 (1H, m), 4.85 (1H, m), 4.68 (1H, m), 4.31 (1H, d), 4.21 (1H, t), 4.10 (2H, m), 3.79 (3H, m), 2.91 (14H, m), 1.13 (22H, t). 31P NMR (DMSO-d6) 45° C. δ 1.80, −1.05.
  • The HPLC retention time of 9a is 7.25 min compared to 9.3 min for c-di-GMP using a gradient of 2 to 20% CH3CN in 10 mM triethylammonium acetate over 20 min on a C-18 column (3 micron, 100×4.6 mm, 0.6 ml/min.) The HRMS (FT-ICR) confirmed the expected elemental formula: [M-H] calcd for C20H23N10O14P2 689.0876; found 689.0874. The 31-P NMR of 9a showed two peaks (integrating 1:1) at 2.03 and -0.95 ppm consistent with a 2′,5′/3′,5′ mixed linkage (both c[G(3′,5′)pG(3′,5′)p] and c[G(2′,5′)pG(2′,5′)p], for example, would give only one 31-P NMR signal due to symmetry). Direct evidence for the regiochemistry of the phosphodiester linkages was obtained by 1H-1H COSY in combination with phosphorous decoupling experiments, and by 1H-31P HMBC two-dimensional NMR (FIGS. 3b and 3c ). The anomeric (H-1) protons appear as overlapping doublet of doublets (or triplet) at 5.82 ppm. The “A” designation was given to the downfield anomeric (H-1) proton and “B” to the anomeric proton slightly upfield of that. Starting with the anomeric proton in both the “A” and “B” ribose a 1H-1H COSY experiment (FIG. 3b ) allowed assignment of H-2A (4.96 ppm), H-3A (4.31 ppm), as well as H-2B (4.67 ppm) and H-3B (4.84 ppm). Irradiation of the downfield phosphorous (2.03 ppm) converted the H-3B multiplet to a doublet, while irradiation of the upfield phosphorous (−0.95 ppm) resulted in a simplification of the complex multiplet of H-2A. In both decoupling experiments simplification of the 5′ ribose methylene multiplet was also observed. Two-dimensional 1H-31P HMBC confirmed the result of the decoupling experiments. The 1H-1H COSY results in combination with phosphorous decoupling and 1H-31P HMBC experiments thus provide direct evidence that the regiochemistry of the phosphodiester linkages is 2′,5′/3′,5′ and that 9a is cyclic [G(2′,5′)pG(3′,5′)p].
  • Example 3. General Experimental for the ML-CDA Series (FIG. 2 b): Synthesis of Cyclic [A(2′,5′)pA(3′,5′)p] Na Salt 21 (See Compound FIG. 2 c)
  • 1) Preparation of 13.
  • To a solution of 5 g (5.15 mmol) N6-benzoyl-5′-O-(4,4′-dimethoxytrityl)-2′-O-tert-butyldimethylsilyl-3′-O-[(2-cyanoethyl)-N,N-diisopropylaminophinyl]adenosine (11) in 25 ml acetonitrile was added 0.18 ml (10 mmole) water and 1.20 g (6.2 mmole) pyridinium trifluoroacetate. After 5 minutes stirring at room temperature 25 ml tert-butylamine was added and the reaction stirred for 15 minutes at room temperature. The solvents were removed under reduced pressure to give 12 as a foam which was then co-evaporated with acetonitrile (2×50 ml), then dissolved in 60 ml dichloromethane. To this solution was added water (0.9 ml, 50 mmole) and 60 ml of 6% (v/v) dichloroacetic acid (44 mmol) in dichloromethane. After 10 minutes at room temperature the reaction was quenched by the addition of pyridine (7.0 ml, 87 mmol), and concentrated to an oil which was dried by three co-evaporations with 40 ml anhydrous acetonitrile, the last time leaving 13 in a volume of 12 ml.
  • 2) Preparation of a dry solution of 14.
  • N6-benzoyl-5′-O-(4,4′-dimethoxytrityl)-3′-O-tert-butyldimethylsilyl-2′-O-[(2-cyanoethyl)-N,N-diisopropylaminophinyl]adenosine (14, 6.4 g, 6.6 mmole) was dissolved in 40 ml anhydrous acetonitrile and dried by three co-evaporations with 40 ml anhydrous acetonitrile, the last time leaving 20 ml. Ten 3 Å molecular sieves were added and the solution stored under argon until use.
  • 3) Preparation of 2′,5′-monothio-linear dimer 16.
  • Azeo dried 14 (6.4 g, 6.6 mmole) in 20 ml acetonitrile was added via syringe to a solution of 13 (5.15 mmol) in 12 ml of anhydrous acetonitrile. After 5 minutes stirring at room temperature, 1.14 g (5.6 mmol) of 3-((N,N-dimethylaminomethylidene)amino)-3H-1,2,4-dithiazole-5-thione (DDTT) was added and the reaction stirred for 30 minutes at room temperature. The reaction was concentrated and the residual oil dissolved in 80 ml dichloromethane. Water (0.9 ml, 50 mmol) and 80 ml of 6% (v/v) dichloroacetic acid (58 mmol) in dichloromethane was added, and the reaction stirred for 10 minutes at room temperature. 50 ml pyridine was added to quench the dichloroacetic acid. The solvents were removed under reduced pressure to give crude 16b as a solid.
  • 4) Cyclization and sulfurization of 16b to give the protected cyclic-dithio diastereoisomers 17b and 17c.
  • 16b was dissolved in 150 ml dry pyridine which was concentrated down to a volume of approximately 100 ml. 2-chloro-5,5-dimethyl-1,3,2-dioxaphosphorinane-2-oxide (DMOCP, 3.44 g, 18 mmole) was then added and the reaction stirred for 5 minutes at room temperature. 3.2 ml water was added immediately followed by addition of 3-H-1,2-benzodithiol-3-one (1.3 g, 7.7 mmole), and the reaction stirred for 5 minutes at room temperature. The reaction mix was then poured into 700 ml water containing 20 g NaHCO3 and stirred for 5 minutes at room temperature, then poured into a separatory funnel and extracted with 800 ml 1:1 ethyl acetate:diethyl ether. The aqueous layer was extracted again with 600 ml 1:1 ethyl acetate:diethyl ether. The organic layers were combined and concentrated under reduced pressure to yield approximately 11 g of an oil containing diastereoisomers 17b and 17c.
  • 5) Silica gel column chromatography of the crude mixture containing 17b and 17c.
  • The crude mixture above was dissolved in dichloromethane and applied to a 250 g silica column. The desired diastereoisomers were eluted from the column using a gradient of methanol in dichloromethane (0-10%). Fractions containing the desired diastereoisomers 17b and 17c were combined and concentrated, giving 2.26 g of approximately 50% 17b and 50% 17c.
  • 6) Deprotection of the fully-protected cyclic diastereoisomers 17b and 17c to crude 18b and 18c.
  • 2.26 g of crude 17b and 17c from the silica gel column was transferred to a thick-walled glass pressure tube. 60 ml methanol and 60 ml concentrated aqueous ammonia was added and the tube was heated with stirring in an oil bath at 50° C. for 16 h (recent runs have been 12 h since starting material is consumed at this time). The reaction mixture was cooled to near ambient temperature, sparged with a stream of nitrogen gas for 30 minutes, and then transferred to a large round bottom flask. Most of the volatiles were removed under reduced pressure with caution so as to avoid foaming and bumping. If water was still present the residue was frozen and lyophilized to dryness.
  • 7) Preparative HPLC purification of crude 18b and 18c to give pure 18b.
  • The lyophilized crude mixture containing 18b and 18c was taken up in approximately 50 ml of CH3CN/10 mM aqueous triethylammonium acetate (60/40). After 0.45 micron PTFE filtration, 4-5 ml sample portions were applied to a C-18 Dynamax column (40×250 mm). Elution was performed with a gradient of acetonitrile and 10 mM aqueous triethylammonium acetate (30% to 50% CH3CN over 20 minutes at 50 ml/min flow). Fractions from the preparative HPLC runs containing pure 18b were pooled, evaporated to remove CH3CN and lyophilized to give 360 mg of pure 18b (the RPRP-diastereoisomer) as the bis-triethylammonium salt.
  • 8) Deprotection of the two TBS groups of 18b with triethylamine trihydrofluoride, neutralization with TEAB, solid phase extraction with a C-18 Sep-Pak and lyophilization to give pure 19b as the bis-triethylammonium salt.
  • 8a) To 270 mg (0.24 mmol) of 18b was added 5.0 ml of neat triethylamine trihydrofluoride. The mixture was stirred at room temperature for approximately 40 h. After confirming completion of reaction by analytical HPLC, the sample was neutralized by dropwise addition into 45 ml of chilled, stirred 1M triethylammonium bicarbonate. The neutralized solution was desalted on a Waters C-18 Sep-Pak and the product eluted with CH3CN/10 mM aqueous triethylammonium acetate (5:1). The CH3CN was evaporated under reduced pressure and the remaining aqueous solution was frozen and lyophilized. Multiple rounds of lyophilization from water gave 122 mg (57%) of dithio-(Rp,Rp)-[cyclic-A(2′,5′)pA(3′,5′)p](19b) as the bis-triethylammonium salt.
  • 8b) 90 mg (0.08 mmol) of 18b was coevaporated three times with 10 ml dry acetonitrile. The dried residue was taken up in 0.4 ml anhydrous pyridine. The flask with a vent needle was placed in a 50° C. oil bath, and 0.62 ml triethylamine trihydorfluoride and 1.0 ml triethylamine were added simultaneously to the stirring mixture. The mixture was stirred at 50° C. for two hours. After confirming completion of reaction by analytical HPLC, the sample was neutralized by dropwise addition into 25 ml of chilled, stirred 1M triethylammonium bicarbonate. The neutralized solution was desalted on a Waters C-18 Sep-Pak and the product eluted with CH3CN/10 mM aqueous triethylammonium acetate (1:4). The CH3CN was evaporated under reduced pressure and the remaining aqueous solution was frozen and lyophilized. Multiple rounds of lyophilization from water gave 54 mg (76%) of dithio-(Rp,Rp)-[cyclic-A(2′,5′)pA(3′,5′)p] (19b) as the bis-triethylammonium salt.
  • 8c) A variant of TEA-HF deprotection by heating in neat TEA-HF at 45° C. followed by TEAB neutralization, Sep-Pak desalting and lyophilization.
  • TEA.3HF (1 mL, 6.1 mmol) was added to 18b (41 mg, 0.04 mmol) in a flask equipped with a vent needle and the mixture stirred at 45° C. The reaction progress was monitored by LC and upon consumption of the starting material and mono-TBS analogs (˜2 hr) the mixture was cooled to room temperature. The mixture was slowly pipetted into a solution of 1 M TEAB (4.9 mL) and TEA (1.6 mL) at 0° C. and a slightly basic pH was confirmed by pH paper. The neutralized solution was desalted on a Waters C-18 Sep-Pak (10 g) and the product eluted with 15% CH3CN/10 mM aqueous triethylammonium acetate. Lyophilization gave 21 mg (64%) of 19b (bis-triethylammonium salt) as a white solid. Analysis by analytical HPLC (2-20% Acetonitrile/10 nM TEAA buffer—20 min) showed >95% purity (FIG. 3h ). 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 8.58 (s, 1H), 8.41 (s, 1H), 8.18 (s, 1H), 8.15 (s, 1H), 6.12 (d, J=8.0, 1H), 5.92 (d, J=7.0, 1H), 5.30 (td, J=8.5, 4.0, 1H), 5.24-5.21 (m, 1H), 5.03 (dd, J=7.5, 4.5, 1H), 4.39 (d, J=4, 1H), 4.23 (dd, J=10.5, 4.0, 1H), 4.18 (s, 1H), 4.14-4.08 (m, 2H), 3.85-3.83 (m, 1H), 3.73 (d, J=12.0, 1H), 3.06 (q, J=7.5, 12H), 1.15 (t, J=7.5, 1H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 58.81, 52.54; HRMS (FT-ICR) m/z calcd for C20H24O10N10P2S2 (M-H) 689.0521, found 689.0514.
  • 8d) the work-up of the TEA-HF reaction via acetone precipitation as described in Gaffney et al. 2010 is also possible, but we have obtained somewhat cleaner product using the modifications described in sections 8a-8c above.
  • 10) Conversion to sodium salt
  • The ML-RR-CDA bis-TEA salt (19b) is readily converted to the pharmaceutically acceptable sodium salt (21) by ion exchange as described below.
  • ML-RR-CDA.2Na+ (21). BT AG® 50W-X2 Resin 100-200 Mesh, hydrogen form (100 mg) was slurry packed with DI water into a Bio-spin® column. The excess DI water was drained via gravity. 3 bed volumes of 1 M NaOH (1 mL) was passed through the column via gravity followed by 5 bed volumes of DI water (2 mL). After draining the excess DI water via gravity a solution of ML-RR-CDA.2TEA (19b, 10 mg) in DI water (1 mL) was loaded onto the column. The column was eluted with 5 bed volumes of DI water (2 mL), fractions were collected and checked for UV activity via TLC plate and UV lamp. The fractions of interest were pooled, frozen, and lyophilized over night to give ML-RR-CDA.2Na+ quantitatively. 1H NMR (500 MHz, 45° C., (CD3)2SO-30 μL D2O) δ 8.54 (s, 1H), 8.40 (s, 1H), 8.17 (s, 1H), 8.167 (s, 1H), 6.09 (d, J=8.0, 1H), 5.92 (d, J=8.0, 1H), 5.26 (td, J=8.5, 4.5, 1H), 5.21-5.19 (m, 1H), 5.01 (dd, J=7.5, 4.5, 1H), 4.42 (d, J=4, 1H), 4.23 (dd, J=10.5, 5.0, 1H), 4.17 (s, 1H), 4.15-4.00 (m, 2H), 3.90-3.82 (m, 1H), 3.73-3.70 (m, 1H); 31P NMR (200 MHz, 45° C., (CD3)2SO-30 μL D2O) δ 58.85, 51.53 (FIG. 3d-3g ); HRMS (FT-ICR) m/z calcd for C20H23O10N10P2S2 (M-H) 689.0521, found 689.0503.
  • Direct evidence for the regiochemistry of the phosphodiester linkages was obtained by 1H-1H COSY in combination with 1H-31P HMBC two-dimensional NMR (FIG. 3e-3g ) analogously to the ML-CDG series experimentals.
  • ML-RR-CDG (9b).
  • Compound 9b was synthesized analogously to ML-CDG following the procedures of ML-CDG series experimental with the following modifications (FIG. 2a ): e) DDTT; h) 3-H-1,2-benzodithiol-3-one; n) obtained as the TEA salt, no ion exchange was needed.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 7.98 (s, 1H), 7.94 (s, 1H), 5.85 (d, J=9.0, 1H), 5.80 (d, J=7.5, 1H), 5.25-5.23 (m, 1H), 5.12 (dd, J=8.5, 4.5, 1H), 4.73 (dd, J=8.0, 4.5, 1H), 4.42 (d, J=4.0, 1H), 4.22 (t, J=7.5, 1H), 4.14-4.10 (m, 2H), 3.94-3.90 (m, 2H), 3.77-3.73 (m, 1H), 3.05 (q, J=7.0, 12H), 1.160 (t, J=7.0, 1H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 59.09, 50.37; HRMS (FT-ICR) m/z calcd for C20H23O12N10P2S2 (M-H) 721.0419, found 721.0410.
  • ML-RS-CDG (9c).
  • Compound 9c was synthesized analogously to ML-CDG following the procedures of ML-CDG series experimental with the following modifications (FIG. 2a ): e) DDTT; h) 3-H-1,2-benzodithiol-3-one; k) the [Rp, Sp] diastereomer 8c was collected; n) obtained as the TEA salt, no ion exchange was needed.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 8.01 (s, 1H), 7.98 (s, 1H), 5.86 (d, J=8.5, 1H), 5.79 (d, J=8.0, 1H), 5.29 (dd, J=8.5, 4.0, 1H), 5.20-5.19 (m, 1H), 4.68 (dd, J=8.5, 4.0, 1H), 4.21-4.18 (m, 2H), 4.10-4.05 (m, 3H), 3.71-3.68 (m, 2H), 2.96 (q, J=7.0, 12H), 1.13 (t, J=7.0, 18H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 59.89, 57.17; HRMS (FT-ICR) m/z calcd for C20H24O12N10P2S2 (M-H) 721.041904, found 721.04143.
  • C14-ML-CDG (10):
  • Compound 10 (FIG. 2c ) was synthesized analogously to ML-CDG following the procedures of ML-CDG series experimental with the following modifications (FIG. 2a ): n) myristic anhydride, DMF.
  • To the bis-triethylamine salt of 9a (0.260 g, 0.291 mmol) was added 3.7 ml DMF, 0.3 ml pyridine, and 128 mg (0.292 mmol) of myristic anhydride. The reaction mixture was heated for a total of 5 h at 60° C., cooled to room temperature and quenched with 100 ul of MeOH. The LC trace indicated 25% conversion to a major new product with the remainder of mass appearing in the retention time range of starting material. The mass of the major product was confirmed as the C14-acylated product by LC/MS in negative mode, with m/z (M−1) of 899 (calcd for C34H49N10O15P2: 889.3). After evaporation the residue was taken up in 2 ml CH3CN, 3 ml 0.1 M TEAA and enough MeOH to bring most of material into solution. After a brief spin down via centrifugation to remove a small amount of particulate matter the solution was purified via C18-prep HPLC using a gradient of 25%->50% CH3CN in 10 mM TEAA over 20 min. Fractions containing the desired product were combined and lyophilized to dryness to afford 36 mg of C14-ML-CDG 10 (triethylammonium salt) as a white solid.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 8.00 (s, 1H), 7.90 (s, 1H), 5.98 (d, J=7.5, 1H), 5.83 (d, J=8.5, 1H), 5.76 (dd, J=7.5, 4.5, 1H), 5.15-5.10 (m, 1H), 4.90-4.85 (m, 1H), 4.36 (d, J=4.5, 1H), 4.30-4.27 (m, 1H), 4.07 (s, 1H), 3.94-3.90 (m, 3H), 3.82-3.78 (m, 1H), 3.04 (q, J=7.0, 12H), 2.37-2.23 (m, 2H), 1.51-1.43 (m, 2H), 1.28-1.14 (m, 38H). 0.85 (t, J=7.0, 3H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ−1.36, −2.12; HRMS (FT-ICR) m/z calcd for C34H49O15N10P2(M-H) 899.2860, found 899.2834.
  • ML-CDA (19a).
  • Compound 19a was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications (FIG. 2b ): e) t-BuOOH; h) I2/H2O; n) obtained as the TEA salt, no ion exchange was needed.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 8.44 (s, 1H), 8.37 (s, 1H), 8.16 (s, 1H), 8.14 (s, 1H), 6.08 (d, J=8.0, 1H), 5.90 (d, J=7.5, 1H), 5.10-5.0 (m, 3H), 4.30 (d, J=4.5, 1H), 4.3-4.19 (m, 1H), 4.14 (d, J=1.5, 1H), 4.05 (q, J=11.5, 2H), 3.78-3.75 (m, 2H), 2.90 (q, J=7.5, 18H), 1.08 (t, J=7.0, 27H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 1.67, −0.47; HRMS (FT-ICR) m/z calcd for C20H24O12N10P2(M-H) 657.097763, found 657.09680.
  • ML-RS-CDA (19c).
  • Compound 19c was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications (FIG. 2b ): k) the [Rp, Sp] diastereomer 18c was collected; n) obtained as the TEA salt, no ion exchange was needed.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 8.52 (s, 1H), 8.37 (s, 1H), 8.16 (s, 1H), 8.15 (s, 1H), 6.10 (d, J=8.5, 1H), 5.90 (d, J=7.5, 1H), 5.45 (dd, J=8.5, 4.5, 1H), 5.31-5.26 (m, 1H), 5.00 (dd, J=8.5, 4.5, 1H), 4.41-4.36 (m, 1H), 4.22 (d, J=5.0, 1H), 4.14-4.07 (m, 3H), 3.70-3.67 (m, 3H), 2.84 (q, J=7.0, 19H), 1.08 (t, J=7.5, 29H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 59.98, 57.35; HRMS (FT-ICR) m/z calcd for C20H24O10N10P2S2 (M−2H+Na) 711.0340, found 711.0316.
  • ML-3′-5′-R-CDA (19e).
  • Compound 19e was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications (FIG. 2b ): e) t-BuOOH; h) 3-H-1,2-benzodithiol-3-one; n) obtained as the TEA salt, no ion exchange was needed.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 8.49 (s, 1H), 8.38 (s, 1H), 8.17 (s, 1H), 8.14 (s, 1H), 6.09 (d, J=8.5, 1H), 5.90 (d, J=7.5, 1H), 5.23 (dd, J=8.0, 5.0, 1H), 5.12-5.04 (m, 2H), 4.31 (d, J=4.5, 1H), 4.21-4.14 (m, 3H), 4.10 (q, J=11.0, 1H), 3.80-3.71 (m, 2H), 2.85 (q, J=7.0, 18H), 1.08 (t, J=7.5, 27H); 31P NMR (200 MHz, 45° C., (CD3)2SO-15 μL D2O) δ 59.32, −0.37; HRMS (FT-ICR) m/z calcd for C20H23O11N10P2S (M-H) 673.0749, found 673.0729.
  • ML-RR-CDA (22) as an Ammonia Salt.
  • Compound 22 was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications (FIG. 2b ): n) BT AG® 50W-X2 Resin 100-200 Mesh, hydrogen form, 1 M NH4OH. 1H NMR (500 MHz, 45° C., (CD3)2SO-30 μL D2O) δ 8.80 (s, 1H), 8.44 (s, 1H), 8.39 (s, 2H), 6.45 (d, J=10.0, 1H), 6.34 (s, 1H), 5.50 (td, J=10.5, 4.5, 1H), 5.21-5.15 (m, 1H), 5.02 (d, J=4.0, 1H), 4.92 (d, J=4.5, 1H), 4.61-4.49 (m, 2H), 4.30-4.27 (m, 2H); 1HRMS (FT-ICR) m/z calcd for C20H23O10N10P2S2 (M-H) 689.0521, found 689.0504.
  • ML-RR-cGAMP (20).
  • Compound 20 (FIG. 2c ) was synthesized analogously to ML-RR-CDA following the procedures of ML-CDA series experimental with the following modifications (FIG. 2b ): d) pyr, 4; n) obtained as the TEA salt, no ion exchange was needed.
  • 1H NMR (500 MHz, 45° C., (CD3)2SO-30 μL D2O) δ 8.34 (s, 1H), 8.15 (s, 1H), 8.01 (s, 1H), 5.91 (d, J=7.5, 1H), 5.86 (d, J=8.5, 1H), 5.29-5.23 (m, 1H), 5.17-5.14 (m, 1H), 5.02 (dd, J=7.5, 4.0, 1H), 4.41 (d, J=4.5, 1H), 4.25 (dd, J=5.0, 10.5, 1H), 4.13-4.03 (m, 3H), 3.95-3.85 (m, 1H), 3.78-3.74 (m, 1H), 2.84 (q, J=7.5, 18H), 1.08 (t, J=7.5, 28H); 31P NMR (200 MHz, 45° C., (CD3)2SO-30 μL D2O) δ 58.81, 50.91; HRMS (FT-ICR) m/z calcd for C20H23O11N10P2S2 (M-H) 705.0470, found 705.0451.
  • Example 4. Ribose 2′- and 3-Substituted Derivatives
  • Examples of derivatives finding use in the present invention are depicted in FIG. 4-6.
  • Example 5. CDN-Induced Type I Interferon Expression
  • To determine the relative level of type I interferon induced in human cells by each of the native and derivative molecules as a signature of adjuvant potency, 4×105 THP1-Blue™ ISG cells (a human monocyte cell line transfected with an IRF-inducible secreted embryonic alkaline phosphatase reporter gene (Invivogen) which express alkaline phosphatase under the control of a promoter comprised of five IFN-stimulated response elements) were incubated with 100 μM of cyclic [G(3′,5′)pG(3′,5′)p] (CDG), cyclic [G(2′,5′)pG(3′,5′)p] (mixed linkage, or ML-CDG), or HBSS for 30 minutes at 37° C. with 5% CO2. After 30 minutes, cells were washed and plated in 96-well dish in RPMI media containing 10% FBS, and incubated at 37° C. with 5% CO2. Cell culture supernatants from each sample were collected after overnight incubation, and 20 μL of the cell culture supernatants was added to 180 μL QUANTI-Blue reagent (Invivogen) and incubated for 45 minutes to evaluate type I interferon protein levels. Readings at Absorbance 655 nm were taken every 3 minutes using a Versa Max kinetic spectrophotometer (Molecular Diagnostics).
  • As shown in FIG. 7, cyclic [G(2′,5′)pG(3′,5′)p] (ML-CDG) induced significantly higher levels of IFN-β than cyclic [G(3′,5′)pG(3′,5′)p] across a broad range of time points. These results demonstrate that a purified preparation of cyclic [G(2′,5′)pG(3′,5′)p] more profoundly activates the innate immune response than does cyclic [G(3′,5′)pG(3′,5′)p] in a human monocyte cell line.
  • To determine the levels of IFN-α, -β and -γ induced by cyclic [G(2′,5′)pG(3′,5′)p](ML-CDG) compared to cyclic [G(3′,5′)pG(3′,5′)p] as a signature of potency to activate innate immunity, 1×106 primary human PBMCs isolated from four independent human donors were incubated in a 96 well U bottom plate for 30 min at 37° C., 5% CO2 with 5 or 0.5 μM of cyclic [G(3′,5′)pG(3′,5′)p] (CDG) or cyclic [G(2′,5′)pG(3′,5′)p] (ML-CDG), 1 μg/mL of Interferon Stimulatory DNA (ISD), or 4 μg/mL of Poly (I:C) utilizing Effectene transfection reagent (Qiagen) to transfer the molecules into the PBMC. ISD (Interferon Stimulating DNA) is TLR independent (Stetston, D. B. et. al. Immunity 24, 93-103, January 2006) and signals through cGAS, and is thus STING-dependent, while Poly (I:C) can signal through both TLR3 and RIG-I pathways, and are thus STING-independent. After 30 minutes, the cells were washed and replaced with RPMI media containing 10% FBS and incubated at 37° C., 5% CO2. After 6 hrs incubation, a portion of the cells were harvested and assessed by real-time quantitative RT-PCR for gene expression of the type I cytokines interferon alpha 2 (IFNA2) and interferon beta 1 (IFNB 1), and the type II cytokine gene interferon gamma (IFNG). Gene expression was determined by real-time quantitative RT-PCR using the PrimePCR RNA purification and cDNA analysis system, and run on the CFX96 gene cycler (all BioRad). Normalized expression was determined for each, which accounts for the different efficiencies of PCR amplification for the target (Etarget) and the reference (Ereference), and transforms the logarithmic scaled raw data unit Cycle Threshold (CT) into the linear unit of Normalized Expression. Reference genes used were GUSB and PGK1, genes confirmed to have a coefficient variable (CV) below 0.5 and M value below 1, and thus did not vary with different treatment conditions. To assess correlative secreted protein levels of these cytokines, supernatants were harvested from the remaining cells after 24 hours incubation and IFN-α and -γ levels were determined by Cytometric Bead Array (CBA, BD Biosciences), while IFN-3 levels were determined by ELISA (PBL).
  • As shown in FIG. 8, gene expression of interferon alpha 2 (IFNA2) was significantly higher for cyclic [G(2′,5′)pG(3′,5′)p] at 5 μM than for cyclic [G(3′,5′)pG(3′,5′)p] at 5 μM across all four donors. Similarly, gene expression of interferon beta 1 (IFNB 1) was significantly higher for cyclic [G(2′,5′)pG(3′,5′)p] at 5 μM than for cyclic [G(3′,5′)pG(3′,5′)p] at 5 μM in all four donors. Gene expression for interferon gamma (IFNG) was induced to a significantly higher level for cyclic [G(2′,5′)pG(3′,5′)p] at 5 μM than for cyclic [G(3′,5′)pG(3′,5′)p] across all four donors. These data demonstrate the increased potency of cyclic [G(2′,5′)pG(3′,5′)p]compared to cyclic [G(3′,5′)pG(3′,5′)p] to induce gene expression of critical innate immune cytokines in a variety of human donors.
  • As shown in FIG. 9(a), the levels of secreted IFN-α induced in primary human PBMCs by cyclic [G(2′,5′)pG(3′,5′)p] at 5 μM are higher than cyclic [G(3′,5′)pG(3′,5′)p] at the same or lower dose across all four donors. In FIG. 9(b), levels of IFN-(3, as assessed by ELISA, for cyclic [G(2′,5′)pG(3′,5′)p] at 5 μM were also higher than with cyclic [G(3′,5′)pG(3′,5′)p]induced levels, as well as for the ISD and Poly I:C controls in all four donors. FIG. 9(c) demonstrates a similar finding for secretion of IFN-γ, as assessed by CBA. At both 5 μM and 0.5 μM, cyclic [G(2′,5′)pG(3′,5′)p] induced higher levels of IFN-γ than cyclic [G(3′,5′)pG(3′,5′)p] at the same doses, and higher levels than the ISD and Poly I:C controls across all four donors. These data demonstrate the increased potency of cyclic [G(2′,5′)pG(3′,5′)p] compared to cyclic [G(3′,5′)pG(3′,5′)p] to stimulate type I and II IFN production, critical to the induction of innate immunity across a broad sampling of human donors.
  • To determine the relative level of IFN-β induced in human cells by each of the native and derivative molecules as a signature of adjuvant potency, 4×105 THP1-Blue cells, a human monocyte cell line transfected with an IRF-inducible secreted embryonic alkaline phosphatase reporter gene (Invivogen), were incubated with 50 μM of cyclic [G(3′,5′)pG(3′,5′)p] (CDG), cyclic [G(2′,5′)pG(3′,5′)p] (mixed linkage, or ML-CDG), Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG), compared to [A(3′,5′)pA(3′,5′)p] (CDA), cyclic [A(2′,5′)pA(3′,5′)p] (mixed linkage, or ML-CDA), Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p](ML RR-CDA), or media control for 30 minutes at 37° C. with 5% CO2. After 30 minutes, cells were washed and plated in 96-well dish in RPMI media containing 10% FBS, and incubated at 37° C. with 5% CO2. Cell culture supernatants from each sample were collected after overnight incubation, and 20 μL of the cell culture supernatants was added to 180 μL QUANTI-Blue reagent (Invivogen) and incubated for 45 minutes. Readings at Absorbance 655 nm were taken at 15 minutes using a SpectraMax spectrophotometer (Molecular Diagnostics).
  • As shown in FIG. 10, the Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) derivative induced significantly higher levels of IFN-β than the unmodified cyclic c-di-GMP (CDG) or modified CDG molecules. Similarly, the Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p](ML RR-CDA) molecule induced significantly higher IFN-β levels that either the unmodified CDA or ML CDA molecules. These results demonstrate that purified preparations of the ML RR-CDN derivatives more profoundly activate the innate immune response than the parental CDN molecules in a human monocyte cell line.
  • To determine the relative ability of the derivative molecules to activate immune responses, CDN compounds were administered to 6-8 week old female BALB/c mice (in a total volume of 100 μL in HBSS) at doses of 50, 5 and 0.5 μM by subcutaneous injection into the base of the tail. Mice were assessed 24 hours later for lymphocyte immune cell activation by fluorescent activated cell sorting (FACS) for upregulation of surface CD69 expression on natural killer (NK) cells, CD4+ and CD8+ T cells, as compared to IgG1 isotype controls.
  • As shown in FIGS. 11(a-c), the Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) molecule induced potent immune activation of NK and T cells in a dose-dependent manner. The Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) molecule also induced NK and T cell activation, although to a lesser extent than the ML RR-CDG molecule. Both the ML RR-CDN molecules induced more immune cell activation that the ML CDN molecules at all doses. These data demonstrate the increased immune activation properties of the ML RR-CDN molecules as compared to the ML CDN molecules, and specifically, highlights the ability of the ML RR-CDG molecule to induce potent immune cell activation.
  • Example 6. Enhanced Resistance of Rp,Rp Dithio CDNs to Phosphodiesterases
  • The induction of type I interferon in human cells was measured to evaluate the potency of untreated and phosphodiesterase-treated oxo, Rp monothio and Rp, Rp dithio derivatives. Five compounds (cyclic[A(3′,5′)pA(3′,5′)p] (CDA), cyclic [A(2′,5′)pA(3′,5′)p](ML-CDA), Rp monothio (Rp, monothio cyclic [A(2′,5′)pA(3′,5′)p] (ML R-CDA), Rp, Rp dithio (Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA), and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) were either treated with 160 μg of snake venom phosphodiesterase (SVPD) from Crotalus adamanteus (Sigma), 2.5 mU of Nuclease P1 (NP1) from Penicillium citrinum (Sigma) or mock treated. 7 μg of each compound was diluted in either SVPD buffer (IX PBS and 0.6 mM MgCl2), NP1 buffer (30 mM Na Acetate, pH 5.3, 2 mM ZnCl2) or left untreated and then incubated for 2 hr at 37° C., followed by boiling for 10 min to inactivate the nucleases. 4×105 THP1-Blue™ ISG cells (a human monocyte cell line transfected with an IRF-inducible secreted embryonic alkaline phosphatase reporter gene (Invivogen) which express alkaline phosphatase under the control of a promoter comprised of five IFN-stimulated response elements) were incubated with 50 μM of mock treated, SVPD treated or NP1-treated molecules. After 30 minutes, cells were washed and plated in a 96-well dish in RPMI media containing 10% FBS, and incubated at 37° C. with 5% CO2. Cell culture supernatants from each sample were collected after 16 hr incubation, and 20 μL of the cell culture supernatants was added to 180 μL QUANTI-Blue reagent (Invivogen) and incubated for 25 minutes to evaluate type I interferon protein levels. Readings at Absorbance 655 nm were measured with a Versa Max spectrophotometer (Molecular Diagnostics).
  • As shown in FIG. 12, the untreated Rp, Rp dithio compounds, Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA) and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) are more potent inducers of type I interferon than the oxo (cyclic[A(3′,5′)pA(3′,5′)p] (CDA) and cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA) and the Rp monothio. (Rp, monothio cyclic[A(2′,5′)pA(3′,5′)p] (ML R-CDA) CDN derivative molecules. We evaluated the activity of the CDN derivatives after treatment with either the phosphodiesterase SVPD, which cleaves both 2′-5′ and 3′-5′ phosphodiester linkages, or with NP1, which selectively digests 3′-5′phosphodiester linkages (Pino, et al, (2008) Journal of Biological Chemistry, 283, 36494-36503). FIG. 12 shows that the Rp, Rp dithio compounds, Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA) and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) retain their potency after SVPD and NP1 treatment, whereas the oxo (cyclic[A(3′,5′)pA(3′,5′)p](CDA) and cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA) lost activity after digestion with both SVPD and NP1. The Rp monothio derivative (Rp, monothio cyclic [A(2′,5′)pA(3′,5′)p] (ML R-CDA) which contains a single thio substitution at the 3′-5 phosphodiester linkage retained activity after NP1 digestion, but was susceptible to SVPD treatment, which cleaves the 2′-5′ phosphodieseter linkage. The differential susceptibility of the oxo, Rp monothio and Rp, Rp dithio derivatives to SVPD or NP1 digestion confirms the structure of the Rp monothio and Rp, Rp dithio derivatives. These results also demonstrate the utility of the Rp, Rp dithio derivatives due to their resistance to digestion with phosphodiesterases, present in sera and/or in host cells, thus resulting in more potent activation of innate immune signaling, and increased therapeutic anti-tumor efficacy in vivo, as shown herein.
  • Example 7. Synthetic CDN Derivative Molecules Potently Activate Signaling of all Human STING Alleles
  • To determine the responsiveness of the five known natural human STING variants (referred to as WT, REF, HAQ, AQ and Q) to the native and derivative molecules, a panel of human embryonic kidney (HEK) 293T cell lines that expressed the human STING alleles was generated. The parental HEK 293T cell line does not express endogenous STING, so the responsiveness of exogenously expressed STING alleles can be evaluated. MSCV2.2 plasmids encoding hSTING(REF)-GFP, hSTING(WT)-GFP, hSTING(HAQ)-GFP, hSTING(Q)-GFP and mSTING(WT)-GFP were obtained from the Vance Laboratory at UC Berkeley. hSTING(AQ)-GFP was derived from hSTING(Q)-GFP using a QuickChange Site-Directed Mutagenesis kit (Stratagene). The sequence of the hSTING(REF) allele is also known as the Barber allele (Ishikawa, H., and Barber, G. N. (2008). Nature 455, 674-678), and has the NCBI Reference Sequence NP_938023.1. The amino acid difference between hSTING(REF) and the other WT, HAQ, AQ and Q human STING alleles are shown in FIG. 13, which is adapted from Yi et al., Plos One 8: e77846 (2013). Stable HEK 293T-derived cell lines expressing each of the individual human STING alleles were generated by FACS sorting of GFP positive cells using a Mo Flo cell sorter at the Cancer Research Laboratory Flow Cytometry Facility at UC Berkeley. 1×104 HEK293T STING cells were seeded in 96-well plates and transiently transfected (using Lipofectamine 2000) with 50 ng of a human IFN-β reporter plasmid (pLuc-IFN-β) expressing the human IFN-β promoter upstream of a luciferase reporter and 10 ng of TK-renilla for normalization. 24 hours later, cells were stimulated with native and synthetic CDN derivative molecules using digitonin permeabalization to ensure uniform uptake. Each STING cell line was stimulated with 10 μM of cyclic [G(3′,5′)pA(3′,5′)p] (cGAMP), cyclic [G(2′,5′)pA(3′,5′)p](ML-cGAMP), Rp, Rp dithio cyclic [G(2′,5′)pA(3′,5′)p] (ML RR-cGAMP), cyclic[A(3′,5′)pA(3′,5′)p] (CDA), Rp, Rp dithio cyclic [A(3′,5′)pA(3′,5′)p] (RR-CDA), cyclic [A(2′,5′)pA(3′,5′)p] (ML-CDA), Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA), cyclic [G(3′,5′)pG(3′,5′)p] (CDG), Rp, Rp dithio cyclic [G(3′,5′)pG(3′,5′)p] (RR-CDG), cyclic [G(2′,5′)pG(3′,5′)p] (ML-CDG) or Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) in 25 μl digitonin buffer (50 mM HEPES, 100 mM KCL, 3 mM MgCl2, 0.1 mM DTT, 85 mM Sucrose, 0.2% BSA, 1 mM ATP, 0.1 mM GTP, 10 μg/ml digitonin). After 20 min, the stimulation mixtures were removed and 200 μl of standard RPMI media was added. After stimulation for 6 hrs, cell lysates were prepared and reporter gene activity measured using the Dual Luciferase Assay System (Promega) on a Spectramax M3 luminometer.
  • FIG. 14 depicts stimulation of HEK293 cell lines encoding human STING variant alleles by measuring the fold induction of the IFNβ-LUC reporter (RLU plotted on y-axis). As shown in FIG. 14, the Rp, Rp dithio mixed linkage compounds, Rp, Rp dithio cyclic [G(2′,5′)pA(3′,5′)p] (ML RR-cGAMP), Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) strongly induce IFN3 reporter activity by all human STING alleles. The refractory human STING alleles, hSTING (REF) and hSTING (Q), responded poorly to stimulation with the native molecules with canonical internucleotide phosphate bridge linkages: cyclic [G(3′,5′)pA(3′,5′)p] (cGAMP), cyclic[A(3′,5′)pA(3′,5′)p] (CDA); and, cyclic [G(3′,5′)pG(3′,5′)p] (CDG). In striking contrast, cell lines expressing the refractory human STING alleles were responsive to stimulation with the synthetic Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA): ML RR-CDA; ML RR-CDG; and, ML RR-cGAMP. Cells expressing mouse STING were responsive to all of the molecules tested, demonstrating that the modified synthetic CDN derivative molecules are relevant for activation of the human STING signaling pathway. These results demonstrate that the Rp, Rp dithio mixed linkage compounds, Rp, Rp dithio cyclic [G(2′,5′)pA(3′,5′)p] (ML RR-cGAMP), Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) potently activate all human STING alleles tested, indicating that these molecules will effectively induce innate immunity across a broad range of the human population.
  • To demonstrate that the synthetic CDN derivative molecules induced the maturation of human dendritic cells (DCs), CD14+ monocytes from human PBMCs were treated for 6 days with 50 ng/ml GM-CSF and 25 ng/ml IL-4. Seven days later, the monocyte-derived DCs were stimulated with either LPS (1 ag/ml) or CDNs (50 μM) added directly to the media. After 48 hrs, surface expression of MHC class I (HLA-ABC), CD80, CD83 and CD86 was determined by FACS gated on the CD11c+ DC population. FIG. 15A depicts bar graphs indicating the average of the mean fluorescence intensity (MFI) following stimulation with the CDN molecules indicated in the figure. Also shown in FIG. 15B are representative histograms of CD80, CD86, CD83 and MHC Class I (HLA-ABC) expression in human DCs. Filled histograms correspond to unstimulated cells, the dotted line represents LPS stimulation, and the solid line represents Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) stimulation. These results demonstrate that synthetic CDN molecules with structures comprising Rp, Rp dithio substitution of the non-bridging oxygen atoms of the internucleotide phosphate bridge in combination with 2′-5, 3′-5′ noncanonical or mixed linkage (ML) phosphate bridge structure activate signaling in all human STING alleles, and potently activate the maturation of human DCs.
  • Example 8. CDN-Induced Antigen-Specific T-Cell Response
  • To determine the OVA-specific CD8 T cell response induced by the different cyclic dinucleotide molecules, C57BL/6 mice (n=5) were immunized subcutaneously with 0 μg (no CDN) or 5 μg or 25 μg [G(2′,5′)pG(3′,5′)p] (mixed linkage or ML-CDG) formulated in 2% squalene-and-water with 10 μg ovalbumin protein. Seven days following the vaccination, blood was collected from each animal, and PBMCs were prepared. 5×104 PBMCs were stimulated overnight in an IFNγ ELISpot assay with media alone (unstimulated) or with 1 μM OVA257-264 peptide in the presence of 1×105 naïve splenocytes as feeder cells. IFNγ ELISpots were developed and quantified using a CTL plate reader and ImmunoSpot software.
  • As shown in FIG. 16, both doses of cyclic [G(2′,5′)pG(3′,5′)p] (ML-CDG) induce OVA-specific CD8 immune responses in C57BL/6 mice. These responses are significantly higher than responses induced by unstimulated controls and by a no CDN control group. These results demonstrate that formulations of cyclic [G(2′,5′)pG(3′,5′)p] (ML-CDG) with an antigen can stimulate antigen-specific CD8 T cell responses in vivo.
  • To determine whether STING signaling is required for c[G(2′,5′)pG(3′,5′)p] (ML-CDG) to induce an OVA-specific CD8 T cell response, C57BL/6 mice (n=3 or 5) and goldenticket mice (n=3) were immunized subcutaneously with either 0 μg (no CDN) or 25 ag c[G(2′,5′)pG(3′,5′)p] (ML-CDG) formulated in 2% squalene-and-water with 10 μg ovalbumin protein. Seven days following the vaccination, blood was collected from each animal, and PBMCs were prepared. 5×104 PBMCs were stimulated overnight in an IFNγ ELISpot assay with media alone (unstimulated) or with 1 μM OVA257-264 peptide in the presence of 1×105 naïve splenocytes as feeder cells. IFNγ ELISpots were developed and quantified using a CTL plate reader and ImmunoSpot software.
  • FIG. 17 shows that c[G(2′,5′)pG(3′,5′)p] (ML-CDG) induces an OVA-specific CD8 T cell response that is dependent on the presence of a functional STING molecule. In the wild-type C57BL/6 mice with a functional STING molecule, formulation of c[G(2′,5′)pG(3′,5′)p](ML-CDG) and ovalbumin protein induces significant OVA257-264 immune responses compared to unstimulated control and a no CDN control. In goldenticket mice, which do not express a functional STING molecule (Sauer, Infection and Immunity 2011), the OVA-specific responses induced by c[G(2′,5′)pG(3′,5′)p] (ML-CDG) are not significantly different than the OVA-specific responses induced by a control formulation that does not include CDN (no CDN). These results indicate that immune response induced by c[G(2′,5′)pG(3′,5′)p] (ML-CDG) requires a functional STING molecule.
  • Example 9. Comparative Data with Various CDN Derivatives
  • To assess the ability of the derivative molecules to promote anti-tumor immunity, B16 melanoma cells (5×104 cells in 100 μL PBS) were implanted subcutaneously on the lower back of 6-8 week old female C57BL/6 mice (8 mice per group). Treatments began when tumors reached a volume of approximately 75 mm3, on day 14 post tumor implantation. The CDN compounds were administered (25 μg in a total volume of 40 μL HBSS) by subcutaneous injection into the center of the tumor using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections. The CDNs tested were cyclic [G(3′,5′)pG(3′,5′)p] (CDG); cyclic [G(2′,5′)pG(3′,5′)p] (mixed linkage, or ML CDG); Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG); cyclic [A(3′,5′)pA(3′,5′)p] (CDA); cyclic [A(2′,5′)pA(3′,5′)p] (mixed linkage, or ML CDA); and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA).
  • As shown in FIG. 18, the ML RR-CDG and ML RR-CDA derivatives induced potent anti-tumor efficacy, as compared to the cyclic ML CDG and cyclic ML CDA cyclic dinucleotide molecules. The ML RR-CDA molecule induced significantly more tumor rejection than the ML CDA derivatice (P=0.0004, student's t-test), and mice remained nearly tumor-free in the ML RR-CDG tumor group by day 44 post tumor implantation. These data demonstrate the enhanced potency of the ML RR-CDN derivatives compared to the ML CDN derivative molecules, and the significant anti-tumor efficacy of the ML RR-CDN molecules in the B16 melanoma mouse model.
  • To further assess the ability of the derivative molecules to promote anti-tumor immunity, CT26 colon carcinoma cells (2×105 cells in 100 μL PBS) were implanted by intravenous injection into 6-8 week old female BALB/c mice and assessed for overall survival. The CDN compounds (25 μg in a total volume of 100 μL HBSS) were administered one day post tumor implantation by subcutaneous injection into the base of the tail. Mice were boosted with an additional injection one week later for a total of two vaccinations.
  • As shown in FIG. 19A, the Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) induced significantly higher survival rates compared to the cyclic [G(2′,5′)pG(3′,5′)p] (ML CDG) molecule (P=0.0018, log-rank test), and the Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p](ML RR-CDA) induced significantly higher survival rates compared to the cyclic [A(2′,5′)pA(3′,5′)p] (ML CDA) molecule (P=0.0005, log-rank test). This demonstrates the significant anti-tumor efficacy of the ML RR-CDN derivatives compared to the ML CDN derivative molecules in a CT26 lung metastasis survival model. These results demonstrate that CDN derivative molecules can be successfully administered subcutaneously.
  • To demonstrate that activation of tumor-initiated T cell priming and anti-tumor efficacy induced by CDN derivative molecules was not limited to a single tumor type and mouse genetic background, the ability of the synthetic CDNs to promote anti-tumor immunity in other tumor models was tested. Either CT26 colon carcinoma cells (1×105 cells in 100 μL PBS) or 4T1 mammary carcinoma cells (1×105 cells in 100 μL PBS) were implanted subcutaneously on the flanks of 6-8 week old female BALB/c mice (8 mice per group). Treatments began when tumors reached a volume of approximately 75 mm3, which was approximately day 14 post tumor implantation. The compounds Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) or Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) compounds (25 μg in a total volume of 40 L HBSS), or HBSS vehicle control, and Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) (50 μg in a total volume of 40 μL HBSS) or HBSS vehicle control, were administered by subcutaneous injection into the center of the tumor using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • As shown in FIG. 19B, ML RR-CDG completely inhibited tumor growth in 7 out of 8 mice, while ML RR-CDA completely inhibited tumor growth of all established CT26 tumors. As shown in FIG. 19C, ML RR-CDA derivative completely inhibited tumor growth of all established 4T1 mammary tumors. These data demonstrate the striking potency and durable anti-tumor efficacy of the synthetic mixed linkage RpRp dithio cyclic dinucleotide (ML RR-CDN) derivatives in multiple tumor models.
  • Example 10. CDN Induced Anti-Tumor Efficacy is STING-Dependent
  • To determine whether the effects of the derivative molecules are STING-dependent, B16 melanoma cells (5×104 cells in 100 μL PBS) were implanted on the right flanks of 6-8 week old female goldenticket STING−/− mice, or wild-type C57BL/6 control mice (5 mice per group). Treatments began when tumors reached a volume of approximately 75 mm3, on day 14 post tumor implantation. The compounds administered were Rp, Rp dithio cyclic [G(2′,5′)pG(3′,5′)p] (ML RR-CDG) (25 μg in a total volume of 40 μL HBSS), Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) (50 μg in a total volume of 40 μL HBSS), the TLR9 agonist CpG 1668 (50 μg in a total volume of 40 μL HBSS), or HBSS vehicle control. Mice were treated by subcutaneous injection into the center of tumor only using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • As shown in FIG. 20A, the derivative ML RR-CDNs induced dramatic tumor inhibition in wild-type C57BL/6 mice as compared to HBSS control, and significantly more tumor inhibition than the TLR9 agonist CpG 1668 (P=0.03, student's t-test). In FIG. 20B, tumor growth was not inhibited by either ML RR-CDG or ML RR-CDA, demonstrating that the anti-tumor efficacy observed in wild-type C57BL/6 mice (FIG. 20A) was entirely dependent on a functional STING signaling pathway. In contrast, tumor growth of CpG 1668 was similar in both wild-type and STING−/− mice, as compared to HBSS control (P=0.03, student's t-test), demonstrating that the action of this compound is STING-independent.
  • Example 11. CDN Derivatives Induce Durable and Effective Anti-Tumor Specific T-Cell Immunity
  • To determine whether the synthetic derivative CDN molecules elicit durable and effective anti-tumor T-cell immunity, 6-8 week old female BALB/c mice (8 mice per group) were implanted with CT26 colon carcinoma cells (1×105 cells in 100 μL PBS). Mice were treated with Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound (50 μg in a total volume of 40 μL HBSS) or HBSS vehicle control, and tumor growth was monitored as per previous example. Mice were bled on day 18 post tumor implantation and PBMCs were isolated by Ficoll gradient (Miltenyi Biotech). 5×104 PBMCs were stimulated overnight in an IFNγ ELISpot assay with media alone (unstimulated), or with 1 μM of the endogenous H-2 Ld-restricted tumor rejection antigen AH1 peptide in the presence of 1×105 naïve splenocytes as feeder cells. IFN-γ ELISpot plates were developed and quantified using a CTL plate reader and ImmunoSpot software. On day 55 post tumor implantation, surviving mice and age-matched naïve control mice were implanted on the contralateral flank with either CT26 or 4T1 (both 1×105 cells in 100 μL PBS) tumor cells (4 mice per group), and monitored for tumor growth.
  • As shown in FIG. 21A, all mice treated with ML RR-CDA rejected the growth of established CT26 colon carcinomas. To demonstrate that the effect was mediated by the CDN-mediated induction of an adaptive T cell immune response, PBMCs on day 18 post tumor induction were assessed for IFN-γ production by ELISpot assay, in response to stimulation with the endogenous tumor antigen AH1. As shown in FIG. 21B, PBMCs isolated from mice treated with ML RR-CDA generated significantly higher IFN-γ in response to AH1 peptide stimulation, as compared to the HBSS-treated control group (P=0.003, student's t-test). Further, in FIG. 21C, surviving mice re-challenged with a contralateral tumor exhibited complete protection against the same CT26 tumor type, while not inhibiting growth of the 4T1 tumor type. These data demonstrate the ability of ML RR-CDA to elicit durable and effective tumor-specific T cell-mediated anti-tumor immunity that results in both rejection of the treated tumor, and a stable tumor-antigen specific memory T cell population that can reject tumor challenge.
  • To determine whether CDN derivative molecules induce effective and durable anti-tumor immunity in an alternate tumor model, 6-8 week old female BALB/c mice (8 mice per group) were implanted with 4T1 mammary carcinoma cells (1×105 cells in 100 μL PBS). Mice were treated with Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound (50 μg in a total volume of 40 μL HBSS), or HBSS vehicle control, as per previous experiment. On day 35 post tumor implantation, surviving mice and age-matched naïve control mice were implanted on the contralateral flank with either CT26 or 4T1 (both 1×105 cells in 100 μL PBS) tumor cells (4 mice per group), and monitored for tumor growth.
  • As shown in FIG. 22A, and demonstrated previously, treatment with ML RR-CDA completely inhibited tumor growth of established 4T1 mammary carcinomas. Further, in FIG. 22B, re-challenge with 4T1 tumor cells on the contralateral flank induced complete protection. Re-challenge with the more immunogenic CT26 tumor also elicited complete protection, indicating that these tumors share similar tumor antigens, providing yet further evidence of the potency of the synthetic CDN derivative molecules.
  • Example 12. Activation of Tumor-Initiated T Cell Priming by Intratumoral Injection with CDN Synthetic Derivatives Induces Abscopal Tumor Inhibition
  • The examples shown herein demonstrate that intratumoral (IT) injection of synthetic CDN derivatives results in striking and durable tumor destruction, due to the STING-dependent activation of pro-inflammatory cytokines, to facilitate the development of effective tumor-specific T cell immunity. The STING-dependent induction of tumor-specific T cell immunity protects animals against subsequent challenge with the autologous tumor. It will be apparent to those skilled in the art that advanced cancer is metastatic, and that to be effective, therapies must inhibit outgrowth of distal masses. Treatment of one or selected lesions that inhibits tumor outgrowth of distal untreated tumor masses is known as an abscopal effect. To test whether IT injection of a selected tumor with synthetic CDN derivative molecules inhibited the tumor outgrowth of a distal untreated tumor, (A) CT26 colon carcinoma cells (1×105 cells in 100 μL PBS) and (B) 4T1 mammary carcinoma cells (1×105 cells in 100 μL PBS) were implanted subcutaneously on the contralateral flanks of 6-8 week old female BALB/c mice (8 mice per group). Treatments began when tumors reached a volume of approximately 75 mm3, on day 13 post tumor implantation. The Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound (50 μg in a total volume of 40 μL HBSS), or HBSS vehicle control, was administered by subcutaneous injection into the center of the primary (right side) tumor only using a 27 gauge needle. Injections were repeated every three days, for a total of three intratumoral injections.
  • As shown in FIG. 23, the Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) compound induced complete inhibition of the treated primary tumor in both CT26 (FIG. 23A) and 4T1 (FIG. 23B) tumor-bearing animals, as compared to HBSS vehicle control. Further, outgrowth of the contralateral (untreated) tumor in both tumor models was also significantly inhibited, as compared to HBSS controls (FIG. 23A P=0.0011, FIG. 23B P=0.0019, student's t-test). These data demonstrate the significant anti-tumor efficacy of the ML RR-CDA derivative when injected into the primary tumor, as well as its significant abscopal anti-tumor immune effects.
  • To determine whether the synthetic CDN derivative molecules promote abscopal anti-tumor immunity in an alternative tumor model and mouse genetic background, 6-8 week old female C57BL/6 mice (8 mice per group) were implanted with B16 melanoma cells (5×104 cells in 100 μL PBS) in the right flank. One week later mice were implanted intravenously with 1×105 B16 melanoma cells to colonize the lung, along with a group of naïve age-matched control mice. When the subcutaneous flank tumor reached approximately 75 mm3 on day 13, mice were treated intratumorally with Rp, Rp dithio cyclic [A(2′,5′)pA(3′,5′)p] (ML RR-CDA) (50 μg in a total volume of 40 μL HBSS) or HBSS vehicle control, for three injections as per previous protocol. On day 28 post subcutaneous tumor implantation (day 21 post i.v. implantation), mice were euthanized and lungs were harvested and fixed (10% Neutral Buffered Formalin), and the number of lung tumor nodules counted using a dissecting microscope.
  • As shown in FIG. 24A, and in previous experiments, treatment with ML RR-CDA significantly inhibited tumor growth of the primary flank tumor, as compared to the HBSS control group (P<0.001, student's t-test). Further, in FIG. 24B and depicted in FIG. 24C, treatment with the CDN derivative significantly inhibited the growth of distal lung tumor nodules, compared to the HBSS and naïve (i.v. only) tumor groups. The results shown here demonstrate that intratumoral (IT) injection of synthetic CDN derivatives results in an abscopal anti-tumor effect, as demonstrated by the destruction of the treated tumor, due to the STING-dependent activation of pro-inflammatory cytokines and development of effective tumor-specific T cell immunity, which then inhibits outgrowth of untreated distal tumors.
  • One skilled in the art readily appreciates that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The examples provided herein are representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention.
  • It is to be understood that the invention is not limited in its application to the details of construction and to the arrangements of the components set forth in the following description or illustrated in the drawings. The invention is capable of embodiments in addition to those described and of being practiced and carried out in various ways. Also, it is to be understood that the phraseology and terminology employed herein, as well as the abstract, are for the purpose of description and should not be regarded as limiting.
  • As such, those skilled in the art will appreciate that the conception upon which this disclosure is based may readily be utilized as a basis for the designing of other structures, methods and systems for carrying out the several purposes of the present invention. It is important, therefore, that the claims be regarded as including such equivalent constructions insofar as they do not depart from the spirit and scope of the present invention.
  • While the invention has been described and exemplified in sufficient detail for those skilled in this art to make and use it, various alternatives, modifications, and improvements should be apparent without departing from the spirit and scope of the invention. The examples provided herein are representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Modifications therein and other uses will occur to those skilled in the art. These modifications are encompassed within the spirit of the invention and are defined by the scope of the claims.
  • It will be readily apparent to a person skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention.
  • All patents and publications mentioned in the specification are indicative of the levels of those of ordinary skill in the art to which the invention pertains. All patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference.
  • The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. Thus, for example, in each instance herein any of the terms “comprising”, “consisting essentially of” and “consisting of” may be replaced with either of the other two terms. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.
  • Other embodiments are set forth within the following claims.

Claims (6)

1. A method of treating cancer in a subject in need thereof, comprising:
administering to the subject one or both of a CTLA-4 pathway antagonist and a PD-1 pathway antagonist; and
co-administering to the subject a cyclic dinucleotide that induces Stimulator of Interferon Genes (“STING”)-dependent type I interferon production.
2. The method of claim 1, wherein the cyclic dinucleotide has the formula:
Figure US20200282049A1-20200910-C00004
where R1 and R2 are each H,
or a pharmaceutically acceptable salt thereof.
3. The method of claim 1, wherein the cyclic dinucleotide and one or both of a CTLA-4 pathway antagonist and a PD-1 pathway antagonist are administered as separate administrations.
4. The method of claim 1, wherein the cyclic dinucleotide and one or both of a CTLA-4 pathway antagonist and a PD-1 pathway antagonist are administered at different times.
5. The method of claim 1, wherein the cyclic dinucleotide and one or both of a CTLA-4 pathway antagonist and a PD-1 pathway antagonist are administered by different routes of administration.
6. The method of claim 1, wherein the cancer is selected from the group consisting of a colorectal cancer, an aero-digestive squamous cancer, a lung cancer, a brain cancer, a liver cancer, a stomach cancer, a sarcoma, a leukemia, a lymphoma, a multiple myeloma, an ovarian cancer, a uterine cancer, a breast cancer, a melanoma, a prostate cancer, a pancreatic carcinoma, and a renal carcinoma.
US16/877,218 2013-05-18 2020-05-18 Compositions and methods for activating "stimulator of interferon gene"-dependent signalling Abandoned US20200282049A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/877,218 US20200282049A1 (en) 2013-05-18 2020-05-18 Compositions and methods for activating "stimulator of interferon gene"-dependent signalling

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361825005P 2013-05-18 2013-05-18
US201361902125P 2013-11-08 2013-11-08
US14/280,667 US9724408B2 (en) 2013-05-18 2014-05-18 Compositions and methods for activating stimulator of interferon gene-dependent signalling
US15/665,378 US10653774B2 (en) 2013-05-18 2017-07-31 Compositions and methods for activating “stimulator of interferon gene”-dependent signalling
US16/877,218 US20200282049A1 (en) 2013-05-18 2020-05-18 Compositions and methods for activating "stimulator of interferon gene"-dependent signalling

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/665,378 Continuation US10653774B2 (en) 2013-05-18 2017-07-31 Compositions and methods for activating “stimulator of interferon gene”-dependent signalling

Publications (1)

Publication Number Publication Date
US20200282049A1 true US20200282049A1 (en) 2020-09-10

Family

ID=51934006

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/280,667 Active US9724408B2 (en) 2013-05-18 2014-05-18 Compositions and methods for activating stimulator of interferon gene-dependent signalling
US15/665,378 Active US10653774B2 (en) 2013-05-18 2017-07-31 Compositions and methods for activating “stimulator of interferon gene”-dependent signalling
US16/877,218 Abandoned US20200282049A1 (en) 2013-05-18 2020-05-18 Compositions and methods for activating "stimulator of interferon gene"-dependent signalling

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/280,667 Active US9724408B2 (en) 2013-05-18 2014-05-18 Compositions and methods for activating stimulator of interferon gene-dependent signalling
US15/665,378 Active US10653774B2 (en) 2013-05-18 2017-07-31 Compositions and methods for activating “stimulator of interferon gene”-dependent signalling

Country Status (37)

Country Link
US (3) US9724408B2 (en)
EP (2) EP2996473B1 (en)
JP (1) JP6453855B2 (en)
KR (1) KR20160009039A (en)
CN (1) CN105228450B (en)
AP (1) AP2015008700A0 (en)
AU (1) AU2014268836B2 (en)
BR (1) BR112015028341A2 (en)
CA (1) CA2904536A1 (en)
CL (1) CL2015002522A1 (en)
CR (1) CR20150616A (en)
CU (1) CU24377B1 (en)
CY (1) CY1122274T1 (en)
DK (1) DK2996473T3 (en)
DO (1) DOP2015000281A (en)
EC (1) ECSP15052938A (en)
ES (1) ES2754269T3 (en)
GT (1) GT201500326A (en)
HK (2) HK1219024A1 (en)
HR (1) HRP20191972T1 (en)
HU (1) HUE046942T2 (en)
IL (1) IL242359B (en)
LT (1) LT2996473T (en)
ME (1) ME03586B (en)
MX (1) MX354057B (en)
MY (1) MY175308A (en)
NI (1) NI201500162A (en)
PE (1) PE20160080A1 (en)
PH (1) PH12015502438B1 (en)
PL (1) PL2996473T3 (en)
PT (1) PT2996473T (en)
RS (1) RS59500B1 (en)
SA (1) SA515370123B1 (en)
SG (1) SG11201508273RA (en)
SI (1) SI2996473T1 (en)
SV (1) SV2015005107A (en)
WO (1) WO2014189805A1 (en)

Families Citing this family (228)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2347774B1 (en) 2005-12-13 2017-07-26 The President and Fellows of Harvard College Scaffolds for cell transplantation
JP6104806B2 (en) 2010-10-06 2017-03-29 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Injectable pore-forming hydrogel for material-based cell therapy
EP2838515B1 (en) 2012-04-16 2019-11-20 President and Fellows of Harvard College Mesoporous silica compositions for modulating immune responses
IN2014MN02492A (en) 2012-06-08 2015-07-17 Aduro Biotech
EP2931738B1 (en) 2012-12-13 2019-02-06 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
SG11201508165VA (en) 2013-04-29 2015-11-27 Sloan Kettering Inst Cancer Compositions and methods for altering second messenger signaling
WO2014179760A1 (en) 2013-05-03 2014-11-06 The Regents Of The University Of California Cyclic di-nucleotide induction of type i interferon
US9549944B2 (en) 2013-05-18 2017-01-24 Aduro Biotech, Inc. Compositions and methods for inhibiting “stimulator of interferon gene”—dependent signalling
MX354057B (en) 2013-05-18 2018-02-09 The Regents Of The Univ Of California Star Compositions and methods for activating "stimulator of interferon gene"-dependent signalling.
WO2015017652A1 (en) 2013-07-31 2015-02-05 Memorial Sloan-Kettering Cancer Center Sting crystals and modulators
EP3049442A4 (en) 2013-09-26 2017-06-28 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
US20160287623A1 (en) * 2013-11-19 2016-10-06 The University Of Chicago Use of sting agonist as cancer treatment
WO2015109391A1 (en) 2014-01-24 2015-07-30 Children's Hospital Of Eastern Ontario Research Institute Inc. Smc combination therapy for the treatment of cancer
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
JP7348708B2 (en) 2014-04-30 2023-09-21 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Combination vaccine device and method for killing cancer cells
US10450341B2 (en) 2014-06-04 2019-10-22 Glaxosmithkline Intellectual Property Development Limited Cyclic di-nucleotides as modulators of STING
CN107206071A (en) 2014-09-13 2017-09-26 诺华股份有限公司 The conjoint therapy of ALK inhibitor
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
ES2952717T3 (en) 2014-10-14 2023-11-03 Novartis Ag Antibody molecules against PD-L1 and uses thereof
US11058758B2 (en) * 2014-11-20 2021-07-13 National Institutes Of Biomedical Innovation, Health And Nutrition TH1-inducing adjuvant comprising combination of different nucleic acid adjuvants, and use of same
EP3233882B1 (en) 2014-12-16 2019-10-30 Kayla Therapeutics Fluorinated cyclic dinucleotides for cytokine induction
GB201501462D0 (en) * 2015-01-29 2015-03-18 Glaxosmithkline Ip Dev Ltd Novel compounds
EP3250250A4 (en) * 2015-01-30 2019-05-22 President and Fellows of Harvard College Peritumoral and intratumoral materials for cancer therapy
US10130663B2 (en) 2015-02-11 2018-11-20 The John Hopkins University Bacteria over-expressing c-di-AMP and therapeutic methods
MY190404A (en) 2015-03-10 2022-04-21 Aduro Biotech Inc Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
JP7094533B2 (en) 2015-04-10 2022-07-04 プレジデント アンド フェローズ オブ ハーバード カレッジ Immune cell capture device and its manufacture and use
DK3326641T3 (en) 2015-04-22 2019-09-30 Curevac Ag RNA-Containing Composition for the Treatment of Tumor Diseases
WO2017019897A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to tim-3
CN114272371A (en) 2015-07-29 2022-04-05 诺华股份有限公司 Combination therapy comprising anti-PD-1 antibody molecules
LT3317301T (en) 2015-07-29 2021-07-26 Novartis Ag Combination therapies comprising antibody molecules to lag-3
CN106692967A (en) * 2015-08-12 2017-05-24 聊城市奥润生物医药科技有限公司 Application of cyclic dinucleotide cGAMP combined PD1 antibody (Nivolumab) in preparation of antitumor drugs
EP3334745B1 (en) * 2015-08-13 2024-05-15 Merck Sharp & Dohme LLC Cyclic di-nucleotide compounds as sting agonists
US11453697B1 (en) 2015-08-13 2022-09-27 Merck Sharp & Dohme Llc Cyclic di-nucleotide compounds as sting agonists
CN106540255A (en) * 2015-09-18 2017-03-29 聊城市奥润生物医药科技有限公司 Ring dinucleotide cGAMP combines application of the Avastin in antitumor
EP3368072A4 (en) 2015-10-28 2019-07-03 Aduro BioTech, Inc. Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
MA44334A (en) 2015-10-29 2018-09-05 Novartis Ag ANTIBODY CONJUGATES INCLUDING A TOLL-TYPE RECEPTOR AGONIST
CN107849084B (en) 2015-12-03 2021-09-14 葛兰素史密斯克莱知识产权发展有限公司 Cyclic purine dinucleotides as STING modulators
KR20180107102A (en) 2015-12-16 2018-10-01 그릿스톤 온콜로지, 인코포레이티드 Identification of new antigens, manufacture, and uses
US20180369268A1 (en) * 2015-12-16 2018-12-27 Aduro Biotech, Inc. Methods for identifying inhibitors of "stimulator of interferon gene"- dependent interferon production
EP3389712B1 (en) 2015-12-17 2024-04-10 Novartis AG Antibody molecules to pd-1 and uses thereof
WO2017122130A1 (en) 2016-01-11 2017-07-20 Novartis Ag Immune-stimulating humanized monoclonal antibodies against human interleukin-2, and fusion proteins thereof
US10723756B2 (en) 2016-01-11 2020-07-28 Innate Tumor Immunity Inc. Cyclic dinucleotides for treating conditions associated with STING activity such as cancer
KR20180097751A (en) 2016-01-11 2018-08-31 인네이트 튜머 이뮤니티, 인코포레이티드 RTI ID = 0.0 &gt; STING &lt; / RTI &gt; activity,
CN106544319A (en) * 2016-01-24 2017-03-29 聊城市奥润生物医药科技有限公司 A kind of compositionss for stimulating maturing dendritic cell and its method for stimulating maturing dendritic cell
JP7138864B2 (en) 2016-02-06 2022-09-20 プレジデント アンド フェローズ オブ ハーバード カレッジ Recapitulation of the hematopoietic niche to reconstitute immunity
KR102530488B1 (en) * 2016-03-18 2023-05-08 이뮨 센서, 엘엘씨 Cyclic di-nucleotide compounds and methods of use
CN106540256A (en) * 2016-03-27 2017-03-29 聊城市奥润生物医药科技有限公司 Ring dinucleotide-application of the liposome conjugated monoclonal antibodies in antitumor
KR20180132783A (en) 2016-04-07 2018-12-12 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Heterocyclic amides useful as protein modulators
KR102527784B1 (en) 2016-04-07 2023-04-28 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Heterocyclic amides useful as protein modulators
WO2017184735A1 (en) 2016-04-19 2017-10-26 Ifm Therapeutics, Inc Nlrp3 modulators
BR112018071347A2 (en) 2016-04-19 2019-02-05 Innate Tumor Immunity Inc nlrp3 modulators
US10696985B1 (en) 2016-06-06 2020-06-30 Vanderbilt University Reversibly crosslinked endosomolytic polymer vesicles for cytosolic drug delivery
WO2018009466A1 (en) 2016-07-05 2018-01-11 Aduro Biotech, Inc. Locked nucleic acid cyclic dinucleotide compounds and uses thereof
WO2018009652A1 (en) * 2016-07-06 2018-01-11 Sperovie Biosciences, Inc. Compounds, compositions, and methods for the treatment of disease
MX2019000216A (en) * 2016-07-06 2019-11-12 Sperovie Biosciences Inc Compounds, compositions, and methods for the treatment of disease.
CN109789092A (en) 2016-07-13 2019-05-21 哈佛学院院长等 Antigen presenting cell simulates bracket and its preparation and application
NL2017267B1 (en) 2016-07-29 2018-02-01 Aduro Biotech Holdings Europe B V Anti-pd-1 antibodies
NL2017270B1 (en) 2016-08-02 2018-02-09 Aduro Biotech Holdings Europe B V New anti-hCTLA-4 antibodies
WO2018029256A1 (en) 2016-08-09 2018-02-15 Aarhus Universitet Modulation of ifi16 and sting activity
EP3922279A1 (en) 2016-08-30 2021-12-15 Dana Farber Cancer Institute, Inc. Drug delivery compositions 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
US10537590B2 (en) 2016-09-30 2020-01-21 Boehringer Ingelheim International Gmbh Cyclic dinucleotide compounds
CR20190168A (en) 2016-10-04 2019-05-17 Merck Sharp & Dohme BENZO[b]THIOPHENE COMPOUNDS AS STING AGONISTS
WO2018065360A1 (en) 2016-10-07 2018-04-12 Biolog Life Science Institute Forschungslabor Und Biochemica-Vertrieb Gmbh Cyclic dinucleotides containing benzimidazole, method for the production of same, and use of same to activate stimulator of interferon genes (sting)-dependent signaling pathways
AU2017341735B2 (en) 2016-10-11 2022-01-13 Helmholtz Center for Infection Research Hepatitis C virus immunogenic compositions comprising as an adjuvant a cyclic dinucleotide or an archaeosome and methods of use thereof
JOP20170188A1 (en) 2016-11-25 2019-01-30 Janssen Biotech Inc Cyclic dinucleotides as sting agonists
JP2018090562A (en) * 2016-12-01 2018-06-14 武田薬品工業株式会社 Cyclic dinucleotide
JOP20170192A1 (en) * 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co Cyclic dinucleotide
WO2018118664A1 (en) * 2016-12-20 2018-06-28 Merck Sharp & Dohme Corp. Combinations of pd-1 antagonists and cyclic dinucleotide sting agonists for cancer treatment
RU2019122598A (en) * 2016-12-20 2021-01-22 Мерк Шарп И Доум Корп. CYCLIC DINUCLEOTIDE STING AGONISTS FOR CANCER TREATMENT
AU2018212788A1 (en) * 2017-01-27 2019-07-25 Janssen Biotech, Inc. Cyclic dinucleotides as STING agonists
ES2891326T3 (en) 2017-01-27 2022-01-27 Janssen Biotech Inc Cyclic dinucleotides as STING agonists
US20190351039A1 (en) 2017-02-01 2019-11-21 Modernatx, Inc. Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides
US20200055883A1 (en) 2017-02-17 2020-02-20 Eisai R&D Management Co., Ltd. Cyclic di-nucleotides derivative for the treatment of cancer
TWI674261B (en) 2017-02-17 2019-10-11 美商英能腫瘤免疫股份有限公司 Nlrp3 modulators
JP2018131427A (en) * 2017-02-17 2018-08-23 国立研究開発法人理化学研究所 Technology for controlling immune cells
US10933078B2 (en) 2017-02-21 2021-03-02 Board Of Regents, The University Of Texas System Cyclic dinucleotides as agonists of stimulator of interferon gene dependent signalling
CN106727331B (en) * 2017-03-13 2022-02-01 杭州星鳌生物科技有限公司 Composition of immunoliposome-cycloddinucleotide, preparation method and application of immunoliposome-cycloducleotide in resisting tumor
CN110651036A (en) 2017-03-20 2020-01-03 健诺西生物科学公司 Method of treatment
JOP20190218A1 (en) 2017-03-22 2019-09-22 Boehringer Ingelheim Int Modified cyclic dinucleotide compounds
AU2018252546A1 (en) 2017-04-13 2019-10-10 Sairopa B.V. Anti-SIRPα antibodies
EP3610022A1 (en) 2017-04-14 2020-02-19 Tollnine, Inc. Immunomodulating polynucleotides, antibody conjugates thereof, and methods of their use
UY37695A (en) 2017-04-28 2018-11-30 Novartis Ag BIS 2’-5’-RR- (3’F-A) (3’F-A) CYCLE DINUCLEOTIDE COMPOUND AND USES OF THE SAME
AR113224A1 (en) 2017-04-28 2020-02-19 Novartis Ag ANTIBODY CONJUGATES INCLUDING A STING AGONIST
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
EP3621624B1 (en) 2017-05-12 2023-08-30 Merck Sharp & Dohme LLC Cyclic di-nucleotide compounds as sting agonists
WO2018210279A1 (en) * 2017-05-16 2018-11-22 科济生物医药(上海)有限公司 Use of toll-like receptor agonist combined with immune effector cell
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
CN110785187B (en) 2017-06-22 2024-04-05 诺华股份有限公司 Antibody molecules against CD73 and uses thereof
WO2018237173A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
AU2018301681B2 (en) 2017-07-14 2022-07-14 Innate Tumor Immunity, Inc. NLRP3 modulators
EP3431484A1 (en) * 2017-07-21 2019-01-23 Ludwig-Maximilians-Universität München A fluorescent cyclic dinucleotide and its use in methods of identifying substances having an ability to modulate the cgas/sting pathway
KR20200032180A (en) * 2017-07-28 2020-03-25 브리스톨-마이어스 스큅 컴퍼니 Cyclic dinucleotide as an anticancer agent
AU2018311965A1 (en) 2017-08-04 2020-02-13 Merck Sharp & Dohme Llc Combinations of PD-1 antagonists and benzo[b]thiophene sting antagonists for cancer treatment
US11285131B2 (en) 2017-08-04 2022-03-29 Merck Sharp & Dohme Corp. Benzo[b]thiophene STING agonists for cancer treatment
JP7311514B2 (en) 2017-08-30 2023-07-19 ベイジン シュエンイー ファーマサイエンシズ カンパニー, リミテッド Cyclic Dinucleotides as Interferon Gene Stimulator Modulators
EP3676279B1 (en) * 2017-08-31 2021-12-01 Bristol-Myers Squibb Company Cyclic dinucleotides as anticancer agents
ES2945140T3 (en) * 2017-08-31 2023-06-28 Bristol Myers Squibb Co Cyclic dinucleotides as anticancer agents
US11638716B2 (en) 2017-08-31 2023-05-02 F-star Therapeutics, Inc. Compounds, compositions, and methods for the treatment of disease
AU2018330188C1 (en) 2017-09-08 2024-02-29 The Board Of Trustees Of The Leland Stanford Junior University ENPP1 inhibitors and their use for the treatment of cancer
WO2019051489A1 (en) 2017-09-11 2019-03-14 Sperovie Biosciences, Inc. Compounds, compositions, and methods for the treatment of disease
US11584774B2 (en) 2017-09-11 2023-02-21 F-star Therapeutics, Inc. Compounds, compositions, and methods for the treatment of disease
BR112020006780A2 (en) 2017-10-05 2020-10-06 Glaxosmithkline Intellectual Property Development Limited modulators of the interferon gene stimulator (sting)
EP3692033A1 (en) 2017-10-05 2020-08-12 GlaxoSmithKline Intellectual Property Development Limited Modulators of stimulator of interferon genes (sting) useful in treating hiv
CN111465989A (en) 2017-10-10 2020-07-28 磨石肿瘤生物技术公司 Identification of neoantigens using hot spots
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019092660A1 (en) 2017-11-10 2019-05-16 Takeda Pharmaceutical Company Limited Sting modulator compounds, and methods of making and using
EA038805B1 (en) * 2017-11-21 2021-10-21 Такеда Фармасьютикал Компани Лимитед Cyclic dinucleotides as sting (stimulator of interferon genes) agonists
EP3714275A4 (en) 2017-11-22 2021-10-27 Gritstone bio, Inc. Reducing junction epitope presentation for neoantigens
US11633473B2 (en) 2017-11-27 2023-04-25 Children's Medical Center Corporation Stimulator of Interferon Genes (STING) ligands and uses thereof
WO2019123340A1 (en) 2017-12-20 2019-06-27 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3' cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019125974A1 (en) 2017-12-20 2019-06-27 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2019136118A2 (en) * 2018-01-04 2019-07-11 Academia Sinica Cell-associating immunologic adjuvants for treatment enhancement
JP2021512144A (en) * 2018-01-26 2021-05-13 ザ リージェンツ オブ ザ ユニヴァーシティ オブ カリフォルニアThe Regents of the University of California Intranasal delivery of tuberculosis vaccine containing cyclic dinucleotide adjuvant
CN111936156A (en) 2018-02-02 2020-11-13 诺华股份有限公司 Combination of STING agonists and IL-15/IL15-Ra for the treatment of cancer
EP3749681A1 (en) * 2018-02-06 2020-12-16 STipe Therapeutics ApS Modulation of p62 and sting activity
TW201946650A (en) 2018-03-12 2019-12-16 美商詹森藥物公司 Vaccines against intra-abdominal infections
CN110272462B (en) * 2018-03-15 2023-03-10 南开大学 Process for preparing compounds
IL309265A (en) 2018-03-23 2024-02-01 Codiak Biosciences Inc Extracellular vesicles comprising sting-agonist
CN111989338A (en) 2018-03-27 2020-11-24 勃林格殷格翰国际有限公司 Modified cyclic dinucleotide compounds
CN111971291A (en) 2018-03-27 2020-11-20 勃林格殷格翰国际有限公司 Cyclic dinucleotide compounds containing 2-aza-hypoxanthine or 6H-pyrazolo [1,5-D ] [1,2,4] triazin-7-one as STING agonists
BR112020020085A8 (en) 2018-04-03 2023-04-11 Merck Sharp & Dohme COMPOUND, PHARMACEUTICAL COMPOSITION, AND METHODS FOR INDUCING AN IMMUNE RESPONSE, FOR INDUCING STING-DEPENDENT TYPE I INTERFERON PRODUCTION, AND FOR TREAT A CELL PROLIFERATION DISORDER
US11702430B2 (en) 2018-04-03 2023-07-18 Merck Sharp & Dohme Llc Aza-benzothiophene compounds as STING agonists
WO2019195658A1 (en) 2018-04-05 2019-10-10 Dana-Farber Cancer Institute, Inc. Sting levels as a biomarker for cancer immunotherapy
TWI818007B (en) * 2018-04-06 2023-10-11 捷克科學院有機化學與生物化學研究所 2'3'-cyclic dinucleotides
CA3093888A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
TW202005654A (en) 2018-04-06 2020-02-01 捷克科學院有機化學與生物化學研究所 2'2'-cyclic dinucleotides
CN112074516A (en) 2018-04-25 2020-12-11 先天肿瘤免疫公司 NLRP3 modulators
CN108310378A (en) * 2018-04-28 2018-07-24 杭州星鳌生物科技有限公司 The preparation and its application of a kind of conjuncted antitumor original new drug of novel immune
TW202014193A (en) 2018-05-03 2020-04-16 捷克科學院有機化學與生物化學研究所 2’3’-cyclic dinucleotides comprising carbocyclic nucleotide
GB201807924D0 (en) 2018-05-16 2018-06-27 Ctxt Pty Ltd Compounds
US20210214459A1 (en) 2018-05-31 2021-07-15 Novartis Ag Antibody molecules to cd73 and uses thereof
CN112512531B (en) 2018-06-01 2024-04-09 卫材R&D管理有限公司 Methods for the treatment of bladder cancer
CA3110102A1 (en) * 2018-06-20 2019-12-26 Yale University Rig-i agonists and treatments using same
JP7482122B2 (en) 2018-07-03 2024-05-13 アイエフエム デュー インコーポレイテッド Compounds and compositions for treating conditions associated with STING activity
TW202014408A (en) 2018-07-03 2020-04-16 美商Ifm Due有限公司 Compounds and compositions for treating conditions associated with sting activity
KR20210030973A (en) * 2018-07-11 2021-03-18 액팀 테라퓨틱스, 인코퍼레이티드 Engineered immunostimulatory bacterial strains and uses thereof
WO2020037091A1 (en) 2018-08-16 2020-02-20 Innate Tumor Immunity, Inc. Imidazo[4,5-c]quinoline derived nlrp3-modulators
CA3099904A1 (en) 2018-08-16 2020-02-20 Eisai R&D Management Co., Ltd. Salts of compounds and crystals thereof
CN112996567A (en) 2018-08-16 2021-06-18 先天肿瘤免疫公司 Imidazo [4,5-C ] quinoline-derived NLRP 3-modulators
MX2021001581A (en) 2018-08-16 2021-04-19 Innate Tumor Immunity Inc SUBSTITUED 4-AMINO-<i>1H</i>-IMIDAZO[4,5-C]QUINOLINE COMPOUNDS AND IMPROVED METHODS FOR THEIR PREPARATION.
EP3841112A1 (en) 2018-08-24 2021-06-30 Codiak BioSciences, Inc. Extracellular vesicles targeting dendritic cells and uses thereof
CN112714649A (en) 2018-09-06 2021-04-27 第一三共株式会社 Novel cyclic dinucleotide derivatives and antibody drug conjugates thereof
WO2020049534A1 (en) 2018-09-07 2020-03-12 Novartis Ag Sting agonist and combination therapy thereof for the treatment of cancer
CN112770771A (en) * 2018-09-17 2021-05-07 丘拉提斯股份有限公司 Immune adjuvant containing interferon gene stimulating protein agonist and vaccine composition
CA3113425A1 (en) * 2018-09-21 2020-03-26 Shanghai De Novo Pharmatech Co., Ltd. Cyclic dinucleotide analogue, pharmaceutical composition thereof, and application
US11110106B2 (en) 2018-10-29 2021-09-07 Venenum Biodesign, LLC Sting agonists for treating bladder cancer and solid tumors
WO2020092379A1 (en) 2018-10-29 2020-05-07 Genocea Biosciences, Inc. Treatment methods
WO2020092127A1 (en) 2018-10-29 2020-05-07 Venenum Biodesign, LLC Novel sting agonists
WO2020092633A1 (en) 2018-10-30 2020-05-07 Vanderbilt University Graft copolymers, methods of forming graft copolymers, and methods of use thereof
CN113348181A (en) 2018-10-31 2021-09-03 诺华股份有限公司 DC-SIGN antibody conjugates comprising a STING agonist
US11596692B1 (en) 2018-11-21 2023-03-07 Incyte Corporation PD-L1/STING conjugates and methods of use
WO2020117624A1 (en) * 2018-12-07 2020-06-11 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
WO2020117623A1 (en) * 2018-12-07 2020-06-11 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
EP3891164A4 (en) * 2018-12-07 2022-08-24 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
JP7224688B2 (en) * 2019-01-10 2023-02-20 南▲開▼大学 Cyclic dinucleotide prodrug molecules, methods of making and uses thereof
JP2022517112A (en) 2019-01-14 2022-03-04 イネイト・テューマー・イミュニティ・インコーポレイテッド NLRP3 modulator
WO2020150113A1 (en) 2019-01-14 2020-07-23 Innate Tumor Immunity, Inc. Substituted quinazolines as nlrp3 modulators, for use in the treatment of cancer
ES2930151T3 (en) 2019-01-14 2022-12-07 Innate Tumor Immunity Inc NLRP3 heterocyclic modulators, for use in cancer treatment
WO2020150115A1 (en) 2019-01-14 2020-07-23 Innate Tumor Immunity, Inc. Nlrp3 modulators
WO2020150417A2 (en) 2019-01-17 2020-07-23 Ifm Due, Inc. Compounds and compositions for treating conditions associated with sting activity
US10998091B2 (en) 2019-02-07 2021-05-04 Sandeep Patil Systems for monitoring compliance with a patch dosage regimen and methods of using the same
JP7350872B2 (en) * 2019-03-07 2023-09-26 インスティチュート オブ オーガニック ケミストリー アンド バイオケミストリー エーエスシーアール,ヴイ.ヴイ.アイ. 3'3'-cyclic dinucleotide and its prodrug
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
CA3134216A1 (en) 2019-03-18 2020-09-24 Janssen Pharmaceuticals, Inc. Bioconjugates of e. coli o-antigen polysaccharides, methods of production thereof, and methods of use thereof
EP4253554A3 (en) 2019-03-18 2024-01-17 Janssen Pharmaceuticals, Inc. Methods of producing bioconjugates of e. coli o-antigen polysaccharides, compositions thereof, and methods of use thereof
EP3941937A2 (en) 2019-03-21 2022-01-26 Codiak BioSciences, Inc. Extracellular vesicles for vaccine delivery
CA3133314A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Extracellular vesicle conjugates and uses thereof
WO2020191369A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
EP3946324A4 (en) 2019-04-04 2022-11-30 Merck Sharp & Dohme LLC Inhibitors of histone deacetylase-3 useful for the treatment of cancer, inflammation, neurodegeneration diseases and diabetes
TW202104214A (en) 2019-04-05 2021-02-01 英商葛蘭素史密斯克藍智慧財產發展有限公司 Chemical compounds
EP3962493A2 (en) 2019-05-03 2022-03-09 Flagship Pioneering Innovations V, Inc. Methods of modulating immune activity/level of irf or sting or of treating cancer, comprising the administration of a sting modulator and/or purinergic receptor modulator or postcellular signaling factor
CA3137119A1 (en) 2019-05-09 2020-11-12 Aligos Therapeutics, Inc. Modified cyclic dinucleoside compounds as sting modulators
MX2021013657A (en) 2019-05-10 2022-02-21 Takeda Pharmaceuticals Co Antibody drug conjugates.
JP2022535743A (en) 2019-05-29 2022-08-10 アイエフエム デュー インコーポレイテッド Compounds and compositions for treating conditions associated with STING activity
WO2021003445A1 (en) 2019-07-03 2021-01-07 Codiak Biosciences, Inc. Extracellular vesicles targeting t cells and uses thereof
EP3993834A1 (en) 2019-07-05 2022-05-11 Tambo, Inc. Trans-cyclooctene bioorthogonal agents and uses in cancer and immunotherapy
GB201910304D0 (en) 2019-07-18 2019-09-04 Ctxt Pty Ltd Compounds
GB201910305D0 (en) 2019-07-18 2019-09-04 Ctxt Pty Ltd Compounds
CN114585623A (en) 2019-08-02 2022-06-03 梅尔莎纳医疗公司 Bis [ N- ((5-carbamoyl) -1H-benzo [ d ] imidazol-2-yl) pyrazole-5-carboxamide ] derivatives and related compounds as STING (interferon gene stimulator) agonists for the treatment of cancer
US20210047425A1 (en) 2019-08-12 2021-02-18 Purinomia Biotech, Inc. Methods and compositions for promoting and potentiating t-cell mediated immune responses through adcc targeting of cd39 expressing cells
WO2021041532A1 (en) 2019-08-26 2021-03-04 Dana-Farber Cancer Institute, Inc. Use of heparin to promote type 1 interferon signaling
EP3785719A1 (en) * 2019-08-28 2021-03-03 Helmholtz-Zentrum für Infektionsforschung GmbH New use of cyclic dinucleotides
US20230103726A1 (en) 2019-09-25 2023-04-06 Codiak Biosciences, Inc. Methods of producing extracellular vesicles
MX2022003570A (en) 2019-09-25 2022-07-11 Codiak Biosciences Inc Extracellular vesicle compositions.
EP4034247A1 (en) 2019-09-25 2022-08-03 Codiak BioSciences, Inc. Sting agonist comprising exosomes for treating neuroimmunological disorders
AU2020351729A1 (en) 2019-09-25 2022-04-21 Lonza Sales Ag STING agonist comprising exosomes combined with IL-12 displaying exosomes for treating a tumour
KR20220107166A (en) 2019-10-02 2022-08-02 얀센 백신스 앤드 프리벤션 비.브이. Staphylococcus Peptides and Methods of Use
WO2021074695A1 (en) 2019-10-16 2021-04-22 Avacta Life Sciences Limited PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES.
EP4069683A1 (en) 2019-12-06 2022-10-12 Mersana Therapeutics, Inc. Dimeric compounds as sting agonists
EP4087842A1 (en) 2020-01-10 2022-11-16 Innate Tumor Immunity, Inc. Nlrp3 modulators
JP7485771B2 (en) 2020-01-16 2024-05-16 ヤンセン ファーマシューティカルズ,インコーポレーテッド FimH mutants, compositions thereof, and uses thereof
CA3169523A1 (en) 2020-02-28 2021-09-02 Jaume Pons Transglutaminase-mediated conjugation
MX2022009597A (en) 2020-03-06 2022-09-02 Daiichi Sankyo Co Ltd Antibody-drug conjugate including novel cyclic dinucleotide derivative.
US20230114434A1 (en) 2020-03-13 2023-04-13 Codiak Biosciences, Inc. Extracellular vesicles for treating neurological disorders
JP2023518414A (en) 2020-03-20 2023-05-01 コディアック バイオサイエンシーズ, インコーポレイテッド Extracellular vesicles for therapy
WO2021206158A1 (en) 2020-04-10 2021-10-14 小野薬品工業株式会社 Method of cancer therapy
US11857618B2 (en) 2020-04-17 2024-01-02 Emory University Boosting immunogenicity of vaccines using saponins and agonists of the intracellular stimulator of interferon genes pathway
JP2023526250A (en) 2020-05-13 2023-06-21 マサチューセッツ インスティテュート オブ テクノロジー Compositions of polymer microdevices and their use in cancer immunotherapy
WO2021237100A1 (en) 2020-05-21 2021-11-25 Codiak Biosciences, Inc. Methods of targeting extracellular vesicles to lung
EP4192506A1 (en) 2020-08-07 2023-06-14 Tambo, Inc. Trans-cyclooctene bioorthogonal agents and uses in cancer and immunotherapy
KR20230061360A (en) 2020-09-02 2023-05-08 다이이찌 산쿄 가부시키가이샤 Novel endo-β-N-acetylglucosaminidase
IL308201A (en) 2020-09-17 2024-01-01 Janssen Pharmaceuticals Inc Multivalent vaccine compositions and uses thereof
CA3192470A1 (en) 2020-09-23 2022-03-31 Tim SOOS Methods of producing extracellular vesicles
US20230366794A1 (en) 2020-09-23 2023-11-16 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2022066928A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2022066883A1 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Extracellular vesicles comprising kras antigens and uses thereof
EP4228681A1 (en) 2020-10-14 2023-08-23 Boehringer Ingelheim International GmbH Combination of a sting agonist and a complex comprising a cell penetrating peptide, a cargo and a tlr peptide agonist
EP3984535A1 (en) 2020-10-16 2022-04-20 Albert-Ludwigs-Universität Freiburg Nlrp3 activators for use in the treatment of infectious diseases or cancer by activating nlrp3 inflammasome
US20240059729A1 (en) 2020-10-20 2024-02-22 Tyligand Bioscience (Shanghai) Limited Multifunctional cyclic dinucleotide and use thereof
JP2023548310A (en) 2020-10-30 2023-11-16 アバクタ・ライフ・サイエンシーズ・リミテッド FAP activated serum half-life extended therapeutic conjugate
MX2023005381A (en) 2020-11-09 2023-05-23 Takeda Pharmaceuticals Co Antibody drug conjugates.
JP2023554396A (en) 2020-12-17 2023-12-27 トラスティーズ オブ タフツ カレッジ FAP-activated radiotheranostics and related uses
KR20230125842A (en) 2021-01-12 2023-08-29 얀센 파마슈티칼즈, 인코포레이티드 FimH mutants, compositions comprising them and uses thereof
TW202241454A (en) 2021-02-01 2022-11-01 日商第一三共股份有限公司 Novel method for producing antibody-immunostimulator conjugate
KR20230164108A (en) 2021-04-01 2023-12-01 얀센 파마슈티칼즈, 인코포레이티드 Production of E. coli O18 bioconjugate
EP4326769A1 (en) 2021-04-20 2024-02-28 Institut Curie Compositions and methods for use in immunotherapy
EP4326321A1 (en) 2021-04-20 2024-02-28 Institut Curie Compositions and methods for use in immunotherapy
WO2022234003A1 (en) 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
US20220313724A1 (en) * 2021-05-17 2022-10-06 Highlight Therapeutics, S.L. Compositions for intratumoral administration and related methods
WO2023056468A1 (en) 2021-09-30 2023-04-06 Codiak Biosciences, Inc. Extracellular vesicle comprising cholesterol tagged sting-agonist
TW202334196A (en) 2021-10-07 2023-09-01 英商阿法克塔生命科學有限公司 Pd-l1 binding polypeptides
CN114106140A (en) * 2021-11-23 2022-03-01 哈尔滨医科大学附属肿瘤医院 Expression specific STING isomer, cell line, preparation method and application
WO2023154799A1 (en) 2022-02-14 2023-08-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination immunotherapy for treating cancer
WO2023218243A1 (en) 2022-05-12 2023-11-16 Avacta Life Sciences Limited Lag-3/pd-l1 binding fusion proteins

Family Cites Families (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9110808D0 (en) 1991-05-17 1991-07-10 Retroscreen Ltd Aids vaccine and method for its production
GB2257704B (en) 1991-07-18 1995-03-01 Erba Carlo Spa Cyclic oligonucleotides phosphorothioates
US5637483A (en) 1991-10-04 1997-06-10 Whitehead Institute For Biomedical Research Irradiated tumor cell vaccine engineered to express GM-CSF
US5904920A (en) 1991-10-04 1999-05-18 Whitehead Institute For Biomedical Research Regulation of systemic immune responses utilizing cytokines and antigens
IT1262895B (en) 1992-03-02 1996-07-22 Protein extracted from cytotoxic strains of Helicobacter pylori, gene expressing it, use of the protein as a vaccine or for diagnostic purposes.
US5635160A (en) 1995-06-07 1997-06-03 The University Of North Carolina At Chapel Hill Dinucleotides useful for the treatment of cystic fibrosis and for hydrating mucus secretions
US6033674A (en) 1995-12-28 2000-03-07 Johns Hopkins University School Of Medicine Method of treating cancer with a tumor cell line having modified cytokine expression
US6277368B1 (en) 1996-07-25 2001-08-21 The Regents Of The University Of California Cancer immunotherapy using autologous tumor cells combined with cells expressing a membrane cytokine
US6183121B1 (en) 1997-08-14 2001-02-06 Vertex Pharmaceuticals Inc. Hepatitis C virus helicase crystals and coordinates that define helicase binding pockets
AU741602B2 (en) 1998-02-02 2001-12-06 Johns Hopkins University School Of Medicine, The A universal immunomodulatory cytokine-expressing bystander cell line and related compositions and methods of manufacture and use
EP1153931B1 (en) 1999-02-15 2005-07-20 Nippon Shinyaku Co., Ltd. Shortened-chain polynucleotides and process for the preparation thereof
US6558670B1 (en) 1999-04-19 2003-05-06 Smithkline Beechman Biologicals S.A. Vaccine adjuvants
DE60027719T2 (en) 1999-06-14 2007-04-26 Cancer Research Technology Ltd. CANCER THERAPY
US20020150588A1 (en) 2000-09-21 2002-10-17 Allison James P. SPAS-1 cancer antigen
EP1889630B1 (en) 2000-10-18 2011-11-23 GlaxoSmithKline Biologicals S.A. Vaccines comprising MAGE antigen linked to protein D fragment
US7279883B2 (en) 2001-01-23 2007-10-09 Lydia L. Sohn Particle analyzer and methods for use thereof
TW200303759A (en) 2001-11-27 2003-09-16 Schering Corp Methods for treating cancer
CN1688192B (en) * 2002-08-19 2012-07-18 科勒制药集团有限公司 Immunostimulatory nucleic acids
AR040996A1 (en) * 2002-08-19 2005-04-27 Coley Pharm Group Inc IMMUNE STIMULATING NUCLEIC ACIDS
US8367716B2 (en) * 2003-07-28 2013-02-05 Karaolis David K R Method for attentuating virulence of microbial pathogens and for inhibiting microbial biofilm formation
AU2004281834A1 (en) 2003-10-15 2005-04-28 Cerus Corporation Listeria-based EphA2 vaccines
EP1677765A1 (en) 2003-10-24 2006-07-12 Alza Corporation Preparation of lipid particles
EP1729781B1 (en) 2004-03-15 2012-10-24 Karaolis, David K. R. A method for inhibiting cancer cell proliferation or increasing cancer cell apoptosis
US7592326B2 (en) 2004-03-15 2009-09-22 Karaolis David K R Method for stimulating the immune, inflammatory or neuroprotective response
US20060286549A1 (en) 2005-05-06 2006-12-21 The Regents Of The University Of California Microfluidic system for identifying or sizing individual particles passing through a channel
US20070059683A1 (en) 2005-09-15 2007-03-15 Tom Barber Veterinary diagnostic system
EP1782826A1 (en) * 2005-11-08 2007-05-09 GBF Gesellschaft für Biotechnologische Forschung mbH PQS and c-diGMP and its conjugates as adjuvants and their uses in pharmaceutical compositions
ES2548240T3 (en) 2005-12-01 2015-10-15 Pronai Therapeutics, Inc. Therapies for cancer and pharmaceutical compositions used therein
CA2659301A1 (en) 2006-07-28 2008-02-07 Applera Corporation Dinucleotide mrna cap analogs
US20080076778A1 (en) 2006-09-05 2008-03-27 Bipar Sciences, Inc. Methods for designing parp inhibitors and uses thereof
WO2009133560A1 (en) 2008-04-29 2009-11-05 Smart Assays Non-hydrolyzable and permeable cyclic bis-[nucleotide monophosphate] derivatives and uses thereof
US20110262485A1 (en) 2008-08-04 2011-10-27 University Of Miami Sting (stimulator of interferon genes), a regulator of innate immune responses
US8840881B2 (en) 2008-08-28 2014-09-23 Aduro Gvax Inc. Methods and compositions for treating prostate cancer or inducing a humoral immune response against prostate cancer
WO2010067262A1 (en) 2008-12-09 2010-06-17 Pfizer Inc. Immunostimulatory oligonucleotides
EP2405758B1 (en) 2009-03-09 2016-04-27 Molecular Express, Inc. Methods and compositions for liposomal formulation of antigens and uses thereof
US8414630B2 (en) 2009-03-10 2013-04-09 Marc Evan Richelsoph Active bone screw
BRPI1011072B1 (en) 2009-06-05 2021-09-28 Infectious Disease Research Institute GLA COMPOUND, VACCINE AND PHARMACEUTICAL COMPOSITIONS INCLUDING SUCH COMPOUND, AS WELL AS THEIR USE TO STIMULATE, INDUCE OR EMPHASIZE AN IMMUNE RESPONSE IN AN INDIVIDUAL
US20120178710A1 (en) 2009-07-01 2012-07-12 Rutgers, The State University Of New Jersey Synthesis of cyclic diguanosine monophosphate and thiophosphate analogs thereof
US8771933B2 (en) 2009-10-06 2014-07-08 Massachusetts Institute Of Technology Continuous-flow deformability-based cell separation
US20110293558A1 (en) 2010-03-22 2011-12-01 Massachusetts Institute Of Technology Material properties of t cells and related methods and compositions
CN102199183B (en) 2010-03-26 2013-12-18 北京大学 C-di-GMP, analogues thereof and preparation method thereof
BR112012027745A2 (en) 2010-04-27 2017-01-10 Univ Johns Hopkins method and immunogenic composition for treatment of neoplasia
US8450293B2 (en) 2010-08-10 2013-05-28 Rutgers, The State University Of New Jersey Synthesis and characterization of C8 analogs of c-di-GMP
JP5998370B2 (en) 2010-11-17 2016-09-28 アデュロ バイオテック Methods and compositions for eliciting an immune response against EGFRVIII
US9061048B2 (en) * 2010-12-15 2015-06-23 The Regents Of The University Of California Cyclic di-AMP induction of type I interferon
WO2012088155A1 (en) 2010-12-22 2012-06-28 Alios Biopharma, Inc. Cyclic nucleotide analogs
US9880084B2 (en) 2011-04-15 2018-01-30 The University Of British Columbia Apparatus for separation of particles
WO2013086331A1 (en) 2011-12-07 2013-06-13 President And Fellows Of Harvard College High efficiency di-nucleotide cyclase
CN104540945A (en) 2012-04-30 2015-04-22 格伦·N·巴伯 Modulating immune responses
IN2014MN02492A (en) * 2012-06-08 2015-07-17 Aduro Biotech
US9090646B2 (en) 2012-12-05 2015-07-28 Rutgers, The State University Of New Jersey Biotinylated compounds
EP2931738B1 (en) 2012-12-13 2019-02-06 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
AU2013363087B2 (en) * 2012-12-19 2018-07-19 Board Of Regents, The University Of Texas System Pharmaceutical targeting of a mammalian cyclic di-nucleotide signaling pathway
SG11201508165VA (en) 2013-04-29 2015-11-27 Sloan Kettering Inst Cancer Compositions and methods for altering second messenger signaling
WO2014179760A1 (en) * 2013-05-03 2014-11-06 The Regents Of The University Of California Cyclic di-nucleotide induction of type i interferon
MX354057B (en) 2013-05-18 2018-02-09 The Regents Of The Univ Of California Star Compositions and methods for activating "stimulator of interferon gene"-dependent signalling.
CN105188373B (en) 2013-05-18 2017-09-22 艾杜罗生物科技公司 Suppress the composition and method of " interferon gene stimulates the protein " dependent signals conduction
US9549944B2 (en) 2013-05-18 2017-01-24 Aduro Biotech, Inc. Compositions and methods for inhibiting “stimulator of interferon gene”—dependent signalling
WO2015017652A1 (en) 2013-07-31 2015-02-05 Memorial Sloan-Kettering Cancer Center Sting crystals and modulators
CA2927009C (en) 2013-10-21 2019-04-23 Drexel University Use of sting agonists to treat chronic hepatitis b virus infection
US20160287623A1 (en) 2013-11-19 2016-10-06 The University Of Chicago Use of sting agonist as cancer treatment
WO2015074145A1 (en) 2013-11-22 2015-05-28 Brock University Use of fluorinated cyclic dinucleotides as oral vaccine adjuvants
US10421971B2 (en) 2014-01-15 2019-09-24 The University Of Chicago Anti-tumor therapy
US10450341B2 (en) 2014-06-04 2019-10-22 Glaxosmithkline Intellectual Property Development Limited Cyclic di-nucleotides as modulators of STING
EP3233882B1 (en) 2014-12-16 2019-10-30 Kayla Therapeutics Fluorinated cyclic dinucleotides for cytokine induction
GB201501462D0 (en) 2015-01-29 2015-03-18 Glaxosmithkline Ip Dev Ltd Novel compounds
EP3334745B1 (en) 2015-08-13 2024-05-15 Merck Sharp & Dohme LLC Cyclic di-nucleotide compounds as sting agonists
CN107849084B (en) 2015-12-03 2021-09-14 葛兰素史密斯克莱知识产权发展有限公司 Cyclic purine dinucleotides as STING modulators

Also Published As

Publication number Publication date
ES2754269T9 (en) 2020-08-19
AU2014268836A1 (en) 2015-09-24
CN105228450B (en) 2019-01-18
IL242359B (en) 2020-11-30
EP2996473A4 (en) 2017-01-04
JP6453855B2 (en) 2019-01-16
ME03586B (en) 2020-07-20
EP3653637A1 (en) 2020-05-20
GT201500326A (en) 2018-11-27
ES2754269T3 (en) 2020-04-16
SG11201508273RA (en) 2015-12-30
DK2996473T3 (en) 2019-11-04
MX2015015800A (en) 2016-03-15
SA515370123B1 (en) 2018-02-06
CU20150158A7 (en) 2016-05-30
US20150056224A1 (en) 2015-02-26
AP2015008700A0 (en) 2015-08-31
CU24377B1 (en) 2018-12-05
LT2996473T (en) 2019-12-10
SI2996473T1 (en) 2019-12-31
WO2014189805A8 (en) 2015-01-22
PL2996473T3 (en) 2020-06-01
US20170333552A1 (en) 2017-11-23
NI201500162A (en) 2016-01-25
PH12015502438A1 (en) 2016-03-28
ECSP15052938A (en) 2019-03-29
CN105228450A (en) 2016-01-06
PE20160080A1 (en) 2016-02-21
KR20160009039A (en) 2016-01-25
AU2014268836B2 (en) 2018-08-02
EP2996473B1 (en) 2019-08-21
DOP2015000281A (en) 2016-02-29
BR112015028341A2 (en) 2017-07-25
CY1122274T1 (en) 2020-11-25
MY175308A (en) 2020-06-18
JP2016520085A (en) 2016-07-11
HUE046942T2 (en) 2020-04-28
PT2996473T (en) 2019-11-18
HRP20191972T1 (en) 2020-02-07
PH12015502438B1 (en) 2016-03-28
US10653774B2 (en) 2020-05-19
RS59500B1 (en) 2019-12-31
MX354057B (en) 2018-02-09
HK1222512A1 (en) 2017-07-07
CL2015002522A1 (en) 2016-12-30
CR20150616A (en) 2016-04-19
SV2015005107A (en) 2018-08-20
WO2014189805A1 (en) 2014-11-27
HK1219024A1 (en) 2017-03-24
CA2904536A1 (en) 2014-11-27
US9724408B2 (en) 2017-08-08
EP2996473A1 (en) 2016-03-23

Similar Documents

Publication Publication Date Title
US20200282049A1 (en) Compositions and methods for activating &#34;stimulator of interferon gene&#34;-dependent signalling
AU2018203682B2 (en) Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
US11040053B2 (en) Compositions and methods for activating “stimulator of interferon gene”13 dependent signalling
US10975114B2 (en) Bis 2′-5′-RR-(3′F-A)(3′F-A) cyclic dinucleotide compound and uses thereof
US11098077B2 (en) Locked nucleic acid cyclic dinucleotide compounds and uses thereof
US10906930B2 (en) Compositions and methods for activating “stimulator of interferon gene”-dependent signalling
NZ707561B2 (en) Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VANCE, RUSSELL E.;REEL/FRAME:052692/0910

Effective date: 20140808

Owner name: ADURO BIOTECH, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DUBENSKY, THOMAS W., JR.;KANNE, DAVID B.;LEONG, MEREDITH LAI LING;AND OTHERS;REEL/FRAME:052692/0900

Effective date: 20140912

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION