US20230130767A1 - Glycolipid compositions and methods of use - Google Patents

Glycolipid compositions and methods of use Download PDF

Info

Publication number
US20230130767A1
US20230130767A1 US17/965,573 US202217965573A US2023130767A1 US 20230130767 A1 US20230130767 A1 US 20230130767A1 US 202217965573 A US202217965573 A US 202217965573A US 2023130767 A1 US2023130767 A1 US 2023130767A1
Authority
US
United States
Prior art keywords
glycolipid
antigen
psa
canceled
composition
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.)
Pending
Application number
US17/965,573
Inventor
Sungwhan Oh
Deniz Erturk-Hasdemir
Dennis L. Kasper
Kailyn L. Stefan
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.)
Harvard College
Original Assignee
Harvard College
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 Harvard College filed Critical Harvard College
Priority to US17/965,573 priority Critical patent/US20230130767A1/en
Assigned to PRESIDENT AND FELLOWS OF HARVARD COLLEGE reassignment PRESIDENT AND FELLOWS OF HARVARD COLLEGE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STEFAN, KAILYN L., ERTURK-HASDEMIR, Deniz, KASPER, DENNIS L., OH, Sungwhan
Publication of US20230130767A1 publication Critical patent/US20230130767A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/739Lipopolysaccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7024Esters of saccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • 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
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H13/00Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids
    • C07H13/02Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by carboxylic acids
    • C07H13/04Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by carboxylic acids having the esterifying carboxyl radicals attached to acyclic carbon atoms
    • C07H13/06Fatty acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells

Definitions

  • the invention relates to glycolipids, compositions, methods of synthesis, isolation and/or purification, and methods of use thereof.
  • PSA Polysaccharide A of Bacteroides fragilis ( B. fragilis ) has been reported to be an immunomodulator with therapeutic and prophylactic activities.
  • PSA has been shown to provide protection from intestinal inflammatory diseases as well as systemic immune-mediated diseases such as the mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE).
  • EAE experimental autoimmune encephalomyelitis
  • PSA is one of eight capsular polysaccharides made by B. fragilis.
  • PSA was recently discovered to possess a lipid moiety.
  • the lipid moiety was hypothesized to anchor the polysaccharide in the B. fragilis outer membrane. It was also recently discovered that this “lipidated PSA” was more potent than non-lipidated PSA (referred to herein as “PSA”) forms provided in the prior art.
  • the invention is based, in part, on the identification and characterization of the glycolipid moiety that is found conjugated to PSA using certain isolation methods.
  • the invention is further premised, in part, on surprising finding that such glycolipid is able to stimulate the innate immune response and in particular activate antigen-presenting cells such as plasmacytoid dendritic cells. Based on this surprising finding, this disclosure contemplates and provides, among other things, use of the glycolipids as adjuvants in for example vaccine formulations.
  • the innate immunostimulatory property of the glycolipid is enhanced upon cleavage of the glycolipid moiety from the polysaccharide chain of PSA.
  • glycolipids of defined chemical structure as well as compositions comprising such glycolipids.
  • Such compositions may be further defined by the purity and/or concentration of glycolipids contained therein.
  • compositions of the glycolipids with antigens such as but not limited to bacterial antigens, including without limitation PSA and lipidated PSA.
  • such compositions comprise lipidated PSA and glycolipid in ratios ranging from 4:1 to 20:1 (w/w), or 4:1 to 15:1 (w/w), or 4:1 to 10:1 (w/w).
  • a glycolipid comprising a tri-acylated, tetra-acylated or penta-acylated diglucosamine, and the diglucosamine may be optionally conjugated to an oligosaccharide.
  • the glycolipid may be isolated intending that it is physically separated from other components including contaminants such as but not limited to components of B. fragilis cells. Isolation may also intend a degree of purity or concentration in a composition.
  • the glycolipid unless otherwise stated is unconjugated to the polysaccharide chain of PSA (i.e., the polysaccharide comprised of repeating tetrasaccharides found in PSA and lipidated PSA).
  • the acylated diglucosamine is conjugated to the oligosaccharide, such oligosaccharide is not the PSA polysaccharide, and such glycolipid is not PSA.
  • the glycolipid may be formulated with such polysaccharide chain of PSA or it may be formulated with lipidated PSA or it may be formulated with another antigen or another polysaccharide bearing moiety, although it will typically be provided in an unconjugated or free form.
  • examples of such formulations include for example micelles or liposomes or other delivery vehicles that are amenable to lipid based agents.
  • the diglucosamine is tetra-acylated or penta-acylated.
  • the tri-acylated, tetra-acylated or penta-acylated diglucosamine comprises acyl chains ranging in length from 14-17 carbons.
  • the tri-acylated, tetra-acylated or penta-acylated diglucosamine comprises acyl chains range in length from 15-17 carbons.
  • the glycolipid is substantially free of other components found in a B. fragilis capsule or found in B. fragilis cells. In some embodiments, the glycolipid is substantially free of LPS. In some embodiments, the glycolipid is substantially free of polysaccharide including for example the repeating tetrasaccharide polysaccharide of PSA.
  • the glycolipid is provided in purified form. In some embodiments, the glycolipid is provided an isolated form. In some embodiments, the glycolipid is provided in a micelle form or a liposome form, alone or optionally with other agents such as antigens, polysaccharides and the like. In some embodiments, the glycolipid is provided in a lyophilized form. Lyophilized forms glycolipid are particularly suitable for long-term storage, ranging from days, weeks, months or even years. In some embodiments, the glycolipid is provided in a form that is suitable for administration to a human, including an orally administered form or a parenterally administered form.
  • the glycolipid is obtained or derived from B. fragilis cells, including B. fragilis cells that overexpress PSA relative to polysaccharide B (PSB), PSC. PSD, PSE. PSF, PSG and PSH.
  • PSB polysaccharide B
  • obtained or derived intends that the glycolipid that is defined structurally herein is rendered using the isolation methods provided herein. It does not however intend that any naturally occurring counterpart of the glycolipid is structurally identical to the structure defined herein.
  • At least one acyl chain is unmodified. In some embodiments, at least one acyl chain is modified, for example modified with a hydroxyl group. In some embodiments, at least one acyl chain is unmodified and at least one of the acyl chains is modified, for example modified with a hydroxyl group. In some embodiments, at least one acyl chain is modified with a hydroxyl group. In some embodiments, at least one acyl chain is C16:0-OH. In some embodiments, at least one acyl chain is C17:0-OH. In some embodiments, at least one acyl chain is C14:0. In some embodiments, at least one acyl chain is C15:0.
  • At least one acyl chain is N-substituted. In some embodiments, at least one acyl chain is O-substituted. In some embodiments, at least one acyl chain is N-substituted and at least one acyl chain is O-substituted. In some embodiments, all of the acyl chains are N-substituted or all of the acyl chains are O-substituted.
  • the diglucosamine is phosphorylated. In some embodiments, the diglucosamine is mono phosphorylated. In some embodiments, the diglucosamine is not phosphorylated.
  • the diglucosamine is conjugated to the oligosaccharide by an acid-labile bond.
  • the acid-labile bond is a ketosidic bond.
  • the oligosaccharide comprises or contains 3-10 sugars (e.g., it may comprise 3 or more sugars).
  • the oligosaccharide however is not the polysaccharide of PSA (i.e., it is not the zwitterionic tetrasaccharide moiety or repeated moiety shown in FIG. 1 ).
  • the oligosaccharide contains 2-10, 2-9, 3-9, 4-9, 5-9, 6-9, 7-9 or 8-9 sugars.
  • the oligosaccharide contains 6-9 sugars.
  • the oligosaccharide comprises galactose and/or glucose and/or fucose residues.
  • the sugars of the oligosaccharide may be identical or different from each other.
  • the oligosaccharide comprises one or two KDO residues.
  • the oligosaccharide is not and does not comprise the repeating tetrasaccharide units of PSA.
  • the glycolipid is formulated with a detergent or a bile salt.
  • the detergent or bile salt is present at or less than 1%, 0.5% or 0.1% (weight of detergent or bile salt/weight of glycolipid).
  • the glycolipid is formulated with an antigen other than a lipidated PSA or a component of such lipidated PSA.
  • the glycolipid is formulated with vaccine additives.
  • the glycolipid is formulated with a non-naturally occurring preservative. In some embodiments, the glycolipid is formulated with a non-naturally occurring stabilizer. In some embodiments, the glycolipid is formulated with human albumin, phenol, glycerin or glycine.
  • the glycolipid is formulated with thimerosal, aluminum hydroxide, benzethonium chloride, formaldehyde, formalin, glutaraldehyde, potassium phosphate, aluminum potassium sulfate, bovine extract, calf serum, ammonium sulfate, aluminum phosphate, non-human cells. Vero (monkey kidney) cells, human cells, MRC-5 (human diploid) cells. MRC-5 cellular proteins, and the like.
  • the glycolipid is formulated for parenteral administration. In some embodiments, the glycolipid is formulated for oral administration.
  • compositions comprising any of the foregoing glycolipids.
  • the composition further comprises an antigen other than a lipidated PSA or a component of such lipidated PSA.
  • the composition comprises any of the foregoing glycolipids and lipidated PSA in non-naturally occurring weight/weight ratios such as but not limited to 1:75, 1:50, 1:25, 1:20, 1:10, 1:5, and the like.
  • the composition further comprises a detergent or a bile salt.
  • the detergent or bile salt is present at or less than 1%, 0.5% or 0.1% (weight of detergent or bile salt/weight of glycolipid).
  • the composition further comprises vaccine additives.
  • the composition further comprises a non-naturally occurring preservative. In some embodiments, the composition further comprises a non-naturally occurring stabilizer. In some embodiments, the composition further comprises human albumin, phenol, glycerin or glycine.
  • the composition further comprises thimerosal, aluminum hydroxide, benzethonium chloride, formaldehyde, formalin, glutaraldehyde, potassium phosphate, aluminum potassium sulfate, bovine extract, calf serum, ammonium sulfate, aluminum phosphate, non-human cells, Vero (monkey kidney) cells, human cells, MRC-5 (human diploid) cells, MRC-5 cellular proteins, and the like.
  • any of the foregoing glycolipids or compositions is formulated for parenteral administration. In some embodiments, any of the foregoing glycolipids or compositions is formulated for oral administration.
  • composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids in a 4:1 to 20:1 ratio (w/w).
  • PSA lipidated polysaccharide A
  • composition comprising a lipidated polysaccharide A (PSA) and at least 0.5% (w/w) of any of the foregoing glycolipids.
  • PSA lipidated polysaccharide A
  • the composition further comprises a pharmaceutically acceptable carrier.
  • the composition further comprises another therapy such as an additional active agent or it is formulated for administration in combination with another therapy such as an additional active agent.
  • the other therapy is or comprises a cancer immunotherapy.
  • the cancer immunotherapy comprises administration of an antibody or antibody fragment thereof.
  • the composition is formulated for parenteral administration. In some embodiments, the composition is formulated for oral administration.
  • composition comprising any one of the foregoing glycolipids and an antigen.
  • the composition is a vaccine.
  • the antigen is a bacterial antigen. In some embodiments, the antigen is attenuated bacteria. In some embodiments, the antigen is a viral antigen. In some embodiments, the antigen is attenuated virus. In some embodiments, the antigen is a fungal antigen. In some embodiments, the antigen is a mycobacterial antigen. In some embodiments, the antigen is a parasitic antigen. In some embodiments, the antigen is a cancer antigen. In some embodiments, the antigen is a human protein. In some embodiments, the antigen is a human polysaccharide. In some embodiments, the composition is formulated for parenteral administration.
  • Also provided herein is a method of enhancing an innate immune response comprising administering, to a subject in need of an immune response, such as an enhanced immune response, and further such as an enhanced innate immune response, an effective amount of any of the foregoing glycolipids.
  • the method further comprises administering a lipidated PSA in combination with another therapy.
  • the other therapy is or comprises a cancer immunotherapy.
  • the cancer immunotherapy comprises administration of an antibody or antibody fragment thereof.
  • Also provided herein is a method of inducing an immune response comprising administering, to a subject in need of immune response induction or modulation, an effective amount of any of the foregoing glycolipids.
  • the method further comprises administering a lipidated PSA in combination with another therapy.
  • the other therapy is or comprises a cancer immunotherapy.
  • the cancer immunotherapy comprises administration of an antibody or antibody fragment thereof.
  • the immune response is an innate immune response. In some embodiments, the immune response is an adaptive immune response.
  • Also provided herein is a method of inducing an immune response comprising administering, to a subject in need of immune response induction or modulation, an effective amount of any of the foregoing glycolipids, wherein the immune response comprises induction of interferon-beta, and the subject is experiencing or is likely to experience an interferon-beta responsive condition (e.g., a condition that has been shown to benefit from administration of exogenous interferon-beta and/or that can benefit from induction of interferon-beta in vivo).
  • an interferon-beta responsive condition e.g., a condition that has been shown to benefit from administration of exogenous interferon-beta and/or that can benefit from induction of interferon-beta in vivo.
  • Such conditions include but are not limited to any of the autoimmune diseases provided herein.
  • the condition is multiple sclerosis (MS).
  • the condition is systemic lupus erythematosus.
  • Also provided herein is a method of inducing an immune response to an antigen comprising administering, to a subject in need of an antigen-specific immune response, an effective amount of any of the foregoing glycolipid and the antigen.
  • the subject has or is at risk of developing a bacterial infection.
  • the antigen is a bacterial antigen. In some embodiments, the antigen is attenuated bacteria.
  • the subject has or is at risk of developing a viral infection.
  • the antigen is a viral antigen. In some embodiments, the antigen is attenuated virus.
  • the subject has or is at risk of developing a fungal infection.
  • the antigen is a fungal antigen.
  • the subject has or is at risk of developing a mycobacterial infection.
  • the antigen is a mycobacterial antigen.
  • the subject has or is at risk of developing a parasitic infection.
  • the antigen is a parasitic antigen.
  • the subject has or is at risk of developing a cancer.
  • the antigen is a cancer antigen.
  • the antigen is a human protein. In some embodiments, the antigen is a human polysaccharide.
  • the method further comprises administering to the subject lipidated PSA.
  • a micelle consisting essentially of any one or any combination of the foregoing glycolipids.
  • compositions comprising a micelle consisting essentially of any one or any combination of the foregoing glycolipids and a detergent or bile salt.
  • the detergent or bile salt is present in a pharmaceutically acceptable amount.
  • the composition is a pharmaceutical composition. In some embodiments, formulated for parenteral administration.
  • the glycolipid is free of non-lipidated PSA. In various embodiments, the glycolipid is free of lipidated PSA.
  • the glycolipid is suitable for administration to a human.
  • the composition is formulated for parenteral or enteral or oral administration to a subject.
  • the composition is formulated for lipophilic delivery, including for example in a liposome or in an oil-based delivery system.
  • the various compositions provided herein may be formulated as a capsule or other discrete dosage form, including those intended for oral or enteral administration.
  • the invention provides an isolated glycolipid comprising a diglucosamine covalently conjugated to 3-5 acyl chains, each independently ranging in length from 14-17 carbons.
  • the glycolipid may be any of the glycolipids provided herein, or a combination thereof.
  • the diglucosamine is covalently conjugated to 3-5 acyl chains.
  • the diglucosamine is covalently conjugated to 4 or 5 acyl chains.
  • the acyl chains range in length from 15-17 carbons. Other embodiments relating to the glycolipids are recited above and apply equally.
  • glycolipids that comprise, consist or consist essentially of a disaccharide, such as a diglucosamine, covalently conjugated to 3-5 acyl chains, and optionally each chain may independently range in length from 4-17 carbons.
  • a disaccharide such as a diglucosamine
  • the invention provides a method comprising administering to a subject in need of immune response modulation a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids in an effective amount.
  • a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids in an effective amount.
  • PSA lipidated polysaccharide A
  • the invention provides a method comprising administering to a subject in need of immune response modulation an effective amount of a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids, in combination with an additional therapy.
  • PSA lipidated polysaccharide A
  • the invention provides a method comprising administering to a subject in need of immune response modulation one or both of a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids; and another therapy, so that the subject receives therapy with the composition and the other therapy in combination.
  • a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids
  • PSA lipidated polysaccharide A
  • the subject has or is at risk of developing cancer. In some embodiments, the subject is diagnosed as suffering from, susceptible to, and/or receiving therapy for cancer.
  • the additional therapy is or comprises a cancer immunotherapy.
  • the cancer immunotherapy is or comprises an antibody or antibody fragment thereof.
  • the step of administering comprises administering according to a regimen that has been demonstrated to achieve statistically significant improvement in immune function when administered to a relevant population of subjects.
  • the relevant population of subjects may comprise subjects diagnosed as suffering from, susceptible to, and/or receiving therapy for cancer.
  • the improvement in immune function comprises modulation of an innate and/or adaptive immune response.
  • the subject in need of immune response modulation is a subject in need of innate immune response modulation. In some embodiments, the subject in need of immune response modulation is a subject in need of adaptive immune response modulation. In some embodiments, the subject in need of immune response modulation is a subject in need of immune response stimulation.
  • improvement in immune function comprises stimulation of an innate and/or adaptive immune response.
  • the step of administering comprises administering an amount of the composition effective to achieve an improvement in immune function as compared to a subject who has not been administered the composition.
  • the acylated diglucosamine such as but not limited to the tetra-acylated diglucosamine or the penta-acylated diglucosamine, may be administered to a subject experiencing or likely to experience a proinflammatory immune response, such as may occur for example during an E. coli infection or an infection by gram negative bacteria.
  • the acylated diglucosamine may be conjugated to an oligosaccharide such as 3-10-mer oligosaccharide.
  • the subject may be defined as one experiencing or likely to experience a gram negative bacterial infection, including but not limited to infection by any of the gram negative bacterial species provided herein.
  • FIG. 1 is a proposed generic structure of isolated B. fragilis lipidated PSA.
  • the structure includes a repeating tetrasaccharide unit having an average molecular weight of about 130 KDa, an oligosaccharide core unit comprising on average less 10 or fewer sugar residues, and a glycolipid.
  • the glycolipid may comprise 3-5 acyl chains that may be N or O linked to a diglucosamine.
  • the diglucosamine may be mono phosphorylated or non-phosphorylated.
  • FIG. 2 is one embodiment of a proposed generic structure of isolated B. fragilis glycolipid antigen (referred to herein as GLA) with no repeating tetrasaccharide attached.
  • GLA B. fragilis glycolipid antigen
  • the structure includes a core oligosaccharide unit attached to a diglucosamine-comprising glycolipid moiety having 5 acyl chains. One glucosamine is phosphorylated.
  • the linkage between the diglucosamine and the core oligosaccharide unit is an acid-labile ketosidic linkage. When the oligosaccharide core unit is linked to the repeating tetrasaccharide units of PSA (not shown), this occurs through a glycosidic linkage.
  • the oligosaccharide core unit may comprise galactose, glucose or fucose sugars. It may further contain KDO residues that connect the diglucosamine to the remaining sugars of the core unit. As illustrated, it comprises 6 galactose units conjugated to 2 KDO residues conjugated, through a ketosidic linkage, to a diglucosamine conjugated to 5 acyl chains.
  • the diglucosamine is mono phosphorylated.
  • the glycolipid may be non-phosphorylated
  • the diglucosamine may be tri- or tetra-acylated, and there may be variation in the oligosaccharide core unit in terms of number and nature of sugars. Accordingly.
  • GLA may minimally comprise the diglucosamine linked to a plurality of acyl chains (e.g., 3-5 chains).
  • FIG. 3 is a bar graph showing that B. fragilis GLA activates TLR2 similar to lipidated PSA.
  • the graph further shows that GLA can activate TLR2 at much lower concentrations as compared to lipidated PSA.
  • the graph shows IL-8 production in HEK cells that are stably transfected with TLR2 only (while, or first bar of each pair of bars) or transfected with TLR2 and TLR1 (diagonal striped, or second bar of each pair of bars) in response to purified lipidated PSA that does not contain any free lipid (referred to as PSA39) and B. fragilis GLA alone.
  • HEK cells over-expressing TLR2 and TLR2/l were stimulated with PSA39 or B.
  • frag GLA for 24 hours and IL-8 levels in the culture supernatant were measured with ELISA.
  • the Figure shows that far less (e.g., about 20 fold less to about 100 fold less) GLA preparation is needed to stimulate the same level of IL-8 expression from the H11EK cells.
  • the 100 ng/ml of GLA induces about 100-fold more IL-8 than does the 10 microgram/ml of PSA39, even though the latter dose contains about 100 nanograms of GLA.
  • TLR2 activation plays a role in innate immunity, so the ability of GLA to stimulate TLR2 indicates that GLA is able to stimulate innate immunity.
  • the ability of lipidated PSA (PSA39) to stimulate TLR2 may be due to its glycolipid moiety which comprises only about 1% of the mass of the lipidated PSA.
  • FIGS. 4 A- 4 C show that PSA with higher levels of GLA provides better protection in an EAE model than PSA alone.
  • the Figures show that a preparation comprising lipidated PSA and GLA (at an approximately 4:1 ratio, w/w, referred to herein as Lot 35L) protects in an EAE model system better than purified lipidated PSA lacking any free glycolipid (Lot 39).
  • C57BLU6 mice were orally administered purified lipidated PSA (Lot 39), or a preparation comprising lipidated PSA and “free” GLA (Lot 35L), or PBS every other day starting from one week before inducing the disease. Mice were challenged subcutaneously with MOG 35-55 in Complete Freund's Adjuvant.
  • FIG. 4 A shows the clinical EAE score of the mice, with a score of 5 indicating death.
  • FIG. 4 B shows the % weight loss of the mice. Significance of the results using Lot 35L to Lot 39 or to PBS is indicated by white asterisks (Lot 39 (8/8) and grey asterisks (PBS (8/8)), respectively.
  • FIG. 4 C shows the percent survival of the mice. Lot 35L, which contains more free GLA as compared to Lot 39, reduced the severity of the disease and the cumulative disease score, and increased survival rate of the mice. The presence of free GLA enhanced the protective activity of lipidated PSA.
  • FIG. 5 provides various metrics using the same EAE mouse model as described for FIGS. 4 A- 4 C to show that PSA with higher levels of GLA provides better protection in the EAE model than PSA alone.
  • the statistical significance of the three parameters in the top row and the parameter in lower left were all p ⁇ 0.01.
  • the upper left plot shows the cumulative disease scores calculated as the sum of all EAE clinical scores for each group of mice.
  • Purified lipidated PSA is indicated as 39 and a mixture of purified lipidated PSA and free GLA, in a roughly 4:1 w/w mixture, is indicated as 35L.
  • about 100 micrograms of lipidated PSA (39) were used per mouse.
  • about 100 micrograms of the 35L preparation were used per mouse, corresponding to about 80 micrograms of lipidated PSA and 20 micrograms of GLA.
  • FIGS. 6 A- 6 D show that GLA acts as an adjuvant for lipidated PSA.
  • the experiment was performed as explained for FIGS. 4 A- 4 C .
  • Lot 39 refers to purified lipidated PSA.
  • GLA alone refers to purified GLA.
  • Lot 39 and GL refers to a preparation made by combining Lot 39 and GLA in roughly a 4:1 w/w ratio. Similar results were obtained using a ratio of about 100:1.
  • FIG. 6 A shows the clinical EAE score of mice subjected to MOG-peptide induced EAE (p ⁇ 0.01). Significance of the results using Lot 39 and GL together compared to GLA alone or Lot 39 alone is indicated by asterisks and circled asterisks (Lot 39), respectively, in FIG. 6 A .
  • FIG. 6 B shows the % weight loss in the mice (p ⁇ 0.05).
  • FIG. 6 C shows the percent survival of the mice. GLA alone failed to confer protection, while a reconstituted mixture of purified lipidated PSA and GLA (at a 4:1 w/w ratio) protected animals from EAE.
  • FIG. 6 D shows the cumulative disease scores calculated as the sum of all EAE clinical scores for each group.
  • FIGS. 7 A- 7 B show that a delipidated PSA preparation (referred to as PSA35 delip) cannot protect in EAE.
  • PSA35 refers to a preparation that comprises lipidated PSA and free GLA.
  • PSA35 delip refers to a preparation of PSA35 that has been acid hydrolyzed to cleave the lipid moiety from lipidated PSA and has also undergone a lipid extraction with organic solvent chloroform in order to remove all free lipid including all released lipid.
  • PSA35 delip likely comprises the repeating tetrasaccharide polysaccharide chain of PSA conjugated to the oligosaccharide core unit. It lacks the diglucosamine-containing glycolipid. The experiment was performed as explained in FIGS.
  • FIG. 7 B shows the cumulative scores calculated as the sum of all EAE clinical scores for each group. Delipidated PSA35 failed to confer protection, while PSA35 protected animals from EAE indicating the importance of the glycolipid in such protection. In these experiments, on the order of about 10 micrograms of the PSA35 preparation was administered per mouse, representing about 8 micrograms of lipidated PSA and about 2 micrograms of GLA. This amount was about 10-fold lower than the amounts used in the experiments of the preceding Figures.
  • FIG. 8 is a schematic of a model of lipidated PSA induced immunomodulation.
  • free GLA are shown to bind to a TLR2/1 dimer via the acyl chains, thereby stimulating an innate immune response.
  • the free GLA may comprise the oligosaccharide core unit.
  • FIG. 9 shows the lipid profile of various lots of PSA (PSA39. PSA40 and PSA41) and GLA (GLA acap which was derived from an unencapsulated mutant and GLA41). Each lot was tested for the presence of tri-acylated, tri-acylated and phosphorylated, tetra-acylated, tetra-acylated and phosphorylated, penta-acylated, and penta-acylated and phosphorylated glycolipid moieties. The bars represent the amount of each glycolipid moiety as a percentage of the total lipid (y-axis). It is clear that each preparation contains the same lipid species but in different percentages. In particular, PSA41 and GLA41 contain the same lipid species, although the lipids do not appear to be similarly distributed between the PSA and GLA lots.
  • FIG. 10 shows an in vitro dose response (IL-10 production) of SpDC+Tcells co-cultured with dendritic cells and exposed to 10, 50 and 250 mg/ml of PSA39, PSA40 and PSA41.
  • PSA41 appears more potent in its ability to induce IL-10 as compared to other two preparations.
  • FIG. 11 shows that interferon- ⁇ is induced by B. fragilis glycolipid whether or not attached to PSA.
  • Model antigen presenting cells in the form of bone marrow derived dendritic cells (BMDCs) were stimulated for 6 hours with B. fragilis polysaccharide A (PSA) or the purified glycolipid anchor (GLA unencapsulated mutant), followed by collection of supernatants.
  • PSA B. fragilis polysaccharide A
  • GLA unencapsulated mutant purified glycolipid anchor
  • a cytokine ELISA was used to detect levels of interferon- ⁇ (IFN ⁇ ) in the supernatants.
  • PSA doses are shown stoichiometrically. The two curves would be superimposed if plotted on a molar basis because GLA is about 1% of a PSA molecule (i.e., 100 micrograms of PSA contain about 1 microgram of GLA).
  • FIG. 12 shows that different acylation structures of B. fragilis GLA induce low levels of pro-inflammatory cytokines such as TNF ⁇ .
  • Model antigen presenting cells in the form of bone marrow derived dendritic cells (BMDCs) were stimulated for 6 hours with tri-, tetra- or penta-acylated B. fragilis GLA or hexa-acylated E. coli lipid A, followed by collection of supernatants.
  • a cytokine ELISA was used to detect levels of TNF ⁇ in the supernatants.
  • the three GLA species differ in their ability to induce TNF ⁇ , but they all induce a much lower level of TNF ⁇ as compared to E. coli lipid A.
  • FIG. 13 shows that tetra-acylated and penta-acylated B. fragilis lipid A antagonize production of pro-inflammatory TNF ⁇ secretion by E. coli hexa-acylated lipid A.
  • Model antigen presenting cells in the form of bone marrow derived dendritic cells (BMDCs) were stimulated for 6 hours with purified tetra-acylated B. fragilis GLA and hexa-acylated E. coli lipid A (in a 5:1 ratio) or with purified penta-acylated B. fragilis GLA and hexa-acylated E. coli lipid A (in a 5:1 ratio), followed by collection of supernatants.
  • BMDCs bone marrow derived dendritic cells
  • a cytokine ELISA was used to detect levels of TNF ⁇ in the supernatants. Both the tetra-acylated and penta-acylated GLA were able to suppress the pro-inflammatory response to lipid A. This suggests a suppressive activity that may be useful, among other things, in treating gram negative bacterial infection.
  • compositions and methods of use relating to a glycolipid antigen comprising a diglucosamine-containing glycolipid minimally comprises a diglucosamine-containing glycolipid.
  • the diglucosamine moiety may be mono phosphorylated or non-phosphorylated (e.g., dephosphorylated).
  • the glycolipid may comprise 3-5 acyl chains that are N- or O-linked to the diglucosamine.
  • the glycolipid may further be conjugated to an oligosaccharide, referred to herein as an oligosaccharide core unit.
  • an oligosaccharide may comprise 10 or fewer sugars.
  • the sugars may be galactose, glucose and/or fucose.
  • the oligosaccharides are typically conjugated to the diglucosamine via an acid-labile linkage such as but not limited to a ketosidic linkage.
  • glycolipid is able to stimulate an innate immune response.
  • This activity has been heretofore unrecognized.
  • the finding is significant since it indicates that lipidated PSA is able to stimulate both innate and adaptive immune responses via different portions of its structure.
  • free glycolipid when combined with lipidated PSA, leads to enhanced protective activity in EAE mouse models. This suggests that free glycolipid is able to enhance immune responses to a greater degree than possible when used in its conjugated form.
  • This disclosure provides methods of use of this glycolipid as an adjuvant together with PSA, whether or not lipidated, as well as a variety of other antigens.
  • This newly discovered adjuvant activity indicates a role of the glycolipid in enhancing innate immunity, including when used alone, as well as in enhancing adaptive immunity when used together with an antigen. Examples of this latter application include use as an adjuvant for vaccines, and well as in immuno-oncology.
  • the glycolipid has been obtained from manipulation of B. fragilis cells.
  • the starting material was a non-naturally, mutant occurring strain of B. fragilis that overexpresses PSA but not other capsular polysaccharides.
  • the purification protocol may include a phenol/water extraction, and DNAse. RNase and pronase treatments. If the material is then treated with deoxycholate (DOC), the glycolipid comprising the oligosaccharide core unit is obtained. DOC treatment therefore separates the glycolipid from lipidated PSA.
  • DOC deoxycholate
  • the material is treated instead with acid hydrolysis, the acid-labile linkage between the oligosaccharide core unit and the acyl-substituted diglucosamine is cleaved and the free acyl-substituted diglucosamine is obtained.
  • a purification is performed that does not involve either DOC treatment or acid hydrolysis (e.g., column chromatography), then the resulting preparation comprises about 20% (w/w) free glycolipid and about 80% (w/w) PSA including lipidated PSA.
  • 35L or PSA35L or Lot 35L is referred to as 35L or PSA35L or Lot 35L.
  • a “free” glycolipid refers to a glycolipid that is not covalently attached to a tetrasaccharide unit of PSA (i.e., it does not comprise the polysaccharide portion of PSA that is comprised of repeating tetrasaccharide units).
  • GLA comprises a glucosamine disaccharide with 3 to 5 acyl chains covalently bound to the sugars. It can be a mono phosphorylated or non-phosphorylated at the diglucosamine.
  • One of the glucosamine residues is linked to an oligosaccharide core of 10 or fewer sugars, including 9, 8, 7 or less sugars, by an acid labile bond such as a ketosidic bond.
  • lipidated PSA preparations identified herein as 35L or PSA35L. These preparations were further analyzed and found to comprise on average about a 4:1 ratio of lipidated PSA to free GLA (w/w).
  • PSA39 purified lipidated PSA preparation referred to herein as PSA39 (DOC treated lipidated PSA preparation that did not contain free lipid ( FIG. 4 A , FIG. 5 upper left corner, and FIG. 4 C ).
  • GLA acts as an adjuvant to induce innate immunity. Such activity may occur through engagement and signaling of the TLR2/TLR1 heterodimer complex, and it likely enhances immune responses to PSA.
  • the findings indicate use of GLA in enhancing immune responses to other antigens including but not limited to bacterial antigens, cancer antigens, and the like.
  • glycolipids are comprised of a diglucosamine substituted with three, four or five acyl chains.
  • the glycolipid in some instances further comprises an oligosaccharide core unit conjugated to the diglucosamine via an acid-labile ketosidic bond. This glycolipid has been shown to activate an innate immune response and to enhance an immune response to lipidated PSA.
  • glycolipid structure will now be described in greater detail in the context of lipidated PSA.
  • a form of lipidated PSA isolated from mutant B. fragilis cells comprises a polysaccharide component conjugated, at its reducing end, an oligosaccharide core unit, and a diglucosamine conjugated to 3-5 acyl chains.
  • the innate immunostimulatory activity appears attributable to the diglucosamine substituted with acyl chains.
  • the oligosaccharide core unit by virtue of its hydrophilicity, helps to render the lipid component more water soluble.
  • the GLA of this disclosure will further comprise the oligosaccharide core unit in order to facilitate its dissolution in an aqueous solution, such as but not limited to a pharmaceutical formulation.
  • the polysaccharide component of lipidated PSA comprises a tetrasaccharide repeating unit shown below. It possesses zwitterionic behavior as conferred by a positive charge on its free amine group and a negative charge on its free carboxyl group (per repeating tetrasaccharide unit). Its naturally occurring state has been reported to comprise over 60 tetrasaccharide repeating units (e.g., up to and including in some instances about 100, or about 200, or about 300 repeated units on average), and it has an average molecular size of about 150 kD (with a range of about 75 kD to 240 kD).
  • the repeating tetrasaccharide unit of PSA has a structure as follows:
  • the tetrasaccharide repeating unit may also be expressed as follows:
  • Synthetic forms of lipidated PSA may comprise comprising various ranges of tetrasaccharide units (e.g., 1-60, 1-50, 1-40, 1-30, 1-20, 1-10, 1-9, 1-8, 1-7, 1-6, or 1-5 tetrasaccharide units).
  • Such shorter variants can be obtained by depolymerizing naturally occurring lipidated PSA or by depolymerizing PSA obtained from lipidated PSA.
  • PSA can be depolymerized using for example chemical means (e.g., using reactive oxygen species or reactive nitrogen species such as but not limited to nitrogen monoxide, as described in Duan and Kasper, Glycobiology, 2011, 21(4):401-409), mechanical means, and/or enzymatic means that are known in the art.
  • the invention further contemplates synthetic forms of lipidated PSA comprising more than 300 repeating tetrasaccharide units, including without limitation 350, 400, 500, 600, 700, 800, 900 or 1000 units or more.
  • the polysaccharide component may be covalently conjugated to the glycolipid, or in certain synthetic forms it may be unconjugated to the glycolipid. If covalently conjugated, it may be conjugated via a glycosidic bond to the oligosaccharide core unit. In other embodiments, it may be conjugated via a ketosidic bond or other acid labile bond or via a bond such as an ester, an amide, or an ether bond to form a non-naturally occurring lipidated PSA.
  • the glycolipid component comprises a diglucosamine substituted with one or more acyl chains, and preferably 3, 4 or 5 acyl chains.
  • An exemplary diglucosamine in the context of a glycolipid is provided in FIG. 2 . It is now recognized in accordance with this disclosure that the diglucosamine is conjugated to the oligosaccharide core unit via a ketosidic bond that is acid-labile and thus susceptible to stringent hydrolysis.
  • the disaccharide may be conjugated to one or more acyl chains, including three, four, or five acyl chains in some instances via for example ester or amide linkages, and thus may be referred to as “O” substituted (or O-linked) or “N” substituted (or N-linked) respectively.
  • each glycolipid or GLA, as used herein
  • the glycolipid may be referred to herein as tri-acylated, tetra-acylated or penta-acylated forms.
  • the acyl chains may range in length from 14 to 17 carbons, in some instances.
  • the acyl chains may be unmodified or they may be modified. If modified, the acyl chains may be hydroxy-modified.
  • the glycolipid may comprise one or more acyl chains characterized as C14:0, C14:0-OH, C15:0, C15:0-OH, C16:0, C16:0-OH, C17:0, and C17:0-OH.
  • a single preparation of glycolipid may yield a number glycolipid species that diffe from each other with respect to acyl chain number.
  • the exact composition of a glycolipid may be determined using mass spectrometry (MS), wherein different glycolipid species give rise to different and discernable spectra.
  • penta-acylated species examples include:
  • glycolipids of this disclosure may comprise any of the foregoing combinations of acyl chains, without limitation:
  • the number of each type of chain may vary, and may include without limitation the following options
  • the invention provides defined glycolipid mixtures, having known, and thus optionally pre-defined, glycolipid content and composition, and optionally known and/or pre-defined polysaccharide (e.g., PSA) to glycolipid ratios or non-PSA antigen to glycolipid ratios.
  • polysaccharide e.g., PSA
  • glycolipids of this disclosure may be characterized in terms of any of these structural features, thereby further distinguishing these products from those of the prior art, and where necessary from naturally occurring products.
  • compositions comprising glycolipids that are only or predominantly (e.g., greater than 50%, or at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%) tri-acylated, or tetra-acylated, or penta-acylated, or some combination thereof including but not limited to tetra- and penta-acylated.
  • Such chemically defined compositions were not heretofore contemplated or possible.
  • the preparations provided herein can be characterized by their content of released or free glycolipids.
  • Such content can be about 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% or less.
  • Such content can be less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, less 0.1%, less than 0.05%, less than 0.001%, less than 0.0005%, less than 0.0001% (w/w of released glycolipid to lipidated PSA).
  • the amount of glycolipid may be determined for example using the gel electrophoresis methods or mass spec methods.
  • the glycolipid may also be considered to be pure (i.e., it is free or substantially free of contaminant).
  • the degree of purity may be at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%, or higher.
  • compositions comprising glycolipids including compositions comprising isolated glycolipid at a purity and/or a concentration that has not been heretofore achieved.
  • compositions comprising or consisting essentially of particular species of glycolipid or particular subsets of species of glycolipid. These species may be characterized and thus distinguished from other species and from bulk glycolipid in terms of their lipid and oligosaccharide components.
  • the lipid components may be characterized by the number, position and type of acyl chains they possess.
  • a composition may comprise at least 70%, 75%, 80%, 85%, 90%, 95%, or mom of a tri-acetylated, tetra-acylated and/or penta-acylated glycolipid.
  • compositions may be defined by their degree of purity, for example with respect to their glycolipid components, or with respect to their content of contaminants such as non-lipidated PSA.
  • compositions may be defined by their concentration of glycolipid components, or by their ratio of glycolipid to other components.
  • the invention further provides additional synthetic, non-naturally occurring species of glycolipids.
  • Isolated intends that the glycolipid (or other agent) is provided in a more pure form or a more concentration form compared to its synthesized or obtained form.
  • An “isolated” glycolipid may be a glycolipid that is prepared or obtained from B. fragilis , and is physically separated from a natural environment (e.g., a B. fragilis cell, components of the B. fragilis cell, and/or components of the B. fragilis cell capsular complex such as but not limited to PSB). It does not intend that any naturally occurring product has the same structure of glycolipid as described herein.
  • the compositions are substantially free of naturally occurring contaminants such as nucleic acids (e.g., DNA and RNA), proteins, and other components of B. fragilis and/or the B. fragilis capsule.
  • substantially free intends that these contaminants represent about or less than 5%, less than 1%, less than 0.5%, or less than 0.1% (or less) by weight (weight of the contaminant to weight of the glycolipid). In some instances, such contaminants may be undetectable.
  • compositions may or may not contain LPS.
  • LPS may be present in an amount of about 0.5% (w/w of LPS relative to other components in a composition).
  • compositions may comprise at least about 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more (w/w) of glycolipid.
  • compositions may comprise at least about 95%, 96%, 97%, 98%, 99%, or more (w/w) of lipidated PSA and less than 5%, less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, or less of free, released glycolipid.
  • the various glycolipid forms herein can be used as immunomodulators including immunostimulators, particularly in view of their enhanced ability to stimulate antigen presenting cells such as plasmacytoid dendritic cells. These forms are contemplated for use in vitro and in vivo.
  • In vitro uses include use as an analytical tool (e.g., for screening assays) and as an assay standard or control (e.g., as a positive control or a comparator in an in vitro assay such as a IL-8 induction assay).
  • In vivo uses include uses in animal models and also in human and non-human subjects to treat or prevent conditions that would benefit from such immunomodulation including immunostimulation. Such conditions include those in which an enhanced innate and/or adaptive immune response is beneficial. Other conditions include but are not limited to autoimmune disorders (e.g., multiple sclerosis and inflammatory bowel disease).
  • the glycolipid component may be used as a single agent, or it may be used in combination with other agents such as antigens or cells such as antigen-presenting cells. It may be used to stimulate antigen-presenting cells in vivo or in vitro.
  • One application may involve obtaining antigen-presenting cells from a subject, stimulating such cells ex vivo with the glycolipid of this disclosure with or without antigen, followed by re-introduction of such cells with and without glycolipid and/or antigen to the subject.
  • the glycolipid may be administered to a subject with and/or without antigen.
  • Such immunization or vaccination protocols may involve several administrations of glycolipid and/or antigen.
  • the administration of the glycolipid and the antigen may occur simultaneously (typically occurs if they are formulated together), or substantially simultaneously (typically occurs if they are formulated and administered separately but still within a short period of time in order to work in concert in a subject), and/or they may be formulated and administered separately (e.g., the glycolipid is administered before or after the antigen, including for example hours, days and/or weeks before or after).
  • the invention further contemplates use of the free polysaccharide component of lipidated PSA (i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid component of lipidated PSA (including where such glycolipid comprises the oligosaccharide core unit) and the free glycolipid component obtained from lipidated PSA.
  • the free polysaccharide component of lipidated PSA i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid component of lipidated PSA (including where such glycolipid comprises the oligosaccharide core unit) and the free glycolipid component obtained from lipidated PSA.
  • polysaccharide and glycolipid may be used together in an unconjugated form.
  • the invention further contemplates use of the free PSA (i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid (including where such glycolipid comprises the oligosaccharide core unit) and free glycolipid obtained from lipidated PSA.
  • the free PSA i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid (including where such glycolipid comprises the oligosaccharide core unit
  • free glycolipid obtained from lipidated PSA i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid (including where such glycolipid comprises the oligosaccharide core unit
  • In vivo uses include but are not limited to those involving human subjects.
  • in vivo uses include administration of the glycolipid and compositions thereof to a non-human subject in order to modulate an immune response, for example as a positive control or a comparator.
  • the glycolipids and compositions thereof may be administered to a subject having or at risk of having an infection such as a bacterial infection, a viral infection, a fungal infection, a parasitic infection, or a mycobacterial infection.
  • the glycolipids may be used and/or administered with another therapeutic such as an anti-bacterial agent, an anti-viral agent, an anti-fungal agent, an anti-parasitic agent, or anti-mycobacterial agent.
  • the glycolipids may be used and/or administered with an antigen in order to induce or enhance an immune response directed to the antigen (i.e., an antigen-specific immune response).
  • antigens include bacterial antigens, viral antigens, fungal antigens, parasitic antigens, and mycobacterial antigens.
  • Anti-bacterial agents include broad spectrum antibiotics, narrow spectrum antibiotics, or limited spectrum antibiotics.
  • the anti-bacterial agent may also be a cell wall synthesis inhibitor, cell membrane inhibitor, protein synthesis inhibitor, nucleic acid synthesis or functional inhibitor or a competitive inhibitor.
  • Anti-viral agents include immunoglobulin, amantadine, interferon, nucleoside analogue, nonnucleoside analogue, bioflavonoid and protease inhibitor, although it is not so limited.
  • the protease inhibitor is indinavir, saquinavir, ritonavir, and nelfinavir.
  • the bioflavonoid is robustaflavone, amentoflavone, or a derivative or salt thereof.
  • the non-nucleoside analogue is selected from the group consisting of delavirdine, nevirapine, efavirenz, alpha-interferon, recombinant CD4, amantadine, rimantadine, ribavirin and vidarabine.
  • Anti-fungal agents include imidazole. FK 463, amphotericin B. BAY 38-9502, MK 991, pradimicin, UK 292, butenafine, chitinase and 501 cream.
  • Anti-parasitic agents include albendazole, amphotericin B, benznidazole, bithionol, chloroquine HCl, chloroquine phosphate, clindamycin, dehydroemetine, diethylcarbamazine, diloxanide furoate, eflornithine, furazolidaone, glucocorticoids, halofantrine, iodoquinol, ivermectin, mebendazole, mefloquine, meglumine antimoniate, melarsoprol, metrifonate, metronidazole, niclosamide, nifurtimox, oxamniquine, paromomycin, pentamidine isethionate, piperazine, praziquantel, primaquine phosphate, proguanil, pyrantel pamoate, pyrimethamine-sulfonamides, pyrimethamine-s
  • Anti-mycobacterial agent include anti-tuberculosis agent such as isoniazid, rifampin, rifabutin, rifapentine, pyrazinamide, ethambutol, (+)calanolide A, ( ⁇ )-calanolide A, ( ⁇ )-soulattrolide, ( ⁇ )-costatolide or ( ⁇ )-7,8-dihydrosoulattrolide.
  • Other anti-mycobacterial agents include streptomycin, dapsone, clarithromycin, ciprofloxacin, clofazamine, azithromycin, ethionamide, amikacin or resorcinomycin A.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a bacterial infection.
  • bacterial infections to be treated according to this disclosure include but are not limited to an E. coli infection, a Staphylococcal infection, a Streptococcal infection, a Pseudomonas infection, Clostridium difficile infection. Legionella infection, Pneumococcus infection, Haemophilus infection, Klebsiella infection, Enterobacter infection, Citrobacter infection, Neisseria infection, Shigella infection, Salmonella infection, Listeria infection, Pasteurella infection, Streptobacillus infection.
  • Spirillum infection Treponema infection, Actinomyces infection, Borrelia infection, Corynebacterium infection, Nocardia infection, Gardnerella infection, Campylobacter infection, Spirochaeta infection, Proteus infection, Bacteroides infection, H. pylori infection, and anthrax infection.
  • the subjects may have or may be at risk of developing a gram-negative bacterial infection.
  • Gram negative bacteria include but are not limited to Escherichia coli ( E. coli ), Pseudomonas aeruginosa, Neisseria meningitides, Neisseria gonorrhoeae, Chlamydia trachomatis, Yersinia pestis , Non-typable Hemophilus influenzae, Hemophilus ducreyi, Helicobacter pylori, Campylobacter jejuni; Bacteroides fragilis . B.
  • the tetra- and penta-acylated glycolipids provided herein, and compositions thereof, are used to treat gram-negative bacterial infections, such as any of the foregoing.
  • the subject is experiencing sepsis.
  • the subject does not have and is not at risk of developing a B. fragilis infection. In some embodiments, the subject does have or is at risk of developing a B. fragilis infection or a condition associated with type of infection.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a mycobacterial infection.
  • mycobacterial infections to be treated according to this disclosure include but arm not limited to tuberculosis or leprosy respectively caused by the M. tuberculosis and M. leprae species.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a viral infection.
  • viral infections to be treated according to this disclosure include but are not limited to a Zika infection, an HIV infection, a Herpes simplex virus 1 infection, a Herpes simplex virus 2 infection, cytomegalovirus infection, hepatitis A virus infection, hepatitis B virus infection, hepatitis C virus infection, human papilloma virus infection, Epstein Barr virus infection, rotavirus infection, adenovirus infection, influenza A virus infection, respiratory syncytial virus infection, varicella-zoster virus infections, small pox infection, monkey pox infection, and SARS infection.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a parasitic infection.
  • parasitic infections to be treated according to this disclosure include but are not limited to candidiasis, ringworm, histoplasmosis, blastomycosis, paracoccidioidomycosis, crytococcosis, aspergillosis, chromomycosis, mycetoma infections, pseudallescheriasis, and tinea versicolor infection.
  • Still other parasitic infections that may be treated using the glycolipids of this disclosure include amebiasis, Trypanosoma cruzi infection, Fascioliasis. Leishmaniasis, Plasmodium infections, Onchocerciasis.
  • Paragonimiasis Trypanosoma brucei infection, Pneumocystis infection, Trichomonas vaginalis infection. Taenia infection, Hymenolepsis infection, Echinococcus infections, Schistosomiasis, neurocysticercosis, Necator americanus infection, and Trichuris trichiura infection.
  • the glycolipids and compositions thereof may be used to enhance immune therapies for a number of indications, both in a therapeutic and a prophylactic sense.
  • Immune therapies include but are not limited to passive immune therapies such as immunoglobulin administration, and active immune therapies such as vaccination with antigens alone or antigens in the context of dendritic cells.
  • the methods are intended to treat or prevent various indications that would benefit from an enhanced immune response.
  • the immune response that is stimulated is a cell-mediated immune response involving antigen presenting cells and optionally T cells such as Treg cells.
  • the immune response is an innate immune response, while in others it is an adaptive immune response, while in yet others it is a combined innate and adaptive immune response.
  • the immune response may, in some instances, be antigen specific.
  • the immune therapy may involve administration of an antibody or antigen-binding antibody fragment.
  • the antibody or antigen-binding fragment thereof may be specific for a tumor vasculature molecule.
  • Tumor vasculature molecules include but are not limited to endoglin, ELAM-1, VCAM-1. ICAM-1, ligand reactive with LAM-1, MHC class H antigens, amino phospholipids such as phosphatidylserine and phosphatidylethanolamine, VEGFR1 (Flt-1) and VEGFR2 (KDR/Flk-1).
  • Antibodies to endoglin include TEC-4 and TEC-11.
  • Antibodies that inhibit VEGF include 2C3 (ATCC PTA 1595).
  • antibodies that are specific for tumor vasculature include antibodies that react to a complex of a growth factor and its receptor such as a complex of FGF and the FGFR or a complex of TGF ⁇ and the TGF ⁇ R.
  • Antibodies of this latter class include GV39 and GV97.
  • the antibody or antibody fragment is selected from the group consisting of trastuzumab, alemtuzumab (B cell chronic lymphocytic leukemia), gemtuzumab ozogamicin (CD33+acute myeloid leukemia), hP67.6 (CD33+acute myeloid leukemia), infliximab (inflammatory bowel disease and rheumatoid arthritis), etanercept (rheumatoid arthritis), rituximab, tositumomab, MDX-210, oregovomab, anti-EGF receptor mAb, MDX-447, anti-tissue factor protein (TF), (Sunol); ior-c5, c5, edrecolomab, ibritumomab tiuxetan, anti-idiotypic mAb mimic of ganglioside GD3 epitope, anti-HLA-Dr10 mAb, anti-
  • Gliomab-H mAb GNT-250 mAb, anti-CD22, CMA 676), anti-idiotypic human mAb to GD2 ganglioside, ior egf/r3, anti-ior c2 glycoprotein mAb, ior c5, anti-FLK-2/FLT-3 mAb, anti-GD-2 bispecific mAb, antinuclear autoantibodies, anti-HLA-DR Ab, anti-CEA mAb, palivizumab, bevacizumab, alemtuzumab.
  • BLyS-mAb, anti-VEGF2, anti-Trail receptor B3 mAb, mAb BR96, breast cancer; and Abx-Cbl mAb.
  • the antibody or antibody fragment may be an anti-HER2 antibody, such as trastuzumab.
  • the antibody or antibody fragment may be an anti-CD20 antibody, such as rituximab.
  • shortening a vaccination course refers to reducing either the number of vaccine administrations (e.g., by injection) or the time between vaccine administrations. This is accomplished by stimulating a more robust immune response in the subject.
  • the method may involve, in one embodiment, administering to a subject in need of immunization a glycolipid in an amount effective to induce an antigen-specific immune response to a vaccine administered in a vaccination course, wherein the vaccination course is shortened by at least one immunization. In other embodiments, the vaccination course is shortened by one immunization, two immunizations, three immunizations, or more.
  • the method may involve, in another embodiment, administering to a subject in need of immunization a glycolipid of this disclosure in an amount effective to induce an antigen-specific immune response to a vaccine administered in a vaccination course, wherein the vaccination course is shortened by at least one day.
  • the vaccination course is shortened by one day, two days, three days, four days, five days, six days, one week, two weeks, three weeks, four weeks, one month, two months or more.
  • the glycolipid is administered substantially simultaneously with the vaccine.
  • Immunizations that can be modified in this way include but are not limited to newborn immunizations for HBV; immunizations at for example two months of age for Polio. DTaP, Hib, HBV, Pneumococcus; immunizations at for example four months of age for Polio, DTaP, Hib, Pneumococcus; immunizations at for example six months of age for Polio, DTaP, Hib, HBV, Pneumococcus; immunizations at for example 12-15 months of age for Hib, Pneumococcus, MMR, Varicella; immunizations at for example 15-18 months of age for DtaP; immunizations at for example 4-6 years of age for Polio, DPT, MMR; immunizations at for example 11-12 years of age for MMR; immunizations at for example 14-16 years of age for tetanus-diphtheria (i.e., Td) (
  • a recommended vaccination course for tetanus/diphtheria includes a primary immunization series given in adults if not received as a child, followed by routine booster doses of tetanus-diphtheria (Td) every 10 years.
  • the method of the invention will allow for a shortened series of vaccinations at the first time point, and may in some instances obviate the need for booster shoots later on.
  • hepatitis vaccination commonly requires three administrations spaced at least two weeks, and sometimes one month, apart in order to develop full immunity. Using the methods of the invention, it is possible to either reduce the number of injections from three to two or one, or to reduce the time in between injections from weeks or months to days or weeks.
  • Vaccination courses that can be shortened by the method of the invention include but are not limited to: HBV: Hepatitis B vaccine (3 total doses currently recommended); Polio: Inactivated polio vaccine (4 total doses currently recommended); DTaP: Diphtheria/tetanus/acellular Pertussis (3-in-1 vaccine; 5 total doses currently recommended); Hib: Haemophilus influenzae type b conjugate vaccine (4 total doses currently recommended); Pneumococcus (Prevnar): Protects against certain forms of Strep.
  • glycolipids can be used together with oral polio vaccine.
  • the glycolipids and compositions thereof may be administered to a subject having or at risk of developing cancer.
  • the cancer may be one for which a cancer antigen is known.
  • cancers that may be treated according to this disclosure include basal cell carcinoma, biliary tract cancer; bladder cancer: bone cancer, brain cancer, breast cancer, cervical cancer; choriocarcinoma; CNS cancer; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer, cancer of the head and neck; gastric cancer; intra-epithelial neoplasm; kidney cancer: larynx cancer; leukemia; acute myeloid leukemia, acute lymphoid leukemia, chronic myeloid leukemia, chronic lymphoid leukemia, liver cancer; small cell lung cancer; non-small cell lung cancer, lymphoma, Hodgkin's lymphoma; Non-Hodgkin's lymphoma; melanoma; myeloma; neuroblastoma; oral cavity cancer; ovarian cancer, pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcom
  • glycolipids and compositions thereof may be administered with another therapeutic agent or modality such as but not limited to antibody or other biologic, chemotherapy, radiation, surgery and the like.
  • the glycolipids and compositions thereof may be administered with a cancer antigen or a cancer immunotherapy such as an antibody specific for a cancer antigen.
  • the chemotherapy may be selected from the group consisting of aldesleukin, asparaginase, bleomycin sulfate, carboplatin, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin hydrochloride, docetaxel, doxorubicin, doxorubicin hydrochloride, epirubicin hydrochloride, etoposide, etoposide phosphate, floxuridine, fludarabine, fluorouracil, gemcitabine, gemcitabine hydrochloride, hydroxyurea, idarubicin hydrochloride, ifosfamide, interferons, interferon- ⁇ 2a, interferon- ⁇ 2b, interferon- ⁇ n3, interferon- ⁇ 1b, interleukins, irinotecan, mechlorethamine hydrochloride,
  • the cancer immunotherapy may include an antibody or antibody fragment specific for a cell surface molecule.
  • Cell surface molecules that may be targeted with the antibody or antibody fragment include but are not limited to HER 2, CD20, CD33, EGF receptor, HLA markers such as HLA-DR, CD52, CD1, CEA, CD22, GD2 ganglioside, FLK2/FLT3, VEGF, VEGFR, and the like.
  • the antibody or antibody fragment may be specific for a cancer antigen.
  • Cancer antigens that may be targeted with the antibody or antibody fragment include but are not limited to HER 2 (p185).
  • Other cancer antigens are described in U.S. Pat. No. 5,776,427.
  • Cancer antigens can be classified in a variety of ways. Cancer antigens include antigens encoded by genes that have undergone chromosomal alteration. Many of these antigens are found in lymphoma and leukemia. Even within this classification, antigens can be characterized as those that involve activation of quiescent genes. These include BCL-1 and IgH (Mantel cell lymphoma). BCL-2 and IgH (Follicular lymphoma), BCL-6 (Diffuse large B-cell lymphoma).
  • TAL-1 and TCR ⁇ or SIL T-cell acute lymphoblastic leukemia
  • c-MYC and IgH or IgL Burkitt lymphoma
  • MUN/IRF4 and IgH Myeloma
  • PAX-5 BSAP
  • cancer antigens that involve chromosomal alteration and thereby create a novel fusion gene and/or protein include RAR ⁇ , PML, PLZF, NPM or NuMA (Acute promyelocytic leukemia), BCR and ABL (Chronic myeloid/acute lymphoblastic leukemia), MLL (HRX) (Acute leukemia). E2A and PBX or HLF (B-cell acute lymphoblastic leukemia). NPM, ALK (Anaplastic large cell leukemia), and NPM, MLF-1 (Myelodysplastic syndrome/acute myeloid leukemia).
  • cancer antigens are specific to a tissue or cell lineage. These include cell surface proteins such as CD20, CD22 (Non-Hodgkin's lymphoma. B-cell lymphoma, chronic lymphocytic leukemia (CLL)), CD52 (B-cell CLL), CD33 (acute myelogenous leukemia (AML)), CD10 (gp100) (common (pre-B) acute lymphocytic leukemia and malignant melanoma), CD3/T-cell receptor (TCR) (T-cell lymphoma and leukemia), CD79/B-cell receptor (BCR) (B-cell lymphoma and leukemia).
  • CLL chronic lymphocytic leukemia
  • AML acute myelogenous leukemia
  • CD10 gp100
  • pre-B acute lymphocytic leukemia and malignant melanoma
  • CD3/T-cell receptor (TCR) T-cell lymphoma and leukemia
  • CD26 epidermal and lymphoid malignancies
  • human leukocyte antigen (HLA)-DR human leukocyte antigen
  • HLA-DP human leukocyte antigen
  • HLA-DQ lymphoid malignancies
  • RCAS1 gynecological carcinomas, bilary adenocarcinomas and ductal adenocarcinomas of the pancreas
  • prostate specific membrane antigen prostate cancer
  • Tissue- or lineage-specific cancer antigens also include epidermal growth factor receptors (high expression) such as EGFR (HER1 or erbB1) and EGFRvIII (brain, lung, breast, prostate and stomach cancer), erbB2 (HER2 or HER2/neu) (breast cancer and gastric cancer), erbB3 (HER3) (adenocarcinoma), and erbB4 (HER4) (breast cancer).
  • epidermal growth factor receptors high expression
  • EGFR HER1 or erbB1
  • EGFRvIII brain, lung, breast, prostate and stomach cancer
  • erbB2 HER2 or HER2/neu
  • HER3 adenocarcinoma
  • HER4 erbB4
  • Tissue- or lineage-specific cancer antigens also include cell-associated proteins such as tyrosinase, melan-A/MART-1, tyrosinase related protein (TRP)-1/gp75 (malignant melanoma), polymorphic epithelial mucin (PEM) (breast tumors), and human epithelial mucin (MUC1) (breast, ovarian, colon and lung cancers).
  • cell-associated proteins such as tyrosinase, melan-A/MART-1, tyrosinase related protein (TRP)-1/gp75 (malignant melanoma), polymorphic epithelial mucin (PEM) (breast tumors), and human epithelial mucin (MUC1) (breast, ovarian, colon and lung cancers).
  • Tissue- or lineage-specific cancer antigens also include secreted proteins such as monoclonal immunoglobulin (multiple myeloma and plasmacytoma), immunoglobulin light chains (Multiple Mycloma), ⁇ -fetoprotcin (liver carcinoma), kallikrcins 6 and 10 (ovarian cancer), gastrin-releasing peptide/bombesin (lung carcinoma), and prostate specific antigen (prostate cancer).
  • monoclonal immunoglobulin multiple myeloma and plasmacytoma
  • immunoglobulin light chains Multiple Mycloma
  • ⁇ -fetoprotcin liver carcinoma
  • kallikrcins 6 and 10 ovarian cancer
  • gastrin-releasing peptide/bombesin lung carcinoma
  • prostate specific antigen prostate cancer
  • CT antigens that are expressed in some normal tissues such as testis and in some cases placenta. Their expression is common in tumors of diverse lineages and as a group the antigens form targets for immunotherapy.
  • tumor expression of CT antigens include MAGE-A1, -A3, -A6, -A12, BAGE, GAGE, HAGE, LAGE-1, NY-ESO-1, RAGE, SSX-1, -2, -3, -4, -5, -6, -7, -8, -9, HOM-TES-14/SCP-1, HOM-TES-85 and PRAME.
  • CT antigens and the cancers in which they are expressed include SSX-2, and -4 (Neuroblastoma).
  • SSX-2 HOM-MEL-40
  • MAGE. GAGE, BAGE and PRAME Malignant melanoma
  • HOM-TES-14/SCP-1 Meningioma
  • SSX-4 Oligodendrioglioma
  • HOM-TES-14/SCP-1 MAGE-3 and SSX-4
  • Astrocytoma SSX member
  • SSX member Head and neck cancer, ovarian cancer, lymphoid tumors, colorectal cancer and breast cancer
  • RAGE-1, -2, -4, GAGE-1, -2, -3, -4, -5, -6, -7 and -8 Head and neck squamous cell carcinoma (HNSCC)
  • HOM-TES14/SCP-1 PRAME
  • SSX-1 and CT-7 Non-Hodgkin's lymphoma
  • PRAME A
  • cancer antigens are not specific to a particular tissue or cell lineage. These include members of the carcinoembryonic antigen (CEA) family: CD66a, CD66b, CD66c, CD66d and CD66e. These antigens can be expressed in many different malignant tumors and can be targeted by immunotherapy. Still other cancer antigens are viral proteins and these include Human papilloma virus protein (cervical cancer), and EBV-encoded nuclear antigen (EBNA)-1 (lymphomas of the neck and oral cancer). Still other cancer antigens are mutated or aberrantly expressed molecules such as but not limited to CDK4 and beta-catenin (melanoma).
  • the subject may be one having or likely to develop an aberrant immune response.
  • the aberrant immune response may be an enhanced immune response and the glycolipid acts to down-regulate the immune response.
  • Enhanced immune responses are typically associated with inflammatory conditions, such as but not limited to autoimmune diseases. Autoimmune diseases, autoimmune disorders and autoimmune conditions are used interchangeably herein.
  • compositions comprising glycolipid as a single agent or in combination with an antigen other than lipidated PSA, may be used to modulate (e.g., down-regulate) certain immune responses in subjects having or at risk of developing autoimmune diseases.
  • autoimmune diseases typically experience one or more “events” or recurrences associated with the autoimmune disease.
  • a subject having inflammatory bowel disease may experience temporally isolated attacks of the disease, characterized by the presence of symptoms or increased severity of symptoms.
  • compositions may be used in such subjects to reduce the likelihood of such future recurrences of the disease or to reduce the severity of symptoms associated with the disease (e.g., pain, fever, discomfort, fatigue, etc.).
  • the compositions may be administered prior to such recurrence, and in this manner may be chronically administered, optionally at a regular frequency. Examples include once a day, once every 2, 3, 4, 5 or 6 days, or once a week, etc.
  • the compositions may be administered to the subject during a recurrence in order to reduce the severity of symptoms or shorten the time of the recurrence.
  • the invention provides a method comprising administering to a subject at risk of a recurrence of a condition associated with inflammation an effective amount of the glycolipid as a single agent or in combination with an antigen such as or alternative to PSA or lipidated PSA.
  • the method may reduce the likelihood of a recurrence of the condition or may reduce the frequency of future recurrences.
  • the method may reduce the severity of symptoms associated with the condition, whether such symptoms are present in the first manifestation, in a recurrence, or chronically.
  • autoimmune diseases include but are not limited to multiple sclerosis, inflammatory bowel disease including Crohn's Disease and ulcerative colitis, rheumatoid arthritis, psoriasis, type I diabetes, uveitis, Celiac disease, pernicious anemia, Srojen's syndrome. Hashimoto's thyroiditis, Graves' disease, systemic lupus erythamatosis, acute disseminated encephalomyelitis, Addison's disease, Ankylosing spondylitis, Antiphospholipid antibody syndrome.
  • Guillain-Barre syndrome idiopathic thrombocytopenic purpura, Goodpasture's syndrome, Myasthenia gravis, Pemphigus, giant cell arteritis, aplastic anemia, autoimmune hepatitis, Kawaski's disease, mixed connective tissue disease, Ord throiditis, polyarthritis, primary biliary sclerosis, Reiter's syndrome, Takaysu's arteritis, vitiligo, warm autoimmune hemolytic anemia, Wegener's granulomatosis, Chagas' disease, chronic obstructive pulmonary disease, and sarcoidosis.
  • the autoimmune disease is multiple sclerosis. In other important embodiments, the autoimmune disease is an inflammatory bowel disease including but not limited to ulcerative colitis and Crohn's disease. In other embodiments, the autoimmune disease may be rheumatoid arthritis or type I diabetes.
  • compositions of the invention may be administered to a subject who has yet to manifest an autoimmune disease (including symptoms thereof) yet is at risk of developing such as disease based on a known genetic or familial predisposition.
  • a subject may have one or more family members that are afflicted with the disease.
  • compositions of the invention are administered to subject having or at risk of developing graft-versus-host disease. Administration may occur prior to, during and/or after transplantation of an organ or tissue (including blood or a blood product) into the subject.
  • compositions may be administered to subjects having or at risk of developing a conditions associated with inflammation.
  • the composition may be administered to a subject having asthma.
  • subjects having asthma typically experience asthmatic attacks or events characterized by impaired breathing.
  • the invention contemplates that the compositions described herein may be administered acutely (e.g., a single large dose) or chronically (e.g., repeated, smaller doses) to asthmatic subjects.
  • the compositions may be administered prior to an asthmatic attack in order to prevent the occurrence of the attack, reduce the frequency of attacks, and/or to lessen the severity of the attack.
  • the compositions may be administered during an attack in order to reduce its severity and/or reduce its duration.
  • compositions described herein are administered prior to, during, and/or immediately following surgery, or any combination thereof including but not limited to prior to and during surgery, in order to prevent the occurrence of such adhesions and/or reduce their severity.
  • the compositions may be administered repeatedly following surgery, including for example every day, every two days, every three days, etc. for a week, two weeks, three weeks, a month, or several months post-surgery.
  • Another condition associated with inflammation is an abscess, including but not limited to an abdominal abscess as may occur upon leakage of intestinal contents into the peritoneum.
  • the subjects being treated may also be administered anti-bacterial agents such as antibiotics.
  • a method comprises administering to a subject having or at risk of developing an abscess an effective amount of the glycolipid (separate from the polysaccharide component of PSA) or compositions thereof.
  • the glycolipid is administered prior to development of an abscess and/or prior to the manifestation of symptoms associated with an abscess.
  • the glycolipid is administered after an abscess has been detected or diagnosed and/or after symptoms associated with an abscess are manifested.
  • a subject intends any subject that would benefit from administration of a composition of the invention or that could be administered the composition of the invention.
  • the subject is a human subject.
  • the subject may also be a companion animal such as a dog or cat, agricultural livestock such as horses, cattle, pigs, sheep, etc., laboratory animals such as mice, rats, rabbits, monkeys, etc., or animals such as those maintained in zoos or otherwise in captivity.
  • a variety of administration routes are available. The particular mode selected will depend, of course, upon the particular condition being treated, the severity of the condition being treated, and the dosage required for therapeutic efficacy.
  • the methods of the invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects.
  • modes of administration include oral, rectal, topical, nasal, inhalation (e.g., inhaler or nebulization), or parenteral routes.
  • parenteral includes subcutaneous, intravenous, intramuscular, intraperitoneal, or infusion.
  • the agents e.g., glycolipids
  • Such compositions or preparations may routinely contain pharmaceutically acceptable carriers, concentrations of salt, buffering agents, preservatives, other immune modulators, and optionally other therapeutic agents.
  • pharmaceutically-acceptable carrier as used herein, and described more fully below, means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other animal.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active agent(s) is combined to facilitate administration, long-term storage, stability and the like.
  • the active agents of the present invention may be comingled with the other components of the pharmaceutical compositions, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • compositions may be presented in unit dosage form and may be prepared by any of the methods known in the art of pharmacy. All methods include the step of bringing the active agents into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active agent(s) into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent.
  • Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion. Solid forms may be coated for example enterically coated.
  • the active agent(s) may be administered per se (neat) or in the form of a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts may be used for in vivo applications as well as in vitro applications.
  • Non-pharmaceutically acceptable salts may be used to prepare pharmaceutically acceptable salts thereof and are not excluded from the scope of the invention.
  • Pharmaceutically acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicyclic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • pharmaceutically acceptable salts can be prepared as alkyline metal or alkyline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt 1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v).
  • Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • compositions suitable for parenteral administration conveniently comprise a sterile aqueous preparation of the active agent(s), which can be isotonic with the blood of the recipient.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Carrier formulations suitable for subcutaneous, intramuscular, intraperitoneal intravenous, etc. administrations may be found in Remington's Pharmaceutical Sciences, Mack Publishing Company. Easton, Pa.
  • the glycolipid is formulated with a detergent such as but not limited to Tween or a bile salt such as but not limited to deoxycholate (e.g., sodium deoxycholate) in order to limit or prevent aggregation.
  • a detergent such as but not limited to Tween or a bile salt such as but not limited to deoxycholate (e.g., sodium deoxycholate) in order to limit or prevent aggregation.
  • a detergent or bile salt may be used at a low concentration such that it is still pharmaceutically acceptable. For example, it may be present at about or less than 0.0001%, 0.0005%, 0.001%, 0.002%, 0.005%, 0.01%, 0.02%, 0.05%, 0.07%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 2%, 3%, 4%, 5%, or more.
  • a therapeutically effective amount is that amount necessary to delay the onset of, inhibit the progression of, or halt altogether the particular condition being treated, including reducing the likelihood, frequency and/or severity of a recurrence of the condition.
  • the effective amount may be that amount which serves to reduce, alleviate, or delay the onset of the symptoms (e.g., pain, fever, etc.) of the disorder being treated or prevented. The effective amount will depend upon the mode of administration, the particular condition being treated and the desired outcome.
  • doses of active agent(s) of the present invention may be from about 0.01 mg/kg per day to 1000 mg/kg per day, preferably from about 0.1 mg/kg to 200 mg/kg and most preferably from about 0.2 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or more days. It is expected that doses ranging from 1-500 mg/kg, and preferably doses ranging from 1-100 mg/kg, and even more preferably doses ranging from 1-50 mg/kg, will be suitable. The preferred amount can be determined by one of ordinary skill in the art in accordance with standard practice for determining optimum dosage levels of the agent. It is generally preferred that a maximum dose is the highest safe dose according to sound medical judgment be used.
  • the total daily dose for a human subject may range from about 50-100 micrograms of the glycolipid.
  • the pharmaceutical preparation may be administered alone or in conjunction with one or more other active agents.
  • the pharmaceutical preparation may be used or administered in conjunction with active agents that are suitable for autoimmune disorders such as multiple sclerosis, Crohn's disease, ulcerative colitis, asthma, rheumatoid arthritis, and the like.
  • agents include anti-inflammatory agents.
  • examples include steroids and corticosteroids such as cortisone; non-steroidal anti-inflammatory drugs such as aspirin, salsalate, celecoxib, diclofenac, etodolac, ibuprofen, indomethacin, ketoprofen, ketorolac, nabumetone, naproxen, oxaprozin, piroxicam, sulindac, and tolmetin; aminosalicylates such as sulfasalazine and 5-aminosalicylates including mesalamine, balsalazide, and olsalazine; azathioprine; mercaptopurine; cyclosporine; beta interferons; glatiramer acetate; dimethyl fumarate; fingolimod; mitoxantrone; disease-modifying antirheumatic drugs (DMARDs) such as methotrexate, leflunomide, hydroxychloro
  • TNF alpha inhibitors such as infliximab (Remicade), adalimumab (Humira), and golimumab (Simponi); natalizumab (Tysabri), vedolizumab (Entyvio); ustekinumab (Stelara); abatacept (Orencia); anakinra (Kineret); certolizumab (Cimzia), etanercept (Enbrel), rituximab (Rituxan), tocilizumab (Actemra), and tofacitinib (Xeljanz).
  • the invention contemplates that the combined use of glycolipid together with standard treatments such as those recited above will allow a lower dose of the standard treatment to be used for the same or better therapeutic effect, and/or will result in reduced incidence and/or severity of side effects associated with such standard treatments.
  • the subject is also administered an anti-bacterial agent such as an antibiotic.
  • the pharmaceutical preparation is formulated or given in conjunction with one or more anti-bacterial agents including antibiotics selected from the group consisting of penicillin G, penicillin V, ampicillin, amoxicillin, bacampicillin, cyclacillin, epicillin, hetacillin, pivampicillin, methicillin, nafcillin, oxacillin, cloxacillin, dicloxacillin, flucloxacillin, carbenicillin, ticarcillin, avlocillin, mezlocillin, piperacillin, amdinocillin, cephalexin, cephradine, cefadoxil, cefaclor, cefazolin, cefuroxime axetil, cefamandole, cefonicid, cefoxitin, cefotaxime, ceftizoxime, cefmnenoxine, ceftriaxone, moxal
  • B. fragilis was grown in anaerobic conditions.
  • the capsular complex from B. fragilis was isolated with hot phenol/water extraction.
  • the polysaccharide fraction was precipitated with ethanol after DNAse. RNase and pronase treatments.
  • the precipitate was subjected to size exclusion chromatography in order to separate polysaccharide constituents.
  • the fractions of interest were analyzed and pooled, then dialyzed and lyophilized. The purity of glycolipid can be assessed by nuclear magnetic resonance spectroscopy and mass spectroscopy.
  • the B. fragilis delta44 mutant strain was derived experimentally from strain 9343 and upon further characterization it was found to over-express PSA relative to PSB.
  • Delta44 was plated onto a blood agar plate and grown overnight at 37° C.
  • a swab from a heavily colonized plate was sub-cultured into a 500 ml starter culture of peptone yeast broth.
  • the starter culture was inoculated into 16 liter culture of the same media and pH was titrated to neutrality with 5M NaOH. An anaerobic gas mix was bubbled into the sealed culture.
  • bacteria were checked by Gram stain and subculture. Organisms were collected by centrifugation at 8,000 ⁇ g for 20 minutes. Bacterial pellets were washed two times with saline yielding approximately one liter of bacterial pellet.
  • the bacterial pellet was suspended in 68° C. melted crystalline phenol to a final concentration of phenol of about 37% v/v (yielding a phenol/water preparation) and mixed for 30 minutes at 68° C. followed by stirring at 4° C. for 48 hours.
  • the phenol/water preparation was aliquoted into glass bottles which were then centrifuged at 1500 rpm. The upper water layer was harvested. Any residual phenol contained in the harvested aqueous phase was extracted with an equal volume of ethyl ether. The ether phase was then removed using a separatory funnel and any residual ether in the aqueous phase was evaporated, yielding the final aqueous phase from the phenol/water preparation.
  • the aqueous phase was dialyzed versus water with multiple changes over 5 days at 4° C. and subsequently lyophilized until it was nearly dry (approximately 5 ml water remaining).
  • a solution of 0.05M Tris with magnesium, calcium and sodium azide (total volume 61 ml) was added to the lyophilized product to bring the total volume to about 66 ml.
  • the polysaccharide fraction was precipitated by adding 5 volumes of ethanol at 4° C. to the mixture. The solution was then centrifuged at 12,000 ⁇ g for 30 minutes to pellet the polysaccharide fraction. The supernatant was removed and the pellet was resuspended in 392 ml type 1 H 2 O.
  • the dissolved fraction was then dialyzed against two changes of 16 liters type 1 H 2 O at 4° C. The volume was reduced by lyophilization to approximately 50 mls.
  • mice are treated with glycolipid (on the order of about 75-100 ⁇ g per mouse) or control (saline, PBS) every three days starting 6 days before EAE induction.
  • mice are challenged subcutaneously with 250 ⁇ g of MOG 33-55 (Peptides International) in 200 ⁇ l of complete Freund's adjuvant (Sigma).
  • MOG 33-55 Peptides International
  • mice receive intraperitoneal injections of 250 ng of Bordetella pertussis toxin (List Biological Laboratories). Disease is scored on an established 0 to 5 scale, with 5 being advanced neurological disease. Mice are monitored and scored daily for disease progression.
  • glycolipids Semi-purified B. fragilis glycolipids or overnight-grown bacterial pellet was resuspended in 100 mM acetic acid/sodium acetate buffer (pH 4.5) with 1% sodium dodecylsulfate (SDS) solution at 1-10 mg/mL concentration. The mixture was heated at 95° C. for 30 minutes and cooled. Solution was added with the same volume of chloroform and methanol, shaken well and spun at 1000 g for 10 mins for phase separation. Lower organic phases, containing hydrolyzed GLA, were collected and dried under nitrogen stream and resuspended in 65% isopropanol solution.
  • SDS sodium dodecylsulfate
  • inventive embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed.
  • inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein.
  • a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified.
  • “at least one of A and B” can refer, in one embodiment, to at least one, optionally including more than one. A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.

Abstract

The invention provides immunostimulatory glycolipids and compositions thereof and methods of use thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 62/362,630, entitled “GLYCOLIPID COMPOSITIONS AND METHODS OF USE,” filed Jul. 15, 2016, the entire contents of which are incorporated by reference herein.
  • FIELD
  • The invention relates to glycolipids, compositions, methods of synthesis, isolation and/or purification, and methods of use thereof.
  • BACKGROUND
  • Polysaccharide A (PSA) of Bacteroides fragilis (B. fragilis) has been reported to be an immunomodulator with therapeutic and prophylactic activities. U.S. Pat. Nos. 5,679,654 and 5,700,787; Tzianabos A O et al. (2000) J Biol Chem 275:6733-40. As an example, PSA has been shown to provide protection from intestinal inflammatory diseases as well as systemic immune-mediated diseases such as the mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). PSA is one of eight capsular polysaccharides made by B. fragilis.
  • PSA was recently discovered to possess a lipid moiety. The lipid moiety was hypothesized to anchor the polysaccharide in the B. fragilis outer membrane. It was also recently discovered that this “lipidated PSA” was more potent than non-lipidated PSA (referred to herein as “PSA”) forms provided in the prior art.
  • SUMMARY
  • The invention is based, in part, on the identification and characterization of the glycolipid moiety that is found conjugated to PSA using certain isolation methods. The invention is further premised, in part, on surprising finding that such glycolipid is able to stimulate the innate immune response and in particular activate antigen-presenting cells such as plasmacytoid dendritic cells. Based on this surprising finding, this disclosure contemplates and provides, among other things, use of the glycolipids as adjuvants in for example vaccine formulations.
  • The innate immunostimulatory property of the glycolipid is enhanced upon cleavage of the glycolipid moiety from the polysaccharide chain of PSA.
  • The disclosure therefore provides, in some aspects, glycolipids of defined chemical structure, as well as compositions comprising such glycolipids. Such compositions may be further defined by the purity and/or concentration of glycolipids contained therein.
  • Also provided are compositions of the glycolipids with antigens such as but not limited to bacterial antigens, including without limitation PSA and lipidated PSA. In some instances, such compositions comprise lipidated PSA and glycolipid in ratios ranging from 4:1 to 20:1 (w/w), or 4:1 to 15:1 (w/w), or 4:1 to 10:1 (w/w).
  • In one aspect, provided herein is a glycolipid comprising a tri-acylated, tetra-acylated or penta-acylated diglucosamine, and the diglucosamine may be optionally conjugated to an oligosaccharide. The glycolipid may be isolated intending that it is physically separated from other components including contaminants such as but not limited to components of B. fragilis cells. Isolation may also intend a degree of purity or concentration in a composition. The glycolipid unless otherwise stated is unconjugated to the polysaccharide chain of PSA (i.e., the polysaccharide comprised of repeating tetrasaccharides found in PSA and lipidated PSA). Thus, it is to be understood that if the acylated diglucosamine is conjugated to the oligosaccharide, such oligosaccharide is not the PSA polysaccharide, and such glycolipid is not PSA. The glycolipid may be formulated with such polysaccharide chain of PSA or it may be formulated with lipidated PSA or it may be formulated with another antigen or another polysaccharide bearing moiety, although it will typically be provided in an unconjugated or free form. Examples of such formulations include for example micelles or liposomes or other delivery vehicles that are amenable to lipid based agents.
  • In some embodiments, the diglucosamine is tetra-acylated or penta-acylated. In some embodiments, the tri-acylated, tetra-acylated or penta-acylated diglucosamine comprises acyl chains ranging in length from 14-17 carbons. In some embodiments, the tri-acylated, tetra-acylated or penta-acylated diglucosamine comprises acyl chains range in length from 15-17 carbons.
  • In some embodiments, the glycolipid is substantially free of other components found in a B. fragilis capsule or found in B. fragilis cells. In some embodiments, the glycolipid is substantially free of LPS. In some embodiments, the glycolipid is substantially free of polysaccharide including for example the repeating tetrasaccharide polysaccharide of PSA.
  • In some embodiments, the glycolipid is provided in purified form. In some embodiments, the glycolipid is provided an isolated form. In some embodiments, the glycolipid is provided in a micelle form or a liposome form, alone or optionally with other agents such as antigens, polysaccharides and the like. In some embodiments, the glycolipid is provided in a lyophilized form. Lyophilized forms glycolipid are particularly suitable for long-term storage, ranging from days, weeks, months or even years. In some embodiments, the glycolipid is provided in a form that is suitable for administration to a human, including an orally administered form or a parenterally administered form.
  • In some embodiments, the glycolipid is obtained or derived from B. fragilis cells, including B. fragilis cells that overexpress PSA relative to polysaccharide B (PSB), PSC. PSD, PSE. PSF, PSG and PSH. As used herein, obtained or derived intends that the glycolipid that is defined structurally herein is rendered using the isolation methods provided herein. It does not however intend that any naturally occurring counterpart of the glycolipid is structurally identical to the structure defined herein.
  • In some embodiments, at least one acyl chain is unmodified. In some embodiments, at least one acyl chain is modified, for example modified with a hydroxyl group. In some embodiments, at least one acyl chain is unmodified and at least one of the acyl chains is modified, for example modified with a hydroxyl group. In some embodiments, at least one acyl chain is modified with a hydroxyl group. In some embodiments, at least one acyl chain is C16:0-OH. In some embodiments, at least one acyl chain is C17:0-OH. In some embodiments, at least one acyl chain is C14:0. In some embodiments, at least one acyl chain is C15:0.
  • In some embodiments, at least one acyl chain is N-substituted. In some embodiments, at least one acyl chain is O-substituted. In some embodiments, at least one acyl chain is N-substituted and at least one acyl chain is O-substituted. In some embodiments, all of the acyl chains are N-substituted or all of the acyl chains are O-substituted.
  • In some embodiments, the diglucosamine is phosphorylated. In some embodiments, the diglucosamine is mono phosphorylated. In some embodiments, the diglucosamine is not phosphorylated.
  • In some embodiments, the diglucosamine is conjugated to the oligosaccharide by an acid-labile bond. In some embodiments, the acid-labile bond is a ketosidic bond.
  • In some embodiments, the oligosaccharide comprises or contains 3-10 sugars (e.g., it may comprise 3 or more sugars). The oligosaccharide however is not the polysaccharide of PSA (i.e., it is not the zwitterionic tetrasaccharide moiety or repeated moiety shown in FIG. 1 ). In some embodiments, the oligosaccharide contains 2-10, 2-9, 3-9, 4-9, 5-9, 6-9, 7-9 or 8-9 sugars. In some embodiments, the oligosaccharide contains 6-9 sugars. In some embodiments, the oligosaccharide comprises galactose and/or glucose and/or fucose residues. Thus, the sugars of the oligosaccharide may be identical or different from each other.
  • In some embodiments, the oligosaccharide comprises one or two KDO residues. The oligosaccharide is not and does not comprise the repeating tetrasaccharide units of PSA.
  • In some embodiments, the glycolipid is formulated with a detergent or a bile salt. In some embodiments, the detergent or bile salt is present at or less than 1%, 0.5% or 0.1% (weight of detergent or bile salt/weight of glycolipid).
  • In some embodiments, the glycolipid is formulated with an antigen other than a lipidated PSA or a component of such lipidated PSA.
  • In some embodiments, the glycolipid is formulated with vaccine additives.
  • In some embodiments, the glycolipid is formulated with a non-naturally occurring preservative. In some embodiments, the glycolipid is formulated with a non-naturally occurring stabilizer. In some embodiments, the glycolipid is formulated with human albumin, phenol, glycerin or glycine.
  • In some embodiments, the glycolipid is formulated with thimerosal, aluminum hydroxide, benzethonium chloride, formaldehyde, formalin, glutaraldehyde, potassium phosphate, aluminum potassium sulfate, bovine extract, calf serum, ammonium sulfate, aluminum phosphate, non-human cells. Vero (monkey kidney) cells, human cells, MRC-5 (human diploid) cells. MRC-5 cellular proteins, and the like.
  • In some embodiments, the glycolipid is formulated for parenteral administration. In some embodiments, the glycolipid is formulated for oral administration.
  • Thus, also provided herein are compositions comprising any of the foregoing glycolipids. In some embodiments, the composition further comprises an antigen other than a lipidated PSA or a component of such lipidated PSA. In some embodiments, the composition comprises any of the foregoing glycolipids and lipidated PSA in non-naturally occurring weight/weight ratios such as but not limited to 1:75, 1:50, 1:25, 1:20, 1:10, 1:5, and the like.
  • In some embodiments, the composition further comprises a detergent or a bile salt. In some embodiments, the detergent or bile salt is present at or less than 1%, 0.5% or 0.1% (weight of detergent or bile salt/weight of glycolipid).
  • In some embodiments, the composition further comprises vaccine additives.
  • In some embodiments, the composition further comprises a non-naturally occurring preservative. In some embodiments, the composition further comprises a non-naturally occurring stabilizer. In some embodiments, the composition further comprises human albumin, phenol, glycerin or glycine.
  • In some embodiments, the composition further comprises thimerosal, aluminum hydroxide, benzethonium chloride, formaldehyde, formalin, glutaraldehyde, potassium phosphate, aluminum potassium sulfate, bovine extract, calf serum, ammonium sulfate, aluminum phosphate, non-human cells, Vero (monkey kidney) cells, human cells, MRC-5 (human diploid) cells, MRC-5 cellular proteins, and the like.
  • In some embodiments, any of the foregoing glycolipids or compositions is formulated for parenteral administration. In some embodiments, any of the foregoing glycolipids or compositions is formulated for oral administration.
  • Also provided herein is a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids in a 4:1 to 20:1 ratio (w/w).
  • Also provided herein is a composition comprising a lipidated polysaccharide A (PSA) and at least 0.5% (w/w) of any of the foregoing glycolipids.
  • In some embodiments, the composition further comprises a pharmaceutically acceptable carrier.
  • In some embodiments, the composition further comprises another therapy such as an additional active agent or it is formulated for administration in combination with another therapy such as an additional active agent. In some embodiments, the other therapy is or comprises a cancer immunotherapy. In some embodiments, the cancer immunotherapy comprises administration of an antibody or antibody fragment thereof.
  • In some embodiments, the composition is formulated for parenteral administration. In some embodiments, the composition is formulated for oral administration.
  • Also provided herein is a composition comprising any one of the foregoing glycolipids and an antigen. In some embodiments, the composition is a vaccine.
  • In some embodiments, the antigen is a bacterial antigen. In some embodiments, the antigen is attenuated bacteria. In some embodiments, the antigen is a viral antigen. In some embodiments, the antigen is attenuated virus. In some embodiments, the antigen is a fungal antigen. In some embodiments, the antigen is a mycobacterial antigen. In some embodiments, the antigen is a parasitic antigen. In some embodiments, the antigen is a cancer antigen. In some embodiments, the antigen is a human protein. In some embodiments, the antigen is a human polysaccharide. In some embodiments, the composition is formulated for parenteral administration.
  • Also provided herein is a method of enhancing an innate immune response comprising administering, to a subject in need of an immune response, such as an enhanced immune response, and further such as an enhanced innate immune response, an effective amount of any of the foregoing glycolipids.
  • In some embodiments, the method further comprises administering a lipidated PSA in combination with another therapy. In some embodiments, the other therapy is or comprises a cancer immunotherapy. In some embodiments, the cancer immunotherapy comprises administration of an antibody or antibody fragment thereof.
  • Also provided herein is a method of inducing an immune response comprising administering, to a subject in need of immune response induction or modulation, an effective amount of any of the foregoing glycolipids.
  • In some embodiments, the method further comprises administering a lipidated PSA in combination with another therapy. In some embodiments, the other therapy is or comprises a cancer immunotherapy. In some embodiments, the cancer immunotherapy comprises administration of an antibody or antibody fragment thereof.
  • In some embodiments, the immune response is an innate immune response. In some embodiments, the immune response is an adaptive immune response.
  • Also provided herein is a method of inducing an immune response comprising administering, to a subject in need of immune response induction or modulation, an effective amount of any of the foregoing glycolipids, wherein the immune response comprises induction of interferon-beta, and the subject is experiencing or is likely to experience an interferon-beta responsive condition (e.g., a condition that has been shown to benefit from administration of exogenous interferon-beta and/or that can benefit from induction of interferon-beta in vivo). Such conditions include but are not limited to any of the autoimmune diseases provided herein. In some embodiments, the condition is multiple sclerosis (MS). In some embodiments, the condition is systemic lupus erythematosus.
  • Also provided herein is a method of inducing an immune response to an antigen comprising administering, to a subject in need of an antigen-specific immune response, an effective amount of any of the foregoing glycolipid and the antigen.
  • In some embodiments, the subject has or is at risk of developing a bacterial infection. In some embodiments, the antigen is a bacterial antigen. In some embodiments, the antigen is attenuated bacteria.
  • In some embodiments, the subject has or is at risk of developing a viral infection. In some embodiments, the antigen is a viral antigen. In some embodiments, the antigen is attenuated virus.
  • In some embodiments, the subject has or is at risk of developing a fungal infection. In some embodiments, the antigen is a fungal antigen.
  • In some embodiments, the subject has or is at risk of developing a mycobacterial infection. In some embodiments, the antigen is a mycobacterial antigen.
  • In some embodiments, the subject has or is at risk of developing a parasitic infection.
  • In some embodiments, the antigen is a parasitic antigen.
  • In some embodiments, the subject has or is at risk of developing a cancer. In some embodiments, the antigen is a cancer antigen.
  • In some embodiments, the antigen is a human protein. In some embodiments, the antigen is a human polysaccharide.
  • In some embodiments, the method further comprises administering to the subject lipidated PSA.
  • Also provided herein is a micelle consisting essentially of any one or any combination of the foregoing glycolipids.
  • Also provided herein is a composition comprising a micelle consisting essentially of any one or any combination of the foregoing glycolipids and a detergent or bile salt. In some embodiments, the detergent or bile salt is present in a pharmaceutically acceptable amount. In some embodiments, the composition is a pharmaceutical composition. In some embodiments, formulated for parenteral administration.
  • In various embodiments, the glycolipid is free of non-lipidated PSA. In various embodiments, the glycolipid is free of lipidated PSA.
  • In various embodiments, the glycolipid is suitable for administration to a human.
  • In various embodiments, the composition is formulated for parenteral or enteral or oral administration to a subject. In various embodiments, the composition is formulated for lipophilic delivery, including for example in a liposome or in an oil-based delivery system. The various compositions provided herein may be formulated as a capsule or other discrete dosage form, including those intended for oral or enteral administration.
  • In another aspect, the invention provides an isolated glycolipid comprising a diglucosamine covalently conjugated to 3-5 acyl chains, each independently ranging in length from 14-17 carbons. The glycolipid may be any of the glycolipids provided herein, or a combination thereof. In various embodiments, the diglucosamine is covalently conjugated to 3-5 acyl chains. In various embodiments, the diglucosamine is covalently conjugated to 4 or 5 acyl chains. In various embodiments, the acyl chains range in length from 15-17 carbons. Other embodiments relating to the glycolipids are recited above and apply equally. It is to be understood that any of the aspects and embodiments described herein relating to glycolipids, including compositions and methods of use, embrace and contemplate glycolipids that comprise, consist or consist essentially of a disaccharide, such as a diglucosamine, covalently conjugated to 3-5 acyl chains, and optionally each chain may independently range in length from 4-17 carbons.
  • In another aspect, the invention provides a method comprising administering to a subject in need of immune response modulation a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids in an effective amount.
  • In another aspect, the invention provides a method comprising administering to a subject in need of immune response modulation an effective amount of a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids, in combination with an additional therapy.
  • In another aspect, the invention provides a method comprising administering to a subject in need of immune response modulation one or both of a composition comprising a lipidated polysaccharide A (PSA) and any of the foregoing glycolipids; and another therapy, so that the subject receives therapy with the composition and the other therapy in combination.
  • In some embodiments, the subject has or is at risk of developing cancer. In some embodiments, the subject is diagnosed as suffering from, susceptible to, and/or receiving therapy for cancer.
  • In some embodiments, the additional therapy is or comprises a cancer immunotherapy. In some embodiments, the cancer immunotherapy is or comprises an antibody or antibody fragment thereof.
  • In some embodiments, the step of administering comprises administering according to a regimen that has been demonstrated to achieve statistically significant improvement in immune function when administered to a relevant population of subjects. The relevant population of subjects may comprise subjects diagnosed as suffering from, susceptible to, and/or receiving therapy for cancer. In some embodiments, the improvement in immune function comprises modulation of an innate and/or adaptive immune response.
  • In some embodiments, the subject in need of immune response modulation is a subject in need of innate immune response modulation. In some embodiments, the subject in need of immune response modulation is a subject in need of adaptive immune response modulation. In some embodiments, the subject in need of immune response modulation is a subject in need of immune response stimulation.
  • In some embodiments, improvement in immune function comprises stimulation of an innate and/or adaptive immune response.
  • In some embodiments, the step of administering comprises administering an amount of the composition effective to achieve an improvement in immune function as compared to a subject who has not been administered the composition.
  • In some aspects, the acylated diglucosamine, such as but not limited to the tetra-acylated diglucosamine or the penta-acylated diglucosamine, may be administered to a subject experiencing or likely to experience a proinflammatory immune response, such as may occur for example during an E. coli infection or an infection by gram negative bacteria. The acylated diglucosamine may be conjugated to an oligosaccharide such as 3-10-mer oligosaccharide. The subject may be defined as one experiencing or likely to experience a gram negative bacterial infection, including but not limited to infection by any of the gram negative bacterial species provided herein.
  • It is to be understood that various foregoing aspects and embodiments overlap. It is intended that the embodiments recited above apply equally to the various aspects recited above.
  • It should be appreciated that all combinations of the foregoing concepts and additional concepts discussed in greater detail below (provided such concepts are not mutually inconsistent) are contemplated as being part of the inventive subject matter disclosed herein. In particular, all combinations of claimed subject matter appearing at the end of this disclosure are contemplated as being part of the inventive subject matter disclosed herein. It should also be appreciated that terminology explicitly employed herein that also may appear in any disclosure incorporated by reference should be accorded a meaning most consistent with the particular concepts disclosed herein.
  • BRIEF DESCRIPTION OF THE FIGURES
  • It is to be understood that the Figures are not necessarily to scale, emphasis instead being placed upon generally illustrating the various concepts discussed herein.
  • FIG. 1 is a proposed generic structure of isolated B. fragilis lipidated PSA. The structure includes a repeating tetrasaccharide unit having an average molecular weight of about 130 KDa, an oligosaccharide core unit comprising on average less 10 or fewer sugar residues, and a glycolipid. The glycolipid may comprise 3-5 acyl chains that may be N or O linked to a diglucosamine. The diglucosamine may be mono phosphorylated or non-phosphorylated.
  • FIG. 2 is one embodiment of a proposed generic structure of isolated B. fragilis glycolipid antigen (referred to herein as GLA) with no repeating tetrasaccharide attached. The structure includes a core oligosaccharide unit attached to a diglucosamine-comprising glycolipid moiety having 5 acyl chains. One glucosamine is phosphorylated. The linkage between the diglucosamine and the core oligosaccharide unit is an acid-labile ketosidic linkage. When the oligosaccharide core unit is linked to the repeating tetrasaccharide units of PSA (not shown), this occurs through a glycosidic linkage. The oligosaccharide core unit may comprise galactose, glucose or fucose sugars. It may further contain KDO residues that connect the diglucosamine to the remaining sugars of the core unit. As illustrated, it comprises 6 galactose units conjugated to 2 KDO residues conjugated, through a ketosidic linkage, to a diglucosamine conjugated to 5 acyl chains. The diglucosamine is mono phosphorylated. As taught herein, though, the glycolipid may be non-phosphorylated, the diglucosamine may be tri- or tetra-acylated, and there may be variation in the oligosaccharide core unit in terms of number and nature of sugars. Accordingly. GLA may minimally comprise the diglucosamine linked to a plurality of acyl chains (e.g., 3-5 chains).
  • FIG. 3 is a bar graph showing that B. fragilis GLA activates TLR2 similar to lipidated PSA. The graph further shows that GLA can activate TLR2 at much lower concentrations as compared to lipidated PSA. The graph shows IL-8 production in HEK cells that are stably transfected with TLR2 only (while, or first bar of each pair of bars) or transfected with TLR2 and TLR1 (diagonal striped, or second bar of each pair of bars) in response to purified lipidated PSA that does not contain any free lipid (referred to as PSA39) and B. fragilis GLA alone. HEK cells over-expressing TLR2 and TLR2/l were stimulated with PSA39 or B. frag GLA for 24 hours and IL-8 levels in the culture supernatant were measured with ELISA. The Figure shows that far less (e.g., about 20 fold less to about 100 fold less) GLA preparation is needed to stimulate the same level of IL-8 expression from the H11EK cells. For example, the 100 ng/ml of GLA induces about 100-fold more IL-8 than does the 10 microgram/ml of PSA39, even though the latter dose contains about 100 nanograms of GLA. TLR2 activation plays a role in innate immunity, so the ability of GLA to stimulate TLR2 indicates that GLA is able to stimulate innate immunity. The ability of lipidated PSA (PSA39) to stimulate TLR2 may be due to its glycolipid moiety which comprises only about 1% of the mass of the lipidated PSA.
  • FIGS. 4A-4C show that PSA with higher levels of GLA provides better protection in an EAE model than PSA alone. The Figures show that a preparation comprising lipidated PSA and GLA (at an approximately 4:1 ratio, w/w, referred to herein as Lot 35L) protects in an EAE model system better than purified lipidated PSA lacking any free glycolipid (Lot 39). C57BLU6 mice were orally administered purified lipidated PSA (Lot 39), or a preparation comprising lipidated PSA and “free” GLA (Lot 35L), or PBS every other day starting from one week before inducing the disease. Mice were challenged subcutaneously with MOG 35-55 in Complete Freund's Adjuvant. On days 0 and 2 after challenge, the mice received intraperitoneal Bordetella pertussis toxin. Mice were monitored and scored daily for disease progression. FIG. 4A shows the clinical EAE score of the mice, with a score of 5 indicating death. FIG. 4B shows the % weight loss of the mice. Significance of the results using Lot 35L to Lot 39 or to PBS is indicated by white asterisks (Lot 39 (8/8) and grey asterisks (PBS (8/8)), respectively. FIG. 4C shows the percent survival of the mice. Lot 35L, which contains more free GLA as compared to Lot 39, reduced the severity of the disease and the cumulative disease score, and increased survival rate of the mice. The presence of free GLA enhanced the protective activity of lipidated PSA.
  • FIG. 5 provides various metrics using the same EAE mouse model as described for FIGS. 4A-4C to show that PSA with higher levels of GLA provides better protection in the EAE model than PSA alone. The statistical significance of the three parameters in the top row and the parameter in lower left were all p<0.01. For example, the upper left plot shows the cumulative disease scores calculated as the sum of all EAE clinical scores for each group of mice. Purified lipidated PSA is indicated as 39 and a mixture of purified lipidated PSA and free GLA, in a roughly 4:1 w/w mixture, is indicated as 35L. In these experiments, about 100 micrograms of lipidated PSA (39) were used per mouse. Similarly about 100 micrograms of the 35L preparation were used per mouse, corresponding to about 80 micrograms of lipidated PSA and 20 micrograms of GLA.
  • FIGS. 6A-6D show that GLA acts as an adjuvant for lipidated PSA. The experiment was performed as explained for FIGS. 4A-4C. Lot 39 refers to purified lipidated PSA. GLA alone refers to purified GLA. Lot 39 and GL refers to a preparation made by combining Lot 39 and GLA in roughly a 4:1 w/w ratio. Similar results were obtained using a ratio of about 100:1. FIG. 6A shows the clinical EAE score of mice subjected to MOG-peptide induced EAE (p<0.01). Significance of the results using Lot 39 and GL together compared to GLA alone or Lot 39 alone is indicated by asterisks and circled asterisks (Lot 39), respectively, in FIG. 6A. FIG. 6B shows the % weight loss in the mice (p<0.05). FIG. 6C shows the percent survival of the mice. GLA alone failed to confer protection, while a reconstituted mixture of purified lipidated PSA and GLA (at a 4:1 w/w ratio) protected animals from EAE. FIG. 6D shows the cumulative disease scores calculated as the sum of all EAE clinical scores for each group.
  • FIGS. 7A-7B show that a delipidated PSA preparation (referred to as PSA35 delip) cannot protect in EAE. PSA35 refers to a preparation that comprises lipidated PSA and free GLA. PSA35 delip refers to a preparation of PSA35 that has been acid hydrolyzed to cleave the lipid moiety from lipidated PSA and has also undergone a lipid extraction with organic solvent chloroform in order to remove all free lipid including all released lipid. PSA35 delip likely comprises the repeating tetrasaccharide polysaccharide chain of PSA conjugated to the oligosaccharide core unit. It lacks the diglucosamine-containing glycolipid. The experiment was performed as explained in FIGS. 4A-4C. FIG. 7A shows the clinical EAE score of mice subjected to MOG-peptide induced EAE (p=0.001 on day 15). FIG. 7B shows the cumulative scores calculated as the sum of all EAE clinical scores for each group. Delipidated PSA35 failed to confer protection, while PSA35 protected animals from EAE indicating the importance of the glycolipid in such protection. In these experiments, on the order of about 10 micrograms of the PSA35 preparation was administered per mouse, representing about 8 micrograms of lipidated PSA and about 2 micrograms of GLA. This amount was about 10-fold lower than the amounts used in the experiments of the preceding Figures.
  • FIG. 8 is a schematic of a model of lipidated PSA induced immunomodulation. In this model, free GLA are shown to bind to a TLR2/1 dimer via the acyl chains, thereby stimulating an innate immune response. Although not shown, the free GLA may comprise the oligosaccharide core unit.
  • FIG. 9 shows the lipid profile of various lots of PSA (PSA39. PSA40 and PSA41) and GLA (GLA acap which was derived from an unencapsulated mutant and GLA41). Each lot was tested for the presence of tri-acylated, tri-acylated and phosphorylated, tetra-acylated, tetra-acylated and phosphorylated, penta-acylated, and penta-acylated and phosphorylated glycolipid moieties. The bars represent the amount of each glycolipid moiety as a percentage of the total lipid (y-axis). It is clear that each preparation contains the same lipid species but in different percentages. In particular, PSA41 and GLA41 contain the same lipid species, although the lipids do not appear to be similarly distributed between the PSA and GLA lots.
  • FIG. 10 shows an in vitro dose response (IL-10 production) of SpDC+Tcells co-cultured with dendritic cells and exposed to 10, 50 and 250 mg/ml of PSA39, PSA40 and PSA41. PSA41 appears more potent in its ability to induce IL-10 as compared to other two preparations.
  • FIG. 11 shows that interferon-β is induced by B. fragilis glycolipid whether or not attached to PSA. Model antigen presenting cells in the form of bone marrow derived dendritic cells (BMDCs) were stimulated for 6 hours with B. fragilis polysaccharide A (PSA) or the purified glycolipid anchor (GLA unencapsulated mutant), followed by collection of supernatants. A cytokine ELISA was used to detect levels of interferon-β (IFNβ) in the supernatants. PSA doses are shown stoichiometrically. The two curves would be superimposed if plotted on a molar basis because GLA is about 1% of a PSA molecule (i.e., 100 micrograms of PSA contain about 1 microgram of GLA).
  • FIG. 12 shows that different acylation structures of B. fragilis GLA induce low levels of pro-inflammatory cytokines such as TNFα. Model antigen presenting cells in the form of bone marrow derived dendritic cells (BMDCs) were stimulated for 6 hours with tri-, tetra- or penta-acylated B. fragilis GLA or hexa-acylated E. coli lipid A, followed by collection of supernatants. A cytokine ELISA was used to detect levels of TNFα in the supernatants. The three GLA species differ in their ability to induce TNFα, but they all induce a much lower level of TNFα as compared to E. coli lipid A.
  • FIG. 13 shows that tetra-acylated and penta-acylated B. fragilis lipid A antagonize production of pro-inflammatory TNFα secretion by E. coli hexa-acylated lipid A. Model antigen presenting cells in the form of bone marrow derived dendritic cells (BMDCs) were stimulated for 6 hours with purified tetra-acylated B. fragilis GLA and hexa-acylated E. coli lipid A (in a 5:1 ratio) or with purified penta-acylated B. fragilis GLA and hexa-acylated E. coli lipid A (in a 5:1 ratio), followed by collection of supernatants. A cytokine ELISA was used to detect levels of TNFα in the supernatants. Both the tetra-acylated and penta-acylated GLA were able to suppress the pro-inflammatory response to lipid A. This suggests a suppressive activity that may be useful, among other things, in treating gram negative bacterial infection.
  • DETAILED DESCRIPTION
  • Provided herein are compositions and methods of use relating to a glycolipid antigen comprising a diglucosamine-containing glycolipid. The glycolipid antigen, referred to herein as GLA, minimally comprises a diglucosamine-containing glycolipid. The diglucosamine moiety may be mono phosphorylated or non-phosphorylated (e.g., dephosphorylated). The glycolipid may comprise 3-5 acyl chains that are N- or O-linked to the diglucosamine.
  • The glycolipid may further be conjugated to an oligosaccharide, referred to herein as an oligosaccharide core unit. Such oligosaccharide may comprise 10 or fewer sugars. The sugars may be galactose, glucose and/or fucose. The oligosaccharides are typically conjugated to the diglucosamine via an acid-labile linkage such as but not limited to a ketosidic linkage.
  • Significantly, it has now been discovered, surprisingly, that the glycolipid is able to stimulate an innate immune response. This activity has been heretofore unrecognized. The finding is significant since it indicates that lipidated PSA is able to stimulate both innate and adaptive immune responses via different portions of its structure. Even more surprising is that free glycolipid, when combined with lipidated PSA, leads to enhanced protective activity in EAE mouse models. This suggests that free glycolipid is able to enhance immune responses to a greater degree than possible when used in its conjugated form. This disclosure provides methods of use of this glycolipid as an adjuvant together with PSA, whether or not lipidated, as well as a variety of other antigens. This newly discovered adjuvant activity indicates a role of the glycolipid in enhancing innate immunity, including when used alone, as well as in enhancing adaptive immunity when used together with an antigen. Examples of this latter application include use as an adjuvant for vaccines, and well as in immuno-oncology.
  • The glycolipid has been obtained from manipulation of B. fragilis cells. The starting material was a non-naturally, mutant occurring strain of B. fragilis that overexpresses PSA but not other capsular polysaccharides. The purification protocol may include a phenol/water extraction, and DNAse. RNase and pronase treatments. If the material is then treated with deoxycholate (DOC), the glycolipid comprising the oligosaccharide core unit is obtained. DOC treatment therefore separates the glycolipid from lipidated PSA. If however the material is treated instead with acid hydrolysis, the acid-labile linkage between the oligosaccharide core unit and the acyl-substituted diglucosamine is cleaved and the free acyl-substituted diglucosamine is obtained. If instead a purification is performed that does not involve either DOC treatment or acid hydrolysis (e.g., column chromatography), then the resulting preparation comprises about 20% (w/w) free glycolipid and about 80% (w/w) PSA including lipidated PSA. One such preparation is used in the experiments described herein, and is referred to as 35L or PSA35L or Lot 35L. As used herein, a “free” glycolipid refers to a glycolipid that is not covalently attached to a tetrasaccharide unit of PSA (i.e., it does not comprise the polysaccharide portion of PSA that is comprised of repeating tetrasaccharide units).
  • Structural analysis showed this lipid to belong to the class of lipid A molecules normally found in Gram negative bacteria. GLA, as it is referred to in this disclosure, comprises a glucosamine disaccharide with 3 to 5 acyl chains covalently bound to the sugars. It can be a mono phosphorylated or non-phosphorylated at the diglucosamine. One of the glucosamine residues is linked to an oligosaccharide core of 10 or fewer sugars, including 9, 8, 7 or less sugars, by an acid labile bond such as a ketosidic bond.
  • In order to enhance the harvest of intact glycolipid, the acid hydrolysis step was eliminated and lots were prepared with lipidated PSA and the free glycolipid. Such preparations are identified herein as 35L or PSA35L. These preparations were further analyzed and found to comprise on average about a 4:1 ratio of lipidated PSA to free GLA (w/w). In the in vivo EAE model, treatment with the 35L preparation delayed the EAE clinical outcome, and reduced the severity and the cumulative scores of the disease as compared to a purified lipidated PSA preparation referred to herein as PSA39 (DOC treated lipidated PSA preparation that did not contain free lipid (FIG. 4A, FIG. 5 upper left corner, and FIG. 4C). Next a mixture of free lipid and lipidated PSA was reconstituted at a 4:1 ratio (w/w), and this mixture was tested in the same experiment. The reconstituted mixture, referred to herein as “Lot 39+GL” conferred protection similar to that seen with 35L (FIGS. 6A, 6C and 6D). Moreover, when the PSA35L preparation (comprising lipidated PSA and free lipid in a 4:1 w/w ratio) was hydrolyzed and delipidated after purification, it lost its ability to protect (FIGS. 7A-7B).
  • Further analysis revealed that GLA activates TLR2/TLR 1 expressing HEK cells in a fashion similar to that seen with lipidated PSA (FIG. 3 ).
  • Taken together, the data suggest that GLA acts as an adjuvant to induce innate immunity. Such activity may occur through engagement and signaling of the TLR2/TLR1 heterodimer complex, and it likely enhances immune responses to PSA. In addition, the findings indicate use of GLA in enhancing immune responses to other antigens including but not limited to bacterial antigens, cancer antigens, and the like.
  • Provided herein is the structural identification and characterization of the glycolipid moiety of lipidated PSA. It has been found in accordance with this disclosure that these glycolipids are comprised of a diglucosamine substituted with three, four or five acyl chains. The glycolipid in some instances further comprises an oligosaccharide core unit conjugated to the diglucosamine via an acid-labile ketosidic bond. This glycolipid has been shown to activate an innate immune response and to enhance an immune response to lipidated PSA.
  • The glycolipid structure will now be described in greater detail in the context of lipidated PSA.
  • Lipidated PSA
  • A form of lipidated PSA isolated from mutant B. fragilis cells comprises a polysaccharide component conjugated, at its reducing end, an oligosaccharide core unit, and a diglucosamine conjugated to 3-5 acyl chains. As described in greater detail herein, the innate immunostimulatory activity appears attributable to the diglucosamine substituted with acyl chains. The oligosaccharide core unit, by virtue of its hydrophilicity, helps to render the lipid component more water soluble. Thus, while the immunostimulatory activity is believed to exist in the acyl chains, typically the GLA of this disclosure will further comprise the oligosaccharide core unit in order to facilitate its dissolution in an aqueous solution, such as but not limited to a pharmaceutical formulation.
  • Polysaccharide Component
  • The polysaccharide component of lipidated PSA, referred to herein as PSA, comprises a tetrasaccharide repeating unit shown below. It possesses zwitterionic behavior as conferred by a positive charge on its free amine group and a negative charge on its free carboxyl group (per repeating tetrasaccharide unit). Its naturally occurring state has been reported to comprise over 60 tetrasaccharide repeating units (e.g., up to and including in some instances about 100, or about 200, or about 300 repeated units on average), and it has an average molecular size of about 150 kD (with a range of about 75 kD to 240 kD).
  • The repeating tetrasaccharide unit of PSA has a structure as follows:
  • Figure US20230130767A1-20230427-C00001
  • The tetrasaccharide repeating unit may also be expressed as follows:
  • Figure US20230130767A1-20230427-C00002
  • Synthetic forms of lipidated PSA may comprise comprising various ranges of tetrasaccharide units (e.g., 1-60, 1-50, 1-40, 1-30, 1-20, 1-10, 1-9, 1-8, 1-7, 1-6, or 1-5 tetrasaccharide units). Such shorter variants can be obtained by depolymerizing naturally occurring lipidated PSA or by depolymerizing PSA obtained from lipidated PSA. PSA can be depolymerized using for example chemical means (e.g., using reactive oxygen species or reactive nitrogen species such as but not limited to nitrogen monoxide, as described in Duan and Kasper, Glycobiology, 2011, 21(4):401-409), mechanical means, and/or enzymatic means that are known in the art.
  • The invention further contemplates synthetic forms of lipidated PSA comprising more than 300 repeating tetrasaccharide units, including without limitation 350, 400, 500, 600, 700, 800, 900 or 1000 units or more.
  • As described herein, the polysaccharide component may be covalently conjugated to the glycolipid, or in certain synthetic forms it may be unconjugated to the glycolipid. If covalently conjugated, it may be conjugated via a glycosidic bond to the oligosaccharide core unit. In other embodiments, it may be conjugated via a ketosidic bond or other acid labile bond or via a bond such as an ester, an amide, or an ether bond to form a non-naturally occurring lipidated PSA.
  • Glycolipid Component
  • The glycolipid component comprises a diglucosamine substituted with one or more acyl chains, and preferably 3, 4 or 5 acyl chains. An exemplary diglucosamine in the context of a glycolipid is provided in FIG. 2 . It is now recognized in accordance with this disclosure that the diglucosamine is conjugated to the oligosaccharide core unit via a ketosidic bond that is acid-labile and thus susceptible to stringent hydrolysis.
  • The disaccharide may be conjugated to one or more acyl chains, including three, four, or five acyl chains in some instances via for example ester or amide linkages, and thus may be referred to as “O” substituted (or O-linked) or “N” substituted (or N-linked) respectively. In some instances, each glycolipid (or GLA, as used herein) comprises three, four or five acyl chains. Accordingly, the glycolipid may be referred to herein as tri-acylated, tetra-acylated or penta-acylated forms.
  • The acyl chains may range in length from 14 to 17 carbons, in some instances. The acyl chains may be unmodified or they may be modified. If modified, the acyl chains may be hydroxy-modified. Thus, in some instances, the glycolipid may comprise one or more acyl chains characterized as C14:0, C14:0-OH, C15:0, C15:0-OH, C16:0, C16:0-OH, C17:0, and C17:0-OH.
  • A single preparation of glycolipid may yield a number glycolipid species that diffe from each other with respect to acyl chain number. The exact composition of a glycolipid may be determined using mass spectrometry (MS), wherein different glycolipid species give rise to different and discernable spectra.
  • Examples of different penta-acylated species include:
  • (1) one chain of C16:0-OH, three chains of C17:0-OH, and one chain of C15:0,
  • (2) two chains of C16:0-OH, two chains of C17:0-OH, and one chain of C15:0,
  • (3) three chains of C16:0 OH, one chain of C17:0-OH, and one chain of C15:0,
  • (4) four chains of C16:0-OH, and one chain of C15:0, and
  • (5) four chains of C16:0-OH, and one chain of C14:0.
  • Various species of tetra-acylated and tri-acylated species are similarly contemplated.
  • It will therefore be appreciated glycolipids of this disclosure, whether isolated from B. fragilis or generated de novo, and whether conjugated or unconjugated to an oligosaccharide core unit, may comprise any of the foregoing combinations of acyl chains, without limitation:
  • (1) C16:0-OH acyl chain(s) only,
  • (2) C17:0-OH acyl chain(s) only,
  • (3) C16:0-OH and C17:0-OH chain(s) only,
  • (4) C16:0-OH and C17:0-OH and C15:0 chain(s) only.
  • (5) C16:0-OH and C17:0-OH and C14:0 chain(s).
  • The number of each type of chain may vary, and may include without limitation the following options
  • (1) 0-4 C16:0-OH chains,
  • (2) 0-4 C17:0-OH chains,
  • (3) 0 or 1 C14:0 chains, and
  • (4) 0 or 1 C15:0 chains.
  • The foregoing examples are not to be considered limiting, and rather the invention contemplates various combinations, and combinations of the foregoing, to be used in compositions provided herein.
  • The invention provides defined glycolipid mixtures, having known, and thus optionally pre-defined, glycolipid content and composition, and optionally known and/or pre-defined polysaccharide (e.g., PSA) to glycolipid ratios or non-PSA antigen to glycolipid ratios. Thus, the glycolipids of this disclosure may be characterized in terms of any of these structural features, thereby further distinguishing these products from those of the prior art, and where necessary from naturally occurring products. For example, based on the teachings provided herein, the invention provides compositions comprising glycolipids that are only or predominantly (e.g., greater than 50%, or at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%) tri-acylated, or tetra-acylated, or penta-acylated, or some combination thereof including but not limited to tetra- and penta-acylated. Such chemically defined compositions were not heretofore contemplated or possible.
  • Isolated Forms
  • The preparations provided herein can be characterized by their content of released or free glycolipids. Such content can be about 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% or less. Such content can be less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, less 0.1%, less than 0.05%, less than 0.001%, less than 0.0005%, less than 0.0001% (w/w of released glycolipid to lipidated PSA). The amount of glycolipid may be determined for example using the gel electrophoresis methods or mass spec methods.
  • The glycolipid may also be considered to be pure (i.e., it is free or substantially free of contaminant). The degree of purity may be at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9%, or higher.
  • Accordingly, the invention provides compositions comprising glycolipids including compositions comprising isolated glycolipid at a purity and/or a concentration that has not been heretofore achieved. Also provided are compositions comprising or consisting essentially of particular species of glycolipid or particular subsets of species of glycolipid. These species may be characterized and thus distinguished from other species and from bulk glycolipid in terms of their lipid and oligosaccharide components. The lipid components may be characterized by the number, position and type of acyl chains they possess. For example, a composition may comprise at least 70%, 75%, 80%, 85%, 90%, 95%, or mom of a tri-acetylated, tetra-acylated and/or penta-acylated glycolipid.
  • The compositions may be defined by their degree of purity, for example with respect to their glycolipid components, or with respect to their content of contaminants such as non-lipidated PSA. The compositions may be defined by their concentration of glycolipid components, or by their ratio of glycolipid to other components.
  • Synthetic Forms
  • The invention further provides additional synthetic, non-naturally occurring species of glycolipids.
  • Glycolipid Compositions
  • Isolated intends that the glycolipid (or other agent) is provided in a more pure form or a more concentration form compared to its synthesized or obtained form. An “isolated” glycolipid may be a glycolipid that is prepared or obtained from B. fragilis, and is physically separated from a natural environment (e.g., a B. fragilis cell, components of the B. fragilis cell, and/or components of the B. fragilis cell capsular complex such as but not limited to PSB). It does not intend that any naturally occurring product has the same structure of glycolipid as described herein.
  • In some embodiments, the compositions are substantially free of naturally occurring contaminants such as nucleic acids (e.g., DNA and RNA), proteins, and other components of B. fragilis and/or the B. fragilis capsule. Substantially free, as used herein, intends that these contaminants represent about or less than 5%, less than 1%, less than 0.5%, or less than 0.1% (or less) by weight (weight of the contaminant to weight of the glycolipid). In some instances, such contaminants may be undetectable.
  • Various compositions may or may not contain LPS. LPS may be present in an amount of about 0.5% (w/w of LPS relative to other components in a composition).
  • Some compositions may comprise at least about 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more (w/w) of glycolipid.
  • Some compositions may comprise at least about 95%, 96%, 97%, 98%, 99%, or more (w/w) of lipidated PSA and less than 5%, less than 4%, less than 3%, less than 2%, less than 1%, less than 0.5%, or less of free, released glycolipid.
  • Methods of Use
  • Also provided are methods of using the various forms of glycolipid in vitro and in vivo. The various glycolipid forms herein can be used as immunomodulators including immunostimulators, particularly in view of their enhanced ability to stimulate antigen presenting cells such as plasmacytoid dendritic cells. These forms are contemplated for use in vitro and in vivo. In vitro uses include use as an analytical tool (e.g., for screening assays) and as an assay standard or control (e.g., as a positive control or a comparator in an in vitro assay such as a IL-8 induction assay). In vivo uses include uses in animal models and also in human and non-human subjects to treat or prevent conditions that would benefit from such immunomodulation including immunostimulation. Such conditions include those in which an enhanced innate and/or adaptive immune response is beneficial. Other conditions include but are not limited to autoimmune disorders (e.g., multiple sclerosis and inflammatory bowel disease).
  • The glycolipid component may be used as a single agent, or it may be used in combination with other agents such as antigens or cells such as antigen-presenting cells. It may be used to stimulate antigen-presenting cells in vivo or in vitro. One application may involve obtaining antigen-presenting cells from a subject, stimulating such cells ex vivo with the glycolipid of this disclosure with or without antigen, followed by re-introduction of such cells with and without glycolipid and/or antigen to the subject. Alternatively, the glycolipid may be administered to a subject with and/or without antigen. Such immunization or vaccination protocols may involve several administrations of glycolipid and/or antigen. The administration of the glycolipid and the antigen may occur simultaneously (typically occurs if they are formulated together), or substantially simultaneously (typically occurs if they are formulated and administered separately but still within a short period of time in order to work in concert in a subject), and/or they may be formulated and administered separately (e.g., the glycolipid is administered before or after the antigen, including for example hours, days and/or weeks before or after).
  • The invention further contemplates use of the free polysaccharide component of lipidated PSA (i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid component of lipidated PSA (including where such glycolipid comprises the oligosaccharide core unit) and the free glycolipid component obtained from lipidated PSA.
  • As another example, the polysaccharide and glycolipid may be used together in an unconjugated form.
  • The invention further contemplates use of the free PSA (i.e., a chain of repeating tetrasaccharide units that is not conjugated to the glycolipid (including where such glycolipid comprises the oligosaccharide core unit) and free glycolipid obtained from lipidated PSA.
  • In vivo uses include but are not limited to those involving human subjects. For example, in vivo uses include administration of the glycolipid and compositions thereof to a non-human subject in order to modulate an immune response, for example as a positive control or a comparator.
  • Infections
  • The glycolipids and compositions thereof may be administered to a subject having or at risk of having an infection such as a bacterial infection, a viral infection, a fungal infection, a parasitic infection, or a mycobacterial infection. In these aspects, the glycolipids may be used and/or administered with another therapeutic such as an anti-bacterial agent, an anti-viral agent, an anti-fungal agent, an anti-parasitic agent, or anti-mycobacterial agent. In these and other aspects, the glycolipids may be used and/or administered with an antigen in order to induce or enhance an immune response directed to the antigen (i.e., an antigen-specific immune response). Such antigens include bacterial antigens, viral antigens, fungal antigens, parasitic antigens, and mycobacterial antigens.
  • Anti-bacterial agents include broad spectrum antibiotics, narrow spectrum antibiotics, or limited spectrum antibiotics. The anti-bacterial agent may also be a cell wall synthesis inhibitor, cell membrane inhibitor, protein synthesis inhibitor, nucleic acid synthesis or functional inhibitor or a competitive inhibitor.
  • Anti-viral agents include immunoglobulin, amantadine, interferon, nucleoside analogue, nonnucleoside analogue, bioflavonoid and protease inhibitor, although it is not so limited. In one embodiment, the protease inhibitor is indinavir, saquinavir, ritonavir, and nelfinavir. In another embodiment, the bioflavonoid is robustaflavone, amentoflavone, or a derivative or salt thereof. In yet another embodiment, the non-nucleoside analogue is selected from the group consisting of delavirdine, nevirapine, efavirenz, alpha-interferon, recombinant CD4, amantadine, rimantadine, ribavirin and vidarabine.
  • Anti-fungal agents include imidazole. FK 463, amphotericin B. BAY 38-9502, MK 991, pradimicin, UK 292, butenafine, chitinase and 501 cream.
  • Anti-parasitic agents include albendazole, amphotericin B, benznidazole, bithionol, chloroquine HCl, chloroquine phosphate, clindamycin, dehydroemetine, diethylcarbamazine, diloxanide furoate, eflornithine, furazolidaone, glucocorticoids, halofantrine, iodoquinol, ivermectin, mebendazole, mefloquine, meglumine antimoniate, melarsoprol, metrifonate, metronidazole, niclosamide, nifurtimox, oxamniquine, paromomycin, pentamidine isethionate, piperazine, praziquantel, primaquine phosphate, proguanil, pyrantel pamoate, pyrimethamine-sulfonamides, pyrimethamine-sulfadoxine, quinacrine HCl, quinine sulfate, quinidine gluconate, spiramycin, stibogluconate sodium (sodium antimony gluconate), suramin, tetracycline, doxycycline, thiabendazole, tinidazole, trimethroprim-sulfamethoxazole, and tryparsamide.
  • Anti-mycobacterial agent include anti-tuberculosis agent such as isoniazid, rifampin, rifabutin, rifapentine, pyrazinamide, ethambutol, (+)calanolide A, (−)-calanolide A, (−)-soulattrolide, (−)-costatolide or (−)-7,8-dihydrosoulattrolide. Other anti-mycobacterial agents include streptomycin, dapsone, clarithromycin, ciprofloxacin, clofazamine, azithromycin, ethionamide, amikacin or resorcinomycin A.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a bacterial infection. Examples of bacterial infections to be treated according to this disclosure include but are not limited to an E. coli infection, a Staphylococcal infection, a Streptococcal infection, a Pseudomonas infection, Clostridium difficile infection. Legionella infection, Pneumococcus infection, Haemophilus infection, Klebsiella infection, Enterobacter infection, Citrobacter infection, Neisseria infection, Shigella infection, Salmonella infection, Listeria infection, Pasteurella infection, Streptobacillus infection. Spirillum infection, Treponema infection, Actinomyces infection, Borrelia infection, Corynebacterium infection, Nocardia infection, Gardnerella infection, Campylobacter infection, Spirochaeta infection, Proteus infection, Bacteroides infection, H. pylori infection, and anthrax infection.
  • In some embodiments, the subjects may have or may be at risk of developing a gram-negative bacterial infection. Gram negative bacteria include but are not limited to Escherichia coli (E. coli), Pseudomonas aeruginosa, Neisseria meningitides, Neisseria gonorrhoeae, Chlamydia trachomatis, Yersinia pestis, Non-typable Hemophilus influenzae, Hemophilus ducreyi, Helicobacter pylori, Campylobacter jejuni; Bacteroides fragilis. B. thetaiotamicron, B vulgatis, Citrobacter rodentium, Haemophilus influenza, Vibrio cholerae, Salmonella shigella, Salmonella enteritidis. Salmonella enterica serovar typhi. Salmonella enterica serovar typhimurium, Klebsiella pneumoniae, Legionella pneumophila, Proteus mirabilis, Enterobacter cloacae, and Serratia marcescens. In some embodiments, the tetra- and penta-acylated glycolipids provided herein, and compositions thereof, are used to treat gram-negative bacterial infections, such as any of the foregoing.
  • In some embodiments, the subject is experiencing sepsis.
  • In some embodiments, the subject does not have and is not at risk of developing a B. fragilis infection. In some embodiments, the subject does have or is at risk of developing a B. fragilis infection or a condition associated with type of infection.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a mycobacterial infection. Examples of mycobacterial infections to be treated according to this disclosure include but arm not limited to tuberculosis or leprosy respectively caused by the M. tuberculosis and M. leprae species.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a viral infection. Examples of viral infections to be treated according to this disclosure include but are not limited to a Zika infection, an HIV infection, a Herpes simplex virus 1 infection, a Herpes simplex virus 2 infection, cytomegalovirus infection, hepatitis A virus infection, hepatitis B virus infection, hepatitis C virus infection, human papilloma virus infection, Epstein Barr virus infection, rotavirus infection, adenovirus infection, influenza A virus infection, respiratory syncytial virus infection, varicella-zoster virus infections, small pox infection, monkey pox infection, and SARS infection.
  • Subjects to be treated according to this disclosure may have or may be at risk of developing a parasitic infection. Examples of parasitic infections to be treated according to this disclosure include but are not limited to candidiasis, ringworm, histoplasmosis, blastomycosis, paracoccidioidomycosis, crytococcosis, aspergillosis, chromomycosis, mycetoma infections, pseudallescheriasis, and tinea versicolor infection. Still other parasitic infections that may be treated using the glycolipids of this disclosure include amebiasis, Trypanosoma cruzi infection, Fascioliasis. Leishmaniasis, Plasmodium infections, Onchocerciasis. Paragonimiasis, Trypanosoma brucei infection, Pneumocystis infection, Trichomonas vaginalis infection. Taenia infection, Hymenolepsis infection, Echinococcus infections, Schistosomiasis, neurocysticercosis, Necator americanus infection, and Trichuris trichiura infection.
  • Immune Therapies
  • The glycolipids and compositions thereof may be used to enhance immune therapies for a number of indications, both in a therapeutic and a prophylactic sense. Immune therapies include but are not limited to passive immune therapies such as immunoglobulin administration, and active immune therapies such as vaccination with antigens alone or antigens in the context of dendritic cells. The methods are intended to treat or prevent various indications that would benefit from an enhanced immune response.
  • In some embodiments, the immune response that is stimulated is a cell-mediated immune response involving antigen presenting cells and optionally T cells such as Treg cells. In some embodiments, the immune response is an innate immune response, while in others it is an adaptive immune response, while in yet others it is a combined innate and adaptive immune response. The immune response may, in some instances, be antigen specific.
  • The immune therapy may involve administration of an antibody or antigen-binding antibody fragment. The antibody or antigen-binding fragment thereof may be specific for a tumor vasculature molecule. Tumor vasculature molecules include but are not limited to endoglin, ELAM-1, VCAM-1. ICAM-1, ligand reactive with LAM-1, MHC class H antigens, amino phospholipids such as phosphatidylserine and phosphatidylethanolamine, VEGFR1 (Flt-1) and VEGFR2 (KDR/Flk-1). Antibodies to endoglin include TEC-4 and TEC-11. Antibodies that inhibit VEGF include 2C3 (ATCC PTA 1595). Other antibodies that are specific for tumor vasculature include antibodies that react to a complex of a growth factor and its receptor such as a complex of FGF and the FGFR or a complex of TGFβ and the TGFβR. Antibodies of this latter class include GV39 and GV97.
  • In a related embodiment, the antibody or antibody fragment is selected from the group consisting of trastuzumab, alemtuzumab (B cell chronic lymphocytic leukemia), gemtuzumab ozogamicin (CD33+acute myeloid leukemia), hP67.6 (CD33+acute myeloid leukemia), infliximab (inflammatory bowel disease and rheumatoid arthritis), etanercept (rheumatoid arthritis), rituximab, tositumomab, MDX-210, oregovomab, anti-EGF receptor mAb, MDX-447, anti-tissue factor protein (TF), (Sunol); ior-c5, c5, edrecolomab, ibritumomab tiuxetan, anti-idiotypic mAb mimic of ganglioside GD3 epitope, anti-HLA-Dr10 mAb, anti-CD33 humanized mAb, anti-CD52 humAb, anti-CD1 mAb (ior t6), MDX-22, celogovab, anti-17-1A mAb, bevacizumab, daclizumab, anti-TAG-72 (MDX-220), anti-idiotypic mAb mimic of high molecular weight proteoglycan (I-Mel-1), anti-idiotypic mAb mimic of high molecular weight proteoglycan (I-Mel-2), anti-CEA Ab, hmAbH11, anti-DNA or DNA-associated proteins (histones) mAb. Gliomab-H mAb, GNT-250 mAb, anti-CD22, CMA 676), anti-idiotypic human mAb to GD2 ganglioside, ior egf/r3, anti-ior c2 glycoprotein mAb, ior c5, anti-FLK-2/FLT-3 mAb, anti-GD-2 bispecific mAb, antinuclear autoantibodies, anti-HLA-DR Ab, anti-CEA mAb, palivizumab, bevacizumab, alemtuzumab. BLyS-mAb, anti-VEGF2, anti-Trail receptor; B3 mAb, mAb BR96, breast cancer; and Abx-Cbl mAb.
  • The antibody or antibody fragment may be an anti-HER2 antibody, such as trastuzumab. The antibody or antibody fragment may be an anti-CD20 antibody, such as rituximab.
  • In still another aspect, provided herein is a method for shortening a vaccination course. As used herein, “shortening a vaccination course” refers to reducing either the number of vaccine administrations (e.g., by injection) or the time between vaccine administrations. This is accomplished by stimulating a more robust immune response in the subject. The method may involve, in one embodiment, administering to a subject in need of immunization a glycolipid in an amount effective to induce an antigen-specific immune response to a vaccine administered in a vaccination course, wherein the vaccination course is shortened by at least one immunization. In other embodiments, the vaccination course is shortened by one immunization, two immunizations, three immunizations, or more. The method may involve, in another embodiment, administering to a subject in need of immunization a glycolipid of this disclosure in an amount effective to induce an antigen-specific immune response to a vaccine administered in a vaccination course, wherein the vaccination course is shortened by at least one day. In other embodiments, the vaccination course is shortened by one day, two days, three days, four days, five days, six days, one week, two weeks, three weeks, four weeks, one month, two months or more. In one embodiment, the glycolipid is administered substantially simultaneously with the vaccine.
  • Immunizations that can be modified in this way include but are not limited to newborn immunizations for HBV; immunizations at for example two months of age for Polio. DTaP, Hib, HBV, Pneumococcus; immunizations at for example four months of age for Polio, DTaP, Hib, Pneumococcus; immunizations at for example six months of age for Polio, DTaP, Hib, HBV, Pneumococcus; immunizations at for example 12-15 months of age for Hib, Pneumococcus, MMR, Varicella; immunizations at for example 15-18 months of age for DtaP; immunizations at for example 4-6 years of age for Polio, DPT, MMR; immunizations at for example 11-12 years of age for MMR; immunizations at for example 14-16 years of age for tetanus-diphtheria (i.e., Td) (with a repeat as a booster every 10 years).
  • As an example, a recommended vaccination course for tetanus/diphtheria includes a primary immunization series given in adults if not received as a child, followed by routine booster doses of tetanus-diphtheria (Td) every 10 years. The method of the invention will allow for a shortened series of vaccinations at the first time point, and may in some instances obviate the need for booster shoots later on. As another example, hepatitis vaccination commonly requires three administrations spaced at least two weeks, and sometimes one month, apart in order to develop full immunity. Using the methods of the invention, it is possible to either reduce the number of injections from three to two or one, or to reduce the time in between injections from weeks or months to days or weeks. Vaccination courses that can be shortened by the method of the invention include but are not limited to: HBV: Hepatitis B vaccine (3 total doses currently recommended); Polio: Inactivated polio vaccine (4 total doses currently recommended); DTaP: Diphtheria/tetanus/acellular Pertussis (3-in-1 vaccine; 5 total doses currently recommended); Hib: Haemophilus influenzae type b conjugate vaccine (4 total doses currently recommended); Pneumococcus (Prevnar): Protects against certain forms of Strep. Pneumoniae (3 total doses recommended); MMR: measles/mumps/rubella (3-in-1 vaccine; 2 total doses recommended): Td: Adult tetanus/diphtheria (2-in-1 vaccine; for use in people over age 7). In another embodiment, the glycolipids can be used together with oral polio vaccine.
  • Cancer
  • The glycolipids and compositions thereof may be administered to a subject having or at risk of developing cancer. The cancer may be one for which a cancer antigen is known.
  • Examples of cancer that may be treated according to this disclosure include basal cell carcinoma, biliary tract cancer; bladder cancer: bone cancer, brain cancer, breast cancer, cervical cancer; choriocarcinoma; CNS cancer; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer, cancer of the head and neck; gastric cancer; intra-epithelial neoplasm; kidney cancer: larynx cancer; leukemia; acute myeloid leukemia, acute lymphoid leukemia, chronic myeloid leukemia, chronic lymphoid leukemia, liver cancer; small cell lung cancer; non-small cell lung cancer, lymphoma, Hodgkin's lymphoma; Non-Hodgkin's lymphoma; melanoma; myeloma; neuroblastoma; oral cavity cancer; ovarian cancer, pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer, renal cancer; cancer of the respiratory system; sarcoma; skin cancer, stomach cancer; testicular cancer; thyroid cancer; uterine cancer; and cancer of the urinary system.
  • The glycolipids and compositions thereof may be administered with another therapeutic agent or modality such as but not limited to antibody or other biologic, chemotherapy, radiation, surgery and the like. The glycolipids and compositions thereof may be administered with a cancer antigen or a cancer immunotherapy such as an antibody specific for a cancer antigen.
  • The chemotherapy may be selected from the group consisting of aldesleukin, asparaginase, bleomycin sulfate, carboplatin, chlorambucil, cisplatin, cladribine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin hydrochloride, docetaxel, doxorubicin, doxorubicin hydrochloride, epirubicin hydrochloride, etoposide, etoposide phosphate, floxuridine, fludarabine, fluorouracil, gemcitabine, gemcitabine hydrochloride, hydroxyurea, idarubicin hydrochloride, ifosfamide, interferons, interferon-α2a, interferon-α2b, interferon-αn3, interferon-α1b, interleukins, irinotecan, mechlorethamine hydrochloride, melphalan, mercatopurine, methotrexate, methotrexate sodium, mitomycin, mitoxantrone, paclitaxel, pegaspargase, pentostatin, prednisone, profimer sodium, procabazine hydrochloride, taxol, taxotere, teniposide, topotecan hydrochloride, vinblastine sulfate, vincristine sulfate or vinorelbine tartrate.
  • The cancer immunotherapy may include an antibody or antibody fragment specific for a cell surface molecule. Cell surface molecules that may be targeted with the antibody or antibody fragment include but are not limited to HER 2, CD20, CD33, EGF receptor, HLA markers such as HLA-DR, CD52, CD1, CEA, CD22, GD2 ganglioside, FLK2/FLT3, VEGF, VEGFR, and the like.
  • The antibody or antibody fragment may be specific for a cancer antigen. Cancer antigens that may be targeted with the antibody or antibody fragment include but are not limited to HER 2 (p185). CD20, CD33. GD3 ganglioside, GD2 ganglioside, carcinoembryonic antigen (CEA), CD22, milk mucin core protein, TAG-72. Lewis A antigen, ovarian associated antigens such as OV-TL3 and MOv18, high Mr melanoma antigens recognized by antibody 9.2.27, HMFG-2, SM-3, B72.3, PR5C5, PR4D2, and the like. Other cancer antigens are described in U.S. Pat. No. 5,776,427.
  • Cancer antigens can be classified in a variety of ways. Cancer antigens include antigens encoded by genes that have undergone chromosomal alteration. Many of these antigens are found in lymphoma and leukemia. Even within this classification, antigens can be characterized as those that involve activation of quiescent genes. These include BCL-1 and IgH (Mantel cell lymphoma). BCL-2 and IgH (Follicular lymphoma), BCL-6 (Diffuse large B-cell lymphoma). TAL-1 and TCRδ or SIL (T-cell acute lymphoblastic leukemia), c-MYC and IgH or IgL (Burkitt lymphoma), MUN/IRF4 and IgH (Myeloma), PAX-5 (BSAP) (Immunocytoma).
  • Other cancer antigens that involve chromosomal alteration and thereby create a novel fusion gene and/or protein include RARα, PML, PLZF, NPM or NuMA (Acute promyelocytic leukemia), BCR and ABL (Chronic myeloid/acute lymphoblastic leukemia), MLL (HRX) (Acute leukemia). E2A and PBX or HLF (B-cell acute lymphoblastic leukemia). NPM, ALK (Anaplastic large cell leukemia), and NPM, MLF-1 (Myelodysplastic syndrome/acute myeloid leukemia).
  • Other cancer antigens are specific to a tissue or cell lineage. These include cell surface proteins such as CD20, CD22 (Non-Hodgkin's lymphoma. B-cell lymphoma, chronic lymphocytic leukemia (CLL)), CD52 (B-cell CLL), CD33 (acute myelogenous leukemia (AML)), CD10 (gp100) (common (pre-B) acute lymphocytic leukemia and malignant melanoma), CD3/T-cell receptor (TCR) (T-cell lymphoma and leukemia), CD79/B-cell receptor (BCR) (B-cell lymphoma and leukemia). CD26 (epithelial and lymphoid malignancies), human leukocyte antigen (HLA)-DR. HLA-DP, and HLA-DQ (lymphoid malignancies), RCAS1 (gynecological carcinomas, bilary adenocarcinomas and ductal adenocarcinomas of the pancreas), and prostate specific membrane antigen (prostate cancer).
  • Tissue- or lineage-specific cancer antigens also include epidermal growth factor receptors (high expression) such as EGFR (HER1 or erbB1) and EGFRvIII (brain, lung, breast, prostate and stomach cancer), erbB2 (HER2 or HER2/neu) (breast cancer and gastric cancer), erbB3 (HER3) (adenocarcinoma), and erbB4 (HER4) (breast cancer).
  • Tissue- or lineage-specific cancer antigens also include cell-associated proteins such as tyrosinase, melan-A/MART-1, tyrosinase related protein (TRP)-1/gp75 (malignant melanoma), polymorphic epithelial mucin (PEM) (breast tumors), and human epithelial mucin (MUC1) (breast, ovarian, colon and lung cancers).
  • Tissue- or lineage-specific cancer antigens also include secreted proteins such as monoclonal immunoglobulin (multiple myeloma and plasmacytoma), immunoglobulin light chains (Multiple Mycloma), α-fetoprotcin (liver carcinoma), kallikrcins 6 and 10 (ovarian cancer), gastrin-releasing peptide/bombesin (lung carcinoma), and prostate specific antigen (prostate cancer).
  • Still other cancer antigens are cancer testis (CT) antigens that are expressed in some normal tissues such as testis and in some cases placenta. Their expression is common in tumors of diverse lineages and as a group the antigens form targets for immunotherapy. Examples of tumor expression of CT antigens include MAGE-A1, -A3, -A6, -A12, BAGE, GAGE, HAGE, LAGE-1, NY-ESO-1, RAGE, SSX-1, -2, -3, -4, -5, -6, -7, -8, -9, HOM-TES-14/SCP-1, HOM-TES-85 and PRAME. Still other examples of CT antigens and the cancers in which they are expressed include SSX-2, and -4 (Neuroblastoma). SSX-2 (HOM-MEL-40), MAGE. GAGE, BAGE and PRAME (Malignant melanoma), HOM-TES-14/SCP-1 (Meningioma), SSX-4 (Oligodendrioglioma), HOM-TES-14/SCP-1, MAGE-3 and SSX-4 (Astrocytoma), SSX member (Head and neck cancer, ovarian cancer, lymphoid tumors, colorectal cancer and breast cancer), RAGE-1, -2, -4, GAGE-1, -2, -3, -4, -5, -6, -7 and -8 (Head and neck squamous cell carcinoma (HNSCC)), HOM-TES14/SCP-1, PRAME, SSX-1 and CT-7 (Non-Hodgkin's lymphoma), and PRAME (Acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) and chronic lymphocytic leukemia (CLL)).
  • Other cancer antigens are not specific to a particular tissue or cell lineage. These include members of the carcinoembryonic antigen (CEA) family: CD66a, CD66b, CD66c, CD66d and CD66e. These antigens can be expressed in many different malignant tumors and can be targeted by immunotherapy. Still other cancer antigens are viral proteins and these include Human papilloma virus protein (cervical cancer), and EBV-encoded nuclear antigen (EBNA)-1 (lymphomas of the neck and oral cancer). Still other cancer antigens are mutated or aberrantly expressed molecules such as but not limited to CDK4 and beta-catenin (melanoma).
  • Autoimmune Diseases
  • Also contemplated are methods of modulating immune responses in a subject by administering to such subject the glycolipid described herein. The subject may be one having or likely to develop an aberrant immune response. The aberrant immune response may be an enhanced immune response and the glycolipid acts to down-regulate the immune response. Enhanced immune responses are typically associated with inflammatory conditions, such as but not limited to autoimmune diseases. Autoimmune diseases, autoimmune disorders and autoimmune conditions are used interchangeably herein.
  • Accordingly, the compositions comprising glycolipid as a single agent or in combination with an antigen other than lipidated PSA, may be used to modulate (e.g., down-regulate) certain immune responses in subjects having or at risk of developing autoimmune diseases. As will be understood by those of ordinary skill in the art, subjects having autoimmune diseases typically experience one or more “events” or recurrences associated with the autoimmune disease. For example, a subject having inflammatory bowel disease may experience temporally isolated attacks of the disease, characterized by the presence of symptoms or increased severity of symptoms. The invention contemplates that the compositions may be used in such subjects to reduce the likelihood of such future recurrences of the disease or to reduce the severity of symptoms associated with the disease (e.g., pain, fever, discomfort, fatigue, etc.). Thus, the compositions may be administered prior to such recurrence, and in this manner may be chronically administered, optionally at a regular frequency. Examples include once a day, once every 2, 3, 4, 5 or 6 days, or once a week, etc. The invention also contemplates that the compositions may be administered to the subject during a recurrence in order to reduce the severity of symptoms or shorten the time of the recurrence.
  • Thus, as an example, the invention provides a method comprising administering to a subject at risk of a recurrence of a condition associated with inflammation an effective amount of the glycolipid as a single agent or in combination with an antigen such as or alternative to PSA or lipidated PSA. The method may reduce the likelihood of a recurrence of the condition or may reduce the frequency of future recurrences. The method may reduce the severity of symptoms associated with the condition, whether such symptoms are present in the first manifestation, in a recurrence, or chronically.
  • Autoimmune diseases are known in the art. Examples of autoimmune diseases include but are not limited to multiple sclerosis, inflammatory bowel disease including Crohn's Disease and ulcerative colitis, rheumatoid arthritis, psoriasis, type I diabetes, uveitis, Celiac disease, pernicious anemia, Srojen's syndrome. Hashimoto's thyroiditis, Graves' disease, systemic lupus erythamatosis, acute disseminated encephalomyelitis, Addison's disease, Ankylosing spondylitis, Antiphospholipid antibody syndrome. Guillain-Barre syndrome, idiopathic thrombocytopenic purpura, Goodpasture's syndrome, Myasthenia gravis, Pemphigus, giant cell arteritis, aplastic anemia, autoimmune hepatitis, Kawaski's disease, mixed connective tissue disease, Ord throiditis, polyarthritis, primary biliary sclerosis, Reiter's syndrome, Takaysu's arteritis, vitiligo, warm autoimmune hemolytic anemia, Wegener's granulomatosis, Chagas' disease, chronic obstructive pulmonary disease, and sarcoidosis.
  • In important embodiments, the autoimmune disease is multiple sclerosis. In other important embodiments, the autoimmune disease is an inflammatory bowel disease including but not limited to ulcerative colitis and Crohn's disease. In other embodiments, the autoimmune disease may be rheumatoid arthritis or type I diabetes.
  • In some instances, the compositions of the invention may be administered to a subject who has yet to manifest an autoimmune disease (including symptoms thereof) yet is at risk of developing such as disease based on a known genetic or familial predisposition. Such a subject may have one or more family members that are afflicted with the disease.
  • In some instances, the compositions of the invention are administered to subject having or at risk of developing graft-versus-host disease. Administration may occur prior to, during and/or after transplantation of an organ or tissue (including blood or a blood product) into the subject.
  • In still other instances, the compositions may be administered to subjects having or at risk of developing a conditions associated with inflammation.
  • As an example, the composition may be administered to a subject having asthma. As will be understood in the art, subjects having asthma typically experience asthmatic attacks or events characterized by impaired breathing. The invention contemplates that the compositions described herein may be administered acutely (e.g., a single large dose) or chronically (e.g., repeated, smaller doses) to asthmatic subjects. Accordingly, in some instances, the compositions may be administered prior to an asthmatic attack in order to prevent the occurrence of the attack, reduce the frequency of attacks, and/or to lessen the severity of the attack. In some instances, the compositions may be administered during an attack in order to reduce its severity and/or reduce its duration.
  • Another condition associated with inflammation is a post-surgical adhesion. The invention contemplates administration of the compositions described herein to subjects having or at risk of developing a post-surgical adhesion. The compositions may be administered prior to, during, and/or immediately following surgery, or any combination thereof including but not limited to prior to and during surgery, in order to prevent the occurrence of such adhesions and/or reduce their severity. The compositions may be administered repeatedly following surgery, including for example every day, every two days, every three days, etc. for a week, two weeks, three weeks, a month, or several months post-surgery.
  • Another condition associated with inflammation is an abscess, including but not limited to an abdominal abscess as may occur upon leakage of intestinal contents into the peritoneum. In these instances, the subjects being treated may also be administered anti-bacterial agents such as antibiotics.
  • Thus, as another example, a method is provided that comprises administering to a subject having or at risk of developing an abscess an effective amount of the glycolipid (separate from the polysaccharide component of PSA) or compositions thereof. In some embodiments, the glycolipid is administered prior to development of an abscess and/or prior to the manifestation of symptoms associated with an abscess. In some embodiments, the glycolipid is administered after an abscess has been detected or diagnosed and/or after symptoms associated with an abscess are manifested.
  • A subject intends any subject that would benefit from administration of a composition of the invention or that could be administered the composition of the invention. In important embodiments, the subject is a human subject. The subject may also be a companion animal such as a dog or cat, agricultural livestock such as horses, cattle, pigs, sheep, etc., laboratory animals such as mice, rats, rabbits, monkeys, etc., or animals such as those maintained in zoos or otherwise in captivity.
  • A variety of administration routes are available. The particular mode selected will depend, of course, upon the particular condition being treated, the severity of the condition being treated, and the dosage required for therapeutic efficacy. The methods of the invention, generally speaking, may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Such modes of administration include oral, rectal, topical, nasal, inhalation (e.g., inhaler or nebulization), or parenteral routes. The term “parenteral” includes subcutaneous, intravenous, intramuscular, intraperitoneal, or infusion.
  • Formulations
  • When administered, the agents (e.g., glycolipids) of this disclosure are formulated as pharmaceutically acceptable compositions or preparations. Such compositions or preparations may routinely contain pharmaceutically acceptable carriers, concentrations of salt, buffering agents, preservatives, other immune modulators, and optionally other therapeutic agents. The term “pharmaceutically-acceptable carrier” as used herein, and described more fully below, means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other animal. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active agent(s) is combined to facilitate administration, long-term storage, stability and the like. The active agents of the present invention may be comingled with the other components of the pharmaceutical compositions, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • The pharmaceutical compositions may be presented in unit dosage form and may be prepared by any of the methods known in the art of pharmacy. All methods include the step of bringing the active agents into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active agent(s) into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product. Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent. Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion. Solid forms may be coated for example enterically coated.
  • The active agent(s) may be administered per se (neat) or in the form of a pharmaceutically acceptable salt. Pharmaceutically acceptable salts may be used for in vivo applications as well as in vitro applications. Non-pharmaceutically acceptable salts may be used to prepare pharmaceutically acceptable salts thereof and are not excluded from the scope of the invention. Pharmaceutically acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicyclic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, pharmaceutically acceptable salts can be prepared as alkyline metal or alkyline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt 1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v). Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • Compositions suitable for parenteral administration conveniently comprise a sterile aqueous preparation of the active agent(s), which can be isotonic with the blood of the recipient. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Carrier formulations suitable for subcutaneous, intramuscular, intraperitoneal intravenous, etc. administrations may be found in Remington's Pharmaceutical Sciences, Mack Publishing Company. Easton, Pa.
  • In some embodiments, the glycolipid is formulated with a detergent such as but not limited to Tween or a bile salt such as but not limited to deoxycholate (e.g., sodium deoxycholate) in order to limit or prevent aggregation. Such detergent or bile salt may be used at a low concentration such that it is still pharmaceutically acceptable. For example, it may be present at about or less than 0.0001%, 0.0005%, 0.001%, 0.002%, 0.005%, 0.01%, 0.02%, 0.05%, 0.07%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 2%, 3%, 4%, 5%, or more.
  • The pharmaceutical preparations, as described above, are administered in effective amounts. For therapeutic applications, it is generally that amount sufficient to achieve a medically desirable result. In general, a therapeutically effective amount is that amount necessary to delay the onset of, inhibit the progression of, or halt altogether the particular condition being treated, including reducing the likelihood, frequency and/or severity of a recurrence of the condition. As an example, the effective amount may be that amount which serves to reduce, alleviate, or delay the onset of the symptoms (e.g., pain, fever, etc.) of the disorder being treated or prevented. The effective amount will depend upon the mode of administration, the particular condition being treated and the desired outcome. It will also depend upon the stage of the condition, the severity of the condition, the age and physical condition of the subject being treated, the nature of concurrent therapy (if any), the duration of the treatment, the specific route of administration and like factors within the knowledge and expertise of the medical practitioner. For prophylactic applications, it is that amount sufficient to delay the onset of, inhibit the progression of, or halt altogether the particular condition being prevented, and may be measured by the amount required to prevent the onset of symptoms.
  • Generally, doses of active agent(s) of the present invention may be from about 0.01 mg/kg per day to 1000 mg/kg per day, preferably from about 0.1 mg/kg to 200 mg/kg and most preferably from about 0.2 mg/kg to about 20 mg/kg, in one or more dose administrations daily, for one or more days. It is expected that doses ranging from 1-500 mg/kg, and preferably doses ranging from 1-100 mg/kg, and even more preferably doses ranging from 1-50 mg/kg, will be suitable. The preferred amount can be determined by one of ordinary skill in the art in accordance with standard practice for determining optimum dosage levels of the agent. It is generally preferred that a maximum dose is the highest safe dose according to sound medical judgment be used.
  • In some instances, the total daily dose for a human subject may range from about 50-100 micrograms of the glycolipid.
  • The pharmaceutical preparation may be administered alone or in conjunction with one or more other active agents.
  • The pharmaceutical preparation may be used or administered in conjunction with active agents that are suitable for autoimmune disorders such as multiple sclerosis, Crohn's disease, ulcerative colitis, asthma, rheumatoid arthritis, and the like.
  • An example of such agents include anti-inflammatory agents. Examples include steroids and corticosteroids such as cortisone; non-steroidal anti-inflammatory drugs such as aspirin, salsalate, celecoxib, diclofenac, etodolac, ibuprofen, indomethacin, ketoprofen, ketorolac, nabumetone, naproxen, oxaprozin, piroxicam, sulindac, and tolmetin; aminosalicylates such as sulfasalazine and 5-aminosalicylates including mesalamine, balsalazide, and olsalazine; azathioprine; mercaptopurine; cyclosporine; beta interferons; glatiramer acetate; dimethyl fumarate; fingolimod; mitoxantrone; disease-modifying antirheumatic drugs (DMARDs) such as methotrexate, leflunomide, hydroxychloroquine and sulfasalazine.
  • Another example of such agents include antibodies or antibody fragments. Examples include TNF alpha inhibitors such as infliximab (Remicade), adalimumab (Humira), and golimumab (Simponi); natalizumab (Tysabri), vedolizumab (Entyvio); ustekinumab (Stelara); abatacept (Orencia); anakinra (Kineret); certolizumab (Cimzia), etanercept (Enbrel), rituximab (Rituxan), tocilizumab (Actemra), and tofacitinib (Xeljanz).
  • The invention contemplates that the combined use of glycolipid together with standard treatments such as those recited above will allow a lower dose of the standard treatment to be used for the same or better therapeutic effect, and/or will result in reduced incidence and/or severity of side effects associated with such standard treatments.
  • In some embodiments, the subject is also administered an anti-bacterial agent such as an antibiotic. In one embodiment the pharmaceutical preparation is formulated or given in conjunction with one or more anti-bacterial agents including antibiotics selected from the group consisting of penicillin G, penicillin V, ampicillin, amoxicillin, bacampicillin, cyclacillin, epicillin, hetacillin, pivampicillin, methicillin, nafcillin, oxacillin, cloxacillin, dicloxacillin, flucloxacillin, carbenicillin, ticarcillin, avlocillin, mezlocillin, piperacillin, amdinocillin, cephalexin, cephradine, cefadoxil, cefaclor, cefazolin, cefuroxime axetil, cefamandole, cefonicid, cefoxitin, cefotaxime, ceftizoxime, cefmnenoxine, ceftriaxone, moxalactam, cefotetan, cefoperazone, ceftazidime, imipenem, clavulanate, timentin, sulbactam, neomycin, erythromycin, metronidazole, chloramphenicol, clindamycin, lincomycin, vancomycin, trimethoprim-sulfamethoxazole, aminoglycosides, quinolones, tetracyclines and rifampin.
  • The following Examples are included for purposes of illustration and are not intended to limit the scope of the invention.
  • EXAMPLES Example 1. Isolation of Glycolipid
  • Briefly, B. fragilis was grown in anaerobic conditions. The capsular complex from B. fragilis was isolated with hot phenol/water extraction. The polysaccharide fraction was precipitated with ethanol after DNAse. RNase and pronase treatments. The precipitate was subjected to size exclusion chromatography in order to separate polysaccharide constituents. The fractions of interest were analyzed and pooled, then dialyzed and lyophilized. The purity of glycolipid can be assessed by nuclear magnetic resonance spectroscopy and mass spectroscopy.
  • The isolation and purification process is provided below in greater detail.
  • The B. fragilis delta44 mutant strain was derived experimentally from strain 9343 and upon further characterization it was found to over-express PSA relative to PSB. Delta44 was plated onto a blood agar plate and grown overnight at 37° C. A swab from a heavily colonized plate was sub-cultured into a 500 ml starter culture of peptone yeast broth. The starter culture was inoculated into 16 liter culture of the same media and pH was titrated to neutrality with 5M NaOH. An anaerobic gas mix was bubbled into the sealed culture.
  • After an overnight culture maintained at pH 7, bacteria were checked by Gram stain and subculture. Organisms were collected by centrifugation at 8,000×g for 20 minutes. Bacterial pellets were washed two times with saline yielding approximately one liter of bacterial pellet.
  • The bacterial pellet was suspended in 68° C. melted crystalline phenol to a final concentration of phenol of about 37% v/v (yielding a phenol/water preparation) and mixed for 30 minutes at 68° C. followed by stirring at 4° C. for 48 hours. The phenol/water preparation was aliquoted into glass bottles which were then centrifuged at 1500 rpm. The upper water layer was harvested. Any residual phenol contained in the harvested aqueous phase was extracted with an equal volume of ethyl ether. The ether phase was then removed using a separatory funnel and any residual ether in the aqueous phase was evaporated, yielding the final aqueous phase from the phenol/water preparation.
  • The aqueous phase was dialyzed versus water with multiple changes over 5 days at 4° C. and subsequently lyophilized until it was nearly dry (approximately 5 ml water remaining). A solution of 0.05M Tris with magnesium, calcium and sodium azide (total volume 61 ml) was added to the lyophilized product to bring the total volume to about 66 ml.
  • To the dissolved product was added 10 ml of Tris buffer with DNase (0.07 mg/ml) and RNase (0.33 mg/ml). The entire suspension was filtered through a 0.45 micron filter and the filtrate was stirred at 37° C. The DNase/RNase treatment was repeated by adding fresh enzymes to the mixture, at similar concentrations, and stirred for two hours.
  • The mixture was then combined with 25 mg pronase in 10 ml Tris/magnesium/calcium solution, and the mixture stirred for 24 hours at 37° C. This step was repeated.
  • The polysaccharide fraction was precipitated by adding 5 volumes of ethanol at 4° C. to the mixture. The solution was then centrifuged at 12,000×g for 30 minutes to pellet the polysaccharide fraction. The supernatant was removed and the pellet was resuspended in 392 ml type 1 H2O.
  • The dissolved fraction was then dialyzed against two changes of 16 liters type 1 H2O at 4° C. The volume was reduced by lyophilization to approximately 50 mls.
  • Twenty ml aliquots were chromatographed on a 5×200 cm column of S400 suspended in PBS and 1% sodium deoxycholate, and fractions were collected. Fractions were tested by double diffusion in agar with an antibody that reacts with both lipidated and non-lipidated PSA to determine where lipidated PSA eluted. Aliquots were tested for UV absorption at 280 nm and it was determined that these fractions had no UV absorbable material (e.g., no nucleic acids, no proteins).
  • Fractions were then pooled, concentrated and dialyzed against type 1 H2O on a Minitan concentrator (Millipore) with 10,000 mw cutoff membranes until conductivity of 100 ml was less than 50 ρS. The final product was then lyophilized.
  • Polysaccharide and glycolipid purity and structure was determined by proton nuclear magnetic resonance spectroscopy on a 600 MHz spectrometer and mass spectroscopy. For MALDI-TOF-TOF and LC-MS analysis, the sample was resuspended to 10 μg/p L in 2% acetic acid and heated at 90 degrees for 90 minutes. For MALDI-TOF-TOF analysis, samples were mixed 1:1 to 1% matrix (CHCA or DHB) solution and directly loaded to stainless steel MALDI plate. For LC-MS analysis, samples did or did not undergo liquid-liquid extraction (chloroform-water) step, and then were dried and resuspended in 50:50 isopropanol:acetonitrile and injected. The acid treatment in this step results in the cleavage of the oligosaccharide core unit from the acyl-substituted diglucosamine.
  • An animal model of multiple sclerosis (EAE) may be used to study the immunological activity of glycolipid in vivo. In this model, mice are treated with glycolipid (on the order of about 75-100 μg per mouse) or control (saline, PBS) every three days starting 6 days before EAE induction. Mice are challenged subcutaneously with 250 μg of MOG33-55 (Peptides International) in 200 μl of complete Freund's adjuvant (Sigma). On days 0 and 2 after challenge, mice receive intraperitoneal injections of 250 ng of Bordetella pertussis toxin (List Biological Laboratories). Disease is scored on an established 0 to 5 scale, with 5 being advanced neurological disease. Mice are monitored and scored daily for disease progression.
  • Example 2. Preparation of Lipids for Chemical Analysis
  • For the experiments shown in FIGS. 9-13 , the following procedure was used to prepare glycolipids. Semi-purified B. fragilis glycolipids or overnight-grown bacterial pellet was resuspended in 100 mM acetic acid/sodium acetate buffer (pH 4.5) with 1% sodium dodecylsulfate (SDS) solution at 1-10 mg/mL concentration. The mixture was heated at 95° C. for 30 minutes and cooled. Solution was added with the same volume of chloroform and methanol, shaken well and spun at 1000 g for 10 mins for phase separation. Lower organic phases, containing hydrolyzed GLA, were collected and dried under nitrogen stream and resuspended in 65% isopropanol solution. These samples were injected to reverse-phase HPLC (Phenomenex Kintex C8, 150 mm×4.6 mm×5 um) with the 65-90% gradient of IPA with 10 mM ammonium formate. Fractions were collected, re-analyzed with LC-MS for quantitation and resuspended in DMSO or 0.05% tween-20 PBS solution for in vitro/in vivo experiments.
  • EQUIVALENTS
  • While several inventive embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the function and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the inventive embodiments described herein. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the inventive teachings is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routing experimentation, many equivalents to the specific inventive embodiments described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed. Inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the inventive scope of the present disclosure.
  • All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.
  • All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.
  • The indefinite articles “a” and “an.” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”
  • The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of” or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e. “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.
  • As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one. A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
  • It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
  • In the claims, as well as in the specification above, all transitional phrases such as “comprising.” “including,” “carrying.” “having,” “containing,” “involving.” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of” and “consisting essentially of” shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of patent Examining Procedures. Section 2111.03.

Claims (32)

1. A glycolipid comprising
a tri-acylated, tetra-acylated or penta-acylated diglucosamine conjugated to an oligosaccharide.
2. (canceled)
3. The glycolipid of claim 1, wherein the tri-acylated, tetra-acylated or penta-acylated diglucosamine comprises acyl chains ranging in length from 14-17 carbons.
4. (canceled)
5. The glycolipid of claim 1, substantially free of other components found in a B. fragilis capsule, substantially free of LPS, substantially free of polysaccharide, in purified form, in an isolated form, and/or in a lyophilized form.
6-9. (canceled)
10. The glycolipid of claim 1, in a micelle form or a liposome form.
11-12. (canceled)
13. The glycolipid of claim 1, obtained or derived from B. fragilis cells that overexpress PSA relative to polysaccharide B (PSB), PSC, PSD, PSE, PSF, PSG and PSH.
14. The glycolipid of claim 1, wherein at least one acyl chain is unmodified.
15-17. (canceled)
18. The glycolipid of claim 1, wherein at least one acyl chain is C16:0-OH; C17:0-OH; C14:0; C15:0; N-substituted; or O-substituted.
19-24. (canceled)
25. The glycolipid of claim 1, wherein the diglucosamine is phosphorylated.
26. The glycolipid of claim 1, wherein the diglucosamine is conjugated to the oligosaccharide by an acid-labile bond.
27. (canceled)
28. The glycolipid of claim 1, wherein the oligosaccharide contains 3-10 sugars.
29. The glycolipid of claim 1, wherein the oligosaccharide comprises galactose and/or glucose and/or fucose residues and/or 1 to 2 KDO residues.
30. (canceled)
31. A composition comprising the glycolipid of claim 1, and any one selected from the group consisting of:
(i) a detergent or a bile salt;
(ii) a non-naturally occurring preservative;
(iii) a non-naturally occurring stabilizer;
(iv) human albumin, phenol, glycerin or glycine;
(v) thimerosal, aluminum hydroxide, benzethonium chloride, formaldehyde, formalin, glutaraldehyde, potassium phosphate, aluminum potassium sulfate, bovine extract, calf serum, ammonium sulfate, aluminum phosphate, non-human cells, Vero (monkey kidney) cells, human cells, MRC-5 (human diploid) cells, and/or MRC-5 cellular proteins;
(vi) a lipidated polysaccharide A (PSA), wherein the lipidated polysaccharide A (PSA) and the glycolipid are in a 4:1 to 20:1 ratio (w/w);
(vii) a lipidated polysaccharide A (PSA), wherein there is at least 0.5% (w/w) of the glycolipid;
(viii) an antigen; and
(ix) an antigen and a lipidated PSA.
32. The composition of claim 31, wherein the detergent or bile salt is present at or less than 1%, 0.5% or 0.1% (weight of detergent or bile salt as a percentage of weight of glycolipid).
33-36. (canceled)
37. The composition of claim 31, formulated for parenteral administration or oral administration.
38-48. (canceled)
49. The composition of claim 31 wherein the antigen is a bacterial antigen, attenuated bacteria, a viral antigen, an attenuated virus, a fungal antigen, a mycobacterial antigen, a parasitic antigen, a cancer antigen, a human protein, or a human polysaccharide.
50-82. (canceled)
83. A micelle consisting essentially of glycolipid of claim 1.
84. A composition comprising
a micelle consisting essentially of glycolipid of claim 1 and
a detergent or bile salt.
85. The composition of claim 84, wherein the detergent or bile salt is present in a pharmaceutically acceptable amount.
86. The composition of claim 84, wherein the composition is a pharmaceutical composition.
87. The composition of claim 84, formulated for parenteral administration.
88-95. (canceled)
US17/965,573 2016-07-15 2022-10-13 Glycolipid compositions and methods of use Pending US20230130767A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/965,573 US20230130767A1 (en) 2016-07-15 2022-10-13 Glycolipid compositions and methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662362630P 2016-07-15 2016-07-15
PCT/US2017/042282 WO2018014012A1 (en) 2016-07-15 2017-07-15 Glycolipid compositions and methods of use
US201916317794A 2019-01-14 2019-01-14
US17/965,573 US20230130767A1 (en) 2016-07-15 2022-10-13 Glycolipid compositions and methods of use

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US16/317,794 Division US11491181B2 (en) 2016-07-15 2017-07-15 Glycolipid compositions and methods of use
PCT/US2017/042282 Division WO2018014012A1 (en) 2016-07-15 2017-07-15 Glycolipid compositions and methods of use

Publications (1)

Publication Number Publication Date
US20230130767A1 true US20230130767A1 (en) 2023-04-27

Family

ID=60951879

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/317,794 Active 2037-08-28 US11491181B2 (en) 2016-07-15 2017-07-15 Glycolipid compositions and methods of use
US17/965,573 Pending US20230130767A1 (en) 2016-07-15 2022-10-13 Glycolipid compositions and methods of use

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/317,794 Active 2037-08-28 US11491181B2 (en) 2016-07-15 2017-07-15 Glycolipid compositions and methods of use

Country Status (4)

Country Link
US (2) US11491181B2 (en)
EP (1) EP3484441A4 (en)
CA (1) CA3030974A1 (en)
WO (1) WO2018014012A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3337321A4 (en) 2015-08-19 2019-07-17 President and Fellows of Harvard College Lipidated psa compositions and methods
CN111956608A (en) * 2020-04-17 2020-11-20 福建医科大学附属协和医院 NPM binding protein coupled micelle with acute lymphocytic leukemia resisting activity and preparation method thereof

Family Cites Families (137)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL36670A (en) 1971-04-21 1974-09-10 Sela M Therapeutic basic copolymers of amino acids
US3723506A (en) 1971-08-11 1973-03-27 Univ Sherbrooke Oxidation of sugars
DE2917104A1 (en) 1979-04-27 1980-11-06 Hoechst Ag METHOD FOR REDUCING THE VISCOSITY OF CELLULOSE ETHERS BY OZONE AND ITS USE
JPS56128721A (en) 1980-03-12 1981-10-08 Eisai Co Ltd Antiallergic agent
US4356170A (en) 1981-05-27 1982-10-26 Canadian Patents & Development Ltd. Immunogenic polysaccharide-protein conjugates
EP0143840B1 (en) 1983-05-06 1990-08-29 Wisconsin Alumni Research Foundation Monosaccharide compounds having immunostimulating activity
JPS618945A (en) 1984-06-25 1986-01-16 Nec Corp Semiconductor integrated circuit device
SE456346B (en) 1984-07-23 1988-09-26 Pharmacia Ab GEL TO PREVENT ADHESION BETWEEN BODY TISSUE AND SET FOR ITS PREPARATION
US4619995A (en) 1984-12-24 1986-10-28 Nova Chem Limited N,O-carboxymethyl chitosan and preparative method therefor
US5700906A (en) 1985-07-12 1997-12-23 New York University Immunogenic peptide antigen corresponding to plasmodium vivax circumsporozoite protein
US4684727A (en) 1985-07-29 1987-08-04 Schering Corporation Zwitterionic 1,8-naphthyridine and pyrazino[2,3-b]pyridine containing compounds useful as anti-allergic, anti-inflammatory and cycloprotective agents
DE3750125T2 (en) 1986-03-26 1994-10-27 Genexpress Aps BIOLOGICAL INSULATION.
US4775626A (en) 1986-05-23 1988-10-04 Syntex (U.S.A.) Inc. Method and compositions for protecting anerobic microorganisms
US4819617A (en) 1986-09-04 1989-04-11 University Of Florida Viscoelastic material for ophthalmic surgery
US5514581A (en) 1986-11-04 1996-05-07 Protein Polymer Technologies, Inc. Functional recombinantly prepared synthetic protein polymer
DE3704389A1 (en) 1987-02-12 1988-08-25 Blutspendedienst Dt Rote Kreuz Process for the preparation of lymphokines by induction of lymphoid cells
US4835252A (en) 1987-02-26 1989-05-30 The Salk Institute Biotechnology/Industrial Associates, Inc. Vasoactive intestinal peptide analogs
US5215896A (en) 1987-03-20 1993-06-01 Creative Biomolecules, Inc. Leader sequences for the production of recombinant proteins
US6174999B1 (en) 1987-09-18 2001-01-16 Genzyme Corporation Water insoluble derivatives of polyanionic polysaccharides
US4937270A (en) 1987-09-18 1990-06-26 Genzyme Corporation Water insoluble derivatives of hyaluronic acid
US5140016A (en) 1988-05-31 1992-08-18 University Of Florida Method and composition for preventing surgical adhesions using a dilute solution of polymer
US5126141A (en) 1988-11-16 1992-06-30 Mediventures Incorporated Composition and method for post-surgical adhesion reduction with thermo-irreversible gels of polyoxyalkylene polymers and ionic polysaccharides
DE3840044A1 (en) 1988-11-27 1990-06-07 Behringwerke Ag GLYCOSPHINGOLIPIDS WITH A COUPLABLE GROUP IN THE SPHINGOID PART, THEIR PRODUCTION AND USE
US5196510A (en) 1988-12-29 1993-03-23 Cytogen Corporation Molecular recognition units
FI895708A0 (en) 1989-02-10 1989-11-29 Alko Ab Oy VATTENLOESLIG SOENDERDELNINGSPRODUKT.
US4952524A (en) 1989-05-05 1990-08-28 At&T Bell Laboratories Semiconductor device manufacture including trench formation
US5158939A (en) 1989-07-21 1992-10-27 Wisconsin Alumni Research Foundation Method of stimulating the immune systems of animals and compositions useful therefor
DE69019164T2 (en) 1989-12-14 1995-09-07 Ca Nat Research Council IMPROVED MENINGOCOCCAL POLYSACCHARIDE CONJUGATE VACCINE.
US5268454A (en) 1991-02-08 1993-12-07 La Jolla Pharmaceutical Company Composition for inducing humoral anergy to an immunogen comprising a t cell epitope-deficient analog of the immunogen conjugated to a nonimmunogenic carrier
US5130417A (en) 1990-04-30 1992-07-14 Washington University Entamoeba histolytical immunogenic protein and cdna clone
US5331573A (en) 1990-12-14 1994-07-19 Balaji Vitukudi N Method of design of compounds that mimic conformational features of selected peptides
KR940006696B1 (en) 1991-01-16 1994-07-25 금성일렉트론 주식회사 Manufacturing method of isolation layer of semiconductor device
CA2059693C (en) 1991-01-28 2003-08-19 Peter J. Kniskern Polysaccharide antigens from streptococcus pneumoniae
GB2254083A (en) 1991-03-28 1992-09-30 Italfarmaco Spa Anticoagulants from e.coli saccharide
GR920100122A (en) 1991-04-05 1993-03-16 Ethicon Inc Ionically crosslinked carboxyl-containing polysaccharides for adhension prevention.
US5605938A (en) 1991-05-31 1997-02-25 Gliatech, Inc. Methods and compositions for inhibition of cell invasion and fibrosis using dextran sulfate
US5705178A (en) 1991-05-31 1998-01-06 Gliatech, Inc. Methods and compositions based on inhibition of cell invasion and fibrosis by anionic polymers
US5936076A (en) 1991-08-29 1999-08-10 Kirin Beer Kabushiki Kaisha αgalactosylceramide derivatives
US20050181021A1 (en) 1992-02-14 2005-08-18 Robert Lamb Vitamin E phosphate/phosphatidycholine liposomes to protect from or ameliorate cell damage
US5350941A (en) 1992-09-23 1994-09-27 Texas Instruments Incorporated Trench isolation structure having a trench formed in a LOCOS structure and a channel stop region on the sidewalls of the trench
IL107103A (en) 1992-09-24 2001-03-19 Brigham & Womens Hospital Multivalent vaccine based on conjugated molecule comprising a capsular polysaccharide linked to a protein and a method for the preparation thereof
US5433794A (en) 1992-12-10 1995-07-18 Micron Technology, Inc. Spacers used to form isolation trenches with improved corners
US5728385A (en) 1993-08-12 1998-03-17 Classen Immunotherapies, Inc. Method and composition for an early vaccine to protect against both common infectious diseases and chronic immune mediated disorders or their sequelae
US5705335A (en) 1993-11-26 1998-01-06 Hendry; Lawrence B. Design of drugs involving receptor-ligand-DNA interactions
IL113812A (en) 1994-05-24 2000-06-29 Yeda Res & Dev Copolymer-1 pharmaceutical compositions containing it and its use
JPH07326664A (en) 1994-05-31 1995-12-12 Fuji Electric Co Ltd Filling method of dielectric isolation trench of wafer
US5565204A (en) 1994-08-24 1996-10-15 American Cyanamid Company Pneumococcal polysaccharide-recombinant pneumolysin conjugate vaccines for immunization against pneumococcal infections
US5700787A (en) 1994-09-02 1997-12-23 Brigham & Women's Hospital, Inc. Capsular polysaccharide immunomodulator
US5679654A (en) 1994-09-02 1997-10-21 Brigham & Women's Hospital, Inc. Capsular polysaccharide immunomodulator
ES2314978T3 (en) 1994-11-02 2009-03-16 Nps Pharmaceuticals, Inc. SPECIFIC SODIUM CHANNELS OF THE PERIPHERAL NER5VIOSE SYSTEM, DNA CODING THEM, EXPLORATION OF PHARMACES AND METHODS TO OBTAIN AND USE THEM.
US5858964A (en) 1995-04-14 1999-01-12 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising synthetic peptide copolymer for prevention of GVHD
US5679658A (en) 1995-05-08 1997-10-21 Chitogenics, Inc. N,O-carbonxymethylchitosan for prevention of surgical adhesions
US5851808A (en) 1997-02-28 1998-12-22 Baylor College Of Medicine Rapid subcloning using site-specific recombination
WO1998042718A1 (en) 1997-03-26 1998-10-01 The Brigham And Women's Hospital, Inc. Method for generating saccharide fragments
DK0971961T3 (en) 1997-04-04 2003-03-24 Fidia Advanced Biopolymers Srl N-sulfated hyaluronic acid compounds, derivatives thereof and a process for their preparation
US6749831B1 (en) * 1997-05-16 2004-06-15 Medical Defense Technology, Llc Vaccine against lipopolysaccharide core
US5929049A (en) 1997-08-08 1999-07-27 Dade Behring Marburg Gmbh Polysaccharide conjugates of biomolecules
EP0908179A1 (en) 1997-08-23 1999-04-14 Werner Bollag Treatment of cell-mediated immune diseases
US6015787A (en) 1997-11-04 2000-01-18 New England Medical Center Hospitals, Inc. Cell-permeable protein inhibitors of calpain
AU3066599A (en) 1998-03-03 1999-10-18 University Of Southern California Use of cytokines and mitogens to inhibit graft versus host disease
AU752942B2 (en) 1998-04-13 2002-10-03 Massachusetts Institute Of Technology Comb copolymers for regulating cell-surface interactions
ITPD980169A1 (en) 1998-07-06 2000-01-06 Fidia Advanced Biopolymers Srl AMIDES OF HYALURONIC ACID AND ITS DERIVATIVES AND PROCESS FOR THEIR PREPARATION.
EP1459757A1 (en) 1999-04-02 2004-09-22 The Brigham And Women's Hospital, Inc. Immunomodulating polymers
BR0009531A (en) 1999-04-02 2001-12-26 Brigham & Womens Hospital Immunomodulation polymers
US7083777B1 (en) 1999-04-02 2006-08-01 The Brigham And Women's Hospital, Inc. Immunomodulating polymers
JP2001055335A (en) 1999-08-18 2001-02-27 Toshio Sato Medicine for treating allergic dermatitis
US6995237B1 (en) 1999-10-27 2006-02-07 Cel-Sci Corporation Preparation and composition of peptides useful for treatment of autoimmune and transplant related graft versus host conditions
US6358939B1 (en) 1999-12-21 2002-03-19 Northern Lights Pharmaceuticals, Llc Use of biologically active vitamin D compounds for the prevention and treatment of inflammatory bowel disease
AU2001272369A1 (en) 2000-07-17 2002-01-30 Chr. Hansen A/S Methods and formultations with probiotic microorganisms and medicaments
AUPQ899700A0 (en) 2000-07-25 2000-08-17 Borody, Thomas Julius Probiotic recolonisation therapy
US6670146B2 (en) 2000-10-04 2003-12-30 Schering Corporation Regulatory T cells; methods
SV2003000753A (en) 2000-12-05 2003-06-16 Brigham & Womens Hospital USE OF ZWITTERIONIC POLYSACARIDS FOR THE SPECIFIC MODULATION OF IMMUNE PROGRESS
JPWO2002055091A1 (en) 2001-01-12 2004-05-13 天藤製薬株式会社 Antiallergic agent
EP1361863A2 (en) 2001-02-13 2003-11-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem Carotenoid-loaded liposomes
AU2002245636B2 (en) 2001-03-09 2004-10-14 Id Biomedical Corporation Of Quebec A novel proteosome-liposaccharide vaccine adjuvant
US20050101012A1 (en) 2001-03-12 2005-05-12 Gerold Schuler CD4+CD25+ regulatory T cells from human blood
US20040185057A1 (en) 2001-06-15 2004-09-23 Kirkby Nikolai Soren Therapeutical vaccination
EP1308167A1 (en) 2001-11-06 2003-05-07 Pickl, Winfried, Ao. Univ. Prof. Dr. Antigen presenting vesicles
GB0130123D0 (en) 2001-12-17 2002-02-06 Microbiological Res Agency Outer membrane vesicle vaccine and its preparation
US20030147865A1 (en) 2002-02-07 2003-08-07 Benoit Salomon Cell therapy using immunoregulatory T-cells
EP1476197B1 (en) 2002-02-22 2009-01-07 National Research Council Of Canada Synthesis of lipopolysaccharide-protein conjugate vaccines via the lipid a region following removal of the glycosidic phosphate residue
AU2003217685A1 (en) 2002-03-08 2003-09-22 Eli Lilly And Company Immunomodulatory polymeric antigens for treating inflammatory pathogies
ATE541036T1 (en) 2002-03-13 2012-01-15 Brigham & Womens Hospital METHOD FOR OVEREXPRESSING ZWITTERIONIC POLYSACCHARIDES
US7829289B2 (en) 2002-05-14 2010-11-09 Institut National De La Sante Et De Recherche Medicale T cell subpopulation regulating gut immunity
GB0213622D0 (en) 2002-06-13 2002-07-24 Glaxosmithkline Biolog Sa Vaccine Corporation
JP5219329B2 (en) 2002-06-28 2013-06-26 ネステク、リミテッド Pharmaceutical composition for prevention or treatment of diarrhea
US20060160153A1 (en) 2002-12-03 2006-07-20 Jean-Pierre Abastado Method to measure a t cell response and its uses to qualify antigen-presenting cells
JP2006522135A (en) 2003-03-31 2006-09-28 ザ ブライアム アンド ウィミンズ ホスピタル インコーポレーテッド Zwitterionic immunomodulators for the treatment of asthma and allergies
ES2529736T3 (en) 2003-04-10 2015-02-25 Novartis Vaccines And Diagnostics, Inc. Immunogenic composition comprising a SARS coronavirus spicular protein
WO2005010215A2 (en) 2003-07-17 2005-02-03 Tolerrx, Inc. Methods for identifying tolerance modulatory compounds and uses therefor
US6846807B1 (en) 2003-07-25 2005-01-25 Board Of Regents, The University Of Texas System Colorectal neoplasia prophylaxis
WO2005019417A2 (en) 2003-08-14 2005-03-03 The Bio Balance Corporation Bacterial strains, compositions including same and probiotic use thereof
DE10353593A1 (en) 2003-11-17 2005-06-23 Klinikum der Universität München Großhadern-Innenstadt Leptin antagonist and method for the quantitative measurement of leptin
US7220738B2 (en) 2003-12-10 2007-05-22 Activbiotics, Inc. Rifamycin analogs and uses thereof
ATE539757T1 (en) 2004-11-02 2012-01-15 Univ Leland Stanford Junior METHOD FOR INHIBITING NKT CELLS
US8426146B2 (en) 2005-06-03 2013-04-23 University of Pittsburgh—of the Commonwealth System of Higher Education Multiparameteric method for assessing immune system status
CA2624660C (en) 2005-10-06 2016-07-12 Probi Ab Use of lactobacillus for treatment of autoimmune diseases
EP1969001A2 (en) 2005-11-22 2008-09-17 Novartis Vaccines and Diagnostics, Inc. Norovirus and sapovirus antigens
US8206726B2 (en) 2006-02-06 2012-06-26 The Brigham And Women's Hospital, Inc. Zwitterionic polysaccharides for promotion of immune system maturation and health
CN100366737C (en) 2006-03-01 2008-02-06 哈尔滨美华生物技术股份有限公司 Production of curing lactase
WO2007106073A2 (en) 2006-03-02 2007-09-20 University Of Massachusetts Modified pathogens for use as vaccines
US20090252708A1 (en) 2006-05-18 2009-10-08 Biobalance Llc Biotherapeutic compositions comprising probiotic escherichia coli and uses thereof
CA2655392A1 (en) 2006-05-31 2007-12-06 The Regents Of The University Of California Cd127 expression inversely correlates with foxp3 and suppressive function of cd4+ tregs
PT103495B (en) 2006-06-05 2017-05-12 Faculdade De Farmácia Da Univ De Lisboa PRO-DRUGS OF ORGANIC ACIDS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THE PRE-DRUGS
PL2484375T3 (en) * 2006-09-26 2018-09-28 Infectious Disease Research Institute Vaccine composition containing synthetic adjuvant
GB0620705D0 (en) 2006-10-18 2006-11-29 Opsona Therapeutics Compounds for the modulation of toll-like receptor activity and assay methods for the identification of said compounds
HUE030386T2 (en) 2007-01-31 2017-05-29 Yeda Res & Dev Redirected, genetically-engineered t regulatory cells and their use in suppression of autoimmune and inflammatory disease
TW200904340A (en) 2007-05-11 2009-02-01 Mannatech Inc Processing of natural polysaccharides by selected non-pathogenic microorganisms and methods of making and using the same
EP2679996A1 (en) 2007-05-31 2014-01-01 AbbVie Inc. Biomarkers predictive of the responsiveness to TNF-alfa inhibitors in autoimmune disorders
US20100221755A1 (en) 2007-06-15 2010-09-02 University Of Rochester Use of antibody secreting cell elispot to assess antibody responses following antigen exposure
CA2698668A1 (en) 2007-09-07 2009-03-19 University Of Georgia Research Foundation, Inc. Synthetic lipid a derivative
US20110002965A1 (en) 2007-11-09 2011-01-06 Round June L Immunomodulating compounds and related compositions and methods
WO2009062132A2 (en) 2007-11-09 2009-05-14 California Institute Of Technology Immunomodulating compounds and related compositions and methods
TWI468188B (en) 2008-01-30 2015-01-11 Univ Tokushima Anti-cancer effect enhancers consisting of oxaliplatin liposomal preparations and anticancer agents comprising the formulation
FR2930264B1 (en) 2008-04-18 2013-02-22 Gervais Danone Sa NEW STRAIN OF LACTOBACILLUS PARACASEI SUBSP. PARACASEI WITH ANTIMICROBIAL AND IMMUNOMODULATORY PROPERTIES
US8586029B2 (en) 2008-06-04 2013-11-19 Trustees Of Dartmouth College Prevention or treatment of immune-relevant disease by modification of microfloral populations
US20110229513A1 (en) * 2008-09-05 2011-09-22 Cox Andrew D LPS Based Vaccines
US20100081956A1 (en) 2008-09-30 2010-04-01 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Method, composition, and system to control pH in pulmonary tissue of a subject
WO2010124256A2 (en) 2009-04-23 2010-10-28 California Institute Of Technology Methods and systems for identifying immunomodulatory substances
US20100311686A1 (en) 2009-06-03 2010-12-09 Kasper Lloyd H Nutraceutical composition and methods for preventing or treating multiple sclerosis
US8580278B2 (en) 2009-06-03 2013-11-12 Trustees Of Dartmouth College Nutraceutical composition and methods for preventing or treating multiple sclerosis
HUE040658T2 (en) 2010-04-07 2019-03-28 California Inst Of Techn Vehicle for delivering a compound to a mucous membrane and related compositions, methods and systems
US20110287048A1 (en) 2010-05-20 2011-11-24 Round June L Antigen Specific Tregs and related compositions, methods and systems
AU2011261528A1 (en) 2010-06-01 2013-01-10 Moore Research Enterprises Llc Cellular constituents from Bacteroides, compositions thereof, and therapeutic methods employing Bacteroides or cellular constituents thereof
US20140030807A1 (en) 2010-07-20 2014-01-30 Trustees Of Dartmouth College Method for stimulating foxp3+ regulatory t cell expression of cd39
ES2662412T3 (en) 2010-10-07 2018-04-06 California Institute Of Technology Probiotic therapies for autism
WO2012103532A1 (en) 2011-01-28 2012-08-02 Mazmanian Sarkis K Combinatorial vitamin d and probiotic therapy for inflammatory bowel disease
EP2731617A4 (en) 2011-07-12 2015-07-01 Brigham & Womens Hospital Lipid-containing psa compositions, methods of isolation and methods of use thereof
WO2013019896A1 (en) 2011-08-01 2013-02-07 Symbiotix Biotherapies, Inc. Platform for identifying and/or characterizing immunomodulatory agents
EP2763540A4 (en) 2011-09-09 2015-04-22 Univ Yale Compositions and methods for assessing and treating inflammatory diseases and disorders
ES2662844T3 (en) 2011-10-03 2018-04-10 California Institute Of Technology Generation of mutant strain producing only PSA
US20140072534A1 (en) 2012-08-30 2014-03-13 California Institute Of Technology Novel therapy for multiple sclerosis using vitamin d and gut bacteria
WO2014138696A1 (en) 2013-03-07 2014-09-12 University Of Maryland, Baltimore Immunotherapeutic potential of modified lipooligosaccharides/lipid a
CA2911826C (en) 2013-05-10 2022-08-23 California Institute Of Technology Probiotic prevention and treatment of colon cancer
KR20170016315A (en) 2014-03-25 2017-02-13 더 가번먼트 오브 더 유나이티드 스테이츠 오브 아메리카 에즈 리프리젠티드 바이 더 세크리터리 오브 더 아미 Methods for enhancing the immunostimulation potency of aluminum salt-adsorbed vaccines
ES2555160B1 (en) 2014-06-24 2016-10-25 Aptus Biotech, S.L. Specific aptamers of TLR-4 and their uses
EP3337321A4 (en) 2015-08-19 2019-07-17 President and Fellows of Harvard College Lipidated psa compositions and methods

Also Published As

Publication number Publication date
EP3484441A4 (en) 2020-03-18
WO2018014012A1 (en) 2018-01-18
CA3030974A1 (en) 2018-01-18
US20190290680A1 (en) 2019-09-26
US11491181B2 (en) 2022-11-08
EP3484441A1 (en) 2019-05-22

Similar Documents

Publication Publication Date Title
US20230130767A1 (en) Glycolipid compositions and methods of use
Hudak et al. Glycotherapy: new advances inspire a reemergence of glycans in medicine
Sun et al. Carbohydrates as T-cell antigens with implications in health and disease
JP6622238B2 (en) Methods for inhibiting protein fucosylation in vivo using fucose analogs
US9539281B2 (en) Lipid-containing PSA compositions, methods of isolation and methods of use thereof
Labrada et al. GM3 (Neu5Gc) ganglioside: an evolution fixed neoantigen for cancer immunotherapy
US20150299290A1 (en) Immunogenic vaccine
JP2021118699A (en) Glycan-interacting compounds and methods of use
JP6774941B2 (en) Immunogenic / Therapeutic Sugar Conjugate Compositions and Their Use
TW201521766A (en) Compound and compositions of carbohydrate vaccine and uses thereof
US20210315923A1 (en) Lipidated psa compositions and methods
US20130149331A1 (en) Rhamnose and forssman conjugated immunogenic agents
US8129518B2 (en) Synthetic polysaccharide antigens for immunological intervention in disease
AU2017201649B2 (en) Xenoantigen-displaying anti-cancer vaccines and method of making
US11491215B2 (en) Antigenic combinations against Francisella bacteria and related nanolipoprotein particles, compositions, methods and systems
WO2014200345A1 (en) Method for antigen-specific tolerance induction in humans using the small heat shock protein alpha b-crystallin.
JP7092308B2 (en) Lipid A
Kleski Progress of Entirely Carbohydrate Conjugates in Cancer Immunotherapeutics? Syntheses and Developments
Kaplonek Improving the Immunoprotective Effect of Carbohydrate Vaccine Against Bacterial Pneumonia
US20050095675A1 (en) Mucin peptide with immunoenhancing properties
KR20020013470A (en) Method for inducing cellular immunity and cells with induced cellular immunity
CA3147314A1 (en) Methods of making and using lipooligosaccharide compositions and vaccines
Sun Structural and Host Factors Influencing Vaccine Responses
Apostolopoulos et al. Carbohydrate-Based Targets and Vehicles for Cancer and Infectious Diseases Vaccines
Singh Glycan-Lectin duet, Novel strategies for DC-targeting

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: PRESIDENT AND FELLOWS OF HARVARD COLLEGE, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OH, SUNGWHAN;ERTURK-HASDEMIR, DENIZ;KASPER, DENNIS L.;AND OTHERS;SIGNING DATES FROM 20171107 TO 20200716;REEL/FRAME:062356/0670