US20150024037A1 - Xenoantigen-Displaying Anti-Cancer Vaccines and Method of Making - Google Patents

Xenoantigen-Displaying Anti-Cancer Vaccines and Method of Making Download PDF

Info

Publication number
US20150024037A1
US20150024037A1 US14/378,805 US201314378805A US2015024037A1 US 20150024037 A1 US20150024037 A1 US 20150024037A1 US 201314378805 A US201314378805 A US 201314378805A US 2015024037 A1 US2015024037 A1 US 2015024037A1
Authority
US
United States
Prior art keywords
composition
cys
moiety
lys
lipid
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
US14/378,805
Inventor
Steven J. Sucheck
Katherine A. Wall
Sourav Sarkar
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 Toledo
Original Assignee
University of Toledo
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Toledo filed Critical University of Toledo
Priority to US14/378,805 priority Critical patent/US20150024037A1/en
Assigned to THE UNIVERSITY OF TOLEDO reassignment THE UNIVERSITY OF TOLEDO ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SARKAR, Sourav, SUCHECK, STEVEN J., WALL, KATHERINE A.
Publication of US20150024037A1 publication Critical patent/US20150024037A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF TOLEDO
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/001172Sialyl-Thomson-nouvelle antigen [sTn]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/00117Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0012Lipids; Lipoproteins
    • 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
    • A61K47/48046
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6925Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a microcapsule, nanocapsule, microbubble or nanobubble
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/60Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton with the carbon atom of at least one of the carboxyl groups bound to nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4727Mucins, e.g. human intestinal mucin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1275Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Streptococcus (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3092Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated mucins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6018Lipids, e.g. in lipopeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • A61K2039/627Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier characterised by the linker
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues

Definitions

  • the invention relates to compositions having a xenoantigen that is incorporated onto a target antigen and method of making the same.
  • a vaccine composition comprised of a lipid-linked antigen in non-covalent association with a lipid-linked xenoantigen, where both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in a liposomal formulation.
  • humoral or antibody responses are important in pathogen neutralization and can be very effective in some conditions such as cancer and infectious diseases.
  • These therapeutic cancer vaccines are a new class of active specific immunotherapy agents that trigger a targeted immune response against cancer. It would be useful to have efficient methods for the synthesis of such vaccines in addition to those presently available.
  • composition comprising a first lipid (lipid a ) moiety and an alkyne amide moiety having a Formula IV.
  • the first lipid (lipid a ) moiety comprises a Toll-like receptor (TLR) agonist ligand selected from one or more of: TLR2, TLR1, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13, TLR14, TLR15 and TLR16.
  • TLR Toll-like receptor
  • the TLR2 ligand comprises: dipalmitoyl-S-glyceryl-cys-(Pam 2 Cys-); tripalmitoyl-S-glyceryl-cys-(Pam 3 Cys-); dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam 2 Cys-Ser-(Lys) 4 ) [SEQ ID NO: 2]; tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam 3 Cys-Ser-(Lys) 4 ) [SEQ ID NO: 3]; or MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-asp-glu-ser-asn-ile-ser-phe-lys-glu-lys (Pam 2 CGNNDESNISF
  • the Formula IV comprises an alkyne-functionalized Pam 3 Cys amide composition (6), wherein Pam is a dipalmitoyl-S-glyceryl-moiety.
  • a method of synthesizing an alkyne-functionalized composition of claim 1 comprising: deprotecting an ester comprising a Fmoc moiety to form a free acid; coupling the free acid of step (a) with an amine; and, removing the Fmoc moiety, and coupling the remaining moiety with palmitic acid to yield an alkyne-functionalized composition.
  • step (b) coupling the free acid of step (a) with propargyl amine in presence of benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), 1-hydroxy-benzotriazole (HOBt) and N,N-diisopropylethylamine (DIPEA) to yield composition (5); and,
  • lipidated glycopeptide composition comprising: a first lipid (lipida) moiety, a first linker (linkera) moiety, and an antigen moiety having a Formula II.
  • the first lipid (lipida) moiety comprises a Toll-like receptor (TLR) agonist ligand selected from one or more of: TLR2, TLR1, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13, TLR14, TLR15 and TLR16.
  • TLR Toll-like receptor
  • the first lipid (lipida) moiety comprises a TLR2 ligand comprising:
  • the antigen moiety comprises: an anti-cancer composition, an anti-bacterial composition, an anti-viral composition, a protein, an isolated DNA, an isolated RNA, an isolated carbohydrate, or an isolated lipid.
  • the antigen moiety comprises a TACA that interacts with B-cell receptors.
  • the TACA comprises one or more glycoproteins and glycolipids on a cancer cell.
  • the composition comprises mucin 1 (MUC1) variable number tandem repeats (VNTRs) conjugated to tumor-associated carbohydrate antigens (TACA).
  • MUC1 mucin 1
  • VNTRs variable number tandem repeats conjugated to tumor-associated carbohydrate antigens
  • the TACA comprises: TF, Tn, sialyl Tn (sTn), or sialyl Lewis a (sLea) antigens.
  • the TACA comprises: an autologous or heterologous helper T-cell epitope, wherein the autologous or heterologous helper T-cell epitope comprises a sequence expressed on a tumor cell.
  • the he autologous or heterologous helper T-cell epitope comprises MUC1 VNTR having one of the following amino acid sequences:
  • the threonine in the sequence GST or PDT is synthetically modified to incorporate ⁇ -GalNAc-O-Thr (Tn) TACA.
  • the first linker comprises a dialkyl-substituted heteroaryl C1-n alkyl of Formula VI, wherein the “A” group comprises: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n-; and n is a positive integer.
  • the “A” group comprises a C1-5 alkyl chain.
  • composition comprising the compound (9).
  • composition comprising the compound (17).
  • composition comprising the compound (21).
  • a method of synthesizing a lipidated glycopeptide composition comprising reacting a composition of Formula IV, with a composition Formula V.
  • a method of synthesizing a Pam3Cys-MUC-1 VNTR-TACA conjugate comprising: synthesizing a 20-amino acid tandem repeat of MUC1; and modifying a glycopeptide with a terminal azido group to make a ‘click’ conjugation to a Pam3Cys alkyne.
  • the 20-amino acid tandem repeat of MUC1 comprises a GS( ⁇ -GalNAc-O-T)A epitope.
  • the 20-amino acid tandem repeat of MUC1 comprises a PD( ⁇ -GalNAc-O-T)R epitope.
  • composition comprising a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety, having the Formula VII.
  • the second lipid (lipidb) moiety contains a structure of the Formula IX.
  • the second linker comprises: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n-; wherein n is a positive integer.
  • the second linker comprises a tetraethyleneglycol (TEG) of Formula VIII.
  • the xenoantigen moiety comprises a xenoantigen linked to a lipid.
  • the xenoantigen binds a natural antibody (NA).
  • NA natural antibody
  • the NA comprises an autoantigen that is present in blood.
  • the NA comprises IgM, IgG, or IgA isotypes in human blood.
  • the NA comprises an anti-carbohydrate NA in human serum.
  • the xenoantigen moiety contains a structure comprising: an ⁇ - or ⁇ -linked L-rhamnose epitope, a ⁇ -linked ⁇ -Gal disaccharide epitope, or an ⁇ - or ⁇ -linked Forssmann disaccharide epitope:
  • a vaccine composition comprising: an antigen composition comprising:
  • a xenoantigen composition comprising: a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety; and,
  • a vaccine composition wherein a lipid-linked antigen is in non-covalent association with a lipid-linked xenoantigen, and wherein both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in a liposomal formulation.
  • the liposomal formulation comprises 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and cholesterol in a ratio of from about 80:20 to about 70:30, respectively.
  • DPPC 1,2-dipalmitoyl-sn-glycero-3-phosphocholine
  • a vaccine composition of claim 37 wherein: the antigen composition comprises a Pam3Cys-MUC1 VNTR-TACA conjugate; the second linker (linkerb) moiety comprises a tetraethyleneglycol (TEG) portion; and the xenoantigen moiety comprises ⁇ - or ⁇ -linked L-rhamnose.
  • the antigen composition comprises a Pam3Cys-MUC1 VNTR-TACA conjugate
  • the second linker (linkerb) moiety comprises a tetraethyleneglycol (TEG) portion
  • TEG tetraethyleneglycol
  • an anti-cancer vaccine comprising a vaccine composition as described herein
  • an anti-viral vaccine comprising a vaccine composition as described herein.
  • an anti-bacterial vaccine comprising a vaccine composition as described herein.
  • the vaccine composition further includes at least one immunologic adjuvant.
  • the immunologic adjuvant comprises one or more of: a saponin, monophosphoryl lipid A, 3′-O-deacylated monophosphoryl lipid A, and interleukin 12.
  • a method for eliciting an immune response to a cancer cell surface antigen in a subject with cancer comprising administering to the subject an antigen-xenoantigen-liposome composition of claim 37 in sufficient dose to elicit the immune response to the cancer cell surface antigen, wherein the antigen-liposome-xenoantigen composition comprises the cancer cell surface antigen, and wherein the immune response is sufficient for treating the cancer.
  • the both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in the liposomal formulation.
  • the cancer cell surface antigen is expressed only in cancer cells in the subject.
  • the cancer cell surface antigen is expressed only by cancer cells.
  • the cancer cell surface antigen comprises a peptide epitope.
  • the cell surface antigen comprises MUC1 mucin.
  • the subject is a human or non-human mammal.
  • the antigen-liposome-xenoantigen composition is administered intranasally, intramuscularly, subcutaneously, intravenously, or orally to the subject.
  • the method further comprises measuring humoral and/or cellular immune responses to the cancer cell surface antigen, and administering a further dose to elicit the immune response, if necessary.
  • a method for selectively killing cancer cells expressing a cancer cell surface antigen in a subject in need thereof comprising administering to the subject an antigen-liposome-xenoantigen composition of claim 27 , under conditions that result in production in the subject of antibodies against the cancer cell surface antigen, wherein the antibodies produced bind the cancer cell surface antigen on cancer cells in the subject, thereby killing cancer cells that express the cancer cell surface antigen.
  • both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in the liposomal formulation.
  • the cancer cell surface antigen is expressed only in cancer cells in the subject.
  • the cancer cell surface antigen is expressed only by cancer cells.
  • a method for improving immunogenicity of vaccines comprising incorporating at least one ⁇ - or ⁇ -linked L-Rha epitope by direct conjugation or by non-covalent association with at least one liposomal vaccine formulation to increase the immunogenicity of the vaccine by a NA-dependant antigen uptake mechanism.
  • an alkyne functionalized composition (IV) comprising: deprotecting an ester having a Fmoc moiety to form a free acid; coupling the free acid with an amine; and, removing the Fmoc moiety, and coupling the remaining moiety with palmitic acid to yield the alkyne functionalized composition.
  • the antigen moiety comprises: an anti-cancer composition, an anti-bacterial composition, or an anti-viral composition.
  • the antigen moiety comprises: a TACA that interacts with B-cell receptors.
  • the TACA comprises an autologous helper T-cell epitope.
  • a method of synthesizing component 1) above comprising reacting an alkyne-functionalized amide derivative with a modified peptide antigen.
  • the xenoantigen contains a structure comprised of: ⁇ - or ⁇ -linked L-rhamnose, a ⁇ -linked ⁇ -Gal epitope, or an ⁇ - or ⁇ -linked Forssmann disaccharide epitope.
  • a method for eliciting an immune response to a cancer cell surface antigen in a subject with cancer comprising administering to the subject an antigen-liposome-xenoantigen composition described herein, in sufficient dose to elicit the immune response to the cancer cell surface antigen.
  • FIG. 2 Schematic illustration of a liposomal vaccine composition comprised of: an antigen composition comprised of a first lipid (lipid a ) moiety, a first linker (linker a ) moiety, and an antigen moiety; and, a second lipid (lipid b ) moiety, a second linker (linker b ) moiety, and a xenoantigen moiety; at least one liposome forming lipid; and, optionally, one or more adjuvants.
  • an antigen composition comprised of a first lipid (lipid a ) moiety, a first linker (linker a ) moiety, and an antigen moiety
  • lipid b second lipid
  • linker b linker
  • xenoantigen moiety at least one liposome forming lipid
  • adjuvants optionally, one or more adjuvants.
  • FIG. 3 Schematic illustration of a lipid-linked antigen component.
  • FIG. 4 Schematic illustration of a dialkyl-substituted heteroaryl C 1-n alkyl component.
  • FIG. 5 Schematic illustration of a composition IV reacting with a composition V to form a composition II.
  • FIG. 6 Schematic illustration of a linker a component.
  • FIG. 7 Schematic illustration of a lipid-linked xenoantigen component.
  • FIG. 8 TEG composition used in a linker b component.
  • FIG. 9 Schematic illustration of a structure contained in a lipid b portion of a lipid-linked xenoantigen component.
  • FIG. 10 Scheme 1: Schematic illustration of synthesis of a L-Rhamnose-TEG-Cholesterol (3).
  • FIG. 11 Scheme 2: Schematic illustration of synthesis of an alkyne functionalized Pam 3 Cys composition (6).
  • FIG. 12 Scheme 3: Schematic illustration of synthesis of a Pam 3 Cys-MUC-1 VNTR-TACA conjugate (9) [SEQ ID NOS: 8, 9 and 7], respectively, in order of appearance.
  • FIGS. 13A-13C Size characterization of liposomes at 1/10,000 dilution: FIG. 13 A—Batch 1 liposomes; FIG. 13 B—Batch 2 liposomes; and FIG. 13 C—Batch 3 liposomes.
  • FIGS. 14A-14B Photographs showing size characterization of liposomes: SEM images at 5 kV acceleration voltage.
  • FIG. 14A Batch 1 liposomes under 50,000 ⁇ magnification.
  • FIG. 14B Batch 1 liposomes under 250,000 ⁇ magnification.
  • FIGS. 15A-15C Fluorescence microscope images with Batch 1 liposomes under 60 ⁇ magnification.
  • FIG. 15A Images with control antibodies (antibodies isolated from preimmunization serum) 1st, 2nd and 3rd images: brightfield, FITC and overlay.
  • FIG. 15B Images with anti-rhamnose antibodies, 1st, 2nd and 3rd images: brightfield, FITC and overlay.
  • FIG. 15C Images with anti-MUC 1 antibodies, 1st, 2nd and 3rd images: brightfield, FITC and overlay.
  • FIG. 16 1 H NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl-2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 17 13 C NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl-2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 18 1 H-gCosy of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl 2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 19 HRMS of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl 2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 20 1 H NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 21 13 C NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 22 1 H-gCosy of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 23 HRMS of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 24 1 H NMR of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 25 13 C NMR of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 26 1 H gCosy of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 27 HRMS of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 28 1 H NMR of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 29 13 C NMR of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 30 1 H-gCosy of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 31 HRMS of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 32 HR-MALDI-TOF of Glycopeptide Azide (9) [SEQ ID NO:8].
  • FIG. 33 HR-MALDI-TOF of Glycopeptide Azide (8) [SEQ ID NO: 10].
  • FIG. 34 HR-MALDI-TOF of Lipopeptide (9) [SEQ ID NO:7].
  • FIG. 35 Scheme showing synthetic route of 2-Aminoethyle- ⁇ -L-Rhamnopyranoside (13).
  • FIG. 36 Anti-Rha Antibody Isotype Titers after 4th Boost with Rha-Ficoll (group A) or Rha-OVA (group B) at 1/5 or 1/500 Serum Dilutions respectively.
  • FIG. 37 T-Cell Proliferation in BALB/c mice with MUC1-Tn (10) Peptide in Presence and Absence of Dendritic Cells.
  • FIGS. 38A-38 T-cell proliferation measured by [ 3 H]thymidine incorporation in T-cells from mice spleens primed with MUC1-Tn (8) and challenged with Pam 3 Cys-MUC1-Tn (9) +Rha liposomes in the presence of anti-Rha antibodies (abs) or control abs [anti Rha(OVA) and anti Rha(Ficoll) abs are the antibodies isolated from the serum of Rha-OVA and Rha-Ficoll immunized mice, respectively].
  • FIG. 38B Stepwise immunization plan. Groups A1, A2, B1, and B2 each represent four groups of female BALB/c mice.
  • Stage I groups A2 and B2 were immunized with Rha-Ficoll/Alum whereas groups A1 and B1 were non-immunized.
  • Stage II vaccination; groups A1 and A2 vaccinated and boosted with Pam 3 Cys-MUC1-Tn liposomes whereas groups B2 and B2 were vaccinated with Pam 3 Cys-MUC1-Tn+Rha liposomes.
  • FIGS. 39A-39B Group average of anti-Rha antibody titers after fourth boost with Rha-Ficoll/Alum.
  • FIG. 39B Group average of anti-MUC1-Tn antibody titers after first boost with Pam 3 Cys-MUC1-Tn liposomes or Pam 3 Cys-MUC1-Tn+Rha liposomes.
  • FIG. 40 Anti-MUC1-Tn Antibody Isotype Titers after first boost with Pam 3 Cys-MUC1-Tn liposomes or Pam 3 Cys-MUC1-Tn+Rha liposomes at 1/50 Serum Dilutions.
  • FIGS. 41A-41B FIG. 41A ) Competitive binding of anti-MUC1-Tn antibodies with bound MUC1-Tn in presence of free MUC1-Tn (8).
  • FIG. 41B Groups average of anti-Tn antibody titer after first boost with Pam 3 Cys-MUC1-Tn liposomes or Pam 3 Cys-MUC1-Tn+Rha liposomes.
  • FIGS. 42A-42B Binding of anti-MUC1-Tn antibodies to human leukemia U266 cells.
  • FIG. 42A Second antibody alone and with mouse anti-human MUC1 antibodies;
  • FIG. 42B with 1/5 dilution of non-immunized mouse serum, and with 1/5 dilution of group B2 mouse serum.
  • FIG. 43 1 H NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 44 13 C NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 45 1 H-gCosy of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 46 HRMS of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 47 1 H NMR of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 48 13 C NMR of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 49 1 H-gCosy of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 50 HRMS of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 51 ESI MS of 2-Aminoethyl ⁇ -L-rhamnopyranoside (13).
  • FIG. 52 Scheme showing the synthesis of Pam 3 Cys-MUC-1 VNTR conjugate (17) [SEQ ID NOS: 11, 11 and 12], respectively, in order of appearance.
  • FIG. 53 HRMS (MALDI-TOF) spectrum of peptide (16).
  • FIG. 54 HPLC trace of peptide (16).
  • FIG. 55 HRMS (MALDI-TOF) spectrum of lipopeptide (17).
  • FIG. 56 Synthesis of Pam 3 Cys-MUC-1 VNTR conjugate (21) [SEQ ID NOS: 13, 14 15], respectively, in order of appearance.
  • FIG. 57 HRMS (MALDI-TOF) of glycopeptide (19).
  • FIG. 58 HPLC trace of glycopeptide (19).
  • FIG. 59 HRMS (MALDI-TOF) of glycopeptide (20).
  • FIG. 60 HRMS (MALDI-TOF) of Pam 3 Cys-MUC-1 VNTR conjugate (21).
  • antibody broadly refers to monoclonal antibodies (including full length monoclonal antibodies) and “antibody fragments” that exhibit a desired biological activity. “Antibody fragments” can comprise a portion of a full length antibody, generally the antigen binding or variable region thereof. Non-limiting examples of antibody fragments include Fab, Fab′, and Fv fragments; diabodies; linear antibodies; and single-chain antibody molecules.
  • monoclonal antibody broadly refers to antibodies that are highly specific, being directed against a single antigenic site.
  • antibody also broadly includes naturally occurring antibodies (NAs) as well as non-naturally occurring antibodies, including, for example, single chain antibodies, chimeric, bifunctional and humanized antibodies, as well as antigen-binding fragments thereof.
  • NAs naturally occurring antibodies
  • non-naturally occurring antibodies can be constructed using solid phase peptide synthesis, can be produced recombinantly or can be obtained, for example, by screening combinatorial libraries consisting of variable heavy chains and variable light chains.
  • subject/s generally refers to any animal that generates an adaptive immune response and can include mammals, birds and reptiles. Examples of subjects can include, but are not limited to, humans, non-human primates, dogs, cats, horses, cows, goats, guinea pigs, mice, rats and rabbits, as well as any other domestic or commercially valuable animal.
  • nucleic acid/s generally encompass both RNA and DNA, including cDNA, genomic DNA, mRNA, synthetic (e.g., chemically synthesized) DNA and chimeras of RNA and DNA.
  • the nucleic acid can be double-stranded or single-stranded. Where single-stranded, the nucleic acid can be a sense strand or an antisense strand.
  • the nucleic acid can be synthesized using oligonucleotide analogs or derivatives (e.g., inosine or phosphorothioate nucleotides). Such oligonucleotides can be used, for example, to prepare nucleic acids that have altered base-pairing abilities or increased resistance to nucleases.
  • C 1-n alkyl can be any linear or branched alkyl group containing 1 to n carbon atoms.
  • the term“C 1-6 alkyl” can comprise groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl(3-methylbutyl), neopentyl(2,2-dimethylpropyl), hexyl, isohexyl(4-methylpentyl) and the like.
  • C 2-n alkyl is to be understood to mean any linear or branched alkyl group containing 2 to n carbon atoms.
  • C 2-6 alkyl can comprise groups such as ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl(3-methylbutyl), neopentyl(2,2-dimethylpropyl), hexyl, isohexyl(4-methylpentyl) and the like.
  • dialkyl-substituted aryl C 1-n alkyl is to be understood to mean an aryl group substituted at two positions wherein the substituents are composed of a C 1-n alkyl group, as defined above.
  • the aryl group may be optionally substituted with at least one substituent selected from the group consisting of hydroxyl, C 1-2 alkoxy and halogen.
  • Examples of dialkyl-substituted aryl C 1-n alkyl groups include, but are not limited to:
  • dialkyl-substituted heteroaryl C 1-n alkyl is to be understood to mean a heteroaryl group substituted at two positions wherein the substituents are composed of a C 1-n alkyl group, as defined above.
  • the aryl group may be optionally substituted with at least one substituent selected from the group consisting of hydroxyl, C 1-2 alkoxy and halogen.
  • substituted heteroaryl C 1-n alkyl groups include, but are not limited to:
  • xenoantigen is to be understood to mean at least one foreign antigen capable of binding natural antibodies.
  • xenoantigens include, but are not limited to, the ⁇ -gal epitope, L-Rha, and the Forssman disaccharide.
  • peptide may be any peptide comprising natural or non-natural amino acids; and if chiral, in its L or D configurations or as racemate.
  • non-natural amino acids include, but are not limited to: homocysteine, homoarginine, cylcohexylalanine, ornithine, and C ⁇ -dibenzylglycine.
  • lipid is defined to include any of a broad range of substances that is characteristically insoluble in water and extractable with an organic solvent. This broad class of compounds is known to those of skill in the art, and as the term “lipid” is used herein, it is not limited to any particular structure. Examples include compounds which contain long-chain aliphatic hydrocarbons and their derivatives. A lipid may be naturally occurring or synthetic (i.e., designed or produced by man).
  • Biological lipids are well known in the art and include, for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof.
  • neutral fats for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof.
  • lipids are also encompassed by the compositions and methods described herein.
  • composition having xenoantigen that is incorporated onto a target antigen.
  • a vaccine composition comprising a lipid-linked antigen in non-covalent association with a lipid-linked xenoantigen, where both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in a liposomal formulation.
  • TACAs tumor-associated carbohydrate antigens
  • MUC1 heavily glycosylated glycoprotein mucin 1
  • TACAs tumor-associated carbohydrate antigens
  • MUC1 heavily glycosylated glycoprotein mucin 1
  • the non-covalent association of the L-Rhamnose xenoantigen in a liposome with the other components of a vaccine independently embedded in the liposome eliminates the need to chemically synthesize a complex xenoantigen-antigen conjugate molecule.
  • the peptide sequences described herein act as an autologous helper T-cell epitope.
  • the use of foreign carrier proteins and peptides as conjugates provide helper T-cell epitopes unrelated to the tumor and act as heterologous helper T-cell epitopes.
  • the MUC1 sequence described herein is shown to be a potent candidate for generating antibodies.
  • TACAs tumor associated carbohydrate antigens
  • TACAs found on the heavily glycosylated glycoprotein mucin 1 are useful targets for anticancer active immunotherapy.
  • MUC1 glycoprotein mucin 1
  • TACAs includes TF, Tn, STn, sLe a antigens:
  • the above TACAs (which are found on tumor cells) are useful as the “B-cell epitopes” in the design of the vaccines described herein.
  • the MUC1 glycoprotein is also shed into the serum and is a tumor marker for cancer.
  • patients possessing naturally occurring anti-MUC1 antibodies demonstrate better disease-specific survival.
  • anti-MUC1 antibodies control hematogenous tumor dissemination and outgrowth.
  • TACAs present on MUC1 variable number tandem repeat (VNTR) domain may help to break the immune system's self tolerance to MUC1, at least in human MUC1-expressing transgenic mice while unglycosylated human MUC1 VNTRs generated a weaker immune response. While not wishing to be bound by theory, it is now also believed that this is because the glycopeptide is possibly seen by the immune system as a more foreign epitope in comparison to the unglycosylated MUC1 which is more self-like.
  • APCs antigen presenting cells
  • the effectiveness of vaccines can be increased by the incorporation of a xenoantigen.
  • the xenoantigen Gal ⁇ 1-3Gal ⁇ 1 ( ⁇ Gal) can be used as an in vivo generated immune complex between the ⁇ Gal epitopes and the natural antibodies (NA) against ⁇ Gal in serum.
  • the Fc portion on this immune complex is recognized by the Fc ⁇ receptors on the APCs, thereby leading to the overall internalization of the antigen facilitating presentation by the major histocompatibility complex (MHC) on APCs.
  • MHC major histocompatibility complex
  • NA can also enhance uptake by APCs using other receptors such as complement receptors.
  • NAs that can bind antigens, including autoantigens, normally present in the blood.
  • the NAs which can be of the IgM, IgG, or IgA isotype in humans, exhibit specific binding to a variety of different antigens, including proteins, DNA, numerous neutral and anionic phospholipids, carbohydrates, and a variety of other lipids, including neutral and anionic glycolipids, cholesterol, and squalene.
  • a large fraction of NAs-to-phospholipid is circulating in vivo as immune complexes to phospholipids.
  • the lipids and phospholipids present in liposomes can complex NAs as well.
  • Antibodies against both ⁇ / ⁇ L-Rha are more prevalent than those against ⁇ -Gal.
  • a vaccine composition that incorporates the non-covalent association of the L-Rha xenoantigen in a liposome with one or more other components of the vaccine composition that are independently embedded in the liposome.
  • This incorporation eliminates the need to chemically synthesize a complex xenoantigen-antigen conjugate molecule.
  • any lipidated antigen can be introduced into the vaccine composition.
  • One non-limiting example is the L-Rha motif which binds anti-L-Rha NAs.
  • the Fc portion of the NA of an in vivo generated immune complex is recognized by the Fc ⁇ receptors located on the surface of the APCs or by complement and complement receptors, thereby leading to the better internalization of the vaccine.
  • the T-cell proliferation is enhanced when the antigen and xenoantigen are co-localized on a liposome in the presence of APCs and anti L-Rha antibodies.
  • the xenoantigen is conjugated to a lipid (such as cholesterol) by means of a linker (such as tretraethylene glycol (TEG)), which allows the xenoantigen to be displayed on the surface of the liposome along with the lipidated-tumor, bacterial or viral antigen.
  • a linker such as tretraethylene glycol (TEG)
  • the linker between the xenoantigen and the cholesterol lipid allows for a xenoantigen-NA binding interaction.
  • an antigen is required for the development of a vaccine (e.g., an anti-cancer vaccine, an anti-viral vaccine, or an anti-bacterial vaccine).
  • an antigen is defined as an entity or foreign molecule that, when introduced into the body, triggers the production of an antibody by the immune system.
  • the term “antigen” also refers to any molecule or molecular fragment that can be bound by a major histocompatibility complex (MHC) and presented to a T-cell receptor.
  • MHC major histocompatibility complex
  • TACAs Tumors of epithelial origin provide a variety of TACAs that can act as antigens by interacting with B-cell receptors. These include TACAs identified on the glycoproteins and glycolipids that decorate the cancer cells.
  • TACAs include: the TF, Tn, sialyl Tn (sTn) and sialyl Lewis a (sLe a ) antigens, whose structures are shown above. These structures as well as many other TACAs can produce a considerable humoral immune response.
  • peptides and glycopeptides can interact with B-cell receptors to produce antibodies.
  • TACAs alone as cancer vaccines, however, is their weak immunogenicity.
  • Generation of high amounts of high affinity IgG antibodies against the cancer antigen depends upon the combined interaction of the B-cells and the helper T-cells, and therefore requires the vaccine to contain a peptide epitope which can be displayed on the major histocompatibility complex (MHC) class-II or class-I molecules that are present on the surface of APCs, such as DCs.
  • MHC major histocompatibility complex
  • TACAs alone cannot activate the helper T-cells, resulting in poor immunogenicity.
  • TACAs To counteract this problem involved the conjugation of TACAs to large carrier proteins like keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA), TLR agonists and zwitterionic polysaccharides.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • TLR agonists TLR agonists
  • zwitterionic polysaccharides TACAs to large carrier proteins
  • one drawback to the protein conjugation strategy is that the major immune response is towards the carrier protein, thus resulting in a suppressed immune response for the cancer antigen.
  • the protein conjugates provide the peptide sequences, which protein sequences can then be displayed as helper T-cell epitopes on MHC class-II molecules.
  • the resulting complexes activate CD4 + helper T-cells which then interact with antibody-producing B-cells allowing the B-cells to produce higher affinity antibodies.
  • TACAs For example, smaller 20-amino acid peptides which can bind MHC class-II can be conjugated to TACAs. These conjugates can produce high amounts of high affinity IgG anti-TACA antibodies.
  • This strategy provides an advantage over the use of large carrier proteins as helper T-cell epitope, as the immune system can now focus more resources on the available TACA antigens rather than producing antibodies to the carrier protein.
  • the use of peptide sequences acts as an “autologous helper T-cell epitope.”
  • autologous helper T-cell epitopes can be sequences expressed on the tumor.
  • the use of foreign carrier proteins and peptides as conjugates thus provide helper-T cell epitopes unrelated to the tumor, and thus act as “heterologous helper T-cell epitopes.”
  • an autologous helper T-epitope is the MUC1 VNTR consisting of the 20 amino acid sequence PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO: 1].
  • the threonine in the sequence GST is synthetically modified to incorporate the ⁇ -GalNAc-O-Thr (Tn) TACA.
  • This region in the MUC1 sequence is a more potent candidate for generating high antibody titers against Tn than the unglycosylated MUC1 VNTR.
  • Glycopeptides formed by the conjugation of B-cell epitopes and helper T-cell epitopes often show moderate immunogenicity. A major reason for such moderate immunogenicity is the inappropriate maturation of the DCs.
  • TLR Toll-like receptor
  • TLR2 interactive lipopeptides include: dipalmitoyl-S-glyceryl-cys-(Pam 2 Cys); tripalmitoyl-S-glyceryl-cys-(Pam 3 Cys-); dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam 2 Cys-Ser-(Lys) 4 ) [SEQ ID NO: 2], tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys-lys (Pam 3 Cys-Ser-(Lys) 4 ) [SEQ ID NO: 3], and MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-asp-glu-ser-asn-ile-ser-phe-lys-glu-lys (Pam 2 CGNNDESNISFKE
  • Non-limiting examples of palmitoylated lipids [SEQ ID NOS: 3, 2 and 4], respectively, in order of appearance, that may interact with TLR2 thus include:
  • Pam 2 Cys-Ser-(Lys) 4 [SEQ ID NO:2] acts as TLR2/TLR6 agonists
  • Pam 3 Cys-Ser-(Lys) 4 [SEQ ID NO:3] acts as a TLR2/1 agonist
  • Lipopeptide MALP-2 acts a TLR2/6 ligand.
  • dipalmitoyl-S-glyceryl-cys (Pam 2 Cys) are minimal structural epitopes common to palmitoylated TLR2 ligands.
  • the lipid component of a vaccine composition serves three purposes: first, the lipid component acts as a vaccine adjuvant capable of interacting with Toll-Like receptors; second, the lipid component facilitates the formulation of the glycolipopeptide into liposomes anchoring the glycopeptide epitope to the inner and outer surfaces of the liposomes; and third, the lipidation facilitates cross-presentation of peptide antigens by APCs.
  • NAs natural antibodies
  • a desired target epitope e.g., the xenoantigen L-Rha epitope
  • the bound NAs thus facilitate the opsonization of the liposomal vaccine by APCs.
  • Covalent attachment of xenoantigens to lipids through the use of a suitable linker thus facilitates the incorporation of these epitopes into the liposomes.
  • the xenoantigen is covalently attached to a lipid through the use of a suitable linker.
  • the length of the linker between the xenoantigen and lipid can be optimized to achieve a desired xenoantigen-NA interaction.
  • the conjugation of a lipid to the xenoantigen allows for the display of multiple xenoantigens on the surface of the liposomes, thereby facilitating multivalent NA-antibody binding to the liposome.
  • the Fc portion of the NA in the in vivo generated immune complex is thus recognized by the Fc ⁇ receptors on APCs, thereby leading to opsonization of the vaccine by APCs.
  • the composition of the vaccine contains non-covalently associated components: i) a lipid-linked antigen; ii) a composition of lipids capable of forming liposomes; iii) a lipid-linked xenoantigen; and, optionally, iv) one or more independent adjuvant molecules.
  • a lipid-linked antigen is comprised of an antigen linked to a first lipid (also called lipid a herein).
  • the antigen can be a peptide or glycopeptide of 1 to 100 amino acids in length.
  • the antigen may also include a B-cell epitope conjugated to a peptide or glycopeptide sequence that is capable of providing a T-cell epitope.
  • the peptide, glycopeptide, peptide-B-cell, or glycopeptides-B-cell epitope conjugate is linked to lipid a .
  • composition II The generalized composition of the lipid a -linked antigen component is the structure of composition II, shown in FIG. 3 .
  • the antigen contains a glycopeptide having the 20 amino acid sequence PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO:1].
  • the T(Tn) in the sequence is the ⁇ -GalNAc-O-T (Tn) TACA.
  • the antigen contains a glycopeptide having the 20 amino acid sequence TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5].
  • the antigen contains a glycopeptides having the 20 amino acid sequence TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6].
  • the T(Tn) in the sequence is the ⁇ -GalNAc-O-T (Tn) TACA.
  • the linker a component can be: a chain of C 2-n alkyl, dialkyl-substituted aryl C 1-n alkyl, dialkyl-substituted heteroaryl C 1-n alkyl, or —CH 2 CH 2 (OCH 2 CH 2 ) n —, where n is a positive integer.
  • the linker a component comprises a dialkyl-substituted heteroaryl C 1-n alkyl of formula III, as shown in FIG. 4 .
  • the lipid a portion of the formula III contains a lipid of one of the following structures: Pam 2 Cys-Ser-(Lys) 4 , Pam 3 Cys-Ser-(Lys) 4 , MALP-2, Pam 3 Cys, or Pam 2 Cys.
  • the first lipid (lipid a ) component of the formula III can contain a lipid having the structure Pam 3 Cys, or the structure Pam 2 Cys.
  • composition IV shows in FIG. 5
  • composition V forms a composition of composition II.
  • the lipid a moiety of the composition IV can be a structure of the Formulae [SEQ ID NOS: 3, 2 and 4], respectively, in order of appearance:
  • the “A” group therein represents a linker.
  • the linker can be comprised of: a chain of C 1-n alkyl, dialkyl substituted aryl C 1-n alkyl, or —CH 2 CH 2 (OCH 2 CH 2 ) n —; n is a positive integer.
  • A comprises a C 1-5 alkyl chain.
  • the antigen moiety can be a peptide or glycopeptide that is covalently bonded at the N-terminus to a carbonyl group on the linker, thus forming an amide bond.
  • the antigen contains a glycopeptide having a 20 amino acid sequence PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO:1], where the T(Tn) in the sequence GSTA is the ⁇ -GalNAc-O-Thr TACA.
  • the antigen contains a glycopeptide having the 20 amino acid sequence TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5].
  • the antigen contains a glycopeptide having the 20 amino acid sequence TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6].
  • the T(Tn) in the sequence is the ⁇ -GalNAc-O-T (Tn) TACA.
  • composition II Upon reaction of composition IV with composition V in the presence of an appropriate catalyst, a composition II is formed.
  • the linker a moiety can be a dialkyl-substituted heteroaryl C 1-n alkyl of formula VI, as shown in FIG. 6 .
  • the “A” group can be a linker comprised of: a chain of C 1-n alkyl, dialkyl substituted aryl C 1-n alkyl, or —CH 2 CH 2 (OCH 2 CH 2 ) n —; n is a positive integer.
  • the “A” group comprises a C 1-5 alkyl chain.
  • the lipid-linked xenoantigen can be a xenoantigen linked to a lipid.
  • the xenoantigen can be any structure which binds NAs.
  • the xenoantigen is linked to a second lipid (lipid b ) through a second linker (linker b ).
  • the generalized composition of the lipid-linked xenoantigen component is structure of composition VII as shown in FIG. 7 .
  • the xenoantigen contains a structure comprised of: alpha- or beta-linked L-rhamnose, a beta-linked alpha-gal epitope, or an alpha- or beta-linked Forssmann disaccharide epitope.
  • the xenoantigen contains a structure consisting of alpha- or beta-linked L-rhamnose.
  • the linker b component can be comprised of: a chain of C 1-n alkyl, dialkyl substituted aryl C 1-n alkyl, or —CH 2 CH 2 (OCH 2 CH 2 ) n —; where n is a positive integer.
  • the linker b component has a tetraethyleneglycol (TEG) of formula VIII, as shown in FIG. 8 .
  • the lipid b portion can contain a structure of the formula IX, as shown in FIG. 9 .
  • the liposomal forming lipids can be comprised of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and cholesterol in a ratio of from about 80:20 to about 70:30, respectively.
  • DPPC 1,2-dipalmitoyl-sn-glycero-3-phosphocholine
  • an immunologic adjuvant may or may not be included as part of the composition of the vaccine.
  • the immunologic adjuvants may be, but are not limited to: a saponin (e.g., QS21), monophosphoryl lipid A, 3′-O-deacylated monophosphoryl lipid A, or interleukin 12.
  • compositions of the present invention comprise an effective amount of a compound(s) or composition(s) disclosed herein, and/or additional agents, dissolved or dispersed in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to molecular entities and compositions that produce no adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human.
  • the preparation of a pharmaceutical composition that contains at least one compound or additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference.
  • preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards.
  • a vaccine composition disclosed herein may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection.
  • Vaccine compositions disclosed herein can be administered intravenously, intradermally, transdermally, intrathecally, intraarterially, intraperitoneally, intranasally, intravaginally, intrarectally, topically, intramuscularly, subcutaneously, mucosally, in utero, orally, topically, locally, via inhalation (e.g., aerosol inhalation), by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference).
  • the actual dosage amount of a composition disclosed herein administered to an animal or human patient can be determined by physical and physiological factors such as body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • compositions may comprise, for example, at least about 0.1% of an active compound.
  • an active compound may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein.
  • the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
  • a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg/body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc. can be administered, based on the numbers described above.
  • a vaccine composition herein and/or additional agents is formulated to be administered via an alimentary route.
  • Alimentary routes include all possible routes of administration in which the composition is in direct contact with the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered orally, buccally, rectally, or sublingually.
  • these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsules, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • a vaccine composition described herein may be administered via a parenteral route.
  • parenteral includes routes that bypass the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered, for example but not limited to, intravenously, intradermally, intramuscularly, intraarterially, intrathecally, subcutaneous, or intraperitoneally (U.S. Pat. Nos. 6,753,514, 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363 are each specifically incorporated herein by reference in their entirety).
  • Solutions of the vaccine compositions disclosed herein as free bases or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, specifically incorporated herein by reference in its entirety).
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (i.e., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • a coating such as lecithin
  • surfactants for example
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption such as, for example, aluminum monostearate or gelatin.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal administration.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580).
  • Sterile injectable solutions are prepared by incorporating the vaccine compositions in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized compositions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • some methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • a powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
  • the vaccine compositions may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or via inhalation.
  • topical i.e., transdermal
  • mucosal administration intranasal, vaginal, etc.
  • inhalation via inhalation.
  • compositions for topical administration may include the vaccine compositions formulated for a medicated application such as an ointment, paste, cream or powder.
  • Ointments include all oleaginous, adsorption, emulsion and water-soluble based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only.
  • Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin. Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram.
  • compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base.
  • Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the vaccine composition and provide for a homogenous mixture.
  • Transdermal administration of the vaccine compositions may also comprise the use of a “patch.”
  • the patch may supply one or more vaccine compositions at a predetermined rate and in a continuous manner over a fixed period of time.
  • the vaccine compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
  • Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in their entirety).
  • the delivery of drugs using intranasal microparticle resins (Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725,871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts and could be employed to deliver the vaccine compositions described herein.
  • transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety), and could be employed to deliver the vaccine compositions described herein.
  • the vaccine compositions disclosed herein may be delivered via an aerosol.
  • aerosol refers to a colloidal system of finely divided solid or liquid particles dispersed in a liquefied or pressurized gas propellant.
  • the typical aerosol for inhalation consists of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent.
  • Suitable propellants include hydrocarbons and hydrocarbon ethers.
  • Suitable containers will vary according to the pressure requirements of the propellant.
  • Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms.
  • Silica (230-400 mesh) for flash column chromatography was obtained from Sorbent Technologies; thin-layer chromatography (TLC) precoated plates were from EMD. TLCs (silica gel 60, f 254 ) were visualized under UV light or by charring (5% H 2 SO 4 -MeOH). Flash column chromatography was performed on silica gel (230-400 mesh) using solvents as received.
  • Tris [(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA), preloaded Fmoc-L-Ala-Wang resin and all other Fmoc-L-amino acids were procured from Anaspec (San Jose, Calif.).
  • FITC goat anti-mouse IgG/IgM and purified mouse anti-human CD227 (anti-human MUC1) were obtained from BD-biosciences (San Jose, Calif.).
  • Scanning electron microscope imaging was done on a JEOL JSM-7500F field scanning electron microscope. Dynamic light scattering measurements was done DynaPro Titan temperature controlled microsampler (Wyatt Technology Corporation). Fluorescence microscopy was done on a Nikon TiU microscope.
  • Cholesterol tetraethylene glycol (1) was glycosylated with peracetyl L-rhamnose in presence of boron trifluoride etherate to afford peracetyl rhamnose-TEG-cholesterol (2) (32%) which was deacetylated under Zémplen conditions to generate L-rhamnose-TEG-cholesterol (3) (85%). See FIG. 10 , illustrating Scheme 1.
  • Compound (3) anchors the Rha epitopes on the surface of the liposomes thereby facilitating anti-Rha antibody binding.
  • reaction mixture was diluted with CH 2 Cl 2 (25 mL) and washed with saturated NaHCO 3 (25 mL), water (25 mL) and brine (25 mL) after which the organic layer was dried over anhydrous Na 2 SO 4 . Excess solvent was evaporated under reduced pressure and the residue was purified by silica gel flash column chromatography using 30% EtOAc in hexanes as solvent to afford (2) as a light yellow solid (0.51 g, 32%).
  • Fmoc group in compound (5) was removed by treatment with a mixture of acetonitrile-dichloromethane-diethyl amine (2:1:2) followed by subsequent palmitoylation by coupling with palmitic acid, PyBOP, HOBt and DIPEA to afford our target alkyne functionalized Pam 3 Cys amide derivative (6) (80% over 2 steps), as shown in FIG. 11 , Scheme 2.
  • Compound (6) which is a TLR-2 agonist, serves the purpose of an immunoadjuvant for a vaccine, and also anchors the MUC1-Tn conjugate on the surface of the liposome.
  • a 20-amino acid tandem repeat of MUC1 which included the GS( ⁇ -GalNAc-O-T)A epitope was synthesized.
  • the glycopeptide was modified with a terminal azido group in order to make a ‘click’ conjugation to the Pam 3 Cys alkyne.
  • the glycopeptide azide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) starting from preloaded Fmoc-L-Ala Wang resin using solid-phase chemistry, as shown in FIG. 12 , Scheme 3.
  • the peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent. A six-fold excess of N ⁇ -Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of N ⁇ -Fmoc group was accomplished by treatment with piperidine in DMF. After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of TFA-thioanisole-ethanedithiol-water-phenol (88:3:5:2:2). The cocktail mixture was filtered through a Quick Snap column, purified by C18 reverse phase HPLC and lyophilized to afford composition (7b).
  • composition (7b) The acetyl groups in composition (7b) were deprotected by treatment with 6 mmol sodium methoxide in methanol.
  • the product was purified by Bio-Gel (P-2, fine 45-90 ⁇ m) size exclusion chromatography using deionized water as solvent. Lyophilization of the fractions afforded composition (8) (100%) as a white powder.
  • composition (9) was purified by LH20 using MeOH-dichloromethane (1:1) as solvent. The eleutants were lyophilized to afford composition (9) as a white solid (100%).
  • the glycopeptide azide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) using solid phase chemistry.
  • the peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent starting with a preloaded Fmoc-alanyl-Wang resin.
  • a six-fold excess of N ⁇ -Fmoc amino acid esters of HOBt in NMP were used in the synthesis.
  • a 1:1 ratio of amino acid to DIC was used in all the coupling reactions.
  • Deprotection of N ⁇ -Fmoc group was accomplished by treatment with 25% piperidine in dimethylformamide twice; first for 5 minutes and then a second time for 25 minutes to afford composition (7a).
  • the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of 88% TFA, 3% thioanisole, 5% ethanedithiol, 2% water and 2% phenol. 4 mL of cleavage cocktail was added to the dried peptide-resin in a 15 mL glass vial blanketed with nitrogen. Cleavage was carried out for 2.5 hrs with gentle magnetic stirring. At the end cleavage time, the cocktail mixture was filtered on a Quick-Snap column. The filtrate was collected in 20 mL ice-cold butane ether.
  • a modified reagent K cocktail consisting of 88% TFA, 3% thioanisole, 5% ethanedithiol, 2% water and 2% phenol. 4 mL of cleavage cocktail was added to the dried peptide-resin in a 15 mL glass vial blanketed with nitrogen. Cleavage was
  • the peptide was allowed to precipitate for an hour at ⁇ 200° C., centrifuged, and washed twice with ice-cold methyl-t-butyl ether. The precipitate was dissolved in 25% acetonitrile and lyophilized to complete dry powder affording composition (7b). Quality of peptides was analyzed by analytical reverse phase HPLC and MALDI-TOF (matrix assisted laser desorption ionization time-of-flight) mass spectrometer, model 4800 from Applied Biosystems. HR-MALDI-MS: [M+H] m/z calcd for C 100 H 155 N 29 O 37 , 2355.1172. Found, 2355.1753.
  • composition (7b) (5 mg, 2.24 ⁇ mol) was dissolved in 2 mL of dry methanol and 12 ⁇ L of freshly prepared 1 M sodium methoxide was added and the reaction mixture was stirred at ambient temperature under N 2 atmosphere for 2 h. The reaction mixture was neutralized with solid carbon dioxide. The reaction mixture was concentrated and purified by Bio-Gel (P-2, fine 45-90 ⁇ m) size exclusion chromatography using deionized water as solvent. Lyophilization of the eleutants afforded composition (8) as a white powder (4.7 mg, 100%).
  • HR-MALDI-MS [M+H] m/z calcd for C 94 H 149 N 29 O 34 , 2229.0895. Found 2229.0959.
  • the liposomes were formulated by the extrusion method in a total lipid concentration of 30 mM.
  • 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) was used for the preparation of the liposomes.
  • Lipid stock solutions were prepared by dissolving each lipid into chloroform inside glass vials. Aliquots of the stock solutions were mixed in proportions in another small glass vial to give a solution with a total lipid concentration of 30 mM in a total volume of 2 mL.
  • Chloroform was removed by subjecting the lipid solutions to a constant stream of nitrogen.
  • the resulting lipid films were dried under vacuum overnight.
  • the suspensions of the lipids in the buffer were agitated at 43° C. for 40 mins.
  • the suspensions were subjected to 10 freeze-thaw cycles (dry ice/acetone and water at 40° C.).
  • Final liposomes were prepared by extrusion (21 times) using a LipoFast Basic fitted with a 100 nm polycarbonate membrane to control the liposome size.
  • the homogeneity, stability as well as size characterization of the liposomes were evaluated by scanning electron microscope (SEM) imaging ( FIGS. 14A-14B ) and dynamic light scattering (DLS) measurements ( FIGS. 13A-13C ).
  • the binding assay proved that the L-rhamnose and the MUC1 VNTR-TACA conjugate were displayed on the surface of the liposomes. No such antibody binding (both anti-rhamnose and mouse anti-human-MUC1) were observed for the control Batch 3 liposomes.
  • Size determination of the liposomes was done by scanning electron microscope (SEM) imaging and dynamic light scattering (DLS) measurements.
  • SEM scanning electron microscope
  • DLS dynamic light scattering
  • NMR spectra and HRMS data of compositions (2), (3), (5) and (6) are shown in FIG. 16 through FIG. 31 .
  • FIG. 16 1 H NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl-2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 17 13 C NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl-2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 18 1 H-gCosy of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl 2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 19 HRMS of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl 2,3,4-tri-O-acetyl- ⁇ -L-Rhamnopyranoside (2).
  • FIG. 20 1 H NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 21 13 C NMR of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 22 1 H-gCosy of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 23 HRMS of (5-Cholesten-3 ⁇ -yloxy)-3n 3 9 -trixaundecanyl Rhamnopyranoside (3).
  • FIG. 24 1 H NMR of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 25 13 C NMR of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 26 1 HgCosy of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 27 HRMS of N-Propargyl Pam 2 FmocCys Amide Derivative (5).
  • FIG. 28 1 H NMR of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 29 13 C NMR of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 30 1 H-gCosy of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • FIG. 31 HRMS of N-Propargyl Pam 3 Cys Amide Derivative (6).
  • compositions (7b), (8) and (9) are shown in FIG. 32 through FIG. 34 .
  • FIG. 32 HR-MALDI-TOF of Glycopeptide Azide (7b).
  • FIG. 33 HR-MALDI-TOF of Glycopeptide Azide (8).
  • FIG. 34 HR-MALDI-TOF of Lipopeptide (9).
  • reaction mixture was diluted with CH 2 Cl 2 (15 mL) and washed with water (2 ⁇ 20 mL), saturated NaHCO 3 (2 ⁇ 20 mL) and brine (20 mL), after which the organic layer was dried over anhydrous Na 2 SO 4 . Excess solvent was removed under reduced pressure and the crude material was purified by silica gel flash column chromatography (3.3 ⁇ 8.5 cm). Elution with 1:5 EtOAc/hexanes afforded (11) as a colorless solid (1.78 g, 83%).
  • Ficoll 400 (1.00 g, 0.0025 mmol) was dissolved in acetate buffer (10 mL, pH 4.7) and NaIO 4 (0.01 g, 0.047 mmol) was added and the reaction mixture was stirred at ambient temperature for 2 h in the dark. Excess NaIO 4 was removed by dialysis against the acetate buffer (pH 4.7) through dialysis tubing with a molecular weight cutoff value of 10000 Da with six to seven changes of the buffer at 4° C. The oxidized Ficoll 400 was transferred to a round bottom flask and excess solvent was evaporated to dryness under reduced pressure.
  • the epitope ratio of (14) was calculated to be 9.44 (Rha:Ficoll) by hydrolysis of (14) followed by derivatization with 4-amino-N-[2-(diethylamino)ethyl]benzamide (DEAEAB) and comparison of the UV-HPLC peak area with a standard curve obtained from DEAEAB derivative of (12) by the methods described by Dalpathado and coworkers. Briefly, the standard curve was generated by refluxing compound (12) (0.007 g, 0.031 mmol) with 1 M HCl at 100° C. for 4 h and the reaction mixture was evaporated to dryness. The residue was dissolved in tetrahydrofuran (2 mL).
  • FIG. 43 1 H NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 44 13 C NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 45 1 H-gCosy of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 46 HRMS of 2-Azidoethyl-2,3,4-Tri-O-acetyl- ⁇ -L-rhamnopyranoside (11).
  • FIG. 47 1 H NMR of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 48 13 C NMR of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 49 1 H-gCosy of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 50 HRMS of 2-Azidoethyl ⁇ -L-rhamnopyranoside (12).
  • FIG. 51 ESI MS of 2-Aminoethyl ⁇ -L-rhamnopyranoside (13).
  • a 20-amino acid (TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5]) tandem repeat of MUC1 was synthesized.
  • the peptide was modified with a terminal azido group in order to make a ‘click’ conjugation to the Pam 3 Cys alkyne with a terminal azido group.
  • the peptide was synthesized by a Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) starting from preloaded Fmoc-L-Val Wang resin using solid-phase chemistry, as shown in FIG. 52 .
  • the peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent. A six-fold excess of N ⁇ -Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of N ⁇ -Fmoc group was accomplished by treatment with piperidine in DMF. After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of TFA-thioanisole-ethanedithiol-water-phenol (88:3:5:2:2). The cocktail mixture was filtered through a Quick Snap column, purified by C18 reverse phase HPLC, and lyophilized to afford composition (16) as a white powder.
  • composition (17) was purified by neutralization by 7.5% citric acid solution followed by extraction with chloroform. Evaporation of the chloroform extract afforded the composition (17) as a white solid.
  • the peptidazide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) using solid phase chemistry.
  • the peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent starting with a preloaded Fmoc-valinyl-Wang resin.
  • a six-fold excess of N ⁇ -Fmoc amino acid esters of HOBt in NMP were used in the synthesis.
  • a 1:1 ratio of amino acid to DIC was used in all the coupling reactions.
  • Deprotection of N ⁇ -Fmoc group was accomplished by treatment with 25% piperidine in dimethylformamide twice; first for 5 minutes and then a second time for 25 minutes to afford composition (15).
  • the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of 88% TFA, 3% thioanisole, 5% ethanedithiol, 2% water, and 2% phenol. 4 mL of cleavage cocktail was added to the dried peptide-resin in a 15 mL glass vial blanketed with nitrogen. Cleavage was carried out for 2.5 hrs with gentle magnetic stirring. At the end cleavage time, the cocktail mixture was filtered on a Quick-Snap column. The filtrate was collected in 20 mL ice-cold butane ether.
  • a modified reagent K cocktail consisting of 88% TFA, 3% thioanisole, 5% ethanedithiol, 2% water, and 2% phenol. 4 mL of cleavage cocktail was added to the dried peptide-resin in a 15 mL glass vial blanketed with nitrogen. Clea
  • the peptide was allowed to precipitate for an hour at ⁇ 200° C., centrifuged, and washed twice with ice-cold methyl-t-butyl ether. The precipitate was dissolved in 25% acetonitrile and lyophilized to complete dry powder affording composition (16). Quality of peptides was analyzed by analytical reverse phase HPLC and MALDI-TOF (matrix assisted laser desorption ionization time-of-flight) mass spectrometer, model 4800 from Applied Biosystems. HR-MALDI-MS: [M+H] m/z calcd for C 86 H 136 N 28 O 29 , 2026.17. Found, 2026.137.
  • FIGS. 53 and 54 show the HRMS (MALDI-TOF) spectrum and HPLC trace, respectively, of peptide (16).
  • FIG. 55 shows the HRMS (MALDI-TOF) spectrum of lipopeptide (17).
  • a 20-amino acid (TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6]) tandem repeat of MUC1 which included the PD( ⁇ -GalNAc-O-T)R epitope was synthesized.
  • the glycopeptides was modified with a terminal azido group in order to make a ‘click’ conjugation to the Pam 3 Cys alkyne.
  • the glycopeptides azide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) starting from preloaded Fmoc-L-Val Wang resin using solid-phase chemistry, as shown in FIG. 56 .
  • the peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent. A six-fold excess of N ⁇ -Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of N ⁇ -Fmoc group was accomplished by treatment with piperidine in DMF. After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of TFA-thioanisole-ethanedithiol-water-phenol (88:3:5:2:2). The cocktail mixture was filtered through a Quick Snap column, purified by C18 reverse phase HPLC and lyophilized to afford composition (19).
  • composition (19) The acetyl groups in composition (19) were deprotected by treatment with 6 mmol sodium methoxide in methanol.
  • the product was purified by Bio-Gel (P-2, fine 45-90 ⁇ m) size exclusion chromatopgraphy using deionized water as solvent. Lyophilization of the fractions afforded composition (20) (100%) as a white powder.
  • composition (19) (1 eqv) Conjugation of the composition (19) (1 eqv) with the composition (6) (3 eqv) in the presence of copper(I) iodide (12 eqv), tris [(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA) (3 eqv), diisopropyl ethyl amine (DIEA) (12 eqv) and sodium ascorbate (12 eqv) in H 2 O-THF-DMF (1:1:2) as solvent at 20° C. thus afforded the target, Pam 3 Cys-MUC1 VNTR-TACA conjugate (21), after 16 h.
  • Composition (21) was purified by neutralization by 7.5% citric acid solution followed by extraction with chloroform. Evaporation of the chloroform extract afforded the composition (21) as light yellow solid.
  • composition (19) (5 mg, 2.24 ⁇ mol) was dissolved in 2 mL of dry methanol. 12 ⁇ L of freshly prepared 1 M sodium methoxide was added to the mixture and the reaction mixture was stirred at ambient temperature under N 2 atmosphere for 3 h. The reaction mixture was neutralized with solid carbon dioxide. The reaction mixture was concentrated and purified by Bio-Gel (P-2, fine 45-90 ⁇ m) size exclusion chromatography using deionized water as solvent. Lyophilization of the eleutants afforded composition (20) as a white powder (4.7 mg, 100%).
  • HR-MALDI-MS [M+H] m/z calcd for C 94 H 149 N 29 O 34 , 2229.09. Found 2026.336.
  • FIGS. 57 and 58 show the HRMS (MALDI-TOF) spectrum and HPLC trace of glycopeptide (19).
  • FIG. 59 is the HRMS (MALDI-TOF) spectrum of glycopeptides (20).
  • FIG. 60 is the HRMS (MALDI-TOF) spectrum of Pam 3 Cys-MUC-1 VNTR conjugate (21).
  • Also provided herein is a method for eliciting an immune response against a cancer cell surface antigen in a subject in need thereof.
  • the method comprises administering to the subject an antigen-liposome-xenoantigen vaccine composition in sufficient dose to elicit the immune response to the cancer cell surface antigen, wherein the antigen-liposome-xenoantigen vaccine composition comprises the cancer cell surface antigen and a liposome.
  • the cancer cell surface antigen is expressed only in cancer cells in the subject, such that the immune response specific for the antigen would not attack healthy tissues/organs of the subject.
  • the antigen that is expressed on the cancer cell surface is a mutant protein present only on cancer cells, and the immune response is specific for the mutant protein epitope comprising the mutation.
  • the mutation can be a protein mutation, or abnormal sugar or lipid structures on cancer cell surface.
  • Rha-Ficoll and the Rha-OVA immunized mice were bled on day 66 and the sera was pooled.
  • IgG fractions from each pool were prepared by precipitation at 40% saturation of ammonium sulfate. The mixtures were incubated overnight and centrifuged at 10000 ⁇ g for 10 minutes and then resuspended in 0.5 mL water. The antibody solutions were concentrated and buffer was changed twice with PBS using an Ultrafree 0.5 centrifugal filter device (Millipore, Billerica, Mass.) having a molecular cut off of 50000 D.
  • spleen cell culture medium DMEM with 10% fetal calf serium
  • Single cell suspension was prepared using modified sterile glass homogenizers. The cells were washed three times with culture medium and brought to 5 ⁇ 10 6 cells/mL. 100 ⁇ L of the spleen cell suspensions were added to 96 well plates (5 ⁇ 10 5 cells per well).
  • the dendritic cell (DC) suspension cultured from the bone marrow of a BALB/c mouse was pulsed with the antigen by incubating with the Rha-displaying MUC1-Tn liposomes at antigen concentrations of 8.8 ⁇ 10 ⁇ 3 -1.1 ⁇ g/mL at 37° C. for 4 h together with anti-Rha antibodies generated from either Rha-Ficoll or Rha-OVA immunized mice sera (5 ⁇ Ci/mL, 25 ⁇ L per well) and incubated overnight at 37° C.
  • the cells were harvested on glass-fiber filters and incorporation was determined by measurements on a Top Count scintillation counter.
  • mice used for this study were divided into four groups A1, A2, B1, and B2, containing 5 mice each.
  • Groups A1 and B1 served as the control groups and were not immunized.
  • Groups A2 and B2 were injected subcutaneously (day 0) with a 100 ⁇ L subcutaneous injections Rha-Ficoll/Alum on days 14, 28, 42, and 56 (100 ⁇ g of Rha-Ficoll per mouse, each boost).
  • the mice in each group A1, A2, B1, and B2 were bled on day 66 and the collected sera were tested for anti-Rha antibodies.
  • 96 well plates (Immulon 4 HBX) were coated with Rha-BSA conjugate (6) (2 ⁇ g/mL) in 0.01 M PBS and incubated overnight at 4° C. The plates were washed 5 times with PBS containing 0.1% Tween-20. Blocking was achieved by incubating the plates for 1 h at room temperature with BSA in 0.01 M PBS (1 mg/mL). The plates were then washed 5 times and incubated for 1 h with serum dilutions in PBS.
  • HRP Horseradish Peroxidase
  • Vaccinations were performed on day 77. Two separate liposomal formulations were prepared with DPPC (80%), cholesterol (20%) and Pam 3 Cys-MUC1-Tn (9) (2 nmol) (Pam 3 Cys-MUC1-Tn liposomes) and DPPC (80%), cholesterol (10%), Rha-TEG-cholesterol 3 (10%) and Pam 3 Cys-MUC1-Tn (9) (2 nmol) (Pam 3 Cys-MUC1-Tn+Rha liposomes) in total lipid concentrations of 30 mmol.
  • Groups A1 and A2 were cavvinated with 100 ⁇ L subcutaneous injections of the Pam 3 Cys-MUC1-Tn liposomes (2 nmol of peptide per mouse) and groups B1 and B2 were vaccinated with 100 ⁇ L subcutaneous injections of the Pam 3 Cys-MUC1-Tn+Rha liposomes (2 nmol peptide per mouse).
  • the mice were boosted on day 91 with either the Pam 3 Cys-MUC1-Tn liposomes (groups A1 and A2, 2 nmol peptide per mouse) or the Pam 3 Cys-MUC1-Tn+Rha liposomes (groups B1 and B2).
  • the mice were bled on day 101 and the sera collected were tested for anti-MUC1-Tn and anti-Tn antibodies.
  • subclass specific IgG1, IgG2s, IgG2b, IgG3, 1gA, and IgM
  • rabbit anti-mouse antibody Zymed Laboratories mouse monoAb-ID kit
  • the plates were washed and incubated with HRP-goat anti-rabbit IgG (H+L) for 1 h at room temperature.
  • the plates were washed and ABTS substrate buffer (diluted 50 times) was added and allowed to react for 30 min. Absorbances were recorded at 405 nm and compared for each antibody subclass in each group.
  • a 96-well plate (Immulon 4 HBX) was coated with MUC1-Tn conjugate (8) (15 ⁇ L/mL) in PBS and incubated overnight at 4° C. The plate was washed 5 times with PBS containing 0.1% Tween-20. Blocking was achieved by incubating the plate for 1 h at room temperature with BSA in M PBS (1 mg/mL). The plate was then washed 5 times and incubated for 1 h with serum dilutions of 1/100 in PBS with or without prior mixing with varying concentrations of free MUC1-Tn (8) from 0, 10 ⁇ 5 , 10 ⁇ 4 , 10 ⁇ 3 M in PBS.
  • HRP Horseradish Peroxidase
  • U266 cells (American Type Culture Collection, Manassas, Va.), were cultured in RPMI 1640 with 15% fetal calf serum (FCS). Cells were stained with purified mouse anti-human MUC1 antibodies (CD227, 0.5 ⁇ g), non-immune BALB/c mice serum (1/5 dilution) and group B2 mice serum (1/5 dilution). The cells were then stained with FITC-conjugated goat anti-mouse IgG+IgM (0.5 ⁇ g) and fluorescence was quantified with a BD FACS Calibur.
  • FCS fetal calf serum
  • mice Two groups of five female BALB/c mice each were immunized on day 0 with Rha-Ficoll/Alum adjuvant (group A) or Rha-OVA/complete Freund's adjuvant (CFA) (group B). The mice were boosted three more times on days 14, 28, and 42 with either Rha-Ficoll/Alum (group A) or rha-OVA/incomplete Freund's adjuvant (ICF) (group B). Sera were collected separately from groups A and B after the third boost and the anti-Rha antibodies in the sera from the two groups of mice were isotyped by screening against Rha-BSA. See FIG. 36 .
  • Rha-Ficoll and Rha-OVA produced the anti-Rha antibody subclasses in different proportions.
  • Anti-Rha antibodies from Rha-OVA immunization were dominated by IgG1 (65%) while Rha-Ficoll immunization produced antibodies which comprised mainly IgG3 (48%) and IgM (25%).
  • IgG1 and IgG3 act similarly in that they both stimulate high affinity Fc ⁇ RI receptors which trigger responses from macrophages.
  • IgG1 also stimulates low affinity Fc ⁇ RIIB receptors which inhibit the signals from the Fc ⁇ RI and B cell receptors thereby diminishing B-cell activity and immunogenicity of macrophages.
  • the anti-Rha antibody isotypes from Rha-Ficoll immunized mice serum resembled those naturally occurring in the human serum which is presumed to be generated through a T-independent response.
  • T-cell proliferation assays were performed to determine if the combination of anti-Rha antibodies and Rha-modified liposomal vaccine would potentiate a T-cell proliferative response.
  • the proliferation assay conditions were optimized.
  • BALB/c mice were immunized (day 0) and boosted (days 14, 28, and 42) with 100 ⁇ L emulsions of MUC1-Tn (8)/Sigma adjuvant system (SAS) (50 ⁇ g peptide per mouse, each injection).
  • SAS Sigma adjuvant system
  • mice were sacrificed (day 49), the spleens were removed, and single cell suspensions were prepared and incubated with MUC1-Tn (8.8 ⁇ 10 ⁇ 3 -1.1 ⁇ g/mL) alone or with syngeneic bone marrow dendritic cells (DCs) previously pulsed with the same doses of antigen.
  • DCs showed enhanced proliferation, as shown in FIG. 37 .
  • spleen cells from BALB/c mice immunized as above were prepared.
  • DCs from BALB/c bone marrow were pulsed with the antigen by incubating with Pam 3 Cys-MUC1-Tn+Rha liposomes at antigen concentrations of 8.8 ⁇ 10 ⁇ 3 -0.22 ⁇ g/mL together with antibodies isolated from either Rha-Ficoll or Rha-OVA immunized mice or nonimmune mice.
  • the pulsed DCs were added to the spleen cells and proliferation assessed after 3 days.
  • the spleen T-cells proliferated better in the presence of anti-Rha antibodies (from both Rha-Ficoll and Rha-OVA immunized mice serum) than in the presence of control serum antibodies over the antigen concentration range of 8.8 ⁇ 10 ⁇ 3 -0.22 ⁇ g/mL.
  • the T-cell proliferation was higher in the presence of anti-Rha antibodies generated against Rha-Ficoll (6328, 6045, and 6521 counts per minute (cpm) at antigen concentrations of 8.8 ⁇ 10 ⁇ 3 , 0.044, and 0.22 ⁇ g/mL) than those against Rha-OVA (5018, 4926, and 4880 cpm at antigen concentrations of 8.8 ⁇ 10 ⁇ 3 , 0.044, and 0.22 ⁇ g/mL), even though the titer of anti-Rha antibodies was higher in the serum of Rha-OVA immunized mice.
  • Rha-modified antigen was more effectively internalized and presented by the APCs in the presence of anti-Rha antibodies, particularly those less inhibitory isotypes characteristic of natural antibodies and generated by Rha-Ficoll immunization. Therefore, BALB/c mice in which anti-Rha antibodies are generated with Rha-Ficoll (14) immunization are an appropriate model for the immunogenicity of the Rha-conjugated MUC1-Tn liposomes.
  • Groups A1, A2, B1, and B2 Four groups of five female BALB/c mice each (groups A1, A2, B1, and B2) (6-8 weeks old) were used for this vaccination study. Groups A2 and B2 were immunized (day 0) and boosted (days 14, 28, 42, and 56) with 100 ⁇ L equivolume emulsion of Rha-Ficoll (prepared in PBS) and alum adjuvant. Groups A1 and B1 served as the control groups and were deprived of the Rha-Ficoll/Alum immunization. See FIG. 38B .
  • mice were bled on day 66 and the ELISA performed by screening the sera from the different groups against Rha-BSA showed that the anti-Rha antibody titers in groups A2 and B2 were 25-fold higher than the control groups. See FIG. 39 .
  • immunization with Rha-Ficoll confirmed the generation of anti-Rha antibodies in the experimental groups of mice.
  • Two separate liposomal formulations were prepared. The first contained DPPC, cholesterol and Pam 3 Cys-MUC1-Tn (9) (2 nmol) (Pam 3 Cys-MUC1-Tn liposomes) and the second contained DPPC, cholesterol, Rha-TEG-cholesterol (3) and Pam 3 Cys-MUC1-Tn (9) (2 nmol) (Pam 3 CysMUC1-Tn+Rha liposomes). In both formulations the total lipid concentration was 30 mmol. The vaccination was performed on day 77.
  • mice were boosted on day 91 with either the Pam 3 CysMUC1-Tn liposome (groups A1 and A2, 2 nmol peptide per mouse) or the Pam 3 CysMUC1-Tn+Rha liposome (groups B1 and B2). The mice were bled on day 101 and the sera evaluated for anti-MUC1-Tn and anti-Tn antibodies. See FIG. 39B .
  • Anti-MUC1-Tn antibody titers were determined by screening the sera against the MUC1-Tn conjugate (8). ( FIG. 39B ). The data showed that groups A1, A2, and B1 had similar absorbance at 1/25, 1/50, 1/100, and 1/200 serum dilutions. This proved that prior immunization with Rha-Ficoll does not affect the response to a non-Rha conjugated vaccine (groups A1 and A2). In addition, the Rha epitopes on the vaccine do not alter the inherent immunogenicity of the MUC1-Tn epitopes on the vaccine (groups A1 and B1).
  • the anti-MUC1-Tn titers for group B2 showed an 8-fold increase compared to groups A1, A2, and B1, which was mediated by the anti-Rha antibody-dependent antigen-uptake.
  • Group B2 had an anti-MUC1-Tn titer of approximately 1/300, where titer is defined as the highest dilution giving a signal >0.1 above background.
  • the anti-Muc1-Tn antibodies from each group were isotyped, resulting in group B2 showing an increase in IgG1, IgG2a, IgG2b, and IgM isotypes relative to the other three groups. See FIG. 40 .
  • the specificity of the antibodies towards MUC1-Tn antigen was determined by a competitive binding experiment. See FIG.
  • the antibody titer generated solely against the TACA was determined by screening serum dilutions from every group against a Tn-BSA conjugate. See FIG. 41B .
  • a >8-fold increase in the anti-Tn antibody titers for group B2 was also observed in comparison to groups A1, A2, and B1. This was again attributed to the better uptake of the antigen in the presence of the anti-Rha antibodies by an antibody-dependent antigen-uptake mechanism.
  • the anti-MUC1-Tn antibody titers were higher than the corresponding anti-Tn antibody titers for the same serum dilutions for every group, assuming similar levels of antigen on the plate.
  • the absorbances at 620 nm for the anti-MUC1-Tn and the anti-Tn measurements at 1/100 serum dilutions were 0.922 and 0.509, respectively. This observation demonstrates that the Rha-displaying MUC1-Tn vaccine successfully generates antibodies against both the MUC1 peptide and the TACA.
  • mice serum The ability of the anti-MUC1-Tn antibodies in the vaccinated mice serum to bind to MUC1-Tn on human tumor cells was demonstrated with U266 human leukemia cells. These cells express MUC1 on their surface as shown by binding with mouse anti-human MUC1 antibodies (CD 227). See FIG. 42A . Serum from group B2 mice also recognized the MUC1 on the tumor cells with similar efficiency as the CD 227 antibodies relative to non-immunized mouse serum. See FIG. 42B . This demonstrates that the antibodies generated against the glycopeptides recognize the MUC1 protein in its native environment.
  • a fully synthetic two-component vaccine containing the lipopeptide adjuvant Pam 3 Cys appended to a 20-amino acid MUC1 peptide containing the TACA GalNAc-O-Thr (Tn) was synthesized and was successfully formulated into liposomes along with an Rha cholesterol conjugate.
  • the resulting liposomes were homogenous in size and were stable at 4° C. for two days.
  • Binding studies with both anti-Rha and mouse anti-human MUC1 antibodies revealed that the Rha and the MUC1 glycopeptide epitopes were surface displayed on the liposomes.
  • a Rha-Ficoll conjugate (14) was synthesized for the generation of anti-Rha antibodies in mice.
  • the anti-MUC1-Tn antibodies in the serum of the vaccinated mice recognized the aberrant MUC1 on human leukemia U266 cells.
  • this vaccine successfully triggered both T-cell and humoral immunity enhanced by anti-Rha antibody dependant antigen uptake. Because this vaccine uses separate rhamnose and anti-genic epitope components, the vaccine can easily be targeted to different antigens or epitopes by changing the peptide without having to change the other components.
  • anti-viral or anti-bacterial vaccines can also be made from them methods and compositions described herein.

Abstract

Compositions, methods of making, and methods of using, xenoantigen-displaying anticancer vaccines are described. In another broad aspect, there is provided herein a method of synthesizing an alkynefunctionalized composition of claim 1, comprising: deprotecting an ester comprising a Fmoc moiety to form a free acid; coupling the free acid of step (a) with an amine; and, removing the Fmoc moiety, and coupling the remaining moiety with palmitic acid to yield an alkyne-functionalized composition.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application Ser. No. 61/599,925 filed Feb. 16, 2012, the entire disclosure of which is expressly incorporated herein by reference.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
  • The invention was made with U.S. Government support under Grant Number GM094734 awarded by the National Institutes of Health. The United States Government has certain rights in the invention.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted via EFS-web and is hereby incorporated by reference in its entirety. The ASCII copy, created on Feb. 12, 2013, is named 42053665_SEQ_LIST_D2012-14.txt, and is 6,494 bytes in size.
  • TECHNICAL FIELD AND INDUSTRIAL APPLICABILITY OF THE INVENTION
  • The invention relates to compositions having a xenoantigen that is incorporated onto a target antigen and method of making the same. In particular, there is provided herein a vaccine composition comprised of a lipid-linked antigen in non-covalent association with a lipid-linked xenoantigen, where both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in a liposomal formulation.
  • BACKGROUND
  • The use of vaccines against a variety of agents is an important objective for disease control worldwide. It is understood that the protective immunity afforded by vaccines against specific antigen is achieved by humoral, cellular and mucosal immune responses.
  • For example, humoral or antibody responses are important in pathogen neutralization and can be very effective in some conditions such as cancer and infectious diseases. These therapeutic cancer vaccines are a new class of active specific immunotherapy agents that trigger a targeted immune response against cancer. It would be useful to have efficient methods for the synthesis of such vaccines in addition to those presently available.
  • SUMMARY
  • In a first aspect, there is provided herein a composition comprising a first lipid (lipida) moiety and an alkyne amide moiety having a Formula IV.
  • In certain embodiments, the first lipid (lipida) moiety comprises a Toll-like receptor (TLR) agonist ligand selected from one or more of: TLR2, TLR1, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13, TLR14, TLR15 and TLR16.
  • In certain embodiments, the TLR2 ligand comprises: dipalmitoyl-S-glyceryl-cys-(Pam2Cys-); tripalmitoyl-S-glyceryl-cys-(Pam3Cys-); dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam2Cys-Ser-(Lys)4) [SEQ ID NO: 2]; tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam3Cys-Ser-(Lys)4) [SEQ ID NO: 3]; or MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-asp-glu-ser-asn-ile-ser-phe-lys-glu-lys (Pam2CGNNDESNISFKEK)] [SEQ ID NO: 4].
  • In certain embodiments, the Formula IV comprises an alkyne-functionalized Pam3Cys amide composition (6), wherein Pam is a dipalmitoyl-S-glyceryl-moiety.
  • In another broad aspect, there is provided herein a method of synthesizing an alkyne-functionalized composition of claim 1, comprising: deprotecting an ester comprising a Fmoc moiety to form a free acid; coupling the free acid of step (a) with an amine; and, removing the Fmoc moiety, and coupling the remaining moiety with palmitic acid to yield an alkyne-functionalized composition.
  • In another broad aspect, there is provided herein a method of synthesizing an alkyne-functionalized Pam3Cys amide composition (6), comprising:
  • a) deprotecting 0-palmitoylated Fmoc L-cystine tert-butyl ester (4) to form a free acid;
  • b) coupling the free acid of step (a) with propargyl amine in presence of benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), 1-hydroxy-benzotriazole (HOBt) and N,N-diisopropylethylamine (DIPEA) to yield composition (5); and,
  • c) removing a Fmoc group of composition (5) by treatment with a mixture of acetonitrile-dichloromethane-diethyl amine, followed by subsequent palmitoylation by coupling with palmitic acid, PyBOP, HOBt and DIPEA to yield the alkyne-functionalized Pam3Cys amide composition (6).
  • In another broad aspect, there is provided herein a lipidated glycopeptide composition comprising: a first lipid (lipida) moiety, a first linker (linkera) moiety, and an antigen moiety having a Formula II.
  • In certain embodiments, the first lipid (lipida) moiety comprises a Toll-like receptor (TLR) agonist ligand selected from one or more of: TLR2, TLR1, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13, TLR14, TLR15 and TLR16.
  • In certain embodiments, the first lipid (lipida) moiety comprises a TLR2 ligand comprising:
  • dipalmitoyl-S-glyceryl-cys-(Pam2Cys-);
    tripalmitoyl-S-glyceryl-cys-(Pam3Cys-);
    [SEQ ID NO: 2]
    dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys
    (Pam2Cys-Ser-(Lys)4);
    [SEQ ID NO: 3]
    tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys
    (Pam3Cys-Ser-(Lys)4);
    or
    [SEQ ID NO: 4]
    MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-
    asp-glu-ser-asn-ile-ser-phe-lys-glu-lys
    (Pam2CGNNDESNISFKEK)].
  • In certain embodiments, the the antigen moiety comprises: an anti-cancer composition, an anti-bacterial composition, an anti-viral composition, a protein, an isolated DNA, an isolated RNA, an isolated carbohydrate, or an isolated lipid.
  • In certain embodiments, the the antigen moiety comprises a TACA that interacts with B-cell receptors.
  • In certain embodiments, the the TACA comprises one or more glycoproteins and glycolipids on a cancer cell.
  • In certain embodiments, the composition comprises mucin 1 (MUC1) variable number tandem repeats (VNTRs) conjugated to tumor-associated carbohydrate antigens (TACA).
  • In certain embodiments, the the TACA comprises: TF, Tn, sialyl Tn (sTn), or sialyl Lewis a (sLea) antigens.
  • In certain embodiments, the the TACA comprises: an autologous or heterologous helper T-cell epitope, wherein the autologous or heterologous helper T-cell epitope comprises a sequence expressed on a tumor cell.
  • T In certain embodiments, the he autologous or heterologous helper T-cell epitope comprises MUC1 VNTR having one of the following amino acid sequences:
  • [SEQ ID NO: 1]
    PDTRPAPGST(Tn)APPAHGVTSA;
    [SEQ ID NO: 5]
    TSAPDTRPAPGSTAPPAHGV;
    or,
    [SEQ ID NO: 6]
    TSAPDT(Tn)RPAPGSTAPPAHGV.
  • In certain embodiments, the the threonine in the sequence GST or PDT is synthetically modified to incorporate α-GalNAc-O-Thr (Tn) TACA.
  • In certain embodiments, the the first linker (linkera) comprises a dialkyl-substituted heteroaryl C1-n alkyl of Formula VI, wherein the “A” group comprises: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n-; and n is a positive integer.
  • In certain embodiments, the the “A” group comprises a C1-5 alkyl chain.
  • In another broad aspect, there is provided herein a composition comprising the compound (9).
  • In another broad aspect, there is provided herein a composition comprising the compound (17).
  • In another broad aspect, there is provided herein a composition comprising the compound (21).
  • In another broad aspect, there is provided herein a method of synthesizing a lipidated glycopeptide composition, comprising reacting a composition of Formula IV, with a composition Formula V.
  • In another broad aspect, there is provided herein a method of synthesizing a Pam3Cys-MUC-1 VNTR-TACA conjugate, comprising: synthesizing a 20-amino acid tandem repeat of MUC1; and modifying a glycopeptide with a terminal azido group to make a ‘click’ conjugation to a Pam3Cys alkyne.
  • In certain embodiments, the the 20-amino acid tandem repeat of MUC1 comprises a GS(α-GalNAc-O-T)A epitope.
  • In certain embodiments, the the 20-amino acid tandem repeat of MUC1 comprises a PD(α-GalNAc-O-T)R epitope.
  • In another broad aspect, there is provided herein a composition comprising a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety, having the Formula VII.
  • In certain embodiments, the the second lipid (lipidb) moiety contains a structure of the Formula IX.
  • In certain embodiments, the the second linker (linkerb) comprises: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n-; wherein n is a positive integer.
  • In certain embodiments, the the second linker (linkerb) comprises a tetraethyleneglycol (TEG) of Formula VIII.
  • In certain embodiments, the the xenoantigen moiety comprises a xenoantigen linked to a lipid.
  • In certain embodiments, the the xenoantigen binds a natural antibody (NA).
  • In certain embodiments, the NA comprises an autoantigen that is present in blood.
  • In certain embodiments, the the NA comprises IgM, IgG, or IgA isotypes in human blood.
  • In certain embodiments, the the NA comprises an anti-carbohydrate NA in human serum.
  • In certain embodiments, the the xenoantigen moiety contains a structure comprising: an α- or β-linked L-rhamnose epitope, a β-linked α-Gal disaccharide epitope, or an α- or β-linked Forssmann disaccharide epitope:
  • In another broad aspect, there is provided herein a vaccine composition, comprising: an antigen composition comprising:
  • 1) a first lipid (lipida) moiety, a first linker (linkera) moiety, and an antigen moiety;
  • 2) a xenoantigen composition comprising: a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety; and,
  • 3) at least one liposomal formulation.
  • In another broad aspect, there is provided herein a vaccine composition, wherein a lipid-linked antigen is in non-covalent association with a lipid-linked xenoantigen, and wherein both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in a liposomal formulation.
  • In certain embodiments, there is no chemically synthesized complex xenoantigen-antigen conjugate molecule.
  • In certain embodiments, the the liposomal formulation comprises 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and cholesterol in a ratio of from about 80:20 to about 70:30, respectively.
  • In another broad aspect, there is provided herein a vaccine composition of claim 37, wherein: the antigen composition comprises a Pam3Cys-MUC1 VNTR-TACA conjugate; the second linker (linkerb) moiety comprises a tetraethyleneglycol (TEG) portion; and the xenoantigen moiety comprises α- or β-linked L-rhamnose.
  • In another broad aspect, there is provided herein an anti-cancer vaccine comprising a vaccine composition as described herein
  • In another broad aspect, there is provided herein an anti-viral vaccine comprising a vaccine composition as described herein.
  • In another broad aspect, there is provided herein an anti-bacterial vaccine comprising a vaccine composition as described herein.
  • In certain embodiments, the vaccine composition further includes at least one immunologic adjuvant.
  • In certain embodiments, the immunologic adjuvant comprises one or more of: a saponin, monophosphoryl lipid A, 3′-O-deacylated monophosphoryl lipid A, and interleukin 12.
  • In another broad aspect, there is provided herein a method for eliciting an immune response to a cancer cell surface antigen in a subject with cancer, comprising administering to the subject an antigen-xenoantigen-liposome composition of claim 37 in sufficient dose to elicit the immune response to the cancer cell surface antigen, wherein the antigen-liposome-xenoantigen composition comprises the cancer cell surface antigen, and wherein the immune response is sufficient for treating the cancer.
  • In certain embodiments, the both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in the liposomal formulation.
  • In certain embodiments, the cancer cell surface antigen is expressed only in cancer cells in the subject.
  • In certain embodiments, the cancer cell surface antigen is expressed only by cancer cells.
  • In certain embodiments, the cancer cell surface antigen comprises a peptide epitope.
  • In certain embodiments, the cell surface antigen comprises MUC1 mucin.
  • In certain embodiments, the subject is a human or non-human mammal.
  • In certain embodiments, the antigen-liposome-xenoantigen composition is administered intranasally, intramuscularly, subcutaneously, intravenously, or orally to the subject.
  • In certain embodiments, the method further comprises measuring humoral and/or cellular immune responses to the cancer cell surface antigen, and administering a further dose to elicit the immune response, if necessary.
  • In another broad aspect, there is provided herein a method for selectively killing cancer cells expressing a cancer cell surface antigen in a subject in need thereof, comprising administering to the subject an antigen-liposome-xenoantigen composition of claim 27, under conditions that result in production in the subject of antibodies against the cancer cell surface antigen, wherein the antibodies produced bind the cancer cell surface antigen on cancer cells in the subject, thereby killing cancer cells that express the cancer cell surface antigen.
  • In certain embodiments, both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in the liposomal formulation.
  • In certain embodiments, the cancer cell surface antigen is expressed only in cancer cells in the subject.
  • In certain embodiments, the cancer cell surface antigen is expressed only by cancer cells.
  • In another broad aspect, there is provided herein a method for improving immunogenicity of vaccines, the method comprising incorporating at least one α- or β-linked L-Rha epitope by direct conjugation or by non-covalent association with at least one liposomal vaccine formulation to increase the immunogenicity of the vaccine by a NA-dependant antigen uptake mechanism.
  • In another broad aspect, there is provided herein a method of synthesizing an alkyne functionalized composition (IV), comprising: deprotecting an ester having a Fmoc moiety to form a free acid; coupling the free acid with an amine; and, removing the Fmoc moiety, and coupling the remaining moiety with palmitic acid to yield the alkyne functionalized composition.
  • In certain embodiments, the antigen moiety comprises: an anti-cancer composition, an anti-bacterial composition, or an anti-viral composition.
  • In certain embodiments, the antigen moiety comprises: a TACA that interacts with B-cell receptors. In certain embodiments, the TACA comprises an autologous helper T-cell epitope.
  • In one broad aspect, there is provided herein a method of synthesizing component 1) above, comprising reacting an alkyne-functionalized amide derivative with a modified peptide antigen.
  • In another broad aspect, there is provided herein the composition (VII), where the xenoantigen binds a natural antibody (NA). In certain embodiment, the xenoantigen contains a structure comprised of: α- or β-linked L-rhamnose, a β-linked α-Gal epitope, or an α- or β-linked Forssmann disaccharide epitope.
  • In another broad aspect, there is provided herein a method for eliciting an immune response to a cancer cell surface antigen in a subject with cancer, comprising administering to the subject an antigen-liposome-xenoantigen composition described herein, in sufficient dose to elicit the immune response to the cancer cell surface antigen.
  • Other systems, methods, features, and advantages of the present invention will be or will become apparent to one with skill in the art upon examination of the following drawings and detailed description. It is intended that all such additional systems, methods, features, and advantages be included within this description, be within the scope of the present invention, and be protected by the accompanying claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The patent or application file may contain one or more drawings executed in color and/or one or more photographs. Copies of this patent or patent application publication with color drawing(s) and/or photograph(s) will be provided by the Patent Office upon request and payment of the necessary fee.
  • FIG. 1: Schematic representation of Fc-Fcγ receptor interaction in the in vivo generated immune complex and APC leading to enhanced antigen uptake and presentation on MHC I or MHC II (APC=antigen presenting cells, e.g., a dendritic cell).
  • FIG. 2: Schematic illustration of a liposomal vaccine composition comprised of: an antigen composition comprised of a first lipid (lipida) moiety, a first linker (linkera) moiety, and an antigen moiety; and, a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety; at least one liposome forming lipid; and, optionally, one or more adjuvants.
  • FIG. 3: Schematic illustration of a lipid-linked antigen component.
  • FIG. 4: Schematic illustration of a dialkyl-substituted heteroaryl C1-n alkyl component.
  • FIG. 5: Schematic illustration of a composition IV reacting with a composition V to form a composition II.
  • FIG. 6: Schematic illustration of a linkera component.
  • FIG. 7: Schematic illustration of a lipid-linked xenoantigen component.
  • FIG. 8: TEG composition used in a linkerb component.
  • FIG. 9: Schematic illustration of a structure contained in a lipidb portion of a lipid-linked xenoantigen component.
  • FIG. 10: Scheme 1: Schematic illustration of synthesis of a L-Rhamnose-TEG-Cholesterol (3).
  • FIG. 11: Scheme 2: Schematic illustration of synthesis of an alkyne functionalized Pam3Cys composition (6).
  • FIG. 12: Scheme 3: Schematic illustration of synthesis of a Pam3Cys-MUC-1 VNTR-TACA conjugate (9) [SEQ ID NOS: 8, 9 and 7], respectively, in order of appearance.
  • FIGS. 13A-13C: Size characterization of liposomes at 1/10,000 dilution: FIG. 13A—Batch 1 liposomes; FIG. 13B—Batch 2 liposomes; and FIG. 13C—Batch 3 liposomes.
  • FIGS. 14A-14B: Photographs showing size characterization of liposomes: SEM images at 5 kV acceleration voltage. FIG. 14A: Batch 1 liposomes under 50,000× magnification. FIG. 14B: Batch 1 liposomes under 250,000× magnification.
  • FIGS. 15A-15C: Fluorescence microscope images with Batch 1 liposomes under 60× magnification. FIG. 15A: Images with control antibodies (antibodies isolated from preimmunization serum) 1st, 2nd and 3rd images: brightfield, FITC and overlay. FIG. 15B: Images with anti-rhamnose antibodies, 1st, 2nd and 3rd images: brightfield, FITC and overlay. FIG. 15C: Images with anti-MUC 1 antibodies, 1st, 2nd and 3rd images: brightfield, FITC and overlay.
  • FIG. 16: 1H NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl-2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 17: 13C NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl-2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 18: 1H-gCosy of (5-Cholesten-3α-yloxy)-3n3 9- trixaundecanyl 2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 19: HRMS of (5-Cholesten-3α-yloxy)-3n3 9- trixaundecanyl 2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 20: 1H NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 21: 13C NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 22: 1H-gCosy of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 23: HRMS of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 24: 1H NMR of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 25: 13C NMR of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 26: 1H gCosy of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 27: HRMS of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 28: 1H NMR of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 29: 13C NMR of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 30: 1H-gCosy of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 31: HRMS of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 32: HR-MALDI-TOF of Glycopeptide Azide (9) [SEQ ID NO:8].
  • FIG. 33: HR-MALDI-TOF of Glycopeptide Azide (8) [SEQ ID NO: 10].
  • FIG. 34: HR-MALDI-TOF of Lipopeptide (9) [SEQ ID NO:7].
  • FIG. 35: Scheme showing synthetic route of 2-Aminoethyle-α-L-Rhamnopyranoside (13).
  • FIG. 36: Anti-Rha Antibody Isotype Titers after 4th Boost with Rha-Ficoll (group A) or Rha-OVA (group B) at 1/5 or 1/500 Serum Dilutions respectively.
  • FIG. 37: T-Cell Proliferation in BALB/c mice with MUC1-Tn (10) Peptide in Presence and Absence of Dendritic Cells.
  • FIGS. 38A-38: FIG. 38A) T-cell proliferation measured by [3H]thymidine incorporation in T-cells from mice spleens primed with MUC1-Tn (8) and challenged with Pam3Cys-MUC1-Tn (9) +Rha liposomes in the presence of anti-Rha antibodies (abs) or control abs [anti Rha(OVA) and anti Rha(Ficoll) abs are the antibodies isolated from the serum of Rha-OVA and Rha-Ficoll immunized mice, respectively]. FIG. 38B) Stepwise immunization plan. Groups A1, A2, B1, and B2 each represent four groups of female BALB/c mice. Stage I: groups A2 and B2 were immunized with Rha-Ficoll/Alum whereas groups A1 and B1 were non-immunized. Stage II: vaccination; groups A1 and A2 vaccinated and boosted with Pam3Cys-MUC1-Tn liposomes whereas groups B2 and B2 were vaccinated with Pam3Cys-MUC1-Tn+Rha liposomes.
  • FIGS. 39A-39B: FIG. 39A) Group average of anti-Rha antibody titers after fourth boost with Rha-Ficoll/Alum. FIG. 39B) Group average of anti-MUC1-Tn antibody titers after first boost with Pam3Cys-MUC1-Tn liposomes or Pam3Cys-MUC1-Tn+Rha liposomes.
  • FIG. 40: Anti-MUC1-Tn Antibody Isotype Titers after first boost with Pam3Cys-MUC1-Tn liposomes or Pam3Cys-MUC1-Tn+Rha liposomes at 1/50 Serum Dilutions.
  • FIGS. 41A-41B: FIG. 41A) Competitive binding of anti-MUC1-Tn antibodies with bound MUC1-Tn in presence of free MUC1-Tn (8). FIG. 41B) Groups average of anti-Tn antibody titer after first boost with Pam3Cys-MUC1-Tn liposomes or Pam3Cys-MUC1-Tn+Rha liposomes.
  • FIGS. 42A-42B: Binding of anti-MUC1-Tn antibodies to human leukemia U266 cells. FIG. 42A) Second antibody alone and with mouse anti-human MUC1 antibodies; FIG. 42B) with 1/5 dilution of non-immunized mouse serum, and with 1/5 dilution of group B2 mouse serum.
  • FIG. 43: 1H NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 44: 13C NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 45: 1H-gCosy of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 46: HRMS of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 47: 1H NMR of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 48: 13C NMR of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 49: 1H-gCosy of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 50: HRMS of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 51: ESI MS of 2-Aminoethyl α-L-rhamnopyranoside (13).
  • FIG. 52: Scheme showing the synthesis of Pam3Cys-MUC-1 VNTR conjugate (17) [SEQ ID NOS: 11, 11 and 12], respectively, in order of appearance.
  • FIG. 53: HRMS (MALDI-TOF) spectrum of peptide (16).
  • FIG. 54: HPLC trace of peptide (16).
  • FIG. 55: HRMS (MALDI-TOF) spectrum of lipopeptide (17).
  • FIG. 56: Synthesis of Pam3Cys-MUC-1 VNTR conjugate (21) [SEQ ID NOS: 13, 14 15], respectively, in order of appearance.
  • FIG. 57: HRMS (MALDI-TOF) of glycopeptide (19).
  • FIG. 58: HPLC trace of glycopeptide (19).
  • FIG. 59: HRMS (MALDI-TOF) of glycopeptide (20).
  • FIG. 60: HRMS (MALDI-TOF) of Pam3Cys-MUC-1 VNTR conjugate (21).
  • DETAILED DESCRIPTION
  • Throughout this disclosure, various publications, patents and published patent specifications are referenced by an identifying citation. The disclosures of these publications, patents and published patent specifications are hereby incorporated by reference into the present disclosure to more fully describe the state of the art to which this invention pertains.
  • GENERAL DEFINITIONS
  • The terms “a,” “an,” and “the” include the plural referents unless the context clearly dictates otherwise.
  • The term “antibody” broadly refers to monoclonal antibodies (including full length monoclonal antibodies) and “antibody fragments” that exhibit a desired biological activity. “Antibody fragments” can comprise a portion of a full length antibody, generally the antigen binding or variable region thereof. Non-limiting examples of antibody fragments include Fab, Fab′, and Fv fragments; diabodies; linear antibodies; and single-chain antibody molecules.
  • The term “monoclonal antibody” broadly refers to antibodies that are highly specific, being directed against a single antigenic site.
  • The term “antibody” also broadly includes naturally occurring antibodies (NAs) as well as non-naturally occurring antibodies, including, for example, single chain antibodies, chimeric, bifunctional and humanized antibodies, as well as antigen-binding fragments thereof. For example, such non-naturally occurring antibodies can be constructed using solid phase peptide synthesis, can be produced recombinantly or can be obtained, for example, by screening combinatorial libraries consisting of variable heavy chains and variable light chains.
  • The term “subject/s” generally refers to any animal that generates an adaptive immune response and can include mammals, birds and reptiles. Examples of subjects can include, but are not limited to, humans, non-human primates, dogs, cats, horses, cows, goats, guinea pigs, mice, rats and rabbits, as well as any other domestic or commercially valuable animal.
  • The term/s “nucleic acid/s” generally encompass both RNA and DNA, including cDNA, genomic DNA, mRNA, synthetic (e.g., chemically synthesized) DNA and chimeras of RNA and DNA. The nucleic acid can be double-stranded or single-stranded. Where single-stranded, the nucleic acid can be a sense strand or an antisense strand. The nucleic acid can be synthesized using oligonucleotide analogs or derivatives (e.g., inosine or phosphorothioate nucleotides). Such oligonucleotides can be used, for example, to prepare nucleic acids that have altered base-pairing abilities or increased resistance to nucleases.
  • It is to be further understood that the term “C1-n alkyl” can be any linear or branched alkyl group containing 1 to n carbon atoms. For example, the term“C1-6 alkyl” can comprise groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl(3-methylbutyl), neopentyl(2,2-dimethylpropyl), hexyl, isohexyl(4-methylpentyl) and the like.
  • The term “C2-n alkyl” is to be understood to mean any linear or branched alkyl group containing 2 to n carbon atoms. For example, the term “C2-6 alkyl” can comprise groups such as ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl(3-methylbutyl), neopentyl(2,2-dimethylpropyl), hexyl, isohexyl(4-methylpentyl) and the like.
  • The term “dialkyl-substituted aryl C1-n alkyl” is to be understood to mean an aryl group substituted at two positions wherein the substituents are composed of a C1-n alkyl group, as defined above. The aryl group may be optionally substituted with at least one substituent selected from the group consisting of hydroxyl, C1-2 alkoxy and halogen. Examples of dialkyl-substituted aryl C1-n alkyl groups include, but are not limited to:
  • Figure US20150024037A1-20150122-C00001
  • The term “dialkyl-substituted heteroaryl C1-n alkyl” is to be understood to mean a heteroaryl group substituted at two positions wherein the substituents are composed of a C1-n alkyl group, as defined above. The aryl group may be optionally substituted with at least one substituent selected from the group consisting of hydroxyl, C1-2 alkoxy and halogen. Examples of substituted heteroaryl C1-n alkyl groups include, but are not limited to:
  • Figure US20150024037A1-20150122-C00002
  • Accordingly, the term “xenoantigen” is to be understood to mean at least one foreign antigen capable of binding natural antibodies. Examples of xenoantigens include, but are not limited to, the α-gal epitope, L-Rha, and the Forssman disaccharide.
  • The term “peptide” may be any peptide comprising natural or non-natural amino acids; and if chiral, in its L or D configurations or as racemate. Examples of non-natural amino acids include, but are not limited to: homocysteine, homoarginine, cylcohexylalanine, ornithine, and Cαα-dibenzylglycine.
  • As used herein, the term “lipid” is defined to include any of a broad range of substances that is characteristically insoluble in water and extractable with an organic solvent. This broad class of compounds is known to those of skill in the art, and as the term “lipid” is used herein, it is not limited to any particular structure. Examples include compounds which contain long-chain aliphatic hydrocarbons and their derivatives. A lipid may be naturally occurring or synthetic (i.e., designed or produced by man). Biological lipids are well known in the art and include, for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof. Of course, compounds other than those specifically described herein that are understood by one of skill in the art as lipids are also encompassed by the compositions and methods described herein.
  • GENERAL DESCRIPTION
  • In a first broad aspect, there is provided herein a composition having xenoantigen that is incorporated onto a target antigen.
  • In another broad aspect, there is provided herein a vaccine composition comprising a lipid-linked antigen in non-covalent association with a lipid-linked xenoantigen, where both the lipid-linked antigen and the lipid-linked xenoantigen are independently embedded in liposomes in a liposomal formulation.
  • In another broad aspect, there is provided herein tumor-associated carbohydrate antigens (TACAs) that are on the heavily glycosylated glycoprotein mucin 1 (MUC1).
  • In another aspect, the tumor-associated carbohydrate antigens (TACAs) that are on the heavily glycosylated glycoprotein mucin 1 (MUC1) are useful as targets for anticancer active immunotherapy.
  • In another aspect, there is provided herein the non-covalent association of the L-Rhamnose xenoantigen in a liposome with the other components of a vaccine independently embedded in the liposome. This non-covalent association eliminates the need to chemically synthesize a complex xenoantigen-antigen conjugate molecule.
  • The peptide sequences described herein act as an autologous helper T-cell epitope. The use of foreign carrier proteins and peptides as conjugates provide helper T-cell epitopes unrelated to the tumor and act as heterologous helper T-cell epitopes.
  • In one embodiment, the MUC1 sequence described herein is shown to be a potent candidate for generating antibodies.
  • The glycoconjugates on the surface of cancer cells are expressed in abnormal quantities and show modifications in the structure of the carbohydrate moieties linked to the peptide or lipid components of the conjugates. These aberrant carbohydrate epitopes are specifically known as tumor associated carbohydrate antigens (TACAs). TACAs demonstrate potential as markers for cancer detection and disease progression and therefore are under intense investigation for their role in the development of anti-cancer vaccines.
  • The TACAs found on the heavily glycosylated glycoprotein mucin 1 (MUC1) are useful targets for anticancer active immunotherapy. In most cancers of epithelial origin, e.g., breast, colorectal, and prostate, this glycoprotein becomes highly over expressed and loses its apical distribution, becoming expressed over the entire cell surface. In the transformed state, the glycan chains of MUC1 are typically shorter and show increased sialylation relative to normal. A number of TACAs have been identified from MUC1. TACAs includes TF, Tn, STn, sLea antigens:
  • Figure US20150024037A1-20150122-C00003
  • In certain embodiments, the above TACAs (which are found on tumor cells) are useful as the “B-cell epitopes” in the design of the vaccines described herein.
  • The MUC1 glycoprotein is also shed into the serum and is a tumor marker for cancer. For example, patients possessing naturally occurring anti-MUC1 antibodies demonstrate better disease-specific survival. It is believed that anti-MUC1 antibodies control hematogenous tumor dissemination and outgrowth. It is also now believed that that TACAs present on MUC1 variable number tandem repeat (VNTR) domain may help to break the immune system's self tolerance to MUC1, at least in human MUC1-expressing transgenic mice while unglycosylated human MUC1 VNTRs generated a weaker immune response. While not wishing to be bound by theory, it is now also believed that this is because the glycopeptide is possibly seen by the immune system as a more foreign epitope in comparison to the unglycosylated MUC1 which is more self-like.
  • Xenoantigens:
  • Many carbohydrate antigens are not highly immunogenic. One approach to increasing the immunogenicity of antigens is to target the antigens to antigen presenting cells (APCs). The effectiveness of vaccines can be increased by the incorporation of a xenoantigen. For example, the xenoantigen Galα1-3Galβ1 (αGal) can be used as an in vivo generated immune complex between the αGal epitopes and the natural antibodies (NA) against αGal in serum. The Fc portion on this immune complex is recognized by the Fcγ receptors on the APCs, thereby leading to the overall internalization of the antigen facilitating presentation by the major histocompatibility complex (MHC) on APCs. NA can also enhance uptake by APCs using other receptors such as complement receptors. Also, there are a variety of NAs that can bind antigens, including autoantigens, normally present in the blood.
  • The NAs, which can be of the IgM, IgG, or IgA isotype in humans, exhibit specific binding to a variety of different antigens, including proteins, DNA, numerous neutral and anionic phospholipids, carbohydrates, and a variety of other lipids, including neutral and anionic glycolipids, cholesterol, and squalene. For example, a large fraction of NAs-to-phospholipid is circulating in vivo as immune complexes to phospholipids. Thus, not only can the xenoantigens complex NAs, but also the lipids and phospholipids present in liposomes can complex NAs as well.
  • In addition, there are other, and highly abundant, anti-carbohydrate NAs in human serum. The most abundant NA detectable was specific for β-linked L-rhamnose (L-Rha) with the fourth most abundant being against α-linked L-Rha; for example:
  • Figure US20150024037A1-20150122-C00004
  • Antibodies against both α/β L-Rha are more prevalent than those against α-Gal.
  • In one aspect, there is described herein a method where the incorporation of α/β-linked L-Rha epitopes into vaccines (by direct conjugation or by non-covalent association with liposomal vaccine formulations) increases immunogenicity by NA-dependant antigen uptake mechanism.
  • In another aspect, there is described herein a vaccine composition that incorporates the non-covalent association of the L-Rha xenoantigen in a liposome with one or more other components of the vaccine composition that are independently embedded in the liposome. This incorporation eliminates the need to chemically synthesize a complex xenoantigen-antigen conjugate molecule. Thus, any lipidated antigen can be introduced into the vaccine composition. One non-limiting example is the L-Rha motif which binds anti-L-Rha NAs.
  • The Fc portion of the NA of an in vivo generated immune complex is recognized by the Fcγ receptors located on the surface of the APCs or by complement and complement receptors, thereby leading to the better internalization of the vaccine.
  • Also, the T-cell proliferation is enhanced when the antigen and xenoantigen are co-localized on a liposome in the presence of APCs and anti L-Rha antibodies.
  • In one embodiment described herein, the xenoantigen is conjugated to a lipid (such as cholesterol) by means of a linker (such as tretraethylene glycol (TEG)), which allows the xenoantigen to be displayed on the surface of the liposome along with the lipidated-tumor, bacterial or viral antigen.
  • In such embodiments, the linker between the xenoantigen and the cholesterol lipid allows for a xenoantigen-NA binding interaction.
  • Antigens:
  • For the development of a vaccine (e.g., an anti-cancer vaccine, an anti-viral vaccine, or an anti-bacterial vaccine), an antigen is required. Broadly, an antigen is defined as an entity or foreign molecule that, when introduced into the body, triggers the production of an antibody by the immune system. The term “antigen” also refers to any molecule or molecular fragment that can be bound by a major histocompatibility complex (MHC) and presented to a T-cell receptor.
  • Tumors of epithelial origin provide a variety of TACAs that can act as antigens by interacting with B-cell receptors. These include TACAs identified on the glycoproteins and glycolipids that decorate the cancer cells. Non-limiting examples of TACAs include: the TF, Tn, sialyl Tn (sTn) and sialyl Lewis a (sLea) antigens, whose structures are shown above. These structures as well as many other TACAs can produce a considerable humoral immune response.
  • In addition, peptides and glycopeptides can interact with B-cell receptors to produce antibodies.
  • One of the major disadvantages of using TACAs alone as cancer vaccines, however, is their weak immunogenicity. Generation of high amounts of high affinity IgG antibodies against the cancer antigen depends upon the combined interaction of the B-cells and the helper T-cells, and therefore requires the vaccine to contain a peptide epitope which can be displayed on the major histocompatibility complex (MHC) class-II or class-I molecules that are present on the surface of APCs, such as DCs. In general, TACAs alone cannot activate the helper T-cells, resulting in poor immunogenicity. Previous efforts to counteract this problem involved the conjugation of TACAs to large carrier proteins like keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA), TLR agonists and zwitterionic polysaccharides. However, one drawback to the protein conjugation strategy is that the major immune response is towards the carrier protein, thus resulting in a suppressed immune response for the cancer antigen.
  • The protein conjugates provide the peptide sequences, which protein sequences can then be displayed as helper T-cell epitopes on MHC class-II molecules. The resulting complexes activate CD4+ helper T-cells which then interact with antibody-producing B-cells allowing the B-cells to produce higher affinity antibodies.
  • For example, smaller 20-amino acid peptides which can bind MHC class-II can be conjugated to TACAs. These conjugates can produce high amounts of high affinity IgG anti-TACA antibodies. This strategy provides an advantage over the use of large carrier proteins as helper T-cell epitope, as the immune system can now focus more resources on the available TACA antigens rather than producing antibodies to the carrier protein.
  • In one aspect, the use of peptide sequences acts as an “autologous helper T-cell epitope.” Such autologous helper T-cell epitopes can be sequences expressed on the tumor. The use of foreign carrier proteins and peptides as conjugates thus provide helper-T cell epitopes unrelated to the tumor, and thus act as “heterologous helper T-cell epitopes.”
  • One non-limiting example of an autologous helper T-epitope is the MUC1 VNTR consisting of the 20 amino acid sequence PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO: 1]. In a particular example, the threonine in the sequence GST is synthetically modified to incorporate the α-GalNAc-O-Thr (Tn) TACA. This region in the MUC1 sequence is a more potent candidate for generating high antibody titers against Tn than the unglycosylated MUC1 VNTR.
  • Lipid Attachment to Antigen
  • Glycopeptides formed by the conjugation of B-cell epitopes and helper T-cell epitopes often show moderate immunogenicity. A major reason for such moderate immunogenicity is the inappropriate maturation of the DCs.
  • Described herein is a method which overcomes this problem, where a lipid component is incorporated into a vaccine capable of imparting self adjuvanating properties. Toll-like receptor (TLR) agonists interact with TLRs to facilitate the maturation of dendritic cells. The maturation of the dendritic cells can be achieved by the involvement of a variety of TLR subclasses, as for example: TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13, TLR14, TLR15 and TLR16.
  • Non-limiting examples of TLR2 interactive lipopeptides include: dipalmitoyl-S-glyceryl-cys-(Pam2Cys); tripalmitoyl-S-glyceryl-cys-(Pam3Cys-); dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam2Cys-Ser-(Lys)4) [SEQ ID NO: 2], tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam3Cys-Ser-(Lys)4) [SEQ ID NO: 3], and MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-asp-glu-ser-asn-ile-ser-phe-lys-glu-lys (Pam2CGNNDESNISFKEK)] [SEQ ID NO: 4].
  • Non-limiting examples of palmitoylated lipids [SEQ ID NOS: 3, 2 and 4], respectively, in order of appearance, that may interact with TLR2 thus include:
  • Figure US20150024037A1-20150122-C00005
  • Pam2Cys-Ser-(Lys)4 [SEQ ID NO:2], for example, acts as TLR2/TLR6 agonists, while Pam3Cys-Ser-(Lys)4 [SEQ ID NO:3] acts as a TLR2/1 agonist. Lipopeptide MALP-2 acts a TLR2/6 ligand. Tripalmitoyl-S-glyceryl-cys (Pam3Cys), and dipalmitoyl-S-glyceryl-cys (Pam2Cys), are minimal structural epitopes common to palmitoylated TLR2 ligands.
  • Synthesis of Two-component Vaccine
  • Lipid Components
  • In certain embodiments, the lipid component of a vaccine composition serves three purposes: first, the lipid component acts as a vaccine adjuvant capable of interacting with Toll-Like receptors; second, the lipid component facilitates the formulation of the glycolipopeptide into liposomes anchoring the glycopeptide epitope to the inner and outer surfaces of the liposomes; and third, the lipidation facilitates cross-presentation of peptide antigens by APCs.
  • Xenoantigen Component
  • Another aspect is the ability to bind natural antibodies (NAs) specific for a desired target epitope (e.g., the xenoantigen L-Rha epitope). The bound NAs thus facilitate the opsonization of the liposomal vaccine by APCs. Covalent attachment of xenoantigens to lipids through the use of a suitable linker thus facilitates the incorporation of these epitopes into the liposomes.
  • Linker Components
  • In some embodiments, the xenoantigen is covalently attached to a lipid through the use of a suitable linker. Further, in particular embodiments, the length of the linker between the xenoantigen and lipid can be optimized to achieve a desired xenoantigen-NA interaction. The conjugation of a lipid to the xenoantigen allows for the display of multiple xenoantigens on the surface of the liposomes, thereby facilitating multivalent NA-antibody binding to the liposome.
  • The Fc portion of the NA in the in vivo generated immune complex is thus recognized by the Fcγ receptors on APCs, thereby leading to opsonization of the vaccine by APCs.
  • Vaccine Compositions
  • In certain embodiments, the composition of the vaccine contains non-covalently associated components: i) a lipid-linked antigen; ii) a composition of lipids capable of forming liposomes; iii) a lipid-linked xenoantigen; and, optionally, iv) one or more independent adjuvant molecules. These components form a well defined liposomal vaccine composition I, as shown in FIG. 2. The lipid-linked antigen is comprised of an antigen linked to a first lipid (also called lipida herein). In certain embodiments, the antigen can be a peptide or glycopeptide of 1 to 100 amino acids in length. The antigen may also include a B-cell epitope conjugated to a peptide or glycopeptide sequence that is capable of providing a T-cell epitope. The peptide, glycopeptide, peptide-B-cell, or glycopeptides-B-cell epitope conjugate is linked to lipida.
  • The generalized composition of the lipida-linked antigen component is the structure of composition II, shown in FIG. 3.
  • In one embodiment, the antigen contains a glycopeptide having the 20 amino acid sequence PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO:1]. The T(Tn) in the sequence is the α-GalNAc-O-T (Tn) TACA. In another embodiment, the antigen contains a glycopeptide having the 20 amino acid sequence TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5]. In another embodiment, the antigen contains a glycopeptides having the 20 amino acid sequence TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6]. The T(Tn) in the sequence is the α-GalNAc-O-T (Tn) TACA.
  • The linkera component can be: a chain of C2-n alkyl, dialkyl-substituted aryl C1-n alkyl, dialkyl-substituted heteroaryl C1-n alkyl, or —CH2CH2(OCH2CH2)n—, where n is a positive integer. In one embodiment, the linkera component comprises a dialkyl-substituted heteroaryl C1-n alkyl of formula III, as shown in FIG. 4. In certain embodiments, the lipida portion of the formula III contains a lipid of one of the following structures: Pam2Cys-Ser-(Lys)4, Pam3Cys-Ser-(Lys)4, MALP-2, Pam3Cys, or Pam2Cys. In particular embodiments, the first lipid (lipida) component of the formula III can contain a lipid having the structure Pam3Cys, or the structure Pam2Cys.
  • Synthesis of Toll-Receptor Ligands
  • Another aspect is the synthesis of a composition IV (shown in FIG. 5) which, when reacted with an appropriate catalyst in the presence of a modified peptide antigen of composition V, forms a composition of composition II.
  • In certain embodiments, the lipida moiety of the composition IV can be a structure of the Formulae [SEQ ID NOS: 3, 2 and 4], respectively, in order of appearance:
  • Figure US20150024037A1-20150122-C00006
  • Referring again to FIG. 5 and the composition V, the “A” group therein represents a linker. In certain embodiments, the linker can be comprised of: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n—; n is a positive integer. In a particular embodiment, A comprises a C1-5 alkyl chain.
  • Further, in composition V, the antigen moiety can be a peptide or glycopeptide that is covalently bonded at the N-terminus to a carbonyl group on the linker, thus forming an amide bond. In a particular embodiment, the antigen contains a glycopeptide having a 20 amino acid sequence PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO:1], where the T(Tn) in the sequence GSTA is the α-GalNAc-O-Thr TACA. In another embodiment, the antigen contains a glycopeptide having the 20 amino acid sequence TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5]. In another embodiment, the antigen contains a glycopeptide having the 20 amino acid sequence TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6]. The T(Tn) in the sequence is the α-GalNAc-O-T (Tn) TACA.
  • Upon reaction of composition IV with composition V in the presence of an appropriate catalyst, a composition II is formed. In certain embodiments, the linkera moiety can be a dialkyl-substituted heteroaryl C1-n alkyl of formula VI, as shown in FIG. 6.
  • In formula VI, the “A” group can be a linker comprised of: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n—; n is a positive integer. In a particular embodiment, the “A” group comprises a C1-5 alkyl chain.
  • Referring now to FIG. 7, the lipid-linked xenoantigen can be a xenoantigen linked to a lipid. The xenoantigen can be any structure which binds NAs. The xenoantigen is linked to a second lipid (lipidb) through a second linker (linkerb). The generalized composition of the lipid-linked xenoantigen component is structure of composition VII as shown in FIG. 7.
  • In one embodiment, the xenoantigen contains a structure comprised of: alpha- or beta-linked L-rhamnose, a beta-linked alpha-gal epitope, or an alpha- or beta-linked Forssmann disaccharide epitope. In a particular embodiment, the xenoantigen contains a structure consisting of alpha- or beta-linked L-rhamnose.
  • The linkerb component can be comprised of: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n—; where n is a positive integer. In a particular embodiment, the linkerb component has a tetraethyleneglycol (TEG) of formula VIII, as shown in FIG. 8.
  • In a particular embodiment, the lipidb portion can contain a structure of the formula IX, as shown in FIG. 9. In one particular embodiment, the liposomal forming lipids can be comprised of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and cholesterol in a ratio of from about 80:20 to about 70:30, respectively.
  • It is to be understood that, in certain embodiments, an immunologic adjuvant may or may not be included as part of the composition of the vaccine. For example, in certain embodiments, the immunologic adjuvants may be, but are not limited to: a saponin (e.g., QS21), monophosphoryl lipid A, 3′-O-deacylated monophosphoryl lipid A, or interleukin 12.
  • Pharmaceutical Preparations
  • Pharmaceutical compositions of the present invention comprise an effective amount of a compound(s) or composition(s) disclosed herein, and/or additional agents, dissolved or dispersed in a pharmaceutically acceptable carrier. The phrases “pharmaceutical” or “pharmacologically acceptable” refers to molecular entities and compositions that produce no adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human. The preparation of a pharmaceutical composition that contains at least one compound or additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference. Moreover, for animal (e.g., human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards.
  • A vaccine composition disclosed herein may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection. Vaccine compositions disclosed herein can be administered intravenously, intradermally, transdermally, intrathecally, intraarterially, intraperitoneally, intranasally, intravaginally, intrarectally, topically, intramuscularly, subcutaneously, mucosally, in utero, orally, topically, locally, via inhalation (e.g., aerosol inhalation), by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference).
  • The actual dosage amount of a composition disclosed herein administered to an animal or human patient can be determined by physical and physiological factors such as body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • In certain embodiments, pharmaceutical compositions may comprise, for example, at least about 0.1% of an active compound. In other embodiments, an active compound may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein. Naturally, the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
  • In other non-limiting examples, a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg/body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc., can be administered, based on the numbers described above.
  • In certain embodiments, a vaccine composition herein and/or additional agents is formulated to be administered via an alimentary route. Alimentary routes include all possible routes of administration in which the composition is in direct contact with the alimentary tract. Specifically, the pharmaceutical compositions disclosed herein may be administered orally, buccally, rectally, or sublingually. As such, these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsules, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • In further embodiments, a vaccine composition described herein may be administered via a parenteral route. As used herein, the term “parenteral” includes routes that bypass the alimentary tract. Specifically, the pharmaceutical compositions disclosed herein may be administered, for example but not limited to, intravenously, intradermally, intramuscularly, intraarterially, intrathecally, subcutaneous, or intraperitoneally (U.S. Pat. Nos. 6,753,514, 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363 are each specifically incorporated herein by reference in their entirety).
  • Solutions of the vaccine compositions disclosed herein as free bases or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy injectability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (i.e., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption such as, for example, aluminum monostearate or gelatin.
  • For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal administration. In this connection, sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologics standards.
  • Sterile injectable solutions are prepared by incorporating the vaccine compositions in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized compositions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, some methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. A powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
  • In other embodiments, the vaccine compositions may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or via inhalation.
  • Pharmaceutical compositions for topical administration may include the vaccine compositions formulated for a medicated application such as an ointment, paste, cream or powder. Ointments include all oleaginous, adsorption, emulsion and water-soluble based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only. Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin. Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram. Possible bases for compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base. Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the vaccine composition and provide for a homogenous mixture. Transdermal administration of the vaccine compositions may also comprise the use of a “patch.” For example, the patch may supply one or more vaccine compositions at a predetermined rate and in a continuous manner over a fixed period of time.
  • In certain embodiments, the vaccine compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in their entirety). Likewise, the delivery of drugs using intranasal microparticle resins (Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725,871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts and could be employed to deliver the vaccine compositions described herein. Likewise, transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety), and could be employed to deliver the vaccine compositions described herein.
  • It is further envisioned the vaccine compositions disclosed herein may be delivered via an aerosol. The term aerosol refers to a colloidal system of finely divided solid or liquid particles dispersed in a liquefied or pressurized gas propellant. The typical aerosol for inhalation consists of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent. Suitable propellants include hydrocarbons and hydrocarbon ethers. Suitable containers will vary according to the pressure requirements of the propellant. Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms.
  • EXAMPLES
  • The present invention is further defined in the following Examples, in which all parts and percentages are by weight and degrees are Celsius, unless otherwise stated. It should be understood that these Examples, while indicating preferred embodiments of the invention, are given by way of illustration only. From the above discussion and these Examples, one skilled in the art can ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions. All publications, including patents and non-patent literature, referred to in this specification are expressly incorporated by reference. The following examples are intended to illustrate certain embodiments of the invention and should not be interpreted to limit the scope of the invention as defined in the claims, unless so specified.
  • General Methods
  • All fine chemicals such as L-rhamnose, cholesterol, p-toluene sulfonyl chloride, copper sulfate, etc., and anhydrous solvents, such as anhydrous methanol, were purchased from Acros Organics. 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) was obtained from Avanti Polar Lipids Inc. (Alabaster, Ala.). Boron trifluoride etherate was obtained from Aldrich. The chemicals were used without further purification. All solvents were obtained from Fisher and used as received except dichloromethane, which was dried and distilled following the standard procedures. Silica (230-400 mesh) for flash column chromatography was obtained from Sorbent Technologies; thin-layer chromatography (TLC) precoated plates were from EMD. TLCs (silica gel 60, f254) were visualized under UV light or by charring (5% H2SO4-MeOH). Flash column chromatography was performed on silica gel (230-400 mesh) using solvents as received.
  • 1H NMR was recorded either on a Varian VXRS 400 MHz or an INOVA 600 MHz spectrometer in CDCl3 or CD3OD using residual CHCl3 and CHD2OH as internal references, respectively.
  • 13C NMR was recorded on a Varian VXRS 100 MHz or an INOVA 150 MHz in CDCl3 using the triplet centered at δ 77.23 or CD3 OD using the septet centered at δ 49.0 as internal reference. High resolution mass spectrometry (HRMS) was performed on a TOF mass spectrometer. The peptide was synthesized on a Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.). Tris [(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA), preloaded Fmoc-L-Ala-Wang resin and all other Fmoc-L-amino acids were procured from Anaspec (San Jose, Calif.). FITC goat anti-mouse IgG/IgM and purified mouse anti-human CD227 (anti-human MUC1) were obtained from BD-biosciences (San Jose, Calif.). Scanning electron microscope imaging was done on a JEOL JSM-7500F field scanning electron microscope. Dynamic light scattering measurements was done DynaPro Titan temperature controlled microsampler (Wyatt Technology Corporation). Fluorescence microscopy was done on a Nikon TiU microscope.
  • Example 1 Synthesis of L-Rhamnose-TEG-Cholesterol (3)
  • Cholesterol tetraethylene glycol (1) was glycosylated with peracetyl L-rhamnose in presence of boron trifluoride etherate to afford peracetyl rhamnose-TEG-cholesterol (2) (32%) which was deacetylated under Zémplen conditions to generate L-rhamnose-TEG-cholesterol (3) (85%). See FIG. 10, illustrating Scheme 1. Compound (3) anchors the Rha epitopes on the surface of the liposomes thereby facilitating anti-Rha antibody binding.
  • (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl-2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2)
  • To a solution of 1,2,3,4-tetra-O-acetyl rhamnopyranose (0.64 g, 1.92 mmol) in CH2Cl2 (3 mL) was added (5-cholesten-3α-yloxy)-3n3 9-trixoundecan-1-ol (1) (1.30 g, 2.30 mmol) in CH2Cl2 and the mixture was cooled to 0° C. BF3.OEt2 (486 mL, 3.84 mmol) was added dropwise to the reaction mixture and the resulting solution was stirred at ambient temperature under N2 atmosphere. The reaction was monitored by TLC (EtOAc:hexanes=1:1) and appeared complete after 18 h. The reaction mixture was diluted with CH2Cl2 (25 mL) and washed with saturated NaHCO3 (25 mL), water (25 mL) and brine (25 mL) after which the organic layer was dried over anhydrous Na2SO4. Excess solvent was evaporated under reduced pressure and the residue was purified by silica gel flash column chromatography using 30% EtOAc in hexanes as solvent to afford (2) as a light yellow solid (0.51 g, 32%). 1H NMR (600 MHz, CDCl3): δ 0.67 (s, 3H, cholesterol), 0.85-1.15 (23H, cholesterol), 1.21 (d, 3H, J=6 Hz, C-5 CH3), 1.24-1.52 (12H, cholesterol), 1.80-1.95 (5H, cholesterol), 1.98 (s, 3H, COCH3), 2.05 (s, 3H, COCH3), 2.15 (s, 3H, COCH3), 3.17 (m, 1H, —OCH-cholesterol), 3.63-3.66 (16H, —CH2—CH2O-TEG), 3.92 (m, 1H, H-5), 4.77 (d, 1H, T=1.8 Hz, H-1), 5.06 (t, 1H, J=10.2 Hz, H-4), 5.26 (dd, 1H, J=1.8, 3.6 Hz, H-2), 5.30 (dd, 1H, J=4.2, 9.9 Hz, H-3), 5.33 (m, 1H, —C═CH-cholesterol). 13C NMR (100.56 MHz, CDCl3): δ 12.04, 17.61, 18.90, 19.58, 20.95, 21.03, 21.14, 21.25, 22.76, 23.02, 24.01, 24.48, 28.21, 28.43, 28.53, 29.90, 32.07, 32.13, 35.97, 36.37, 37.07, 37.42, 39.23, 39.70, 39.96, 42.50, 50.36, 53.63, 56.32, 56.96, 66.46, 67.30, 67.46, 69.30, 70.03, 70.24, 71.35, 79.68, 97.74 (C−1), 121.74 (C═C), 141.15 (C═C), 170.18 (COCH3), 170.25 (COCH3), 170.32 (COCH3). HRMS [M+Na] m/z: calcd for C47H78NaO12, 857.5391. Found, 857.5396.
  • (5-Cholesten-3α-yloxy)-3n3 2-trixaundecanyl Rhamnopyranoside (3)
  • To a solution of (2) (0.45 g, 0.54 mmol) in MeOH (10 mL) was added metallic sodium (0.03 g) and the resulting solution was stirred at ambient temperature under N2 atmosphere. The reaction was monitored by TLC (5% MeOH in CH2Cl2) and appeared complete after 1 h. The solution was neutralized by Amberlite H+ exchange resin. Excess solvent was evaporated under reduced pressure and the residue was purified by silica gel flash column chromatography using 5% MeOH in CH2Cl2 as solvent to afford (3) as a yellowish white solid (0.32 g, 85%). 1H NMR (600 MHz, CDCl3): δ 0.68 (s, 3H, cholesterol), 0.86-1.25 (24H, cholesterol), 1.32 (d, 3H, J=6 Hz, C-5 CH3), 1.44-1.53 (16H, cholesterol), 2.83 (s, 1H, C-4 OH), 3.08 (d, 1H, T=3 Hz, H-1), 3.20 (m, 1H, —O—CH-cholesterol), 3.43 (t, 1H, T=9.6 Hz, H-4), 3.62-3.71 (16H, —CH2—CH2O-TEG), 3.73 (m, 1H, H-5), 3.83 (dd, 1H, T=3, 6.9 Hz, H-3), 3.98 (s, 1H, H-2), 4.87 (s, 1H, C-2 OH), 5.31 (s, 1H, C-3 OH), 5.35 (m, 1H, —C═CH-cholesterol). 13C NMR (100.56 MHz, CDCl3): δ 12.07, 17.83, 18.92, 19.60, 21.27, 22.78, 23.04, 24.03, 24.50, 28.23, 28.44 (2), 32.08, 32.15, 35.99, 36.39, 37.06, 37.39, 39.09, 39.73, 39.97, 42.53, 50.36, 56.34, 56.97, 66.74, 67.32, 68.07, 70.48, 70.63, 70.75, 70.81, 70.93, 70.97, 71.05, 71.80, 73.81, 79.87, 99.98 (C−1), 121.96 (C═C-cholesterol), 140.98 (C═C-cholesterol). HRMS [M+Na] m/z: calcd for C41H72NaO9, 731.5074. Found, 731.5090.
  • Example 2 Synthesis of Alkyne Functionalized Pam3Cys (6)
  • O-palmitoylated Fmoc L-cystine tert-butyl ester (4) was deprotected by a brief treatment with trifluoroacetic acid (TFA). The free acid was coupled with propargyl amine in the presence of benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), 1-hydroxy-benzotriazole (HOBt) and N,N-diisopropylethylamine (DIPEA) to yield (5) (66% over 2 steps). Fmoc group in compound (5) was removed by treatment with a mixture of acetonitrile-dichloromethane-diethyl amine (2:1:2) followed by subsequent palmitoylation by coupling with palmitic acid, PyBOP, HOBt and DIPEA to afford our target alkyne functionalized Pam3Cys amide derivative (6) (80% over 2 steps), as shown in FIG. 11, Scheme 2. Compound (6), which is a TLR-2 agonist, serves the purpose of an immunoadjuvant for a vaccine, and also anchors the MUC1-Tn conjugate on the surface of the liposome.
  • N-propargyl Pam2FmocCys Amide Derivative (5)
  • Pam2FmocCys tertiary butyl ester (0.30 g, 0.32 mmol) was dissolved in minimum volume of neat TFA (1 mL) and stirred at ambient temperature under N2 atmosphere. TLC (EtOAc:hexanes=1:4) indicated the completion of the reaction after 1 h. The reaction mixture was evaporated to dryness under vacuum and the residue was dissolved in CH2Cl2 (3 mL). PyBOP (198 m g, 0.38 mmol), HOBt (58 mg, 0.38 mmol), DIPEA (78 μL, 0.47 mmol) and 4 Å mol. sieves (2-3 beads) were added sequentially and the mixture was stirred for 5 minutes at room temperature followed by the addition of propargyl amine (25 μL, 0.38 mmol) and stirred at ambient temperatures under N2 atmosphere. The reaction was monitored by TLC (EtOAc:hexanes=1:4) and appeared complete after 4 h. The reaction mixture was filtered, washed with phosphate buffer (10 mL) and extracted with CH2Cl2 (3×10 mL). The organic layer was dried over anhydrous Na2SO4 and concentrated. The residue was purified by silica gel flash column chromatography using EtOAc-hexanes (1:4) as solvent to afford (5) as a white solid (192 mg, 66%). 1H NMR (600 MHz, CDCl3): δ 0.87 (t, 6H, J=7.2 Hz, Pam-CH3), 1.14-1.65 (m, 52H, Pam-CH2), 1.68 (s, 1H, alkyne-CH), 2.18-2.35 (m, 4H, COCH2), 2.83 (m, 1H, Cys-CHH), 2.89 (dd, 1H, J=7.2, 14.4 Hz, S-glyceryl-OCHH), 3.01 (dd, 1H, J=6, 14.4 Hz, cys-CHH), 4.06 (dd, 1H, J=3, 4.8 Hz, S-glycerylO-CHH), 4.08 (s, 2H, CO—NH—CH2), 4.18 (dd, 1H, J=6, 11.4 Hz, S-glyceryl-O—CHH), 4.23 (t, 1H, J=7.2 Hz, Fmoc-CH), 4.39 (m, 1H, NH—CH—CO), 4.42 (m, 2H, Fmoc-CH2), 5.12 (m, 1H, S-glyceryl-O—CH), 5.73 (d, 1H, J=7.8 Hz, Pam-NH), 6.89 (s, 1H, CO—NHCH2), 7.31-7.81 (m, 8H, Fmoc-ArH). 13C NMR (150.84 MHz, CDCl3): δ 14.35-36.70 (30C, Pam-C), 47.29 (2), 53.32, 63.58, 67.48, 70.61, 71.78, 72.07, 79.09, 79.85, 120.22, 125.30, 127.30 (2), 127.97 (2) 141.49, 141.50, 143.85, 143.89 (Aromatic-C), 170.07, 173.66 (2), 174.04 (Cys-CO). HRMS [M+Na] m/z: calcd for C56H86N2NaO7S, 953.6053. Found, 953.6073.
  • N-propargyl Pam3Cys Amide Derivative (6)
  • Composition (5) (192 mg, 0.21 mmol) was dissolved in a mixture of CH3CN—CH2Cl2-Et2NH (2:1:2, 2.50 mL) and stirred at ambient temperature under N2 atmosphere. TLC (EtOAc:hexanes=1:4) indicated the complete deprotection of the Fmoc group after 2 h. The reaction mixture was evaporated to dryness under vacuum. Palmitic acid (64 mg, 0.25 mmol), PyBOP (128 mg, 0.25 mmol), HOBt (38 mg, 0.25 mmol) were dissolved in CH2Cl2 (3 mL) followed by the addition of DIPEA (51 μL, 0.31 mmol). The mixture was stirred for 5 minutes and added to the residue of the Fmoc deprotected product from compound (5) containing 4 A mol. sieves (2-3 beads). The reaction mixture was stirred at ambient temperature under N2 atmosphere. The reaction was monitored by TLC (EtOAc:hexanes=1:4) and appeared complete after 4 h. The reaction mixture was diluted with CH2Cl2 (15 mL), filtered and evaporated to dryness. The residue was purified by silica gel flash column chromatography using EtOAC-hexanes (1:4) as solvent to afford (6) as a pale yellow solid (156 mg, 80%). 1H NMR (600 MHz, CDCl3): δ 0.88 (t, 12H, J=6.6 Hz, Pam-CH3), 1.10-1.63 (m, 78H, Pam-CH2), 2.23 (s, 1H, Alkyne-CH), 2.24-2.36 (m, 6H, COCH2), 2.71 (dd, 1H, J=7.8, 14.4 Hz, Cys-CHH), 2.86 (m, 6H, COCH2), 2.95 (dd, 1H, J=6, 14 Hz, Cys-CHH), 4.06 (m, 2H, CO—NH—CH2), 4.18 (dd, 1H, J=6.6, 12 Hz, S-glyceryl-O—CHH), 4.40 (dd, 1H, J=3, 12 Hz, S-glyceryl-O—CHH), 4.64 (q, 1H, J=6 Hz, NH—CH—CO), 5.12 (m, 1H, S-flyceryl-OCH), 6.64 (d, 1H, J=8.4 Hz, Pam-NH), 7.02 (t, 1H, J=5.4 Hz, CO—NH—CH2). 13C NMR (150.84 MHz, CDCl3): δ 14.35-42.19 (48C, Pam-C, Cys-CR, S-glyceryl-C, NH—C), 51.30 (Cys-Ca, 63.65, 70.61, 71.98, 79.08, 170.40 (Cys-CO), 173.70, 173.86, 174.06 (Pam-CO). HRMS [M+Na] m/z: calcd for C57H106N2NaO6S, 969.7669. Found, 969.7682.
  • Example 3 Synthesis of Pam3Cys-MUC-1 VNTR-TACA Conjugate (9)
  • A 20-amino acid tandem repeat of MUC1 which included the GS(α-GalNAc-O-T)A epitope was synthesized. The glycopeptide was modified with a terminal azido group in order to make a ‘click’ conjugation to the Pam3Cys alkyne. The glycopeptide azide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) starting from preloaded Fmoc-L-Ala Wang resin using solid-phase chemistry, as shown in FIG. 12, Scheme 3.
  • The peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent. A six-fold excess of Nα-Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of Nα-Fmoc group was accomplished by treatment with piperidine in DMF. After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of TFA-thioanisole-ethanedithiol-water-phenol (88:3:5:2:2). The cocktail mixture was filtered through a Quick Snap column, purified by C18 reverse phase HPLC and lyophilized to afford composition (7b).
  • The acetyl groups in composition (7b) were deprotected by treatment with 6 mmol sodium methoxide in methanol. The product was purified by Bio-Gel (P-2, fine 45-90 μm) size exclusion chromatography using deionized water as solvent. Lyophilization of the fractions afforded composition (8) (100%) as a white powder.
  • Conjugation of the composition (8) (1 eqv) with the composition (6) (3 eqv) in the presence of copper sulfate pentahydrate (12 eqv), Tris [(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA) (12 eqv) and sodium ascorbate (12 eqv) in H2O-MeOH-THF (1:1:2) as solvent at ambient temperatures thus afforded the target, Pam3Cys-MUC1 VNTR-TACA conjugate (9), after 40 h.
  • Composition (9) was purified by LH20 using MeOH-dichloromethane (1:1) as solvent. The eleutants were lyophilized to afford composition (9) as a white solid (100%).
  • Synthesis of Azide (7b)
  • The glycopeptide azide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) using solid phase chemistry. The peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent starting with a preloaded Fmoc-alanyl-Wang resin. A six-fold excess of Nα-Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of Nα-Fmoc group was accomplished by treatment with 25% piperidine in dimethylformamide twice; first for 5 minutes and then a second time for 25 minutes to afford composition (7a).
  • After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of 88% TFA, 3% thioanisole, 5% ethanedithiol, 2% water and 2% phenol. 4 mL of cleavage cocktail was added to the dried peptide-resin in a 15 mL glass vial blanketed with nitrogen. Cleavage was carried out for 2.5 hrs with gentle magnetic stirring. At the end cleavage time, the cocktail mixture was filtered on a Quick-Snap column. The filtrate was collected in 20 mL ice-cold butane ether. The peptide was allowed to precipitate for an hour at −200° C., centrifuged, and washed twice with ice-cold methyl-t-butyl ether. The precipitate was dissolved in 25% acetonitrile and lyophilized to complete dry powder affording composition (7b). Quality of peptides was analyzed by analytical reverse phase HPLC and MALDI-TOF (matrix assisted laser desorption ionization time-of-flight) mass spectrometer, model 4800 from Applied Biosystems. HR-MALDI-MS: [M+H] m/z calcd for C100H155N29O37, 2355.1172. Found, 2355.1753.
  • Synthesis of azide (8)
  • Composition (7b) (5 mg, 2.24 μmol) was dissolved in 2 mL of dry methanol and 12 μL of freshly prepared 1 M sodium methoxide was added and the reaction mixture was stirred at ambient temperature under N2 atmosphere for 2 h. The reaction mixture was neutralized with solid carbon dioxide. The reaction mixture was concentrated and purified by Bio-Gel (P-2, fine 45-90 μm) size exclusion chromatography using deionized water as solvent. Lyophilization of the eleutants afforded composition (8) as a white powder (4.7 mg, 100%). HR-MALDI-MS: [M+H] m/z calcd for C94H149N29O34, 2229.0895. Found 2229.0959.
  • Synthesis of Glycolipopeptide (9)
  • CuSO4.5H2O (134 μg, 0.54 μmol) and TBTA (2.14 mg, 4.04 μmol) were dissolved in H2O-THF (1:1, 0.40 mL) and to it Na-ascorbate (0.80 mg, 4.04 μmol) was added and stirred for 5 minutes. Composition (6) (1.27 mg, 1.35 μmol) in THF (0.40 mL) was added to the reaction mixture and stirred for 15 minutes followed by the addition of a solution of composition (8) (1 mg, 0.45 μmol) in H2O-MeOH (1:3, 0.4 mL). The reaction mixture was stirred at ambient temperature under N2 atmosphere for 40 h. The reaction mixture was concentrated, dissolved in CH2Cl2-MeOH (1:1) and purified by a short LH 20 size exclusion column using CH2Cl2-MeOH (1:1) as solvent. Lyophilization of the eleutants afforded composition (9) as a white solid (1.9 mg, 100%). HR-MALDI-MS: [M+H] m/z calcd for C151H255N31O40S, 3175.593. Found 3175.425. A mass peak corresponding to a protonated methyl ester of the product was also observed.
  • Example 4 Synthesis of a Liposomal Vaccine of Composition I and Control Compositions
  • The liposomes were formulated by the extrusion method in a total lipid concentration of 30 mM. For the preparation of the liposomes, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) was used. Lipid stock solutions were prepared by dissolving each lipid into chloroform inside glass vials. Aliquots of the stock solutions were mixed in proportions in another small glass vial to give a solution with a total lipid concentration of 30 mM in a total volume of 2 mL.
  • Batch 1: DPPC 80%, Cholesterol 10%, Rha-cholesterol 10% and Pam3cys-MUC1-Tn (9) 2 nM.
  • Batch 2: DPPC 80%, cholesterol 20%, Pam3cys-MUC1-Tn (9) 2 nM.
  • Batch 3: DPPC 80%, cholesterol 20%.
  • Chloroform was removed by subjecting the lipid solutions to a constant stream of nitrogen. The resulting lipid films were dried under vacuum overnight. The dried lipid films were hydrated with 2 mL of HEPES buffer (pH=7.4). The suspensions of the lipids in the buffer were agitated at 43° C. for 40 mins. The suspensions were subjected to 10 freeze-thaw cycles (dry ice/acetone and water at 40° C.). Final liposomes were prepared by extrusion (21 times) using a LipoFast Basic fitted with a 100 nm polycarbonate membrane to control the liposome size.
  • General Characterization of Liposomal Vaccine of Composition I and Control Compositions
  • The homogeneity, stability as well as size characterization of the liposomes were evaluated by scanning electron microscope (SEM) imaging (FIGS. 14A-14B) and dynamic light scattering (DLS) measurements (FIGS. 13A-13C).
  • All batches of liposomes were found to be stable at 4° C. for 2 days and were around 100 nm in diameter. Antibody binding study showed positive binding of the Batch 1 liposomes with anti-rhamnose antibodies as well as mouse anti-human-MUC1 (CD 227, BD Biosciences, San Jose, Calif.) antibodies using FITC goat anti-mouse IgG/IgM secondary antibodies and fluorescence imaging of the coated liposomes, as shown in FIG. 15.
  • The binding assay proved that the L-rhamnose and the MUC1 VNTR-TACA conjugate were displayed on the surface of the liposomes. No such antibody binding (both anti-rhamnose and mouse anti-human-MUC1) were observed for the control Batch 3 liposomes.
  • Batch 2 liposomes only demonstrated mouse anti-human-MUC1 antibody binding.
  • Example 5 Liposome Characterization
  • Size Characterization
  • Size determination of the liposomes was done by scanning electron microscope (SEM) imaging and dynamic light scattering (DLS) measurements. For SEM characterization the liposome samples were diluted 1000 times with HEPES buffer (pH=7.4) and freeze dried over copper studs fitted with a carbon conducting tape and the images recorded at an acceleration voltage of 5 kV. DLS measurements were done after dilution of the liposome samples 10000 times with HEPES buffer (pH=7.4).
  • Anti-Rha and Anti-MUC1 Antibody Binding to Surface Exposed Rha and MUC1 Epitopes on Liposomes
  • 106 liposomes from each batch in 50 μL 1×PBS (pH=7.4) were added separately into a 1.5 mL Eppendrof tube followed by 50 μL of primary antibody solution in deionized water containing 5-50 μg/mL of antibodies [either control antibodies (isolated from the serum of non-immunized mice) or anti-Rha antibodies or mouse anti-human CD227 antibodies (anti-human MUC1)] and incubated on ice for 30 mins. 1 mL PBS-0.1% Tween was added to each tube and vortexed. Liposomes were spun at 14000 rpm in Eppendrof centrifuge at 4° C. for 5 mins. The supernatants were carefully discarded and the washing and centrifugation steps were repeated 2 more times for a total of 3 washes. Liposomes were then resuspended in 50 μL of PBS-0.1% Tween. 50 μL of diluted FITC goat anti-mouse IgG/IgM secondary antibody were added (2-30 μg/mL) to the tubes, mixed and covered with aluminum foil to protect from light and incubated on ice for 30 mins. After washing 3 times with PBS-0.1% Tween and centrifugation, the supernatants were removed and pellets were resuspended in 1 mL PBS-0.1% Tween. 10 μL of the resuspended solutions were put on glass slides and imaged under a fluorescent microscope.
  • Example 6
  • NMR spectra and HRMS data of compositions (2), (3), (5) and (6) are shown in FIG. 16 through FIG. 31.
  • FIG. 16: 1H NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl-2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 17: 13C NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl-2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 18: 1H-gCosy of (5-Cholesten-3α-yloxy)-3n3 9- trixaundecanyl 2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 19: HRMS of (5-Cholesten-3α-yloxy)-3n3 9- trixaundecanyl 2,3,4-tri-O-acetyl-α-L-Rhamnopyranoside (2).
  • FIG. 20: 1H NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 21: 13C NMR of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 22: 1H-gCosy of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 23: HRMS of (5-Cholesten-3α-yloxy)-3n3 9-trixaundecanyl Rhamnopyranoside (3).
  • FIG. 24: 1H NMR of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 25: 13C NMR of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 26: 1HgCosy of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 27: HRMS of N-Propargyl Pam2FmocCys Amide Derivative (5).
  • FIG. 28: 1H NMR of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 29: 13C NMR of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 30: 1H-gCosy of N-Propargyl Pam3Cys Amide Derivative (6).
  • FIG. 31: HRMS of N-Propargyl Pam3Cys Amide Derivative (6).
  • The MALDI-TOF spectra data for compositions (7b), (8) and (9) are shown in FIG. 32 through FIG. 34.
  • FIG. 32: HR-MALDI-TOF of Glycopeptide Azide (7b).
  • FIG. 33: HR-MALDI-TOF of Glycopeptide Azide (8).
  • FIG. 34: HR-MALDI-TOF of Lipopeptide (9).
  • Example 7 2-Azidoethyl-2,3,4-Tri-O-Acetyl-α-L-Rhamnopyranoside (11)
  • To a solution of 1,2,3,4-tetra-O-acetylrhamnopyranoside (10) (2.00 g, 6.02 mmol) in CH2Cl2 (5.00 mL) were added 2-azidoethanol (0.79 g, 9.03 mmol) and BF3.OEt2 (1.53 mL, 12.04 mmol) at 0° C. and the resulting solution was stirred at ambient temperature under N2 atmosphere. The reaction was monitored by TLC and appeared to be completed after 12 h. The reaction mixture was diluted with CH2Cl2 (15 mL) and washed with water (2×20 mL), saturated NaHCO3 (2×20 mL) and brine (20 mL), after which the organic layer was dried over anhydrous Na2SO4. Excess solvent was removed under reduced pressure and the crude material was purified by silica gel flash column chromatography (3.3×8.5 cm). Elution with 1:5 EtOAc/hexanes afforded (11) as a colorless solid (1.78 g, 83%). 1H NMR (600 MHz, CDCl3): δ 1.24 (d, 3H, J=6.6 Hz, C-5 CH3), 1.99 (s, 3H, COCH3), 2.06 (s, 3H, COCH3), 2.16 (s, 3H, COCH3), 3.42 (m, 1H, —CHH—N3), 3.48 (m, 1H, —CHH—N3), 3.64 (m, 1H, —O—CHH), 3.87 (m, 1H, —O—CHH), 3.93 (m, 1H, H-5), 4.79 (d, 1H, J=1.8 Hz, H-1), 5.09 (t, 1H, J=10.2 Hz, H-4), 5.27 (dd, 1H, J=1.2, 3.3 Hz, H-2), 5.31 (dd, 1H, J=3.3, 9.9 Hz, H-3). 13C NMR (100 MHz, CDCl3): δ 17.66 (CH3), 20.93, 21.03, 21.13, 50.58, 66.91, 66.99, 69.08, 69.87, 71.09, 97.79 (C−1), 170.09 (C═O), 170.24 (C═O), 170.30 (C═O). HRMS [M+Na] m/z: calcd for C14H21N3O8, 382.1226. Found, 382.1215.
  • 2-Azidoethyl-α-L-Rhamnopyranoside (12)
  • To a solution of (11) (1.53 g, 4.26 mmol) in MeOH (5 mL) was added metallic Na (0.01 g) and the resulting solution was stirred at ambient temperature under N2 atmosphere. The reaction was monitored by TLC and appeared complete after 2 h. Excess solvent was removed under reduced pressure and the crude material was purified by silica gel flash column chromatography (3.3×8.5 cm). Elution with 2:23 MeOH/CH2Cl2 yielded (12) as a colorless solid (0.85 g, 86%). 1H NMR (600 MHz, CDCl3): δ 1.34 (d, 3H, J=6.6 Hz, C-5 CH3), 3.41 (m, 2H, —CH2—N3), 3.49 (t, 1H, J=9.3 Hz, H-4), 3.63 (m, 1H, —O—CHH), 3.69 (m, 1H, H-5), 3.81 (dd, 1H, J=3.3, 9.3 Hz, H-3), 3.89 (m, 1H, —O—CHH), 3.99 (q, 1H, J=1.6 Hz, H-2), 4.83 (d, 1H, J=1.2 Hz, H-1). 13C NMR (100 MHz, CDCl3): δ 17.75 (CH3), 50.71, 66.72, 68.49, 70.98, 71.76, 73.27, 100.02 (C−1). HRMS [M+Na] m/z: calc for C8H15N3O5, 256.0909. Found, 256.0906.
  • 2-Aminoethyl-α-L-Rhamnopyranoside (13)
  • To a solution of (12) (0.42 g, 1.82 mmol) in MeOH (3 mL) was added activated Pd/charcoal (0.025 g) and the resulting solution was stirred at ambient temperature under H2 atmosphere. The reaction was monitored by TLC and appeared to be complete after 12 h. The reaction mixture was diluted with MeOH (2 mL), filtered through Celite and concentrated under reduced pressure to yield (13) as a colorless gel (0.46 g, quantitative) which was used without further purification for subsequent reactions. ESIMS [M+H] m/z: calcd for C8H17NO5, 208.2243. Found, 208.30.
  • 2-Aminoethyl-α-L-Rhamnopyranoside-Ficoll Conjugate (14)
  • Ficoll 400 (1.00 g, 0.0025 mmol) was dissolved in acetate buffer (10 mL, pH 4.7) and NaIO4 (0.01 g, 0.047 mmol) was added and the reaction mixture was stirred at ambient temperature for 2 h in the dark. Excess NaIO4 was removed by dialysis against the acetate buffer (pH 4.7) through dialysis tubing with a molecular weight cutoff value of 10000 Da with six to seven changes of the buffer at 4° C. The oxidized Ficoll 400 was transferred to a round bottom flask and excess solvent was evaporated to dryness under reduced pressure. The residue was dissolved in borate buffer (20 mL, pH 8.0) followed by the addition of (13) (0.05 g, 0.25 mmol) and stirred at ambient temperature for 2 h. To the reaction mixture was added NaBH3CN (0.094 g, 1.50 mmol) and the resulting solution was incubated overnight at 4° C. The mixture was dialyzed through dialysis tubing with a molecular weight cutoff of 10000 Da with six to seven changes in buffer at 4° C. to afford (14). The epitope ratio of (14) was calculated to be 9.44 (Rha:Ficoll) by hydrolysis of (14) followed by derivatization with 4-amino-N-[2-(diethylamino)ethyl]benzamide (DEAEAB) and comparison of the UV-HPLC peak area with a standard curve obtained from DEAEAB derivative of (12) by the methods described by Dalpathado and coworkers. Briefly, the standard curve was generated by refluxing compound (12) (0.007 g, 0.031 mmol) with 1 M HCl at 100° C. for 4 h and the reaction mixture was evaporated to dryness. The residue was dissolved in tetrahydrofuran (2 mL). DEAEAB (0.011 g, 0.037 mmol) and Et3N (0.007 mL, 0.046 mmol) were added and the resulting solution was refluxed for 2 h. The reaction mixture was evaporated to dryness and the residue was dissolved in MeOH (2 mL) followed by the addition of NaB(OAc)3H, and the resulting solution refluxed for 8 h. The solution was evaporated to dryness and the residue was dissolved in MeOH (2 mL) and filtered through a syringe filter. Serial dilutions from this stock solution were prepared and the components were separated on a reverse phase HPLC using a C18 column. Water containing 0.1% TFA (A) and 95% ACN/H2O (B) was used as the mobile phases using a linear gradient (5-20% B in 20 min) at the flow rate of 1 mL/min. Absorbances were recorded at 289 nm. The standard curve was generated by plotting the UV-HPLC peak area against the concentration in mmol of DEAEAB derivative of (12).
  • NMR spectra and HRMS data of compositions (11), (12), and (13) are shown in FIG. 43 through FIG. 51.
  • FIG. 43: 1H NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 44: 13C NMR of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 45: 1H-gCosy of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 46: HRMS of 2-Azidoethyl-2,3,4-Tri-O-acetyl-α-L-rhamnopyranoside (11).
  • FIG. 47: 1H NMR of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 48: 13C NMR of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 49: 1H-gCosy of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 50: HRMS of 2-Azidoethyl α-L-rhamnopyranoside (12).
  • FIG. 51: ESI MS of 2-Aminoethyl α-L-rhamnopyranoside (13).
  • Example 8
  • A 20-amino acid (TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5]) tandem repeat of MUC1 was synthesized. The peptide was modified with a terminal azido group in order to make a ‘click’ conjugation to the Pam3Cys alkyne with a terminal azido group. The peptide was synthesized by a Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) starting from preloaded Fmoc-L-Val Wang resin using solid-phase chemistry, as shown in FIG. 52.
  • The peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent. A six-fold excess of Nα-Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of Nα-Fmoc group was accomplished by treatment with piperidine in DMF. After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of TFA-thioanisole-ethanedithiol-water-phenol (88:3:5:2:2). The cocktail mixture was filtered through a Quick Snap column, purified by C18 reverse phase HPLC, and lyophilized to afford composition (16) as a white powder.
  • Conjugation of the composition (16) (1 eqv) with the composition (6) (3 eqv) in the presence of copper(I) iodide (12 eqv), tris [(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA) (3 eqv), diisopropyl ethyl amine (DIEA) (12 eqv) and sodium ascorbate (12 eqv) in H2O-THF-DMF (1:1:2) as solvent at 20° C. thus afforded the target, Pam3Cys-MUC1 VNTR conjugate (17), after 16 h.
  • Composition (17) was purified by neutralization by 7.5% citric acid solution followed by extraction with chloroform. Evaporation of the chloroform extract afforded the composition (17) as a white solid.
  • Synthesis of Azide (16)
  • The peptidazide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) using solid phase chemistry. The peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent starting with a preloaded Fmoc-valinyl-Wang resin. A six-fold excess of Nα-Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of Nα-Fmoc group was accomplished by treatment with 25% piperidine in dimethylformamide twice; first for 5 minutes and then a second time for 25 minutes to afford composition (15).
  • After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of 88% TFA, 3% thioanisole, 5% ethanedithiol, 2% water, and 2% phenol. 4 mL of cleavage cocktail was added to the dried peptide-resin in a 15 mL glass vial blanketed with nitrogen. Cleavage was carried out for 2.5 hrs with gentle magnetic stirring. At the end cleavage time, the cocktail mixture was filtered on a Quick-Snap column. The filtrate was collected in 20 mL ice-cold butane ether. The peptide was allowed to precipitate for an hour at −200° C., centrifuged, and washed twice with ice-cold methyl-t-butyl ether. The precipitate was dissolved in 25% acetonitrile and lyophilized to complete dry powder affording composition (16). Quality of peptides was analyzed by analytical reverse phase HPLC and MALDI-TOF (matrix assisted laser desorption ionization time-of-flight) mass spectrometer, model 4800 from Applied Biosystems. HR-MALDI-MS: [M+H] m/z calcd for C86H136N28O29, 2026.17. Found, 2026.137.
  • Synthesis of Lipopeptide (17)
  • CuI (134 μg, 0.54 μmol) and TBTA (0.857 mg, 1.62 μmol) were dissolved in H2O-THF (1:1, 0.40 mL) and to it sodium ascorbate (0.80 mg, 4.04 μmol) was added and stirred for 5 minutes. Composition (6) (1.27 mg, 1.35 μmol) in THF (0.40 mL) was added to the reaction mixture and stirred for 15 minutes followed by the addition of a solution of composition (16) (1 mg, 0.49 μmol) in H2O-DMF (1:3, 0.4 mL). The reaction mixture was stirred at 20° C. under N2 atmosphere for 16 h. The reaction mixture was concentrated, dissolved in CHCl3, washed with 7.5% aqueous citric acid solution, dried over sodium sulfate, and the solvent was evaporated to afford composition (17) as a white solid (1.47 mg, 100%). HR-MALDI-MS: [M+H] m/z calcd for C143H242N30O35S, 2972.78. Found 2972.828.
  • FIGS. 53 and 54 show the HRMS (MALDI-TOF) spectrum and HPLC trace, respectively, of peptide (16).
  • FIG. 55 shows the HRMS (MALDI-TOF) spectrum of lipopeptide (17).
  • Example 9
  • A 20-amino acid (TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6]) tandem repeat of MUC1 which included the PD(α-GalNAc-O-T)R epitope was synthesized. The glycopeptides was modified with a terminal azido group in order to make a ‘click’ conjugation to the Pam3Cys alkyne. The glycopeptides azide was synthesized by Fmoc strategy on an Omega 396 synthesizer (Advanced ChemTech, Louisville, Ky.) starting from preloaded Fmoc-L-Val Wang resin using solid-phase chemistry, as shown in FIG. 56.
  • The peptide synthesis was performed by coupling amino acid esters of HOBt using DIC as the coupling agent. A six-fold excess of Nα-Fmoc amino acid esters of HOBt in NMP were used in the synthesis. A 1:1 ratio of amino acid to DIC was used in all the coupling reactions. Deprotection of Nα-Fmoc group was accomplished by treatment with piperidine in DMF. After the synthesis was complete, the peptide was cleaved from the solid support and deprotected using a modified reagent K cocktail consisting of TFA-thioanisole-ethanedithiol-water-phenol (88:3:5:2:2). The cocktail mixture was filtered through a Quick Snap column, purified by C18 reverse phase HPLC and lyophilized to afford composition (19).
  • The acetyl groups in composition (19) were deprotected by treatment with 6 mmol sodium methoxide in methanol. The product was purified by Bio-Gel (P-2, fine 45-90 μm) size exclusion chromatopgraphy using deionized water as solvent. Lyophilization of the fractions afforded composition (20) (100%) as a white powder. Conjugation of the composition (19) (1 eqv) with the composition (6) (3 eqv) in the presence of copper(I) iodide (12 eqv), tris [(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA) (3 eqv), diisopropyl ethyl amine (DIEA) (12 eqv) and sodium ascorbate (12 eqv) in H2O-THF-DMF (1:1:2) as solvent at 20° C. thus afforded the target, Pam3Cys-MUC1 VNTR-TACA conjugate (21), after 16 h. Composition (21) was purified by neutralization by 7.5% citric acid solution followed by extraction with chloroform. Evaporation of the chloroform extract afforded the composition (21) as light yellow solid.
  • Synthesis of Azide (20)
  • Composition (19) (5 mg, 2.24 μmol) was dissolved in 2 mL of dry methanol. 12 μL of freshly prepared 1 M sodium methoxide was added to the mixture and the reaction mixture was stirred at ambient temperature under N2 atmosphere for 3 h. The reaction mixture was neutralized with solid carbon dioxide. The reaction mixture was concentrated and purified by Bio-Gel (P-2, fine 45-90 μm) size exclusion chromatography using deionized water as solvent. Lyophilization of the eleutants afforded composition (20) as a white powder (4.7 mg, 100%). HR-MALDI-MS: [M+H] m/z calcd for C94H149N29O34, 2229.09. Found 2026.336.
  • Synthesis of Glycolipopeptide (21)
  • CuI (134 μg, 0.54 μmol) and TBTA (0.857 mg, 1.62 μmol) were dissolved in H2O-THF (1:1, 0.40 mL) and to it sodium ascorbate (0.80 mg, 4.04 μmol) was added and stirred for 5 minutes. Composition (6) (1.27 mg, 1.35 μmol) in THF (0.40 mL) was added to the reaction mixture and stirred for 15 minutes followed by the addition of a solution of composition (20) (1 mg, 0.45 μmol) in H2O-DMF (1:3, 0.4 mL). The reaction mixture was stirred at 20° C. under N2 atmosphere for 16 h. The reaction mixture was concentrated, dissolved in CHCl3, washed with 7.5% aqueous citric acid solution, dried over sodium sulfate, and the solvent was evaporated to afford composition (21) as a light yellow solid (1.9 mg, 100%). HR-MALDI-MS: [M+H] m/z calcd for C151H255N31O40S, 3175.86. Found 3175.809.
  • FIGS. 57 and 58 show the HRMS (MALDI-TOF) spectrum and HPLC trace of glycopeptide (19).
  • FIG. 59 is the HRMS (MALDI-TOF) spectrum of glycopeptides (20).
  • FIG. 60 is the HRMS (MALDI-TOF) spectrum of Pam3Cys-MUC-1 VNTR conjugate (21).
  • Methods of Use
  • Also provided herein is a method for eliciting an immune response against a cancer cell surface antigen in a subject in need thereof. The method comprises administering to the subject an antigen-liposome-xenoantigen vaccine composition in sufficient dose to elicit the immune response to the cancer cell surface antigen, wherein the antigen-liposome-xenoantigen vaccine composition comprises the cancer cell surface antigen and a liposome.
  • In certain embodiments, the cancer cell surface antigen is expressed only in cancer cells in the subject, such that the immune response specific for the antigen would not attack healthy tissues/organs of the subject. In certain embodiments, the antigen that is expressed on the cancer cell surface is a mutant protein present only on cancer cells, and the immune response is specific for the mutant protein epitope comprising the mutation. The mutation can be a protein mutation, or abnormal sugar or lipid structures on cancer cell surface.
  • Example 10 T-Cell Proliferation Study Immunization
  • One female BALB/c mouse (6-8 weeks old, The Jackson Laboratory) was primed (day 0) and boosted three time (days 14, 28, and 42) with 100 μL subcutaneous injections of an equivolume emulsion of the MUC1-Tn conjugate (8) (prepared in phosphate buffer saline-PBS) and sigma adjuvant system (SAS) (50 μg of peptide per mouse, each injection).
  • Preparation of Anti-Rha Antibodies
  • The Rha-Ficoll and the Rha-OVA immunized mice were bled on day 66 and the sera was pooled. IgG fractions from each pool were prepared by precipitation at 40% saturation of ammonium sulfate. The mixtures were incubated overnight and centrifuged at 10000×g for 10 minutes and then resuspended in 0.5 mL water. The antibody solutions were concentrated and buffer was changed twice with PBS using an Ultrafree 0.5 centrifugal filter device (Millipore, Billerica, Mass.) having a molecular cut off of 50000 D. Absorbances of the antibody solutions were recorded at 280 nm to calculate the concentrations and the anti-Rha antibody solutions generated and isolated from the Rha-Ficoll and the Rha-OVA immunized mice were each diluted to 1.0 mg/mL.
  • Preparation of Spleen Cell Suspensions and Assay Setup
  • On day 49, the mouse was sacrificed and the spleen was removed and placed in 5 mL of freshly prepared spleen cell culture medium (DMEM with 10% fetal calf serium). Single cell suspension was prepared using modified sterile glass homogenizers. The cells were washed three times with culture medium and brought to 5×106 cells/mL. 100 μL of the spleen cell suspensions were added to 96 well plates (5×105 cells per well). The dendritic cell (DC) suspension cultured from the bone marrow of a BALB/c mouse was pulsed with the antigen by incubating with the Rha-displaying MUC1-Tn liposomes at antigen concentrations of 8.8×10−3-1.1 μg/mL at 37° C. for 4 h together with anti-Rha antibodies generated from either Rha-Ficoll or Rha-OVA immunized mice sera (5 μCi/mL, 25 μL per well) and incubated overnight at 37° C. The cells were harvested on glass-fiber filters and incorporation was determined by measurements on a Top Count scintillation counter.
  • Immunizations
  • The 20 female BALB/c mice used for this study were divided into four groups A1, A2, B1, and B2, containing 5 mice each. Groups A1 and B1 served as the control groups and were not immunized. Groups A2 and B2 were injected subcutaneously (day 0) with a 100 μL subcutaneous injections Rha-Ficoll/Alum on days 14, 28, 42, and 56 (100 μg of Rha-Ficoll per mouse, each boost). The mice in each group A1, A2, B1, and B2 were bled on day 66 and the collected sera were tested for anti-Rha antibodies.
  • ELISA for Measuring Anti-Rha Antibody Titers
  • 96 well plates (Immulon 4 HBX) were coated with Rha-BSA conjugate (6) (2 μg/mL) in 0.01 M PBS and incubated overnight at 4° C. The plates were washed 5 times with PBS containing 0.1% Tween-20. Blocking was achieved by incubating the plates for 1 h at room temperature with BSA in 0.01 M PBS (1 mg/mL). The plates were then washed 5 times and incubated for 1 h with serum dilutions in PBS. Unbound antibody in the serum was removed by washing and the plates were incubated for 1 h at room temperature with Horseradish Peroxidase (HRP) goat anti-mouse IgG+IgM (Jackson Immunoresearch Laboratories) diluted 5000 times in PBS/BSA. The plates were washed with TMB (3,3′,5,5′-tetramethylbenzidine). One component HRP microwell substrate (Bio FX, Owings Mills, Md.) was added and allowed to react for 10 mins. Absorbances were recorded at 620 nm and were plotted against log10 [1/serum dilution].
  • Vaccinations
  • Vaccinations were performed on day 77. Two separate liposomal formulations were prepared with DPPC (80%), cholesterol (20%) and Pam3Cys-MUC1-Tn (9) (2 nmol) (Pam3Cys-MUC1-Tn liposomes) and DPPC (80%), cholesterol (10%), Rha-TEG-cholesterol 3 (10%) and Pam3Cys-MUC1-Tn (9) (2 nmol) (Pam3Cys-MUC1-Tn+Rha liposomes) in total lipid concentrations of 30 mmol. Groups A1 and A2 were cavvinated with 100 μL subcutaneous injections of the Pam3Cys-MUC1-Tn liposomes (2 nmol of peptide per mouse) and groups B1 and B2 were vaccinated with 100 μL subcutaneous injections of the Pam3Cys-MUC1-Tn+Rha liposomes (2 nmol peptide per mouse). The mice were boosted on day 91 with either the Pam3Cys-MUC1-Tn liposomes (groups A1 and A2, 2 nmol peptide per mouse) or the Pam3Cys-MUC1-Tn+Rha liposomes (groups B1 and B2). The mice were bled on day 101 and the sera collected were tested for anti-MUC1-Tn and anti-Tn antibodies.
  • ELISA for Measuring Anti-MUC1-Tn Antibody Titers
  • 96 Well plates (Immulon 4 HBX) were coated with MUC1-Tn conjugate (8) (15 μL/mL) in 0.01 M PBS and incubated overnight at 4° C. The ELISA was continued as described above.
  • ELISA for Measuring Anti-Tn Antibody Titers
  • 96 Well plates (Immulon 4 HBX) were coated with Tn-BSA conjugate (15 μL/mL) in 0.01 M PBS and incubated overnight at 4° C. The ELISA was continued as described above.
  • Anti-MUC1-Tn Antibody Subclass Identification
  • 96 Well plates (Immulon 4 HBX) were coated with MUC1-Tn conjugate (8) (15 μL/mL) in 0.01 M PBS and incubated overnight at 4° C. The plates were washed 4 times with PBS containing 0.1% Tween-20. Blocking was achieved by incubating the plates for 1 h at room temperature with BSA in 0.01 M PBS (1 mg/mL). The plates were then washed 4 times and incubated for 1 h with 1/100 serum dilution in PBS. Unbound antibody in the serum was removed by washing and the plates were incubated overnight at 4° C. with subclass specific (IgG1, IgG2s, IgG2b, IgG3, 1gA, and IgM) rabbit anti-mouse antibody (Zymed Laboratories mouse monoAb-ID kit). The plates were washed and incubated with HRP-goat anti-rabbit IgG (H+L) for 1 h at room temperature. The plates were washed and ABTS substrate buffer (diluted 50 times) was added and allowed to react for 30 min. Absorbances were recorded at 405 nm and compared for each antibody subclass in each group.
  • ELISA for Competitive Binding with Free MUC1-Tn
  • A 96-well plate (Immulon 4 HBX) was coated with MUC1-Tn conjugate (8) (15 μL/mL) in PBS and incubated overnight at 4° C. The plate was washed 5 times with PBS containing 0.1% Tween-20. Blocking was achieved by incubating the plate for 1 h at room temperature with BSA in M PBS (1 mg/mL). The plate was then washed 5 times and incubated for 1 h with serum dilutions of 1/100 in PBS with or without prior mixing with varying concentrations of free MUC1-Tn (8) from 0, 10−5, 10−4, 10−3 M in PBS. Unbound antibody in the serum was removed by washing and the plate was incubated for 1 h at room temperature with Horseradish Peroxidase (HRP) goat anti-mouse IgG+IgM (secondary antibody) diluted 5000 times in PBS/BSA. The plate was washed and TMB 1 component HRP microwell substrate was added and allowed to react for 10 mins. Absorbances were recorded at 620 nm and were plotted against log10 [1/free Tn concentration].
  • Tumor Cell Staining
  • U266 cells (American Type Culture Collection, Manassas, Va.), were cultured in RPMI 1640 with 15% fetal calf serum (FCS). Cells were stained with purified mouse anti-human MUC1 antibodies (CD227, 0.5 μg), non-immune BALB/c mice serum (1/5 dilution) and group B2 mice serum (1/5 dilution). The cells were then stained with FITC-conjugated goat anti-mouse IgG+IgM (0.5 μg) and fluorescence was quantified with a BD FACS Calibur.
  • Comparison of Anti-Rha Antibody Titers Generated Against Rha-Ficoll and Rha-OVA
  • Two groups of five female BALB/c mice each were immunized on day 0 with Rha-Ficoll/Alum adjuvant (group A) or Rha-OVA/complete Freund's adjuvant (CFA) (group B). The mice were boosted three more times on days 14, 28, and 42 with either Rha-Ficoll/Alum (group A) or rha-OVA/incomplete Freund's adjuvant (ICF) (group B). Sera were collected separately from groups A and B after the third boost and the anti-Rha antibodies in the sera from the two groups of mice were isotyped by screening against Rha-BSA. See FIG. 36. The results demonstrated the anti-Rha antibody titers in the Rha-OVA immunized micro groups were 100-fold higher than those from the Rha-Ficoll immunized mice. However, the isotype distribution confirmed that Rha-Ficoll and Rha-OVA produced the anti-Rha antibody subclasses in different proportions. Anti-Rha antibodies from Rha-OVA immunization were dominated by IgG1 (65%) while Rha-Ficoll immunization produced antibodies which comprised mainly IgG3 (48%) and IgM (25%). IgG1 and IgG3 act similarly in that they both stimulate high affinity FcγRI receptors which trigger responses from macrophages. However, IgG1 also stimulates low affinity FcγRIIB receptors which inhibit the signals from the FcγRI and B cell receptors thereby diminishing B-cell activity and immunogenicity of macrophages. The anti-Rha antibody isotypes from Rha-Ficoll immunized mice serum resembled those naturally occurring in the human serum which is presumed to be generated through a T-independent response.
  • T-Cell Proliferation Study
  • T-cell proliferation assays were performed to determine if the combination of anti-Rha antibodies and Rha-modified liposomal vaccine would potentiate a T-cell proliferative response. In the first part of the study, the proliferation assay conditions were optimized. BALB/c mice were immunized (day 0) and boosted (days 14, 28, and 42) with 100 μL emulsions of MUC1-Tn (8)/Sigma adjuvant system (SAS) (50 μg peptide per mouse, each injection). The mice were sacrificed (day 49), the spleens were removed, and single cell suspensions were prepared and incubated with MUC1-Tn (8.8×10−3-1.1 μg/mL) alone or with syngeneic bone marrow dendritic cells (DCs) previously pulsed with the same doses of antigen. DCs showed enhanced proliferation, as shown in FIG. 37. To test the ability of anti-Rha antibodies to enhance antigen presentation, spleen cells from BALB/c mice immunized as above were prepared. DCs from BALB/c bone marrow were pulsed with the antigen by incubating with Pam3Cys-MUC1-Tn+Rha liposomes at antigen concentrations of 8.8×10−3-0.22 μg/mL together with antibodies isolated from either Rha-Ficoll or Rha-OVA immunized mice or nonimmune mice. The pulsed DCs were added to the spleen cells and proliferation assessed after 3 days. The spleen T-cells proliferated better in the presence of anti-Rha antibodies (from both Rha-Ficoll and Rha-OVA immunized mice serum) than in the presence of control serum antibodies over the antigen concentration range of 8.8×10−3-0.22 μg/mL. See FIG. 38. Also, the T-cell proliferation was higher in the presence of anti-Rha antibodies generated against Rha-Ficoll (6328, 6045, and 6521 counts per minute (cpm) at antigen concentrations of 8.8×10−3, 0.044, and 0.22 μg/mL) than those against Rha-OVA (5018, 4926, and 4880 cpm at antigen concentrations of 8.8×10−3, 0.044, and 0.22 μg/mL), even though the titer of anti-Rha antibodies was higher in the serum of Rha-OVA immunized mice. The results strongly suggest that the Rha-modified antigen was more effectively internalized and presented by the APCs in the presence of anti-Rha antibodies, particularly those less inhibitory isotypes characteristic of natural antibodies and generated by Rha-Ficoll immunization. Therefore, BALB/c mice in which anti-Rha antibodies are generated with Rha-Ficoll (14) immunization are an appropriate model for the immunogenicity of the Rha-conjugated MUC1-Tn liposomes.
  • Anti-Rha Antibody Generation
  • Four groups of five female BALB/c mice each (groups A1, A2, B1, and B2) (6-8 weeks old) were used for this vaccination study. Groups A2 and B2 were immunized (day 0) and boosted (days 14, 28, 42, and 56) with 100 μL equivolume emulsion of Rha-Ficoll (prepared in PBS) and alum adjuvant. Groups A1 and B1 served as the control groups and were deprived of the Rha-Ficoll/Alum immunization. See FIG. 38B. The mice were bled on day 66 and the ELISA performed by screening the sera from the different groups against Rha-BSA showed that the anti-Rha antibody titers in groups A2 and B2 were 25-fold higher than the control groups. See FIG. 39. Thus, immunization with Rha-Ficoll confirmed the generation of anti-Rha antibodies in the experimental groups of mice.
  • Vaccination with Rha and non-Rha-Displaying MUCA1-Tn Liposomes
  • Two separate liposomal formulations were prepared. The first contained DPPC, cholesterol and Pam3Cys-MUC1-Tn (9) (2 nmol) (Pam3Cys-MUC1-Tn liposomes) and the second contained DPPC, cholesterol, Rha-TEG-cholesterol (3) and Pam3Cys-MUC1-Tn (9) (2 nmol) (Pam3CysMUC1-Tn+Rha liposomes). In both formulations the total lipid concentration was 30 mmol. The vaccination was performed on day 77. Groups A1 and A2 were given 100 μL subcutaneous injections of the Pam3Cys-MUC1-Tn liposomes (2 nmol of peptide per mouse) and groups B1 and B2 were given 100 μL subcutaneous injections of the Pam3CysMUC1-Tn+Rha liposome (2 nmol peptide per mouse). The mice were boosted on day 91 with either the Pam3CysMUC1-Tn liposome (groups A1 and A2, 2 nmol peptide per mouse) or the Pam3CysMUC1-Tn+Rha liposome (groups B1 and B2). The mice were bled on day 101 and the sera evaluated for anti-MUC1-Tn and anti-Tn antibodies. See FIG. 39B.
  • Anti-MUC1-Tn antibody titers were determined by screening the sera against the MUC1-Tn conjugate (8). (FIG. 39B). The data showed that groups A1, A2, and B1 had similar absorbance at 1/25, 1/50, 1/100, and 1/200 serum dilutions. This proved that prior immunization with Rha-Ficoll does not affect the response to a non-Rha conjugated vaccine (groups A1 and A2). In addition, the Rha epitopes on the vaccine do not alter the inherent immunogenicity of the MUC1-Tn epitopes on the vaccine (groups A1 and B1). The anti-MUC1-Tn titers for group B2 showed an 8-fold increase compared to groups A1, A2, and B1, which was mediated by the anti-Rha antibody-dependent antigen-uptake. Group B2 had an anti-MUC1-Tn titer of approximately 1/300, where titer is defined as the highest dilution giving a signal >0.1 above background. The anti-Muc1-Tn antibodies from each group were isotyped, resulting in group B2 showing an increase in IgG1, IgG2a, IgG2b, and IgM isotypes relative to the other three groups. See FIG. 40. The specificity of the antibodies towards MUC1-Tn antigen was determined by a competitive binding experiment. See FIG. 41A. Serum from every group at 1/100 dilution was incubated with the MUC1-Tn conjugate (8) at concentrations of 0, 10−5, 10−4, and 10−3 M in 0.01 M PBS prior to addition in the ELISA plates coated with the conjugate (8). The absorbances decreased uniformly with increasing concentrations of free MUC1-Tn in the serum dilutions for each group. As an example, the absorbances at 620 nm for the serum dilution of group B2 at free MUC1-Tn concentrations of 0, 10−5, 10−4, and 10−3 M were 0.790, 0.601, 0.577, and 0.512, respectively. These results confirmed the specificity of the anti-MUC1-Tn antibodies towards the respective antigen.
  • The antibody titer generated solely against the TACA was determined by screening serum dilutions from every group against a Tn-BSA conjugate. See FIG. 41B. A >8-fold increase in the anti-Tn antibody titers for group B2 was also observed in comparison to groups A1, A2, and B1. This was again attributed to the better uptake of the antigen in the presence of the anti-Rha antibodies by an antibody-dependent antigen-uptake mechanism. Also observed in this study was that the anti-MUC1-Tn antibody titers were higher than the corresponding anti-Tn antibody titers for the same serum dilutions for every group, assuming similar levels of antigen on the plate. For example, for the group B2, the absorbances at 620 nm for the anti-MUC1-Tn and the anti-Tn measurements at 1/100 serum dilutions were 0.922 and 0.509, respectively. This observation demonstrates that the Rha-displaying MUC1-Tn vaccine successfully generates antibodies against both the MUC1 peptide and the TACA.
  • The ability of the anti-MUC1-Tn antibodies in the vaccinated mice serum to bind to MUC1-Tn on human tumor cells was demonstrated with U266 human leukemia cells. These cells express MUC1 on their surface as shown by binding with mouse anti-human MUC1 antibodies (CD 227). See FIG. 42A. Serum from group B2 mice also recognized the MUC1 on the tumor cells with similar efficiency as the CD 227 antibodies relative to non-immunized mouse serum. See FIG. 42B. This demonstrates that the antibodies generated against the glycopeptides recognize the MUC1 protein in its native environment.
  • Summary
  • As the examples herein describe, a fully synthetic two-component vaccine containing the lipopeptide adjuvant Pam3Cys appended to a 20-amino acid MUC1 peptide containing the TACA GalNAc-O-Thr (Tn) was synthesized and was successfully formulated into liposomes along with an Rha cholesterol conjugate. The resulting liposomes were homogenous in size and were stable at 4° C. for two days. Binding studies with both anti-Rha and mouse anti-human MUC1 antibodies revealed that the Rha and the MUC1 glycopeptide epitopes were surface displayed on the liposomes. A Rha-Ficoll conjugate (14) was synthesized for the generation of anti-Rha antibodies in mice. The in vitro proliferation of MUC1-Tn primed mice spleen T-cells showed increased proliferation to Rha-liposomes in the presence of antibodies from Rha-Ficoll immunized mice relative to nonimmune mice. Vaccination studies with Rha- and non-Rha-displaying MUC1-Tn liposomes in mice either non-immunized or immunized with Rha-Ficoll illustrated that anti-MUC1-Tn and anti-Tn antibodies were >8-fold higher in the groups of mice previously immunized with Rha-Ficoll and later vaccinated with the Pam3Cys-MUC1-Tn+Rha liposomes. The anti-MUC1-Tn antibodies in the serum of the vaccinated mice recognized the aberrant MUC1 on human leukemia U266 cells. Overall, this vaccine successfully triggered both T-cell and humoral immunity enhanced by anti-Rha antibody dependant antigen uptake. Because this vaccine uses separate rhamnose and anti-genic epitope components, the vaccine can easily be targeted to different antigens or epitopes by changing the peptide without having to change the other components. For instance, the skilled practitioner will appreciate that anti-viral or anti-bacterial vaccines can also be made from them methods and compositions described herein.
  • While the invention has been described with reference to various and preferred embodiments, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted for elements thereof without departing from the essential scope of the invention. In addition, many modifications may be made to adapt a particular situation or material to the teachings of the invention without departing from the essential scope thereof.
  • Therefore, it is intended that the invention not be limited to the particular embodiment disclosed herein contemplated for carrying out this invention, but that the invention will include all embodiments falling within the scope of the claims.
  • The publication and other material used herein to illuminate the invention or provide additional details respecting the practice of the invention, are incorporated be reference herein, and for convenience are provided in the following bibliography.
  • Citation of the any of the documents recited herein is not intended as an admission that any of the foregoing is pertinent prior art. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicant and does not constitute any admission as to the correctness of the dates or contents of these documents.

Claims (31)

1. A composition comprising a first lipid (lipida) moiety and an alkyne amide moiety having a Formula IV:
Figure US20150024037A1-20150122-C00007
2. The composition of claim 1, wherein the first lipid (lipida) moiety comprises a Toll-like receptor (TLR) agonist ligand selected from one or more of: TLR2, TLR1, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, TLR13, TLR14, TLR15 and TLR16.
3. The composition of claim 1, wherein the lipid moiety comprises:
dipalmitoyl-S-glyceryl-cys-(Pam2Cys-);
tripalmitoyl-S-glyceryl-cys-(Pam3Cys-);
dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam2Cys-Ser-(Lys)4) [SEQ ID NO: 2];
tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam3Cys-Ser-(Lys)4) [SEQ ID NO: 3]; or
MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-asp-glu-ser-asn-ile-ser-phe-lys-glu-lys (Pam2CGNNDESNISFKEK)] [SEQ ID NO: 4].
4. The composition of claim 1, wherein Formula IV comprises an alkyne-functionalized Pam3Cys amide composition (6) comprising:
Figure US20150024037A1-20150122-C00008
wherein Pam is a dipalmitoyl-S-glyceryl-moiety.
5. A method of synthesizing an alkyne-functionalized composition of claim 1, comprising:
a) deprotecting an ester comprising a Fmoc moiety to form a free acid;
b) coupling the free acid of step (a) with an amine; and,
c) removing the Fmoc moiety, and coupling the remaining moiety with palmitic acid to yield an alkyne-functionalized composition.
6. A method of synthesizing an alkyne-functionalized Pam3Cys amide composition (6) of claim 4, comprising:
a) deprotecting 0-palmitoylated Fmoc L-cystine tert-butyl ester (4) to form a free acid;
b) coupling the free acid of step (a) with propargyl amine in presence of benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP), 1-hydroxy-benzotriazole (HOBt) and N,N-diisopropylethylamine (DIPEA) to yield composition (5); and,
c) removing a Fmoc group of composition (5) by treatment with a mixture of acetonitrile-dichloromethane-diethyl amine,
followed by subsequent palmitoylation by coupling with palmitic acid, PyBOP, HOBt and DIPEA to yield the alkyne-functionalized Pam3Cys amide composition (6).
7. A lipidated glycopeptide composition comprising: a first lipid (lipida) moiety, a first linker (linkera) moiety, and an antigen moiety, having a Formula II:
Figure US20150024037A1-20150122-C00009
8. (canceled)
9. The composition of claim 7, wherein the first lipid (lipida) moiety comprises:
dipalmitoyl-S-glyceryl-cys-(Pam2Cys-);
tripalmitoyl-S-glyceryl-cys-(Pam3Cys-);
dipalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam2Cys-Ser-(Lys)4) [SEQ ID NO: 2];
tripalmitoyl-S-glyceryl-cys-ser-lys-lys-lys-lys (Pam3Cys-Ser-(Lys)4) [SEQ ID NO: 3]; or
MALP-2 dipalmitoyl-S-glyceryl-cys-gly-asn-asn-asp-glu-ser-asn-ile-ser-phe-lys-glu-lys (Pam2CGNNDESNISFKEK)] [SEQ ID NO: 4].
10-12. (canceled)
13. The composition of claim 9 comprising mucin 1 (MUC1) variable number tandem repeats (VNTRs) conjugated to tumor-associated carbohydrate antigens (TACA).
14. The composition of claim 9, wherein the antigen is a TACA, selected from the group consisting of TF, Tn, sialyl Tn (sTn), or sialyl Lewis a (sLea) antigens, having the formulae:
Figure US20150024037A1-20150122-C00010
15. (canceled)
16. The composition of claim 9, wherein the antigen comprises MUC1 VNTR having one of the following amino acid sequences:
PDTRPAPGST(Tn)APPAHGVTSA [SEQ ID NO: 1];
TSAPDTRPAPGSTAPPAHGV [SEQ ID NO: 5]; or
TSAPDT(Tn)RPAPGSTAPPAHGV [SEQ ID NO: 6].
17. (canceled)
18. The composition of claim 7, wherein the first linker (linkera) comprises a dialkyl-substituted heteroaryl C1-n alkyl of Formula VI
Figure US20150024037A1-20150122-C00011
wherein the “A” group comprises: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n—; and n is a positive integer.
19. (canceled)
20. A composition of claim 7, comprising the compound (9):
Figure US20150024037A1-20150122-C00012
21. A composition of claim 7, comprising the compound (17):
Figure US20150024037A1-20150122-C00013
22. A composition of claim 7, comprising the compound (21):
Figure US20150024037A1-20150122-C00014
23-26. (canceled)
27. A composition comprising a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety, having the Formula VII:
Figure US20150024037A1-20150122-C00015
28. The composition of claim 27, wherein the second lipid (lipidb) moiety contains a structure of the Formula IX:
Figure US20150024037A1-20150122-C00016
29. The composition of claim 27, wherein the second linker (linkerb) comprises: a chain of C1-n alkyl, dialkyl substituted aryl C1-n alkyl, or —CH2CH2(OCH2CH2)n—; wherein n is a positive integer.
30-35. (canceled)
36. The composition of the claim 27, wherein the xenoantigen moiety contains a structure comprising: an α- or β-linked L-rhamnose epitope, a β-linked α-Gal disaccharide epitope, or an α- or β-linked Forssmann disaccharide epitope:
Figure US20150024037A1-20150122-C00017
37. A vaccine composition, comprising:
1) an antigen composition comprising:
a first lipid (lipida) moiety, a first linker (linkera) moiety, and an antigen moiety;
2) a xenoantigen composition comprising:
a second lipid (lipidb) moiety, a second linker (linkerb) moiety, and a xenoantigen moiety; and
3) at least one liposomal formulation.
38-40. (canceled)
41. A vaccine composition of claim 37, wherein:
the antigen composition comprises a Pam3Cys-MUC1 VNTR-TACA conjugate;
the second linker (linkerb) moiety comprises a tetraethyleneglycol (TEG) portion; and
the xenoantigen moiety comprises α- or β-linked L-rhamnose.
42-59. (canceled)
60. A method for improving immunogenicity of vaccines, the method comprising incorporating at least one α- or β-linked L-Rha epitope by direct conjugation or by non-covalent association with at least one liposomal vaccine formulation to increase the immunogenicity of the vaccine by a NA-dependant antigen uptake mechanism.
US14/378,805 2012-02-16 2013-02-15 Xenoantigen-Displaying Anti-Cancer Vaccines and Method of Making Abandoned US20150024037A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/378,805 US20150024037A1 (en) 2012-02-16 2013-02-15 Xenoantigen-Displaying Anti-Cancer Vaccines and Method of Making

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261599925P 2012-02-16 2012-02-16
US14/378,805 US20150024037A1 (en) 2012-02-16 2013-02-15 Xenoantigen-Displaying Anti-Cancer Vaccines and Method of Making
PCT/US2013/026271 WO2013123282A1 (en) 2012-02-16 2013-02-15 Xenoantigen-displaying anti-cancer vaccines and method of making

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/026271 A-371-Of-International WO2013123282A1 (en) 2012-02-16 2013-02-15 Xenoantigen-displaying anti-cancer vaccines and method of making

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/362,233 Division US10206988B2 (en) 2012-02-16 2016-11-28 Xenoantigen-displaying anti-cancer vaccines and method of making

Publications (1)

Publication Number Publication Date
US20150024037A1 true US20150024037A1 (en) 2015-01-22

Family

ID=48984725

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/378,805 Abandoned US20150024037A1 (en) 2012-02-16 2013-02-15 Xenoantigen-Displaying Anti-Cancer Vaccines and Method of Making
US15/362,233 Active US10206988B2 (en) 2012-02-16 2016-11-28 Xenoantigen-displaying anti-cancer vaccines and method of making

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/362,233 Active US10206988B2 (en) 2012-02-16 2016-11-28 Xenoantigen-displaying anti-cancer vaccines and method of making

Country Status (5)

Country Link
US (2) US20150024037A1 (en)
EP (1) EP2852413A4 (en)
AU (2) AU2013221448B2 (en)
CA (1) CA2869691A1 (en)
WO (1) WO2013123282A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018058086A1 (en) * 2016-09-26 2018-03-29 The University Of Toledo Synthetic lipopeptide vaccines and immunotherapeutics

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2906249B1 (en) * 2012-10-12 2018-06-27 MedImmune Limited Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
CN111760021B (en) * 2020-05-18 2023-06-16 广州中医药大学(广州中医药研究院) Conjugate containing alpha-galactosylceramide analogue and saccharide antigen, and preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2854896A1 (en) * 2003-05-16 2004-11-19 Mayoly Spindler Lab New surfactants used for preparation of liposomes and cosmetic and/or dermatological compositions comprise polar saccharidal heads, spacer group and lipophilic residue
WO2009025168A1 (en) * 2007-08-20 2009-02-26 Konica Minolta Holdings, Inc. Sugar-modified liposome containing compound for contrast agent and contrast agent

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5725871A (en) 1989-08-18 1998-03-10 Danbiosyst Uk Limited Drug delivery compositions comprising lysophosphoglycerolipid
US5707644A (en) 1989-11-04 1998-01-13 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5466468A (en) 1990-04-03 1995-11-14 Ciba-Geigy Corporation Parenterally administrable liposome formulation comprising synthetic lipids
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5756353A (en) 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
DE69312487T2 (en) 1992-05-18 1997-11-27 Minnesota Mining & Mfg DEVICE FOR TRANSMUCOSAL DELIVERY OF ACTIVE SUBSTANCES
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
IE80468B1 (en) 1995-04-04 1998-07-29 Elan Corp Plc Controlled release biodegradable nanoparticles containing insulin
US6248329B1 (en) * 1998-06-01 2001-06-19 Ramaswamy Chandrashekar Parasitic helminth cuticlin nucleic acid molecules and uses thereof
US6613308B2 (en) 2000-09-19 2003-09-02 Advanced Inhalation Research, Inc. Pulmonary delivery in treating disorders of the central nervous system
KR100484371B1 (en) 2001-10-25 2005-04-20 가부시키가이샤 아텍스 Method of manufacturing the sheet with heating wire
WO2003089574A2 (en) * 2002-04-15 2003-10-30 Biomira, Inc. Synthetic glyco-lipo-peptides as vaccines
CA2674269C (en) 2006-01-03 2018-06-12 University Of Georgia Research Foundation, Inc. Three component glycolipopeptides
US20120039984A1 (en) 2008-07-03 2012-02-16 University Of Georgia Research Foundation, Inc. Glycopeptide and uses thereof
AU2009298802A1 (en) * 2008-09-23 2010-04-08 Alnylam Pharmaceuticals, Inc. Chemical modifications of monomers and oligonucleotides with cycloaddition
US20100240773A1 (en) * 2009-03-23 2010-09-23 Kenneth Korzekwa Multifunctional linkers
CA2800911A1 (en) 2010-06-11 2011-12-15 University Of Georgia Research Foundation, Inc. Immunogenic vaccine

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2854896A1 (en) * 2003-05-16 2004-11-19 Mayoly Spindler Lab New surfactants used for preparation of liposomes and cosmetic and/or dermatological compositions comprise polar saccharidal heads, spacer group and lipophilic residue
WO2009025168A1 (en) * 2007-08-20 2009-02-26 Konica Minolta Holdings, Inc. Sugar-modified liposome containing compound for contrast agent and contrast agent

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Bardonnet, Pierre-Louis, et al., "Cholesteryl oligoethyleneglycol glycosides: Fluidizing effect of their embedment into phospholipid bilayers," Biochemical and Biophysical Research Communications (2005), 329(4), 1186-1192 *
BORONAT, Metabolism of L-Fucose and L-Rhamnose in Escherichia coli:Differences in Induction of Propanediol Oxidoreductase, JOURNAL OF BACTERIOLOGY, July 1981, p. 181-185 *
lipid -- Britannica Online Encyclopedia 8/28/2015 pages 1-22 *
Patent Translate 9/18/2016 19 pages *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018058086A1 (en) * 2016-09-26 2018-03-29 The University Of Toledo Synthetic lipopeptide vaccines and immunotherapeutics
US11007256B2 (en) * 2016-09-26 2021-05-18 The University Of Toledo Synthetic lipopeptide vaccines and immunotherapeutics

Also Published As

Publication number Publication date
EP2852413A1 (en) 2015-04-01
AU2013221448B2 (en) 2017-02-23
CA2869691A1 (en) 2013-08-22
WO2013123282A1 (en) 2013-08-22
US20170072037A1 (en) 2017-03-16
AU2017201649A1 (en) 2017-03-30
US10206988B2 (en) 2019-02-19
EP2852413A4 (en) 2016-04-27
AU2017201649B2 (en) 2018-05-17
AU2013221448A1 (en) 2014-09-11

Similar Documents

Publication Publication Date Title
CA2674269C (en) Three component glycolipopeptides
AU758097B2 (en) Trimeric antigenic O-linked glycopeptide conjugates, methods of preparation and uses thereof
AU750701B2 (en) Alpha-O-linked glycoconjugates, methods of preparation and uses thereof
EP2928902B1 (en) Conjugate compounds
Sarkar et al. Synthesis and immunological evaluation of a MUC1 glycopeptide incorporated into l-rhamnose displaying liposomes
US20150299290A1 (en) Immunogenic vaccine
EP2416801B1 (en) Immunogenic composition comprising a tlr ligand and an antigen
WO2006017180A2 (en) Glycopeptide dimers and uses thereof
US20110002983A1 (en) Compound
US10206988B2 (en) Xenoantigen-displaying anti-cancer vaccines and method of making
JP2006514981A (en) GP120-specific antigen, conjugate thereof; method for its preparation and use
Yang et al. Fully synthetic Tn-based three-component cancer vaccine using covalently linked TLR4 ligand MPLA and iNKT cell agonist KRN-7000 as built-in adjuvant effectively protects mice from tumor development
JP2021504439A (en) Vaccine against Klebsiella pneumoniae
US11007256B2 (en) Synthetic lipopeptide vaccines and immunotherapeutics
CN114126419A (en) Saponin-based vaccine adjuvants
US20050222398A1 (en) Glycopeptide conjugates and uses thereof
Pifferi et al. Development of synthetic, self-adjuvanting, and self-assembling anticancer vaccines based on a minimal saponin adjuvant and the tumor-associated MUC1 antigen
US20230321226A1 (en) Synthetic archaeal diether lipids
Sarkar Synthesis and study of anti-tumor vaccines
Karmakar Synthesis and study of MUC1-based anti-tumor vaccines

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNIVERSITY OF TOLEDO, OHIO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SUCHECK, STEVEN J.;WALL, KATHERINE A.;SARKAR, SOURAV;REEL/FRAME:033665/0835

Effective date: 20140903

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF TOLEDO;REEL/FRAME:035377/0712

Effective date: 20150408

STCB Information on status: application discontinuation

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